IMR Press / RCM / Volume 20 / Issue 1 / DOI: 10.31083/j.rcm.2019.01.3189
Open Access Review
Advances in epigenetic regulation of vascular aging
Show Less
1 Department of Internal Medicine-Cardiovascular, The second hospital of Dalian medical university, Liaoning, 116027, China
2 Department of Medical Affair, The second hospital of Dalian medical university, Dalian City, Liaoning, 116027, China
*Correspondence: YufengLiughj@163.com (Yufeng Liu)
Rev. Cardiovasc. Med. 2019, 20(1), 19–25; https://doi.org/10.31083/j.rcm.2019.01.3189
Published: 30 March 2019
Copyright: © 2019 Jin et al. Published by IMR Press.
This is an open access article under the CC BY-NC 4.0 license (https://creativecommons.org/licenses/by-nc/4.0/)
Abstract

Vascular aging is a major risk factor and driver of agerelated cardiovascular diseases (CVD). Atherosclerosis, hypertension, and other CVD lead to vascular dysfunction that involves multiple pathological processes such as oxidative stress, endothelial dysfunction, inflammation, and autophagy. Epigenetics refers to genetic changes that occur when the DNA remains unchanged that include DNA methylation, histone modification, and non-coding RNA. It has been reported that epigenetics plays an effective regulatory role in CVD and affects cardiovascular repair function. Presently, drugs targeting epigenetics have applications in malignant tumors and inflammation. Therefore, exploration of epigenetic mechanisms in vascular aging will allow us to understand the pathogenesis of diseases related to vascular aging. This review focuses on the pathological changes in vascular aging and analyzes the relationship between vascular aging and epigenetics. Additionally, this review focuses on the pathogenesis of vascular aging related diseases from a new perspective in order to develop epigenetic-based treatment strategies for patients with age-related cardiovascular diseases.

Keywords
Vascular aging
epigenetics
endothelial dysfunction
oxidative stress
inflammation
1 Introduction

Vascular aging is a major risk factor of age-related cardiovascular diseases that lead to the gradual decline of vascular function (Katusic and Austin, 2014). It is necessary to explore the mechanism of vascular aging to diagnose and prevent CVD. Although aging is inevitable, pathological vascular aging is a modifiable risk factor (Laurent, 2012). Clinical studies have shown that vascular aging is associated with vascular endothelial dysfunction, hypertension, atherosclerosis (AS), coronary heart disease, and stroke (Lakatta, 2003). Many of these pathologies are related to endothelial cell injury, vascular smooth muscle cell proliferation, inflammation, fibrosis, and calcification (Cahill-Smith and Li, 2014). The pathogenesis of these age-related CVD; however, are not fully understood. In China, the aging population and chronic cardiovascular complications from health and economic burden has increased. It is necessary to investigate the impact of vascular aging on CVD that prevent and repair damaged aging arteries and reduce the risk of vascular age-related diseases (Harvey et al., 2015).

Structural and mechanical stress changes, such as vascular media thickness may be observed in senescent blood vessels. With the increase of age, endothelium-dependent vascular dilatation is continuously attenuated. This results in vascular structure changes that are accompanied by pathological changes such as lumen dilation, endothelial dysfunction, and diffuse intimal thickening. In addition, endothelial barrier permeability decreases, which allows vascular smooth muscle cells to migrate to the subendothelial space producing extracellular matrix proteins. Both the increased elastin degradation and collagen deposition results in vascular plasticity reduction, which leads to vascular remodeling and endothelial homeostasis change (Najjar et al., 2005). Age-related dysfunction of endothelial cells may in turn aggravate media thickness and vascular fibrosis resulting in hypertension, atherosclerosis and vascular obstruction (Brandes et al., 2005).

2 Pathologic mechanism of vascular aging

Clinical studies have shown that the mortality of CVD increases exponentially with age. Age-related intracellular homeostasis changes make elderly blood vessels more vulnerable to pathophysiological conditions. Exploration of age-related vascular pathophysiological mechanisms is helpful to reduce the mortality of vascular age-related diseases (Fig. 1) (Ungvari et al., 2010).

Figure 1.

Vascular age-related molecular signaling pathway (Paneni et al., 2015). Aging, especially vascular aging, is associated with several signaling pathways. FOXO, forkhead box O; AMPK, AMPactivated protein kinase; VCAM-1, vascular cell adhesion molecule1; ICAM-1, intercellular adhesion molecule-1; MCP-1, monocyte chemoattractant protein-1; MnSOD, manganese superoxide dismutase; ecSOD, extracellular superoxide dismutase.

2.1 Oxidative stress and endothelial dysfunction in vascular aging

Studies have shown that vascular aging is mainly characterized by endothelial dysfunction and vascular stiffness. This is associated with decreased bioavailability of endothelial nitric oxide (NO) and increased production of reactive oxygen species (ROS) (Yeo et al., 2015). ROS can cause endothelial dysfunction, which in turn leads to coronary artery disease and stroke in the elderly (Donato et al., 2007). In vascular aging, NO bioavailability is seriously impaired and endothelial nitric oxide synthase (eNOS) expression is decreased. This decreased expression aggravates NO bioavailability that reduces tetrahydrobiopterin (BH4) (Hoffmann et al., 2001), intracellular L-arginine utilization (Berkowitz et al., 2003) and promotes vascular atherosclerosis.

BH4 is a coenzyme of eNOS that regulates catalytic activity (Alp and Channon, 2004). The BH4 reduction in senescent blood vessels directly leads to the conformational change of eNOS from dimer to monomer state resulting in the loss of NO (Farah et al., 2018). Clinical studies have shown that BH4 supplement can improve endothelial function in the elderly, suggesting that BH4 may be a potential target for improving the aging vascular function (Pierce and Larocca , 2008). In addition, there is increasing evidence that arginine II (Arg II) is down-regulated in senescent cells and involved in early vascular aging by changing the endothelial cell and vascular smooth muscle cell (VSMCs) phenotypes. In contrast, silencing Arg II inhibited the aging phenotype of human endothelial cells (Wu et al., 2015). Furthermore, the Arg II gene deletion reduces vascular inflammation in mice fed a high-cholesterol and high-fat diet, prevents atherosclerosis, and improves insulin sensitivity and glucose homeostasis (Pernow and Jung, 2013). These findings suggest that Arg II may be a co-determinant of metabolic disease, atherosclerosis, and aging (Yepuri et al., 2012).

In recent years, the gradual discovery of longevity genes, such as the Klotho gene, has been observed to accelerate the aging phenotype that is associated with vascular endothelial dysfunction and premature atherosclerosis (Kuro-o et al., 1997). Recent studies have shown that mitochondrial oxidative stress also plays an important role in vascular dysfunction caused by aging. Mitochondria-derived H2O2 contributes to the NF-κB activation, which leads to the pro-inflammatory transformation of endothelial gene expression profile. Apoptosis is another important link between mitochondrial ROS production and vascular aging (Ungvari et al., 2007). The mechanism of mitochondrial oxidative stress in vascular endothelial cell senescence may be multifaceted including reduced glutathione content and dysfunction of the electron transport chain (Ungvari et al., 2008). Recent studies have shown that the mitochondrial enzyme p66Shc protein plays an important regulatory role in the production of mitochondrial ROS linking oxidative stress with apoptos is (Napoli et al., 2003). Mice deficient in p66Shc showed a reduction of ROS, which was closely related to a 30% increase in lifespan and improved vascular endothelial function (Camici et al., 2008).

2.2 Inflammation in vascular aging

Recent studies have found that the interaction of inflammation, ROS and endothelial dysfunction plays an important role in the pathogenesis of cardiovascular aging (Wu et al., 2014). Chronic inflammation is closely related to aging and is the main factor to induce the atherosclerosis (Fulop et al., 2017). ROS could activate endothelial cell activation and secretion of inflammatory mediators. Even in physiological aging, there are inflammatory changes in the vascular gene expression profile such as up-regulation of inflammatory cytokines, chemokines, adhesion molecules, and inducible nitric oxide synthase (Csiszar et al., 2003). Plasma concentrations of various inflammatory markers (TNF-α, VCAM-1, IL-6, IL-18, MCP-1) were positively correlated with age and independent of other cardiovascular risk factors (Miles et al., 2008). High levels of inflammatory cytokines are involved in the development of inflammatory microenvironment and vascular dysfunction, therefore promoting senescent endothelial cell apoptosis (Csiszar et al., 2007). In addition, the NF-κB binding may increase the expression of adhesion-inducible nitric oxide synthase in coronary arteries and aorta (Cernadas et al., 1998), leading to atherosclerosis (Hajra et al., 2000). Removing the mitochondrial source of hydrogen peroxide from aging blood vessels decreases NF-κB activation and suggests that mitochondrial oxidative stress can regulate the activity of NF-κB in senescent endothelial cells. An age-relted decline in mitochondrial function may lead to vascular inflammation (Ungvari et al., 2007).

2.3 Autophagy in vascular aging

Autophagy occurs in age-related diseases; however, its mechanism is not clear. Studies have reported that mice are more prone to pulmonary hypertension after the knockout of autophagy related gene LC3-II (Lee et al., 2011). In addition, Beclin1 gene knockout in local vascular tissues resulted in severe vascular remodeling, decreased vascular reendothelialization, and increased apoptosis (Ye et al., 2014). Specific deletion of ATG7 gene in smooth muscle cells can promote intimal hyperplasia and atherosclerotic plaque formation (Grootaert et al., 2015). In contrast, studies have shown that excessive autophagy can damage cells. The autophagy inhibitor 3-MA inhibits autophagy and alleviates vascular endothelial cell injury. In addition, another autophagy inhibitor chloroquine showed protective effects in pulmonary arterial hypertension (Long et al., 2013).

3 Epigenetics in vascular aging

The mechanism of aging is complex and cannot be fully explained. Studies have defined aging as telomere shortening, protein homeostasis disorder, mitochondrial dysfunction, genomic instability, epigenetic modification, reduced nutritional sensitivity, cellular senescence, stem cell failure, and cell communication changes (Lopez-Otin et al., 2013). While most studies have focused on genetic aging, environmental factors may also influence age-related diseases (Costantino et al., 2018). The epigenetic modification in aging has recently been a topic of discussion.

Epigenetics is necessary for the maintenance of tissue-specific gene expression development as well as the cardiovascular homeostasis and gene plasticity changes. The main content maybe divided into three categories: DNA methylation, histone modification, and non-coding RNA changes (Cencioni et al., 2013). Further understanding of the role of epigenetics in age-related cardiovascular diseases may provide additional insights into understanding age-related CVD and to seek relevant intervention strategies.

3.1 DNA methylation

DNA methylation is defined as covalently linking cytosine residues of CpG island with methyl bonds and inhibiting gene transcription by binding to the gene promoter region or by recruiting chromatin modifying enzymes (Fraineau et al., 2015). DNA methylation is catalyzed by three different methyltransferases (DNMTs). DNMT1 maintains methylation during replication and participates in de novo synthesis of DNA methylation along with DNMT3a and DNMT3b (Okano et al., 1999).

DNA methylation is an independent predictor of mortality in addition to some known risk factors for aging such as smoking, diabetes and hypertension (Marttila et al., 2015). Genome-wide studies of senescent stem cells have shown that the low level of gene promoter DNA methylation is associated with cell selfrenewal (Marttila et al., 2015). In addition, the decrease of agedependent DNA methylation level is also accompanied by the high methylation of some special gene loci, such as c-fos, IGF-2, and p16INK4a. Down-regulation of DNMT3A and TET2 (main regulators of DNA methylation state) contributes to the increased stem cell number and decreased differentiation ability (Challen et al., 2011). Hypomethylation of eNOS gene promoter region induces up-regulation of eNOS gene level and increases enzyme activity. Studies have shown that the DNA methylation of p66shc and Jun D leads to endothelial injury and ROS production (Challen et al., 2011). Furthermore, hypomethylation of the promoter region also promotes gene expression associated with inflammation, obesity, cell dysfunction, and vascular injury (Liu et al., 2008).

3.2 Histone modification

Nucleosome is the smallest unit of eukaryotic chromatin, composed of DNA and core regions (H2A, H2B, H3 and H4). Histone modification regulates gene transcription and expression and posttranslational modifications modify chromatin in different ways including acetylation, methylation, ubiquitination, and phosphorylation to regulate chromatin structure and function that affect gene transcription and expression (Kimura, 2013).

3.2.1 Histone acetylation and deacetylation

Histone acetylation is a special epigenetic marker that plays an important role in chromatin structure, function, gene expression, gene transcription, and chromatin remodeling. Histone acetyltransferase (HATs) and histone deacetylase (HDACs) are responsible for acetylation and deacetylation, respectively. Histone acetylation modifications are the first identified histone posttranslational modifications (Cooper and El-Osta, 2010). Studies have shown that DNA methylase (DNMTs), histone methyltransferase (HMTs), and histone acetyltransferase (HATs) are closely related to chromatin remodeling under certain pathophysiological stimuli. HATs and HDACs are reported to regulate endothelial dysfunction and inflammatory responses in type 2 diabetes (Villeneuve and Natarajan, 2010).

Sirtuins, members of the deacetylase family, are widely studied to be key regulators of the aging process in yeast and mammals. SIRT1 is reported to regulate gene expression, metabolism, and aging by deacetylation of a series of enzymes and transcriptional switches such as PGC-1α, NF-κB, eNOS, FOXO, p53, p300/CBP, H3K9 and H3K56 (Beltrami et al., 2012). Studies have shown that SIRT1 overexpression increases endothelial cell migration, while loss of SIRT1 activity leads to down-regulation of key angiogenic factors including Flt1 and CXCR4 (Potente et al., 2007). Besides, SIRT1 is down-regulated in umbilical cord blood endothelial progenitor cells of premature infants, while SIRT1 overexpression alleviates EPCs aging and dysfunction (Vassallo et al., 2014). In addition, SIRT1 improves hyperglycemia induced endothelial dysfunction by inhibiting the vascular p66Shc gene transcription. SIRT1 decreases oxidative stress and inflammation in the vessel by inhibiting NF-κB and PARP activation (Zheng et al., 2012). In adipose tissue of obese patients, SIRT1 is down-regulated leading to histone hyperacetylation, recruitment of macrophages, and increased expression of pro-inflammatory factors such as IL-6, IL1β, TNF-α, IL-13, IL-10 and IL-4. Collectively, these factors result in inflammation (Gillum et al., 2011). SIRT3 is an important regulator of senescent stem cells (Brown et al., 2013). VEGF expression decreased in SIRT3-/- mice results in angiogenesis and clone formation defects. Interestingly, SIRT3 overexpression reverses these abnormalities. Recent studies have shown that H3K9 deacetylase SIRT6 has a protective effect on senescent endothelial cells (Mostoslavsky et al., 2006). In macrophages, HDAC3 regulates the expression of inflammatory genes, while HDAC2 inhibits inflammation (Zeng et al., 2006). In addition, HDACs are also involved in angiogenesis. HDAC7 promotes endothelial cell migration and lumen formation by activating platelet-derived growth factor B and its receptors (Mottet et al., 2007). In vivo and in vitro, HDAC6 and HDAC9 also play a pro-angiogenic role by inducing endothelial cell migration and the formation of functional cavities (Kaluza et al., 2013). In contrast, HDAC5 was found to play an anti-angiogenic role in endothelial cells (Urbich et al., 2009).

3.2.2 Histone methylation and demethylation

Chromatin status and function are different due to the different number of methylation sites and groups. Histone methyltransferase is responsible for the methylation of histone lysine and arginine with different modification sites that drive different effects of gene expression. H3K4 methylation induces gene activation while H3K9 and H3K27 methylation inhibits gene expression. In addition, modification of polymethyl groups on histone lysine leads to different levels of methylation that may have different biological significance. For example, H3K9me and H3K9me2 modifications are often found in genes whose expression is inhibited, while H3K9me3 forms are found in heterochromatin. PDGF-BB-induced changes in the VSMCs phenotype were also associated with histone modification. Recently, with the development of single-cell epigenetic technology, the modification status of single VSMCs in vascular plaque tissues can be tracked (Gomez et al., 2015).

It has been reported that both H3K4me3 and H3K27me3 modification is closely related to lifespan extension. In senescent HSCs, H3K4me3 and H3K27me3 are increased, but H3K9me3 level is down-regulated in MSCs isolated from elderly patients (Goodell and Rando., 2015). Other studies have shown that H3K4 methylation plays an important role in angiogenesis, which is mediated by the SET domain containing enzymes (e.g. SET1A/B, SET7, MLL1/2, MLL3/4) (Ernst and Vakoc, 2012). SET7-mediated lysine methylation regulates important longevity genes such as SIRT1, FoxO3, NF-κB and p53 (Paneni et al., 2013), but negatively regulates the transcription HIF-α thereby inhibiting retinal and tumor angiogenesis. Another histone methylase MLL1 leads to H3K4me3, which plays an important role in embryonic hematopoiesis, hematopoietic stem cell renewal, endothelial progenitor cell differentiation, and endothelial cells (Diehl et al., 2007). In addition, Histone methyltransferase G9a and SUV39H1 agonists could induce H3K9 methylation and inhibit VEGFA expression in HEK293 cells while treatment with G9a small molecule inhibitor, endothelial progenitor cells improved angiogenesis and myocardial repair after myocardial infarction (Snowden et al., 2002). Inhibition of H3K27 demethylase increased lifespan extension by targeting the insulin/IGF-1 signaling pathway (an important longevity pathway). In endothelial cells, VEGF recruits EZH2 to induce H3K27me3, which affects downstream angiogenesis. Interestingly, EZH2 silencing increases endothelial cell migration and promotes capillary network formation, which is considered as a potential therapeutic strategy to promote angiogenesis. Furthermore, EZH2 was recruited by MTA2 and bounded to the promoter region of the TSC2 gene to inhibit the transcription of TSC2 (Wei et al., 2015). Inhibition of histone methyltransferase HYPB/SETD2 induced down-regulation of H3K36me3 and further damaged the migration and cavity formation suggesting that HYPB/SETD2 plays an important role in the vascular reconstruction of embryos (Hu et al., 2010). Other studies have shown that H3K36me3 may be another important target for biological regulation in regenerative medicine. As a result, drugs targeting histone modification enzymes may have an important application in clinical tumor treatment and vascular remodelingrelated diseases (Li et al., 2011; Usui et al., 2014).

3.3 MicroRNAs

Studies have shown that noncoding RNA, microRNA (miRNA) and long non-coding RNA (lncRNA), play an important role in the post-transcriptional regulation of gene expression. The microarray approach was used to reveal multiple miRNAs involved in endothelial cell senescence and angiogenesis. MiRNA expression was significantly up-regulated during aging leading to posttranscriptional inhibition of age-related genes in endothelial phenotypes (D'Andrade and Fulmer-Smentek, 2012). It has been reported that upregulation of mir-34a in senescent endothelial cells and endothelial progenitor cells inhibited cell proliferation that progressed cellular senescence and decreased mice lifespan (Boon et al., 2013). In response to DNA damage, Mir-29 and mir-17-92 are key regulators of chromatin related proteins such as chk1/2, g-H2AX, and ATM. (Ugalde et al., 2011). In addition, mir-29 was up-regulated in senescent endothelial cells and aortic tissue of elderly mice resulting in a decrease in extracellular matrix deposition. Interestingly, mir-29 was down-regulated in myocardial microvascular endothelial cells isolated from type II diabetic rats and promoted angiogenesis by regulating the expression of IGF-1 (Boon et al., 2011). Mir-20a inhibits endothelial cell migration by the inhibition of MKK3 and the activation of p38 MAP kinase (Deng et al., 2017). Downregulation of mir-126 led to cell apoptosis, decreased proliferation, and endothelial cell migration (Mocharla et al., 2013). By analyzing the MicroRNA expression profile of elderly mice, it was found that mir-10a, mir-21, and target genes are key drivers of endothelial progenitor cell dysfunction. Inhibition of these miRNAs resulted in regeneration of endothelial progenitor cells and improved in vivo and in vitro angiogenesis (Zhu et al., 2013). The increased age-dependent mir217 level impaired endothelial angiogenesis by down-regulating SIRT1, FOXO1, and eNOS. On the contrary, inhibition of mir-217 in senescent endothelial cells alleviated aging and increased angiogenesis. The senescent endothelial cells are also associated with the decreased expression of mir-146a (Vasa-Nicotera et al., 2011).

In summary, epigenetics is an important mechanism of aging involving multiple signaling pathways such as inflammation, oxidative damage, mitochondrial dysfunction, and apoptosis. Vascular aging is an important risk factor of CVD and various types of epigenetic modifications interact with each other. For example, the expression of histone modifications such as H3K9Ac and H3K27me3 are down-regulated in aging. Age-related DNA hypermethylation is closely related to the inhibitory histone marker H3K27me3/H3K9me3 (Bracken et al., 2007); DNA hypomethylation is highly enriched at the chromatin marker H3K4me1 (Fernandez et al., 2015). Furthermore, calorie restriction plays an important role in promoting lifespan by regulating epigenetic mechanisms such as DNA methylation and histone modification (Rizza et al., 2014).

4 Concluding Remarks

Nowadays, a large range of environmental stimuli (pollution, high-fat diets, and cardiovascular risk factors) may bring epigenetic alterations such as DNA methylation, histone modification, and non-coding RNA. Gene expression, cell phenotypes, and biological functions could change upon these epigenetic alterations resulting in hypertension, atherosclerosis, restenosis, pulmonary hypertension, and other cardiovascular diseases. The aim of this review was to investigate the specific mechanism of epigenetics in vascular aging. Formulation of epigenetics treatment strategies is one of the major challenges of age-related cardiovascular diseases. With the development of high-throughput sequencing technology, the changing trends of genome-wide DNA methylation, histone modification and non-coding RNA is easier to detect, it is expected to develop epigenetic-based treatment strategies for patients with age-related cardiovascular diseases in the future (Costantino et al., 2018).

Acknowledgments

Thanks to all the peer reviewers and editors for their opinions and suggestions.

Conflict of Interest

The authors declare no competing interests.

References
[1]
Alp, N. J., Channon, K. M . ( 2004) Regulation of endothelial nitric oxide synthase by tetrahydrobiopterin in vascular disease. Arteriosclerosis, Thrombosis, and Vascular Biology 24, 413-420. 10.1161/01.ATV.0000110785.96039.f66f352c3217fd82ef48ad4806c523f090http%3A%2F%2Fcardiovascres.oxfordjournals.org%2Flookup%2Fijlink%3FlinkType%3DABST%26amp%3BjournalCode%3Datvbaha%26amp%3Bresid%3D24%2F3%2F413%26amp%3Batom%3D%252Fcardiovascres%252F76%252F3%252F539.atomhttps://www.ahajournals.org/doi/10.1161/01.ATV.0000110785.96039.f6
[2]
Beltrami, A. P., Cesselli, D., Beltrami, C. A . ( 2012) Stem cell senescence and regenerative paradigms. Clinical Pharmacology & Therapeutics 91, 21-29. aeed840b2366ccf03ad032b891586ccehttp%3A%2F%2Fonlinelibrary.wiley.com%2Fdoi%2F10.1038%2Fclpt.2011.262%2Fpdf
[3]
Berkowitz, D. E., White, R., Li, D., Minhas, K. M., Cernetich, A., Kim, S., Burke, S., Shoukas, A. A., Nyhan, D., Champion, H. C., Hare, J. M . ( 2003) Arginase reciprocally regulates nitric oxide synthase activity and contributes to endothelial dysfunction in aging blood vessels. Circulation 108, 2000-2006. 10.1161/01.CIR.0000092948.04444.C7e7fc41b73be3bef343edf3cecc9d8a1dhttp%3A%2F%2Fwww.europepmc.org%2Fabstract%2FMED%2F14517171https://www.ahajournals.org/doi/10.1161/01.CIR.0000092948.04444.C7
[4]
Boon, R. A., Iekushi, K., Lechner, S., Seeger, T., Fischer, A., Heydt, S., Kaluza, D., Treguer, K., Carmona, G., Bonauer, A., Horrevoets, A. J., Didier, N., Girmatsion, Z., Biliczki, P., Ehrlich, J. R., Katus, H. A., Muller, O. J., Potente, M., Zeiher, A. M., Hermeking, H., Dimmeler, S . ( 2013) MicroRNA-34a regulates cardiac ageing and function. Nature 495, 107-110. 10.1038/nature11919b9d69ab9ec4c34c6034ec29b0ab73d40http%3A%2F%2Feurheartj.oxfordjournals.org%2Flookup%2Fexternal-ref%3Faccess_num%3D23426265%26amp%3Blink_type%3DMED%26amp%3Batom%3D%252Fehj%252Fearly%252F2014%252F09%252F03%252Feurheartj.ehu312.atom
[5]
Boon, R. A., Seeger, T., Heydt, S., Fischer, A., Hergenreider, E., Horrevoets, A. J., Vinciguerra, M., Rosenthal, N., Sciacca, S., Pilato, M., van Heijningen, P., Essers, J., Brandes, R. P., Zeiher, A. M., Dimmeler, S . ( 2011) MicroRNA-29 in aortic dilation: implications for aneurysm formation. Circulation Research 109, 1115-1119. 10.1161/CIRCRESAHA.111.2557370878370a5eee48c5d1b88b4e9fe58fc4http%3A%2F%2Fbiomedgerontology.oxfordjournals.org%2Fcgi%2Fijlink%3FlinkType%3DABST%26amp%3BjournalCode%3Dcircresaha%26amp%3Bresid%3D109%2F10%2F1115https://www.ahajournals.org/doi/10.1161/CIRCRESAHA.111.255737
[6]
Bracken, A. P., Kleine-Kohlbrecher, D., Dietrich, N., Pasini, D., Gargiulo, G., Beekman, C., Theilgaard-Monch, K., Minucci, S., Porse, B. T., Marine, J. C., Hansen, K. H., Helin, K . ( 2007) The Polycomb group proteins bind throughout the INK4A-ARF locus and are disassociated in senescent cells. Genes Development 21, 525-530. 10.1101/gad.415507aef5ccd1e2cf22d3a7821cde1268029chttp%3A%2F%2Fnew.med.wanfangdata.com.cn%2FPaper%2FDetail%3Fid%3DPeriodicalPaper_JJ0210789717http://www.genesdev.org/cgi/doi/10.1101/gad.415507
[7]
Brandes, R. P., Fleming, I., Busse, R . ( 2005) Endothelial aging. Cardiovasc Research 66, 286-294. 10.1016/j.cardiores.2004.12.027https://academic.oup.com/cardiovascres/article-lookup/doi/10.1016/j.cardiores.2004.12.027
[8]
Brown, K., Xie, S., Qiu, X., Mohrin, M., Shin, J., Liu, Y., Zhang, D., Scadden, D. T., Chen, D . ( 2013) SIRT3 reverses aging-associated degeneration. Cell Report 3, 319-327. 10.1016/j.celrep.2013.01.005357d3493fc54881516f214dd3086fd7bhttp%3A%2F%2Feuropepmc.org%2Farticles%2FPMC3582834https://linkinghub.elsevier.com/retrieve/pii/S2211124713000120
[9]
Cahill-Smith, S., Li, J. M . ( 2014) Oxidative stress, redox signalling and endothelial dysfunction in ageing-related neurodegenerative diseases: a role of NADPH oxidase 2. British Journal of Clinical Pharmacology 78, 441-453. 10.1111/bcp.2014.78.issue-3fed46b8aab4b76e9720e912e1e4b57f5http%3A%2F%2Feuropepmc.org%2Fabstract%2Fmed%2F25279404http://doi.wiley.com/10.1111/bcp.2014.78.issue-3
[10]
Camici, G. G., Cosentino, F., Tanner, F. C., Luscher, T. F . ( 2008) The role of p66Shc deletion in age-associated arterial dysfunction and disease states. Journal of Applied Physiology 105, 1628-1631. 10.1152/japplphysiol.90579.200858c4d1cf8f49884322bf4b73524f2ddfhttp%3A%2F%2Feurheartj.oxfordjournals.org%2Flookup%2Fijlink%3FlinkType%3DABST%26amp%3BjournalCode%3Djap%26amp%3Bresid%3D105%2F5%2F1628%26amp%3Batom%3D%252Fehj%252F36%252F8%252F516.atomhttp://www.physiology.org/doi/10.1152/japplphysiol.90579.2008
[11]
Cencioni, C., Spallotta, F., Martelli, F., Valente, S., Mai, A., Zeiher, M., Gaetano, C . ( 2013) Oxidative stress and epigenetic regulation in ageing and age-related diseases. International Journal of Molecular Sciences 14, 17643-17663. 10.4081/pr.2010.e22398960890346a5e578011d862bc0a6fc123fb00http%3A%2F%2Fnew.med.wanfangdata.com.cn%2FPaper%2FDetail%3Fid%3DPeriodicalPaper_PM23989608http://www.mdpi.com/1422-0067/14/9/17643
[12]
Cernadas, M. R., Sanchez de Miguel, L., Garcia-Duran, M., Gonzalez-Fernandez, F., Millas, I., Monton, M., Rodrigo, J., Rico, L., Fernandez, P., de Frutos, T., Rodriguez-Feo, J. A., Guerra, J., Caramelo, C., Casado, S., Lopez, F . ( 1998) Expression of constitutive and inducible nitric oxide synthases in the vascular wall of young and aging rats. Circulation Research 83, 279-286. 10.1161/01.RES.83.3.279484659ef4e7d48cb984dea57ec823249http%3A%2F%2Fcardiovascres.oxfordjournals.org%2Flookup%2Fijlink%3FlinkType%3DABST%26amp%3BjournalCode%3Dcircresaha%26amp%3Bresid%3D83%2F3%2F279%26amp%3Batom%3D%252Fcardiovascres%252F66%252F2%252F374.atomhttps://www.ahajournals.org/doi/10.1161/01.RES.83.3.279
[13]
Challen, G. A., Sun, D., Jeong, M., Luo, M., Jelinek, J., Berg, J. S., Bock, C., Vasanthakumar, A., Gu, H., Xi, Y., Liang, S., Lu, Y., Darlington, G. J., Meissner, A., Issa, J. P., Godley, L. A., Li, W., Goodell, M. A . ( 2011) Dnmt3a is essential for hematopoietic stem cell differentiation. Nature Genetics 44, 23-31. b0f497c02706bcda4f5104f82963d4d9http%3A%2F%2Feuropepmc.org%2Farticles%2FPMC3637952
[14]
Cooper, M. E., El-Osta, A. ( 2010) Epigenetics: mechanisms and implications for diabetic complications. Circulation Research 107, 1403-1413. 10.1161/CIRCRESAHA.110.223552https://www.ahajournals.org/doi/10.1161/CIRCRESAHA.110.223552
[15]
Costantino, S., Camici, G. G., Mohammed, S. A., Volpe, M., Luscher, T. F., Paneni, F . ( 2018) Epigenetics and cardiovascular regenerative medicine in the elderly. International Journal of Cardiology 250, 207-214. 10.1016/j.ijcard.2017.09.1884c47f9e989830132c20b0fe02f133051http%3A%2F%2Fwww.sciencedirect.com%2Fscience%2Farticle%2Fpii%2FS0167527317371176https://linkinghub.elsevier.com/retrieve/pii/S0167527317343358
[16]
Csiszar, A., Labinskyy, N., Smith, K., Rivera, A., Orosz, Z., Ungvari, Z . ( 2007) Vasculoprotective effects of anti-tumor necrosis factor-alpha treatment in aging. The American Journal of Pathology 170, 388-398. 10.2353/ajpath.2007.0607084da037cee34d9fe5d63b4538f3a417e4http%3A%2F%2Fwww.sciencedirect.com%2Fscience%2Farticle%2Fpii%2FS0002944010608620https://linkinghub.elsevier.com/retrieve/pii/S0002944010608620
[17]
Csiszar, A., Ungvari, Z., Koller, A., Edwards, J. G., Kaley, G . ( 2003) Aging-induced proinflammatory shift in cytokine expression profile in coronary arteries. The FASEB Journal 17, 1183-1185. 10.1096/fj.02-1049fjeb7637e0fc19c2c702ff4376d5e38b014http%3A%2F%2Feuropepmc.org%2Fabstract%2FMED%2F12709402http://www.fasebj.org/doi/10.1096/fj.02-1049fje
[18]
D'Andrade, P. N., Fulmer-Smentek, S. ( 2012) Agilent microRNA microarray profiling system. Methods in Molecular Biology 822, 85-102. 10.1007/978-1-61779-427-8http://www.springerlink.com/index/10.1007/978-1-61779-427-8
[19]
Deng, H. T., Liu, H. L., Zhai, B. B., Zhang, K., Xu, G. C., Peng, X. M., Zhang, Q. Z., Li, L. Y . ( 2017) Vascular endothelial growth factor suppresses TNFSF15 production in endothelial cells by stimulating miR-31 and miR-20a expression via activation of Akt and Erk signals. FEBS Open Bio 7, 108-117. 10.1002/feb4.2017.7.issue-1http://doi.wiley.com/10.1002/feb4.2017.7.issue-1
[20]
Diehl, F., Rossig, L., Zeiher, A. M., Dimmeler, S., Urbich, C . ( 2007) The histone methyltransferase MLL is an upstream regulator of endothelial-cell sprout formation. Blood 109, 1472-1478. 10.1182/blood-2006-08-039651e0cdc2f6d6bbd0f8818af55d7839bc7ahttp%3A%2F%2Fcardiovascres.oxfordjournals.org%2Flookup%2Fijlink%3FlinkType%3DABST%26amp%3BjournalCode%3Dbloodjournal%26amp%3Bresid%3D109%2F4%2F1472%26amp%3Batom%3D%252Fcardiovascres%252F90%252F3%252F404.atomhttp://www.bloodjournal.org/cgi/doi/10.1182/blood-2006-08-039651
[21]
Donato, A. J., Eskurza, I., Silver, A. E., Levy, A. S., Pierce, G. L., Gates,, P. E. , Seals,, D. R. ( 2007) Direct evidence of endothelial oxidative stress with aging in humans: relation to impaired endothelium-dependent dilation and upregulation of nuclear factor-kappaB. Circulation Research 100, 1659-1666. 10.1161/01.RES.0000269183.13937.e8https://www.ahajournals.org/doi/10.1161/01.RES.0000269183.13937.e8
[22]
Ernst, P., Vakoc, C. R . ( 2012) WRAD: enabler of the SET1-family of H3K4 methyltransferases. Briefings in Functional Genomics 11, 217-226. 10.1093/bfgp/els0172265269372d66c65012a4986eb919f99ed53f6e6http%3A%2F%2Fbfg.oxfordjournals.org%2Fcontent%2F11%2F3%2F217.abstracthttps://academic.oup.com/bfg/article-lookup/doi/10.1093/bfgp/els017
[23]
Farah, C., Michel, L. Y.M., Balligand, J. L . ( 2018) Nitric oxide signalling in cardiovascular health and disease. Nature Reviews Cardiology 15, 292-316. 10.1038/nrcardio.2017.2244ca5185be95288a076b8ce7a8d387e54http%3A%2F%2Fwww.ncbi.nlm.nih.gov%2Fpubmed%2F29388567
[24]
Fernandez, A. F., Bayon, G. F., Urdinguio, R. G., Torano, E. G., Garcia, M. G., Carella, A., Petrus-Reurer, S., Ferrero, C., MartinezCamblor, P., Cubillo, I., Garcia-Castro, J., Delgado-Calle, J., Perez-Campo, F. M., Riancho, J. A., Bueno, C., Menendez, P., Mentink, A., Mareschi, K., Claire, F., Fagnani, C., Medda, E., Toccaceli, V., Brescianini, S., Moran, S., Esteller, M., Stolzing, A., de Boer, J., Nistico, L., Stazi, M. A., Fraga, M. F . ( 2015) H3K4me1 marks DNA regions hypomethylated during aging in human stem and differentiated cells. Genome Research 25, 2740. cfc55c29dcc47b38f785f21cec59a9bbhttp%3A%2F%2Feuropepmc.org%2Fabstract%2Fmed%2F25271306
[25]
Fraineau, S., Palii, C. G., Allan, D. S., Brand, M . ( 2015) Epigenetic regulation of endothelial-cell-mediated vascular repair. The Federation of European Biochemical Societies 282, 1605-1629. 10.1111/febs.13183255463320855a1ca0a80edb2ab5ef374fb5fe353http%3A%2F%2Fonlinelibrary.wiley.com%2Fdoi%2F10.1111%2Ffebs.13183%2Fpdfhttp://onlinelibrary.wiley.com/doi/10.1111/febs.13183/pdf
[26]
Fulop, T., Larbi, A., Dupuis, G., Le Page, A., Frost, E. H., Cohen, A. A., Witkowski, J. M., Franceschi, C . ( 2017) Immunosenescence and inflamm-aAging as two sides of the same coin: friends or foes? Frontiers in Immunology 8, 1-13. daabb8e1ddc5f31b0006934498373112http%3A%2F%2Feuropepmc.org%2Farticles%2FPMC5767595%2F
[27]
Gillum, M. P., Kotas, M. E., Erion, D. M., Kursawe, R., Chatterjee, P., Nead, K. T., Muise, E. S., Hsiao, J. J., Frederick, D. W., Yonemitsu, S., Banks, A. S., Qiang, L., Bhanot, S., Olefsky, J. M., Sears, D. D., Caprio, S., Shulman, G. I . ( 2011) SirT1 regulates adipose tissue inflammation. Diabetes 60, 3235-3245. 10.2337/db11-061632199534c9fff8b48f4aa0abae3968497cb0b7ahttp%3A%2F%2Fwww.ncbi.nlm.nih.gov%2Fpubmed%2F22110092http://diabetes.diabetesjournals.org/cgi/doi/10.2337/db11-0616
[28]
Gomez, D., Swiatlowska, P., Owens, G. K . ( 2015) Epigenetic control of smooth muscle cell identity and lineage memory. Arteriosclerosis, Thrombosis, and Vascular Biology 35, 2508-2516. 10.1161/ATVBAHA.115.305044fcae77a1cab039dae8ec56a86b0567f7http%3A%2F%2Feuropepmc.org%2Fabstract%2FMED%2F26449751https://www.ahajournals.org/doi/10.1161/ATVBAHA.115.305044
[29]
Goodell, M. A., Rando, T. A . ( 2015) Stem cells and healthy aging. Science 350, 1199-1204. 10.1126/science.aab33881d08668ab1d7581c9897cd977490c81chttp%3A%2F%2Fwww.ncbi.nlm.nih.gov%2Fpubmed%2F26785478http://www.sciencemag.org/cgi/doi/10.1126/science.aab3388
[30]
Grootaert, M. O., da Costa Martins,, P. A., Bitsch, N., Pintelon, I., De Meyer, G. R., Martinet, W., Schrijvers, D. M . ( 2015) Defective autophagy in vascular smooth muscle cells accelerates senescence and promotes neointima formation and atherogenesis. Autophagy 11, 2014-2032. 10.1080/15548627.2015.109648525b5a26f6d5bf65dc63b38abe71e6efahttp%3A%2F%2Fwww.tandfonline.com%2Fdoi%2Fabs%2F10.1080%2F15548627.2015.1096485https://www.tandfonline.com/doi/full/10.1080/15548627.2015.1096485
[31]
Hajra, L., Evans, A. I., Chen, M., Hyduk, S. J., Collins, T., Cybulsky, M. I . ( 2000) The NF-kappa B signal transduction pathway in aortic endothelial cells is primed for activation in regions predisposed to atherosclerotic lesion formation. Proceedings of the National Academy of Sciences of the United States of America 97, 9052-9057. 10.1073/pnas.97.16.9052bbf32e74aa1106fbebc39ac70b736dd8http%3A%2F%2Feuropepmc.org%2Fabstract%2FMED%2F10922059http://www.pnas.org/cgi/doi/10.1073/pnas.97.16.9052
[32]
Harvey, A., Montezano, A. C., Touyz, R. M . ( 2015) Vascular biology of ageing-Implications in hypertension. Journal of Molecular and Cellular Cardiology 83, 112-121. 10.1016/j.yjmcc.2015.04.011https://linkinghub.elsevier.com/retrieve/pii/S0022282815001261
[33]
Hoffmann, J., Haendeler, J., Aicher, A., Rossig, L., Vasa, M., Zeiher, A. M., Dimmeler, S . ( 2001) Aging enhances the sensitivity of endothelial cells toward apoptotic stimuli: important role of nitric oxide. Circulation Research 89, 709-715. 10.1161/hh2001.097796https://www.ahajournals.org/doi/10.1161/hh2001.097796
[34]
Hu, M., Sun, X. J., Zhang, Y. L., Kuang, Y., Hu, C. Q., Wu, W. L., Shen, S. H., Du, T. T., Li, H., He, F., Xiao, H. S., Wang, Z. G., Liu, T. X., Lu, H., Huang, Q. H., Chen, S. J., Chen, Z . ( 2010) Histone H3 lysine 36 methyltransferase Hypb/Setd2 is required for embryonic vascular remodeling. Proceedings of the National Academy of Sciences of the United States of America 107, 2956-2961. 10.1073/pnas.09150331072013362586d9ae1c7135861b64b59bb90599c978http%3A%2F%2Fonlinelibrary.wiley.com%2Fresolve%2Freference%2FPMED%3Fid%3D20133625http://www.pnas.org/cgi/doi/10.1073/pnas.0915033107
[35]
Kaluza, D., Kroll, J., Gesierich, S., Manavski, Y., Boeckel, J. N., Doebele, C., Zelent, A., Rossig, L., Zeiher, A. M., Augustin, H. G., Urbich, C., Dimmeler, S . ( 2013) Histone deacetylase 9 promotes angiogenesis by targeting the antiangiogenic microRNA-17-92 cluster in endothelial cells. Arteriosclerosis, Thrombosis, and Vascular Biology 33, 533-543. 10.1161/ATVBAHA.112.30041569060a4e06f3d8ea4644b858e389d4e8http%3A%2F%2Feuropepmc.org%2Fabstract%2Fmed%2F23288173https://www.ahajournals.org/doi/10.1161/ATVBAHA.112.300415
[36]
Katusic, Z. S., Austin, S. A . ( 2014) Endothelial nitric oxide: protector of a healthy mind. European Heart Journal 35, 888-894. 10.1093/eurheartj/eht5449e6dd932769fa171594c8f7f19fbe235http%3A%2F%2Fpubmedcentralcanada.ca%2Fpmcc%2Farticles%2FPMC3977136%2Fhttps://academic.oup.com/eurheartj/article-lookup/doi/10.1093/eurheartj/eht544
[37]
Kimura, H. ( 2013) Histone modifications for human epigenome analysis. Journal of Human Genetics 58, 439-445. 10.1038/jhg.2013.66267683e1dd58b7c9d4cd338d23769ce9http%3A%2F%2Fbioinformatics.oxfordjournals.org%2Fexternal-ref%3Faccess_num%3D10.1038%2Fjhg.2013.66%26amp%3Blink_type%3DDOI
[38]
Kuro-o, M., Matsumura, Y., Aizawa, H., Kawaguchi, H., Suga, T., Utsugi, T., Ohyama, Y., Kurabayashi, M., Kaname, T., Kume, E., Iwasaki, H., Iida, A., Shiraki-Iida, T., Nishikawa, S., Nagai, R., Nabeshima, Y. I . ( 1997) Mutation of the mouse klotho gene leads to a syndrome resembling ageing. Nature 390, 45-51. 10.1038/3628563f700050a37b5dcc7af30204a39109ahttp%3A%2F%2Fwww.tandfonline.com%2Fservlet%2Flinkout%3Fsuffix%3Dcit0025%26amp%3Bdbid%3D8%26amp%3Bdoi%3D10.1080%252F21675511.2016.1198458%26amp%3Bkey%3D9363890
[39]
Lakatta, E. G . ( 2003) Arterial and cardiac aging: major shareholders in cardiovascular disease enterprises: Part III: cellular and molecular clues to heart and arterial aging. Circulation 107, 490-497. 10.1161/01.CIR.0000048894.99865.02549f2bdd21d0e282e43143403231c96fhttp%3A%2F%2Fepirev.oxfordjournals.org%2Fcgi%2Fijlink%3FlinkType%3DFULL%26amp%3BjournalCode%3Dcirculationaha%26amp%3Bresid%3D107%2F1%2F139https://www.ahajournals.org/doi/10.1161/01.CIR.0000048894.99865.02
[40]
Laurent, S. ( 2012) Defining vascular aging and cardiovascular risk. Journal of Hypertens 30, S3-S8. 10.1097/HJH.0b013e328353e5010e4191e5e634af4dd7aee4f2d05cce72http%3A%2F%2Feuropepmc.org%2Fabstract%2FMED%2F23124102http://content.wkhealth.com/linkback/openurl?sid=WKPTLP:landingpage&an=00004872-201206001-00002
[41]
Lee, S. J., Smith, A., Guo, L., Alastalo, T. P., Li, M., Sawada, H., Liu, X., Chen, Z. H., Ifedigbo, E., Jin, Y., Feghali-Bostwick, C., Ryter, S. W., Kim, H. P., Rabinovitch, M., Choi, A. M . ( 2011) Autophagic protein LC3B confers resistance against hypoxia-induced pulmonary hypertension. American Journal of Respiratory and Critical Care Medicine 183, 649-658. 10.1164/rccm.201005-0746OC84a9f2fded24697a5575689f520cbd41http%3A%2F%2Fpubmedcentralcanada.ca%2Fpmcc%2Farticles%2FPMC3081281%2F%3Flang%3Dfr%26amp%3Bterm%3Dhttp://www.atsjournals.org/doi/abs/10.1164/rccm.201005-0746OC
[42]
Li, L., Zhang, H., Chen, H., Gao, P., Zhu, L., Li, H., Lv, X., Zhang, Q., Zhang, R., Wang, Z., She, Z., Zhang, R., Wei, Y., Du, G., Liu, D., Liang, C . ( 2011) SIRT1 acts as a modulator of neointima formation following vascular injury in mice. Circulation Research 108, 1180-1189. 10.1161/CIRCRESAHA.110.23787521b3411c2a3d7a7915dd3abb72b637a5http%3A%2F%2Fwww.ncbi.nlm.nih.gov%2Fpubmed%2F21474819https://www.ahajournals.org/doi/10.1161/CIRCRESAHA.110.237875
[43]
Liu, L., Li, Y., Tollefsbol, T. O . ( 2008) Gene-environment interactions and epigenetic basis of human diseases. Current Issues in Molecular Biology 10, 25-36. 0bb673a0f6ecd787b9c9b0db024aab5fhttp%3A%2F%2Fpubmedcentralcanada.ca%2Fpmcc%2Farticles%2FPMC2434999%2F
[44]
Long, L., Yang, X., Southwood, M., Lu, J., Marciniak, S. J., Dunmore, B. J., Morrell, N. W . ( 2013) Chloroquine prevents progression of experimental pulmonary hypertension via inhibition of autophagy and lysosomal bone morphogenetic protein type II receptor degradation. Circulation Research 112, 1159-1170. 10.1161/CIRCRESAHA.111.3004834fbcd7df4fe46a3a2a126074a2870759http%3A%2F%2Fjac.oxfordjournals.org%2Fcgi%2Fijlink%3FlinkType%3DABST%26amp%3BjournalCode%3Dcircresaha%26amp%3Bresid%3D112%2F8%2F1159https://www.ahajournals.org/doi/10.1161/CIRCRESAHA.111.300483
[45]
Lopez-Otin, C., Blasco, M. A., Partridge, L., Serrano, M., Kroemer, G . ( 2013) The hallmarks of aging. Cell 153, 1194-1217. 10.1016/j.cell.2013.05.039https://linkinghub.elsevier.com/retrieve/pii/S0092867413006454
[46]
Marttila, S., Kananen, L., Hayrynen, S., Jylhava, J., Nevalainen, T., Hervonen, A., Jylha, M., Nykter, M., Hurme, M . ( 2015) Ageingassociated changes in the human DNA methylome: genomic locations and effects on gene expression. BMC Genomics 16, 1-17. 10.1186/1471-2164-16-1ffdec8f0dffa63845eb2c6b0337ea385http%3A%2F%2Fjcp.bmj.com%2Flookup%2Fexternal-ref%3Faccess_num%3D10.1186%2F1471-2164-16-1%26amp%3Blink_type%3DDOIhttp://bmcgenomics.biomedcentral.com/articles/10.1186/1471-2164-16-1
[47]
Miles, E. A., Rees, D., Banerjee, T., Cazzola, R., Lewis, S., Wood, Q., Oates, R., Tallant, A., Cestaro, B., Yaqoob, P., Wahle, K. W., Calder, P. C . ( 2008) Age-related increases in circulating inflammatory markers in men are independent of BMI, blood pressure and blood lipid concentrations. Atherosclerosis 196, 298-305. 10.1016/j.atherosclerosis.2006.11.002a15e63c015cd3b9cb2df27dcefde5554http%3A%2F%2Feuropepmc.org%2Fabstract%2FMED%2F17118371https://linkinghub.elsevier.com/retrieve/pii/S0021915006006721
[48]
Mocharla, P., Briand, S., Giannotti, G., Dörries, C., Jakob, P., Paneni, F., Lüscher, T., Landmesser, U . ( 2013) AngiomiR-126 expression and secretion from circulating CD34(+) and CD14(+) PBMCs: role for proangiogenic effects and alterations in type 2 diabetics. Blood 121, 226-236. 10.1182/blood-2012-01-407106f0286dda6ee8128128d8e6580f0f9b89http%3A%2F%2Fwww.ncbi.nlm.nih.gov%2Fpubmed%2F23144172http://www.bloodjournal.org/cgi/doi/10.1182/blood-2012-01-407106
[49]
Mostoslavsky, R., Chua, K. F., Lombard, D. B., Pang, W. W., Fischer, M. R., Gellon, L., Liu, P., Mostoslavsky, G., Franco, S., Murphy, M. M., Mills, K. D., Patel, P., Hsu, J. T., Hong, A. L., Ford, E., Cheng, H. L., Kennedy, C., Nunez, N., Bronson, R., Frendewey, D., Auerbach, W., Valenzuela, D., Karow, M., Hottiger, M. O., Hursting, S., Barrett, J. C., Guarente, L., Mulligan, R., Demple, B., Yancopoulos, G. D., Alt, F. W . ( 2006) Genomic instability and aging-like phenotype in the absence of mammalian SIRT6. Cell 124, 315-329. 10.1016/j.cell.2005.11.044dd1341a757ea098e70f3209b4aedd951http%3A%2F%2Fwww.cell.com%2Fretrieve%2Fpii%2FS0092867406000493%3Fscript%3Dtruehttps://linkinghub.elsevier.com/retrieve/pii/S0092867406000493
[50]
Mottet, D., Bellahcene, A., Pirotte, S., Waltregny, D., Deroanne, C., Lamour, V., Lidereau, R., Castronovo, V . ( 2007) Histone deacetylase 7 silencing alters endothelial cell migration, a key step in angiogenesis. Circulation Research 101, 1237-1246. 10.1161/CIRCRESAHA.107.1493770fc9fc164f195905e283e737e4cdba17http%3A%2F%2Feuropepmc.org%2Fabstract%2Fmed%2F17947801https://www.ahajournals.org/doi/10.1161/CIRCRESAHA.107.149377
[51]
Najjar, S. S., Scuteri, A., Lakatta, E. G . ( 2005) Arterial aging: is it an immutable cardiovascular risk factor? Hypertension 46, 454-462. 10.1161/01.HYP.0000177474.06749.98https://www.ahajournals.org/doi/10.1161/01.HYP.0000177474.06749.98
[52]
Napoli, C., Martin-Padura, I., de Nigris, F., Giorgio, M., Mansueto, G., Somma, P., Condorelli, M., Sica, G., De Rosa, G., Pelicci, P . ( 2003) 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. Proceedings of the National Academy of Sciences of the United States of America 100, 2112-2116. 10.1073/pnas.0336359100b0244352d970dcfef07ac4b5e6563701http%3A%2F%2Fcardiovascres.oxfordjournals.org%2Flookup%2Fijlink%3FlinkType%3DABST%26amp%3BjournalCode%3Dpnas%26amp%3Bresid%3D100%2F4%2F2112%26amp%3Batom%3D%252Fcardiovascres%252F73%252F2%252F414.atomhttp://www.pnas.org/cgi/doi/10.1073/pnas.0336359100
[53]
Okano, M., Bell, D. W., Haber, D. A., Li, E . ( 1999) DNA methyltransferases Dnmt3a and Dnmt3b are essential for de novo methylation and mammalian development. Cell 99, 247-257. 10.1016/S0092-8674(00)81656-6105551415e25dc02c4d36c63b3812e334c85abbchttp%3A%2F%2Fwww.biomedcentral.com%2Fpubmed%2F10555141https://linkinghub.elsevier.com/retrieve/pii/S0092867400816566
[54]
Paneni, F., Costantino, S., Cosentino, F . ( 2015) Molecular pathways of arterial aging. Clinical science 128, 69-79. 10.1042/CS2014030225236971289e28847f360f3b336fb6794d36623chttp%3A%2F%2Feuropepmc.org%2Fabstract%2FMED%2F25236971http://www.clinsci.org/cgi/doi/10.1042/CS20140302
[55]
Paneni, F., Volpe, M., Luscher, T. F., Cosentino, F . ( 2013) SIRT1, p66(Shc), and Set7/9 in vascular hyperglycemic memory: bringing all the strands together. Diabetes 62, 1800-1807. 10.2337/db12-1648http://diabetes.diabetesjournals.org/cgi/doi/10.2337/db12-1648
[56]
Pernow, J., Jung, C. ( 2013) Arginase as a potential target in the treatment of cardiovascular disease: reversal of arginine steal? Cardiovascular Research 98, 334-343. 10.1093/cvr/cvt036c5a3f9a088e7425c6ba6fbabfa606adchttp%3A%2F%2Fwww.tandfonline.com%2Fservlet%2Flinkout%3Fsuffix%3DCIT0025%26amp%3Bdbid%3D8%26amp%3Bdoi%3D10.1080%252F14017431.2018.1459823%26amp%3Bkey%3D23417041https://academic.oup.com/cardiovascres/article-lookup/doi/10.1093/cvr/cvt036
[57]
Pierce, G. L., Larocca, T. J . ( 2008) Reduced vascular tetrahydrobiopterin (BH4) and endothelial function with ageing: is it time for a chronic BH4 supplementation trial in middle-aged and older adults? Journal of Physiology 586, 2673-2674. 10.1113/jphysiol.2008.154229http://doi.wiley.com/10.1113/jphysiol.2008.154229
[58]
Potente, M., Ghaeni, L., Baldessari, D., Mostoslavsky, R., Rossig, L., Dequiedt, F., Haendeler, J., Mione, M., Dejana, E., Alt, F. W., Zeiher, A. M., Dimmeler, S . ( 2007) SIRT1 controls endothelial angiogenic functions during vascular growth. Genes & Development 21, 2644-2658. 1431264135bb229f0074cc878815dd6ehttp%3A%2F%2Feurheartj.oxfordjournals.org%2Fcgi%2Fijlink%3FlinkType%3DABST%26amp%3BjournalCode%3Dgenesdev%26amp%3Bresid%3D21%2F20%2F2644
[59]
Rizza, W., Veronese, N., Fontana, L . ( 2014) What are the roles of calorie restriction and diet quality in promoting healthy longevity? Ageing Research Reviews 13, 38-45. 10.1016/j.arr.2013.11.00224291541da3ec2fead384092e0f9825928193159http%3A%2F%2Fwww.ncbi.nlm.nih.gov%2Fpubmed%2F24291541https://linkinghub.elsevier.com/retrieve/pii/S1568163713000901
[60]
Snowden, A. W., Gregory, P. D., Case, C. C., Pabo, C. O . ( 2002) Gene-specific targeting of H3K9 methylation is sufficient for initiating repression in vivo. Current Biology 12, 2159-2166. 10.1016/S0960-9822(02)01391-X5146ee1745b76b5b16b971fbf804640chttp%3A%2F%2Feuropepmc.org%2Fabstract%2Fmed%2F12498693https://linkinghub.elsevier.com/retrieve/pii/S096098220201391X
[61]
Ugalde, A. P., Ramsay, A. J., de la Rosa, J., Varela, I., Marino, G., Cadinanos, J., Lu, J., Freije, J. M., Lopez-Otin, C . ( 2011) Aging and chronic DNA damage response activate a regulatory pathway involving miR-29 and p53. The EMBO Journal 30, 2219-2232. 10.1038/emboj.2011.1247fc599c01417f46708f62c30ea3439cdhttp%3A%2F%2Fonlinelibrary.wiley.com%2Fdoi%2F10.1038%2Femboj.2011.124%2Ffullhttp://emboj.embopress.org/cgi/doi/10.1038/emboj.2011.124
[62]
Ungvari, Z., Labinskyy, N., Gupte, S., Chander, P. N., Edwards, J. G., Csiszar, A . ( 2008) Dysregulation of mitochondrial biogenesis in vascular endothelial and smooth muscle cells of aged rats. American Journal of Physiology-Heart and Circulatory Physiology 294, H2121-2128. 10.1152/ajpheart.00012.200803a1a684de4792f5408f570517fe9be8http%3A%2F%2Fwww.ncbi.nlm.nih.gov%2Fpubmed%2F18326800http://www.physiology.org/doi/10.1152/ajpheart.00012.2008
[63]
Ungvari, Z., Kaley, G., de Cabo, R., Sonntag, W. E., Csiszar, A . ( 2010) Mechanisms of vascular aging: new perspectives. The Journals of Gerontology. Series A, Biological Sciences and Medical Sciences 65, 1028-1041. 10.1093/gerona/glq1132950814dbb8d9d88a6cd349389865becf61009ahttp%3A%2F%2Fwww.ncbi.nlm.nih.gov%2Fpubmed%2F20576649
[64]
Ungvari, Z., Orosz, Z., Labinskyy, N., Rivera, A., Xiangmin, Z., Smith, K., Csiszar, A . ( 2007) Increased mitochondrial H2O2 production promotes endothelial NF-kappaB activation in aged rat arteries. American Journal of Physiology-Heart and Circulatory Physiology 293, H37-H47. 10.1152/ajpheart.01346.2006http://www.physiology.org/doi/10.1152/ajpheart.01346.2006
[65]
Urbich, C, Rossig, L, Kaluza, D, Potente, M, Boeckel, JN, Knau, A, Diehl, F, Geng, JG, Hofmann, W. K., Zeiher, A. M., Dimmeler, S . ( 2009) HDAC5 is a repressor of angiogenesis and determines the angiogenic gene expression pattern of endothelial cells. Blood 113, 5669-5679. 10.1182/blood-2009-01-19648519351956491bf5b8d9c0af7fc28b8efb6ee584c6http%3A%2F%2Feuropepmc.org%2Fabstract%2Fmed%2F19351956http://www.bloodjournal.org/cgi/doi/10.1182/blood-2009-01-196485
[66]
Usui, T., Morita, T., Okada, M., Yamawaki, H . ( 2014) Histone deacetylase 4 controls neointimal hyperplasia via stimulating proliferation and migration of vascular smooth muscle cells. Hypertension 63, 397-403. 10.1161/HYPERTENSIONAHA.113.01843718f367794e648b5dc07f300ea04c861http%3A%2F%2Feuropepmc.org%2Fabstract%2Fmed%2F24166750https://www.ahajournals.org/doi/10.1161/HYPERTENSIONAHA.113.01843
[67]
Vasa-Nicotera, M., Chen, H., Tucci, P., Yang, A. L., Saintigny, G., Menghini, R., Mahe, C., Agostini, M., Knight, R. A., Melino, G., Federici, M . ( 2011) miR-146a is modulated in human endothelial cell with aging. Atherosclerosis 217, 326-330. 10.1016/j.atherosclerosis.2011.03.034882941ccee5b8f377c622ae9591c65cdhttp%3A%2F%2Fwww.ncbi.nlm.nih.gov%2Fpubmed%2F21511256https://linkinghub.elsevier.com/retrieve/pii/S0021915011002954
[68]
Vassallo, P. F., Simoncini, S., Ligi, I., Chateau, A. L., Bachelier, R., Robert, S., Morere, J., Fernandez, S., Guillet, B., Marcelli, M., Tellier, E., Pascal, A., Simeoni, U., Anfosso, F., Magdinier, F., Dignat-George, F., Sabatier, F . ( 2014) Accelerated senescence of cord blood endothelial progenitor cells in premature neonates is driven by SIRT1 decreased expression. Blood 123, 2116-2126. 10.1182/blood-2013-02-48495624518759a022f0b728cc2b780076f6a25929a957http%3A%2F%2Feuropepmc.org%2Fabstract%2Fmed%2F24518759http://www.bloodjournal.org/lookup/doi/10.1182/blood-2013-02-484956
[69]
Villeneuve, L. M., Natarajan, R . ( 2010) The role of epigenetics in the pathology of diabetic complications. American Journal of Physiology-Renal Physiology 299, 14-25. 10.1152/ajprenal.00200.2010204629729257f71a5da5b1e08d3b317f10a302ddhttp%3A%2F%2Feuropepmc.org%2Farticles%2FPMC2904177%2F
[70]
Wei, F. Z., Cao, Z., Wang, X., Wang, H., Cai, M. Y., Li, T., Hattori, N., Wang, D., Du, Y., Song, B., Cao, L. L., Shen, C., Wang, L., Wang, H., Yang, Y., Xie, D., Wang, F., Ushijima, T., Zhao, Y., Zhu, W. G . ( 2015) Epigenetic regulation of autophagy by the methyltransferase EZH2 through an MTOR-dependent pathway. Autophagy 11, 2309-2322. 10.1080/15548627.2015.1117734f1d8c8330672e9109a368d34b7ef3756http%3A%2F%2Fwww.tandfonline.com%2Fdoi%2Fabs%2F10.1080%2F15548627.2015.1117734https://www.tandfonline.com/doi/full/10.1080/15548627.2015.1117734
[71]
Wu, J., Xia, S., Kalionis, B., Wan, W . ( 2014) The role of oxidative stress and inflammation in cardiovascular aging. BioMed Research International 2014, 1-13. 7f9a09b1c5dfc42c4732713aea8ca86bhttp%3A%2F%2Feuropepmc.org%2Fabstract%2Fmed%2F25143940
[72]
Wu, Z., Yi Yu, Y., Liu, C., Xiong, Y., Montani, J. P., Yang, Z., Ming, X. F . ( 2015) Role of p38 mitogen-activated protein kinase in vascular endothelial aging: interaction with Arginase-II and S6K1 signaling pathway. Aging 7, 70-81. 10.18632/aging.v7i143503256b95efa543ccb2b9d58e7e367a7f28d5http%3A%2F%2Feuropepmc.org%2Farticles%2FPMC4350325%2Fhttp://www.aging-us.com/issue/v7i1
[73]
Ye, L. X., Yu, J., Liang, Y. X., Zeng, J. S., Huang, R. X., Liao, S. J . ( 2014) Beclin 1 knockdown retards re-endothelialization and exacerbates neointimal formation via a crosstalk between autophagy and apoptosis. Atherosclerosis 237, 146-154. 10.1016/j.atherosclerosis.2014.08.0521884002a75e722e6d8e1fd964f9a6ba4http%3A%2F%2Feuropepmc.org%2Fabstract%2Fmed%2F25238224https://linkinghub.elsevier.com/retrieve/pii/S0021915014013811
[74]
Yeo, T. W., Lampah, D. A., Kenangalem, E., Tjitra, E., Price, R. N., Weinberg, J. B., Hyland, K., Granger, D. L., Anstey, N. M . ( 2015) Impaired systemic tetrahydrobiopterin bioavailability and increased dihydrobiopterin in adult falciparum malaria: association with disease severity, impaired microvascular function and increased endothelial activation. PLoS Pathog 11, 1-13. 10.1371/journal.ppat.1004667435738657a724fb850e34bfa588ae60472184b5http%3A%2F%2Feuropepmc.org%2Farticles%2FPMC4357386http://europepmc.org/articles/PMC4357386
[75]
Yepuri, G., Velagapudi, S., Xiong, Y., Rajapakse, A. G., Montani, J. P., Ming, X. F., Yang, Z . ( 2012) Positive crosstalk between arginase-II and S6K1 in vascular endothelial inflammation and aging. Aging Cell 11, 1005-1016. 10.1111/acel.1200122928666c56d3e2feabba118807c4d8254a1e58bhttp%3A%2F%2Fwww.ncbi.nlm.nih.gov%2Fpubmed%2F22928666http://doi.wiley.com/10.1111/acel.2012.11.issue-6
[76]
Zeng, L., Xiao, Q., Margariti, A., Zhang, Z., Zampetaki, A., Patel, S., Capogrossi, M. C., Hu, Y., Xu, Q . ( 2006) HDAC3 is crucial in shearand VEGF-induced stem cell differentiation toward endothelial cells. The Journal of Cell Biology 174, 1059-1069. 10.1083/jcb.200605113c2a626bf0a976c9978fd878df5af5efchttp%3A%2F%2Fwww.onacademic.com%2Fdetail%2Fjournal_1000036515961810_505e.htmlhttp://www.jcb.org/lookup/doi/10.1083/jcb.200605113
[77]
Zheng, Z., Chen, H., Li, J., Li, T., Zheng, B., Zheng, Y., Jin, H., He, Y., Gu, Q., Xu, X . ( 2012) Sirtuin 1-mediated cellular metabolic memory of high glucose via the LKB1/AMPK/ROS pathway and therapeutic effects of metformin. Diabetes 61, 217-228. 10.2337/db11-041632376624c547a74ced8e6c05b19d17dda519953http%3A%2F%2Feuropepmc.org%2Fabstract%2FMED%2F22124463http://diabetes.diabetesjournals.org/cgi/doi/10.2337/db11-0416
[78]
Zhu, S., Deng, S., Ma, Q., Zhang, T., Jia, C., Zhuo, D., Yang, F., Wei, J., Wang, L., Dykxhoorn, D. M., Hare, J. M., Goldschmidt-Clermont, P. J., Dong, C . ( 2013) MicroRNA-10A* and MicroRNA-21 modulate endothelial progenitor cell senescence via suppressing high-mobility group A2. Circulation Research 112, 152-164. 10.1161/CIRCRESAHA.112.28001623072816fd4a67ee29288f9c5d44889cba9d90c3http%3A%2F%2Fonlinelibrary.wiley.com%2Fresolve%2Freference%2FPMED%3Fid%3D23072816https://www.ahajournals.org/doi/10.1161/CIRCRESAHA.112.280016
Share
Back to top