Open access peer-reviewed chapter

Targeting Endothelial SIRT1 for the Prevention of Arterial Aging

Written By

Yumeng Guo and Yu Wang

Submitted: 08 May 2017 Reviewed: 08 December 2017 Published: 22 February 2018

DOI: 10.5772/intechopen.73019

From the Edited Volume

Endothelial Dysfunction - Old Concepts and New Challenges

Edited by Helena Lenasi

Chapter metrics overview

1,272 Chapter Downloads

View Full Metrics

Abstract

Cardiovascular diseases are the leading cause of morbidity and mortality in the elderly population all over the world. Arterial aging is the earliest manifestation and a key risk factor for age-induced cardiovascular abnormalities. The longevity regulator Sirtuin 1 (SIRT1) is abundantly expressed in the endothelium of the arteries and elicits potent protective functions against arterial aging. Targeting endothelial SIRT1 represents a promising approach for the prevention and treatment of cardiovascular diseases. This chapter provides an overview of SIRT1’s regulation and function in endothelial cells and discusses the potential applications of targeting endothelial SIRT1 for arterial aging-related cardiovascular diseases.

Keywords

  • sirtuins
  • endothelium
  • cellular senescence
  • vasodilatation
  • arterial remodeling
  • hypertension
  • atherosclerosis

1. Arterial aging

Chronological age is associated with a progressive alteration of arterial structure and function, herein referred to as arterial aging, which contributes to the development of a wide range of cardiovascular diseases including hypertension, atherosclerosis, heart failure, and stroke [1, 2, 3]. Arterial system is composed of three types of arteries including large elastic or conduit arteries, medium-sized muscular arteries, and small arteries referred to resistance arteries. Arterial aging is characterized by endothelial dysfunction and arterial remodeling, indicating a decline in arterial elasticity/distensibility, decreased arterial compliance, and increased arterial stiffness. Physiological alterations of the vascular wall are dynamic and occur throughout life [4]. During aging, gradual thickening of the arterial wall, changes in wall composition (i.e., elastin fragmentation and collagen deposition), and an increase of artery diameter are observed in conduit arteries [2]. Increased intimal-to-media thickness (IMT) is a valid indicator of arterial aging supported by the finding that the IMT of the carotid artery increases twofold to threefold between 20 and 90 years of age [4]. Pulse wave velocity (PWV) is a noninvasive measure of vascular stiffness. Stiffening of the conduit arteries leads to increased aortic pulse pressure and increased PWV, which occurs in both sexes along aging [5].

The endothelium, a monolayer of flattened, polygonal cells lining the inner surface of arteries, plays an important role in regulating arterial structure and function. The endothelium can respond to pathophysiological signals by producing various factors that regulate vascular tone, cellular adhesion, thromboresistance, smooth muscle cell proliferation, and inflammation. During arterial aging, senescence, activation, and dysfunction of endothelial cells (ECs) represent the earliest abnormalities that lead to an impaired endothelium-dependent vasodilatation and adverse arterial wall remodeling [6]. Senescent ECs undergo permanent growth arrest, get enlarged and flattened in morphology, and also display positive staining for senescence-associated β-galactosidase (SA-β-gal) [7]. There are mainly two types of senescence. One is caused by successive cell duplication as a kind of natural aging process termed as “replicative senescence” and characterized by shortening of telomere [8]. The other is called “premature senescence” and induced by several stress conditions such as oxidative stress, radiations, and exposure to oncogenes [9]. Endothelial activation is defined as the initial event in atherogenesis. Circulating proinflammatory molecules including cytokines (i.e., tumor necrosis factor-α (TNF-α)) or modified lipoproteins (i.e., oxidized low-density lipoprotein (oxLDL)) activates ECs to express chemokines, cytokines, and adhesion molecules, thus attracting and recruiting inflammatory cells such as macrophages and T cells. Both endothelial senescence and activation can induce endothelial dysfunction which is reflected by impairment of endothelium-dependent vasorelaxation caused by a loss of nitric oxide (NO) bioavailability in the vessel wall and altered anticoagulant and anti-inflammatory properties of the endothelium. Impaired endothelium-dependent vasodilation in the coronary circulation of humans has profound prognostic implications in that it predicts adverse cardiovascular events and long-term outcomes [1, 2, 10, 11].

Age-related loss of arterial functions has been demonstrated, and underlying mechanisms were studied in human studies. Reduced NO bioavailability in older age was reported by observing diminished forearm vasoconstrictor response to infusion of NO-synthase inhibitor L-NMMA in resistance arteries [12]. In older adults, supplementation of NO precursor, l-arginine, improves coronary artery blood flow response to acetylcholine [13] and skin blood flow response to whole body heating [14]. Moreover, age-related decline in synthesis of tetrahydrobiopterin, a co-factor in NO production, provides further evidence for impairment of vasodilation NO-pathway during aging [15]. In the aspect of vasoconstrictor pathways, a greater lower limb vasodilatation response to endothelin (ET)-receptor blockade in old men was reported [16]. A small but significant age-related impairment in vascular smooth muscle function was also observed in conduit and resistance arteries in a meta-analysis [17].

Advertisement

2. SIRT1 in endothelial cells: expression and regulation

Sirtuin 1 (SIRT1) is the mammalian orthologue of the yeast longevity regulator Sir2 [18]. Members of the Sirtuin family share a highly conserved core domain to catalyze deacetylation, ADP-ribosyltransferation, desuccinylation, and demalonylation [19]. Sirtuins regulate energy homeostasis, stress resistance, circadian rhythmicity, mitochondrial functions, and embryonic development, which in turn contribute to increased lifespan [20].

Human SIRT1 gene is located at chromosome 10q21.3 containing 11 exons with a total length of 33,715 base pair [21]. SIRT1 is composed of 747 amino acids including a core catalytic domain consisting of 275 amino acids and both N- and C-terminal extensions spanning about 240 amino acids [22]. There are two nuclear localization signals and two nuclear exportation signals located in the extensions whose balanced functionality determines the presence of SIRT1 in either the nucleus or cytoplasm and explains the distinct location of SIRT1 among different cell lines and tissues [23].

Regulation of SIRT1 enzymatic activity occurs at various levels including post-translational modification, protein complex formation, transcriptional regulation, and concentrations of enzymatic substrates [19, 24]. Phosphorylation of SIRT1 represents the major form of post-translational modifications. Independent studies report multiple phosphorylation sites by distinct proteins, including c-Jun N-terminal kinase 1 (Ser27/47), cyclin B/cyclin-dependent kinase 1 (Thr530, Ser540), casein kinase 2 (Ser659/661), and adenosine 5′-monophosphate-activated protein kinase (AMPK) (Thr344) [25, 26, 27, 28]. Additional post-translational modifications include methylation by SET7/9 [29], nitrosylation by glyceraldehyde-3-phosphate dehydrogenase [30], and sumoylation by sentrin-specific protease 1 [31]. In addition, several endogenous protein-binding partners of SIRT1 are found to regulate its function via forming protein complex. For example, the active regulator of SIRT1 can bind to amino acids 114–217 in the N-terminus of SIRT1 and stimulate deacetylation of p53 in vivo [32]. On transcriptional level, SIRT1 was reported that nicotinamide phosphoribosyltransferase (NAMPT) upregulated the expression of SIRT1 and SIRT1 antisense long noncoding RNA, thus regulating senescence, proliferation, and migration of endothelial progenitor cells (EPCs) [33]. SIRT1 activity is also thought to be affected by the levels of intracellular co-substrate nicotinamide adenine dinucleotide (NAD+) and its product nicotinamide [34].

With age, SIRT1 expression in ECs is progressively downregulated. Overexpression of SIRT1 in the endothelium prevents cellular senescence, enhances vasodilatory responses, and attenuates aging-induced vascular damages [35, 36, 37]. The subsequent review will summarize the recent progresses related to the molecular regulation of SIRT1 expression in ECs and the anti-vascular aging effects of SIRT1 by focusing on endothelial dysfunction and arterial remodeling.

Advertisement

3. SIRT1 in endothelial cells: molecular targets and biological functions

Apart from histones, SIRT1 can mediate the deacetylation of various signaling substrates to exert vasoprotective functions. SIRT1 is abundant in ECs mediating postnatal blood vessel growth via Foxo1 and helps to maintain endothelial function [38]. In vitro experiments showed that downregulation of SIRT1 using small interfering RNA (siRNA) uniquely inhibited endothelial sprout formation via a three-dimensional assay, while other mammalian sirtuin family members (SIRT2–SIRT7) could not [38]. In addition, the reduction of matrix metalloproteinase-14 (MMP-14), a membrane-anchored MMP essential for tip cell activity during sprouting angiogenesis, was found in siRNA-SIRT1-treated endothelial sprouts [39]. Decreased expression of SIRT1 either by mRNA silencing or pharmacological inhibition could induce premature-senescence-like phenotypes in ECs [40, 41]. SIRT1 displays anti-senescence activity in ECs by inducing the deacetylation of diversified signaling substrates [42]. For example, SIRT1 can deacetylate tumor suppressor protein p53 to downregulate its stability and activity as to promote cell survival in response to cellular stress [43]. SIRT1 also plays an important role in enhancing the endothelial NO synthase (eNOS) transcription and translation by deacetylating eNOS on lysine 496 and 506 to generate more NO, thus enhancing vessel dilatation, mediating vessel tone regulation, and providing athero-protective effects [44, 45]. Recent study demonstrated that SIRT1 activation could help reduce traction forces and reorganize actin localization (increased peripheral actin) in aged ECs, which is also a sign of anti-senescent effect [46]. Moreover, while senescent porcine aortic ECs (PAECs) showed decreased expression of SIRT1 compared to young PAECs, the protein level of liver kinase B1 (LKB1), a serine/threonine kinase and tumor suppressor, was dramatically increased as well as the phosphorylation of its downstream target AMPK (Thr172). In this case, SIRT1 can antagonize LKB1-dependent AMPK activation by promoting the deacetylation, ubiquitination, and proteasome-mediated degradation in order to retard PAEC senescence which also correlated with the Akt survival signaling pathway [41]. Furthermore, it was reported that SIRT1 can bind to the DOC domain of HERC2 [HECT and RLD domain containing E3 ubiquitin protein ligase 2] and then ubiquitinate LKB1 in the nuclear compartment of ECs [37]. SIRT1 can also negatively modulate Notch signaling in ECs via deacetylation of the Notch1 intracellular domain (NICD), in which loss of endothelial SIRT1 activity leads to impaired growth and sprout elongation [47]. Intracellular NAMPT-NAD+-SIRT1 cascade was shown to improve post-ischemic neovascularization through modulation of Notch signaling pathway [48]. Adapter protein p66Shc which can directly stimulate mitochondrial reactive oxygen species (ROS) generation was discovered downregulated by SIRT1 in mice with hyperglycemia-induced endothelial dysfunction [49]. Moreover, in vitro experiments using human aortic ECs (HUVECs) demonstrated that SIRT1 can deacetylate RelA/p65 to diminish tissue factor expression and suppress nuclear factor-κB (NF-κB) signaling, thus preventing atherothrombosis [50]. In EPCs, SIRT1 was implicated to protect against oxidative stress-induced apoptosis by inhibiting Foxo3a via ubiquitination and degradation [36]. microRNA-34a (miR-34a), regulated by p53 and able to control cell cycle arrest, has been reported to promote cardiac, endothelial, and EPC senescence via downregulation of SIRT1 [51]. Also, visfatin (an adipocytokine closely associated with human cell senescence) was reported to attenuate the oxLDL-induced senescence of EPCs by upregulating SIRT1 expression through the PI3K/Akt/ERK pathway [52].

Advertisement

4. Endothelial SIRT1 prevents arterial aging

Various animal studies demonstrated that SIRT1 plays a vital role in anti-endothelial senescence and anti-atherogenesis. Infiltration of monocyte-derived macrophages into the subendothelial space is a crucial step in atherogenesis [53]. SIRT1 can decrease cholesterol uptake especially oxLDL and prevent macrophage foam cell formation via suppressing the expression of scavenger receptor Lox-1 [54] and reducing the expression of various pro-inflammatory molecules including TNF-α, monocyte chemotactic protein-1, and interleukins [55]. A recent discovery showed that treatment of the SIRT1 activator SRT3025 decreased plasma levels of LDL cholesterol and total cholesterol and attenuated atherosclerosis, owing to reduced secretion of hepatic Pcsk9 and enhanced protein expression of LDL receptor in apolipoprotein E-deficient (ApoE−/−) mice [56]. In the meantime, SIRT1 was demonstrated to promote reverse cholesterol (mainly HDLs) transport into macrophages by directly deacetylating and subsequently regulating the transcriptional activity of liver X receptors, which play a significant role in lipid homeostasis and inflammation and can help express ATP-binding cassette transporter 1 that transport cholesterol into pre-βHDL particles [57].

Some studies regarding upstream regulators of SIRT1 including cathepsin, caspase-1, and cyclin-dependent kinase 5 (CDK5) elucidate beneficial roles of SIRT1 in anti-endothelial senescence and anti-atherogenesis [58, 59, 60]. The cysteine cathepsins belong to the leaked lysosomal contents with the viability in cleavage and degradation of SIRT1, which lead to stress-induced premature senescence [58]. Studies on ApoE−/−/caspase-1−/− double knockout mice have shown promising evidences that early hyperlipidemia promoted endothelial activation via a Caspase-1-SIRT1 pathway [59]. In this case, researchers found that inhibition of caspase-1 resulted in SIRT1 accumulation in the ApoE−/− mouse aorta and ApoE−/−/caspase-1−/− mice had attenuated early atherosclerosis, decreased aortic expression of proinflammatory cytokines, and reduced aortic monocyte recruitment, as well as decreased endothelial activation [59]. Another upstream regulator of SIRT1 is CDK5, which was proved to increase the phosphorylation of SIRT1 especially at S47 during cellular senescence [60]. In this study, replacing S47 with nonphosphorable alanin (S47A) elevated, while mutation of S47 to phospho-mimicking aspartic acid (S47D) abolished the beneficial effects of SIRT1 such as anti-senescence, growth promotion, and downregulation of LKB1 expression [60]. Interaction between SIRT1 and telomeric repeat-binding factor 2-interacting protein 1 was abolished when S47 was phosphorylated. NF-κB signaling pathway is activated to induce endothelial inflammation and leads to endothelial senescence and atherosclerosis. Downregulation of CDK5 by either knockdown (by siRNA) or inhibition (by roscovitine) reduced percentage of senescent ECs and attenuated inflammatory gene expression. Meanwhile, long-term treatment of ApoE−/− mice with the CDK5 inhibitor, roscovitine, resulted in attenuated atherosclerosis in aortae [60]. As CDK5R1(p35/p25) is the crucial activator mediating the kinase activity of CDK5 [60, 61], further research will be conducted to unveil the underlying mechanism of CDK5-p35/p25-SIRT1 pathway in ECs.

Limited information is available concerning the role of endothelial SIRT1 in vascular remodeling. In eNOS-deficient mice, overexpression of endothelial SIRT1 prevents hypertension and age-related adverse arterial remodeling [37].

Laminar shear stress is an important stimulus for the endothelium-dependent control of vascular tone and of vascular remodeling processes. In cultured ECs, laminar flow increases both the expression and activity of SIRT1, whereas oscillating flow decreases SIRT1 expression [62]. In mouse arteries, the formation of neointima is accompanied by a progressive downregulation of SIRT1 expression [63]. SIRT1 inhibition in ECs increases the expressions of p53 and its downstream target, plasminogen activator inhibitor-1 (PAI-1), which promotes the formation of neointima and vascular remodeling in response to vascular injury [40].

Loss of vascular smooth muscle cell (VSMC) function is an alarming sign of vascular disease. During the aging process, VSMCs undergo increased dysregulation, apoptosis, and senescence [64]. In VSMCs, SIRT1 can act as a modulator of neointima formation (associated with repression of activator protein-1 (AP-1) activity [63]) and protect against DNA damage. Aging-related loss of SIRT1 expression correlates with lower capacity for vascular repair, abolished stress response, and elevated senescence [63].

Decreased expression of SIRT1 in VSMCs exerts its proatherogenic effects by the failure to deacetylate histones in DNA repair, response to oxidant stress and LDL, and therefore leads to VSMC senescence and apoptosis [63, 65]. As to atherosclerotic plaques, SIRT1 activity has been suggested to deacetylate the regulatory factor for X-box (RFX5) and antagonized repression of collagen type I (COL1A2) transcription in VSMCs, consequently stabilizing the plaque and avoiding rupture [66]. Another most recent finding relevant to destabilization of atherosclerotic plaque is that SIRT1 participated in downregulation of platelet-activating factor receptor (PAFR) in VSMCs through β-arrestin 2-mediated internalization and degradation, resulting in the inhibition of PAF-induced matrix metalloproteinase (MMP-2) generation [67]. In addition, inhibition of miR-138 was found to increase SIRT1 expression in VSMCs separated from diabetic (db/db) mice and in SMC lines C-12511 in recent study, which indicated miR-138 as another potential inhibitory target to attenuate the proliferation and migration of VSMCs and cure atherosclerosis [68]. Furthermore, SIRT1 was also found to inhibit angiotensin II-induced VSMC hypertrophy in rat embryonic aortic VSMCs [69]. Later on, SIRT1 demonstrated antihypertensive activity in transgenic mice with selective overexpression of SIRT1 in VSMCs (SV-Tg). Alleviated vascular remodeling in mouse thoracic and renal aortae induced by angiotensin II is observed, along with significantly decreased transforming growth factor-β1 (TGF-β1) expression, ROS generation, vascular inflammation, and collagen formation in the arterial wall of SV-Tg mice [70]. Similar to contribution in ECs, overexpression of miR-34a can upregulate p21 level and inflammation through SIRT1 downregulation and cause senescence-associated secretory phenotype factors induction (including pro-inflammatory molecules such as cytokines, chemokines, proteases, growth factors, soluble receptors, etc.), promoting VSMC senescence and leading to arterial dysfunction [71].

Advertisement

5. Targeting endothelial SIRT1 for the prevention of arterial aging

Slowing down the vascular aging requires early intervention, lifelong treatment, and site-specific approaches. To reduce arterial stiffness, pharmacological agents, including angiotensin-converting enzyme inhibitors, angiotensin II type 1 blockers, aldosterone antagonists, and statins, are currently available and in clinical use [35]. The evident vasoprotective effects of SIRT1 definitely pose great opportunities and challenges for drug discovery targeting endothelial dysfunction. So far, a few natural and synthetic substances have been demonstrated as SIRT1 activators to promote vascular health. The first potent activator of SIRT1 is resveratrol, a small polyphenol discovered in red wine, which could protect ECs against inflammation, apoptosis, and oxidative stress [72]. It was found to dramatically lower the incidence of cardiovascular diseases in spite of high saturated fat diet, which was termed as “French paradox” [73]. Several natural ingredients extracted from various traditional Chinese herbs were also found to activate SIRT1. Tetramethylpyrazine is proved to reverse high-glucose-induced endothelial dysfunction via SIRT1 [74]. Polydatin can attenuate hemorrhagic shock by upregulating SIRT1 [75]. Quercetin is capable of inhibiting oxidized LDL-induced EC damage by SIRT1 activation [76]. Some other natural polyphenols including fisetin and butein can also activate SIRT1 [77]. Vitamin D protects ECs from irradiation-induced senescence and apoptosis by modulating MAPK/SirT1 axis [78]. On the other hand, a series of SIRT1 activators like SRT2183, SRT1460, SRT1720, SRT2379, SRT501, SRT2104, SRT3025, and BMT0-512 have been synthesized and developed as potential drugs to protect against vascular aging [77, 79].

Despite the fact that SIRT1 is as an optimal therapeutical target for cardiovascular diseases, the dosage of upregulation of SIRT1 should be considered seriously and titrated cautiously in clinical practice. It was reported that 2.5- to 7-fold overexpression of SIRT1 prevented heart from oxidative stress via SIRT1/FOXO, while 12.5-fold overexpression of SIRT1 increased apoptosis and hypertrophy and decreased cardiac function, suggesting that only low to moderate doses of SIRT1 can exert beneficial effects [80]. The aforementioned findings call for more careful evaluation of dosage and possible adverse effects in drug development targeting endothelial SIRT1.

In light of all the above studies, there have already been several potential drugs to target the anti-vascular aging effects of endothelial SIRT1. The entry of SIRT1 activators into human trials is exciting but also highlights the necessity to better understand the SIRT1 specificity, clinical effects, and side effects of these promising activators in vivo.

Advertisement

Acknowledgments

This work was financially supported by Hong Kong Health and Medical Research Fund 13142651.

Advertisement

Conflict of interest

No interest of conflict was declared.

References

  1. 1. Benjamin EJ, Blaha MJ, Chiuve SE, Cushman M, Das SR, Deo R, de Ferranti SD, Floyd J, Fornage M, Gillespie C, Isasi CR, Jimenez MC, Jordan LC, Judd SE, Lackland D, Lichtman JH, Lisabeth L, Liu S, Longenecker CT, Mackey RH, Matsushita K, Mozaffarian D, Mussolino ME, Nasir K, Neumar RW, Palaniappan L, Pandey DK, Thiagarajan RR, Reeves MJ, Ritchey M, Rodriguez CJ, Roth GA, Rosamond WD, Sasson C, Towfighi A, Tsao CW, Turner MB, Virani SS, Voeks JH, Willey JZ, Wilkins JT, Wu JH, Alger HM, Wong SS, Muntner P. American Heart Association Statistics, and S. Stroke Statistics. Heart disease and stroke statistics-2017 update: A report from the American Heart Association. Circulation. 2017;135(10):e146-e603
  2. 2. Thijssen DH, Carter SE, Green DJ. Arterial structure and function in vascular ageing: Are you as old as your arteries? The Journal of Physiology. 2016;594(8):2275-2284
  3. 3. Rubio-Ruiz ME, Perez-Torres I, Soto ME, Pastelin G, Guarner-Lans V. Aging in blood vessels. Medicinal agents FOR systemic arterial hypertension in the elderly. Ageing Research Reviews. 2014;18:132-147
  4. 4. Lakatta EG. The reality of aging viewed from the arterial wall. Artery Research. 2013;7(2):73-80
  5. 5. Nilsson PM, Khalili P, Franklin SS. Blood pressure and pulse wave velocity as metrics for evaluating pathologic ageing of the cardiovascular system. Blood Pressure. 2014;23(1):17-30
  6. 6. Eckers A, Haendeler J. Endothelial cells in health and disease. Antioxidants & Redox Signaling. 2015;22(14):1209-1211
  7. 7. Bai B, Wang Y. Methods to investigate the role of SIRT1 in endothelial senescence. Methods in Molecular Biology. 2013;965:327-339
  8. 8. Wang Y, Liang Y, Vanhoutte PM. SIRT1 and AMPK in regulating mammalian senescence: A critical review and a working model. FEBS Letters. 2011;585(7):986-994
  9. 9. Kuilman T, Michaloglou C, Mooi WJ, Peeper DS. The essence of senescence. Genes & Development. 2010;24(22):2463-2479
  10. 10. Takumi T, Yang EH, Mathew V, Rihal CS, Gulati R, Lerman LO, Lerman A. Coronary endothelial dysfunction is associated with a reduction in coronary artery compliance and an increase in wall shear stress. Heart. 2010;96(10):773-778
  11. 11. Curcio S, Garcia-Espinosa V, Arana M, Farro I, Chiesa P, Giachetto G, Zocalo Y, Bia D. Growing-related changes in arterial properties of healthy children, adolescents, and young adults nonexposed to cardiovascular risk factors: Analysis of gender-related differences. International Journal of Hypertension. 2016;2016:4982676
  12. 12. Taddei S, Galetta F, Virdis A, Ghiadoni L, Salvetti G, Franzoni F, Giusti C, Salvetti A. Physical activity prevents age-related impairment in nitric oxide availability in elderly athletes. Circulation. 2000;101(25):2896-2901
  13. 13. Chauhan A, More RS, Mullins PA, Taylor G, Petch MC, Schofield PM. Aging-associated endothelial dysfunction in humans is reversed by L-arginine. Journal of the American College of Cardiology. 1996;28(7):1796-1804
  14. 14. Holowatz LA, Thompson CS, Kenney WL. L-arginine supplementation or arginase inhibition augments reflex cutaneous vasodilatation in aged human skin. The Journal of Physiology. 2006;574(Pt 2):573-581
  15. 15. Pierce GL, Larocca TJ. Reduced vascular tetrahydrobiopterin (BH4) and endothelial function with ageing: Is it time for a chronic BH4 supplementation trial in middle-aged and older adults? The Journal of Physiology. 2008;586(11):2673-2674
  16. 16. Thijssen DH, Rongen GA, van Dijk A, Smits P, Hopman MT. Enhanced endothelin-1-mediated leg vascular tone in healthy older subjects. Journal of Applied Physiology. 2007;103(3):852-857
  17. 17. Montero D, Pierce GL, Stehouwer CD, Padilla J, Thijssen DH. The impact of age on vascular smooth muscle function in humans. Journal of Hypertension. 2015;33(3):445-453; discussion 453
  18. 18. Wang Y, Xu C, Liang Y, Vanhoutte PM. SIRT1 in metabolic syndrome: Where to target matters. Pharmacology & Therapeutics. 2012;136(3):305-318
  19. 19. Feldman JL, Dittenhafer-Reed KE, Denu JM. Sirtuin catalysis and regulation. Journal of Biological Chemistry. 2012;287(51):42419-42427
  20. 20. Cencioni C, Spallotta F, Mai A, Martelli F, Farsetti A, Zeiher AM, Gaetano C. Sirtuin function in aging heart and vessels. Journal of Molecular and Cellular Cardiology. 2015
  21. 21. Alcaín FJ, Villalba JM. Sirtuin inhibitors. Expert Opinion on Therapeutic Patents. 2009;19(3):283-294
  22. 22. Huhtiniemi T, Wittekindt C, Laitinen T, Leppänen J, Salminen A, Poso A, Lahtela-Kakkonen M. Comparative and pharmacophore model for deacetylase SIRT1. Journal of Computer-Aided Molecular Design. 2006;20(9):589-599
  23. 23. Canto C, Auwerx J. Targeting sirtuin 1 to improve metabolism: All you need is NAD(+)? Pharmacological Reviews. 2012;64(1):166-187
  24. 24. Flick F, Luscher B. Regulation of sirtuin function by posttranslational modifications. Frontiers in Pharmacology. 2012;3:29
  25. 25. Sasaki T, Maier B, Koclega KD, Chruszcz M, Gluba W, Stukenberg PT, Minor W, Scrable H. Phosphorylation regulates SIRT1 function. PLoS One. 2008;3(12):e4020
  26. 26. Lee CW, Wong LL, Tse EY, Liu HF, Leong VY, Lee JM, Hardie DG, Ng IO, Ching YP. AMPK promotes p53 acetylation via phosphorylation and inactivation of SIRT1 in liver cancer cells. Cancer Research. 2012;72(17):4394-4404
  27. 27. Nasrin N, Kaushik VK, Fortier E, Wall D, Pearson KJ, de Cabo R, Bordone L. JNK1 phosphorylates SIRT1 and promotes its enzymatic activity. PLoS One. 2009;4(12):e8414
  28. 28. Zschoernig B, Mahlknecht U. Carboxy-terminal phosphorylation of SIRT1 by protein kinase CK2. Biochemical and Biophysical Research Communications. 2009;381(3):372-377
  29. 29. Liu X, Wang D, Zhao Y, Tu B, Zheng Z, Wang L, Wang H, Gu W, Roeder RG, Zhu WG. Methyltransferase Set7/9 regulates p53 activity by interacting with Sirtuin 1 (SIRT1). Proceedings of the National Academy of Sciences of the United States of America. 2011;108(5):1925-1930
  30. 30. Kornberg MD, Sen N, Hara MR, Juluri KR, Nguyen JV, Snowman AM, Law L, Hester LD, Snyder SH. GAPDH mediates nitrosylation of nuclear proteins. Nature Cell Biology. 2010;12(11):1094-1100
  31. 31. Yang Y, Fu W, Chen J, Olashaw N, Zhang X, Nicosia SV, Bhalla K, Bai W. SIRT1 sumoylation regulates its deacetylase activity and cellular response to genotoxic stress. Nature Cell Biology. 2007;9(11):1253-1262
  32. 32. Kim EJ, Kho JH, Kang MR, Um SJ. Active regulator of SIRT1 cooperates with SIRT1 and facilitates suppression of p53 activity. Molecular Cell. 2007;28(2):277-290
  33. 33. Ming GF, Wu K, Hu K, Chen Y, Xiao J. NAMPT regulates senescence, proliferation, and migration of endothelial progenitor cells through the SIRT1 AS lncRNA/miR-22/SIRT1 pathway. Biochemical and Biophysical Research Communications. 2016;478(3):1382-1388
  34. 34. Jackson MD, Schmidt MT, Oppenheimer NJ, Denu JM. Mechanism of nicotinamide inhibition and transglycosidation by Sir2 histone/protein deacetylases. The Journal of Biological Chemistry. 2003;278(51):50985-50598
  35. 35. Guo Y, Xu A, Wang Y. SIRT1 in endothelial cells as a novel target for the prevention of early vascular aging. Journal of Cardiovascular Pharmacology. 2016;67(6):465-473
  36. 36. Wang YQ, Cao Q, Wang F, Huang LY, Sang TT, Liu F, Chen SY. SIRT1 protects against oxidative stress-induced endothelial progenitor cells apoptosis by inhibiting FOXO3a via FOXO3a ubiquitination and degradation. Journal of Cellular Physiology. 2015;230(9):2098-2107
  37. 37. Bai B, Man AW, Yang K, Guo Y, Xu C, Tse HF, Han W, Bloksgaard M, De Mey JG, Vanhoutte PM, Xu A, Wang Y. Endothelial SIRT1 prevents adverse arterial remodeling by facilitating HERC2-mediated degradation of acetylated LKB1. Oncotarget. 2016;7(26):39065-39081
  38. 38. Potente M, Ghaeni L, Baldessari D, Mostoslavsky R, Rossig L, Dequiedt F, Haendeler J, Mione M, Dejana E, Alt FW. SIRT1 controls endothelial angiogenic functions during vascular growth. Genes & Development. 2007;21(20):2644-2658
  39. 39. Yana I, Sagara H, Takaki S, Takatsu K, Nakamura K, Nakao K, Katsuki M, Taniguchi SI, Aoki T, Sato H. Crosstalk between neovessels and mural cells directs the site-specific expression of MT1-MMP to endothelial tip cells. Journal of Cell Science. 2007;120(9):1607-1614
  40. 40. Ota H, Akishita M, Eto M, Iijima K, Kaneki M, Ouchi Y. Sirt1 modulates premature senescence-like phenotype in human endothelial cells. Journal of Molecular and Cellular Cardiology. 2007;43(5):571-579
  41. 41. Zu Y, Liu L, Lee MY, Xu C, Liang Y, Man RY, Vanhoutte PM, Wang Y. SIRT1 promotes proliferation and prevents senescence through targeting LKB1 in primary porcine aortic endothelial cells. Circulation Research. 2010;106(8):1384-1393
  42. 42. Bai B, Vanhoutte PM, Wang Y. Loss-of-SIRT1 function during vascular ageing: Hyperphosphorylation mediated by cyclin-dependent kinase 5. Trends in Cardiovascular Medicine. 2014;24(2):81-84
  43. 43. Luo J, Nikolaev AY, Imai SI, Chen D, Su F, Shiloh A, Guarente L, Gu W. Negative control of p53 by Sir2α promotes cell survival under stress. Cell. 2001;107(2):137-148
  44. 44. Wallerath T, Li H, Gödtel-Ambrust U, Schwarz PM, Förstermann U. A blend of polyphenolic compounds explains the stimulatory effect of red wine on human endothelial NO synthase. Nitric Oxide. 2005;12(2):97-104
  45. 45. 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. Proceedings of the National Academy of Sciences of the United States of America. 2007;104(37):14855-14860
  46. 46. Cheung TM, Yan JB, Fu JJ, Huang J, Yuan F, Truskey GA. Endothelial cell senescence increases traction forces due to age-associated changes in the Glycocalyx and SIRT1. Cellular and Molecular Bioengineering. 2015;8(1):63-75
  47. 47. Guarani V, Deflorian G, Franco CA, Kruger M, Phng LK, Bentley K, Toussaint L, Dequiedt F, Mostoslavsky R, Schmidt MH, Zimmermann B, Brandes RP, Mione M, Westphal CH, Braun T, Zeiher AM, Gerhardt H, Dimmeler S, Potente M. Acetylation-dependent regulation of endothelial notch signalling by the SIRT1 deacetylase. Nature. 2011;473(7346):234-238
  48. 48. Wang P, Du H, Zhou CC, Song J, Liu X, Cao X, Mehta JL, Shi Y, Su DF, Miao CY. Intracellular NAMPT-NAD+-SIRT1 cascade improves post-ischaemic vascular repair by modulating notch signalling in endothelial progenitors. Cardiovascular Research. 2014;104(3):477-488
  49. 49. Chen HZ, Wan YZ, Liu DP. Cross-talk between SIRT1 and p66Shc in vascular diseases. Trends in Cardiovascular Medicine. 2013;23(7):237-241
  50. 50. Breitenstein A, Stein S, Holy EW, Camici GG, Lohmann C, Akhmedov A, Spescha R, Elliott PJ, Westphal CH, Matter CM. Sirt1 inhibition promotes in vivo arterial thrombosis and tissue factor expression in stimulated cells. Cardiovascular Research. 2010;89(2):464-472
  51. 51. Xu Q, Seeger FH, Castillo J, Iekushi K, Boon RA, Farcas R, Manavski Y, Li YG, Assmus B, Zeiher AM. Micro-RNA-34a contributes to the impaired function of bone marrow-derived mononuclear cells from patients with cardiovascular disease. Journal of the American College of Cardiology. 2012;59(23):2107-2117
  52. 52. Ming GF, Tang YJ, Hu K, Chen Y, Huang WH, Xiao J. Visfatin attenuates the ox-LDL-induced senescence of endothelial progenitor cells by upregulating SIRT1 expression through the PI3K/Akt/ERK pathway. International Journal of Molecular Medicine. 2016;38(2):643-649
  53. 53. Stein S, Matter CM. Protective roles of SIRT1 in atherosclerosis. Cell Cycle. 2011;10(4):640-647
  54. 54. Stein S, Lohmann C, Schäfer N, Hofmann J, Rohrer L, Besler C, Rothgiesser KM, Becher B, Hottiger MO, Borén J. SIRT1 decreases Lox-1-mediated foam cell formation in atherogenesis. European Heart Journal. 2010;31(18):2301-2309
  55. 55. Schug TT, Xu Q, Gao H, Peres-da-Silva A, Draper DW, Fessler MB, Purushotham A, Li X. Myeloid deletion of SIRT1 induces inflammatory signaling in response to environmental stress. Molecular and Cellular Biology. 2010;30(19):4712-4721
  56. 56. Miranda MX, Van Tits LJ, Lohmann C, Arsiwala T, Winnik S, Tailleux A, Stein S, Gomes AP, Suri V, Ellis JL. The Sirt1 activator SRT3025 provides atheroprotection in Apoe−/− mice by reducing hepatic Pcsk9 secretion and enhancing Ldlr expression. European Heart Journal. 2015;36(1):51-59
  57. 57. Kim HJ, Park KG, Yoo EK, Kim YH, Kim YN, Kim HS, Kim HT, Park JY, Lee KU, Jang WG. Effects of PGC-1α on TNF-α-induced MCP-1 and VCAM-1 expression and NF-κB activation in human aortic smooth muscle and endothelial cells. Antioxidants & Redox Signaling. 2007;9(3):301-307
  58. 58. Chen J, Xavier S, Moskowitz-Kassai E, Chen R, Lu CY, Sanduski K, Spes A, Turk B, Goligorsky MS. Cathepsin cleavage of sirtuin 1 in endothelial progenitor cells mediates stress-induced premature senescence. The American Journal of Pathology. 2012;180(3):973-983
  59. 59. Yin Y, Li X, Sha X, Xi H, Li YF, Shao Y, Mai J, Virtue A, Lopez-Pastrana J, Meng S. Early hyperlipidemia promotes endothelial activation via a Caspase-1-Sirtuin 1 pathway. Arteriosclerosis, Thrombosis, and Vascular Biology. 2015;35(4):804-816
  60. 60. Bai B, Liang Y, Xu C, Lee MY, Xu A, Wu D, Vanhoutte PM, Wang Y. Cyclin-dependent kinase 5–mediated hyperphosphorylation of Sirtuin-1 contributes to the development of endothelial senescence and atherosclerosis. Circulation. 2012;126(6):729-740
  61. 61. Ko J, Humbert S, Bronson RT, Takahashi S, Kulkarni AB, Li E, Tsai LH. p35 and p39 are essential for cyclin-dependent kinase 5 function during neurodevelopment. The Journal of Neuroscience. 2001;21(17):6758-6771
  62. 62. Chen Z, Peng IC, Cui X, Li YS, Chien S, Shyy JY. Shear stress, SIRT1, and vascular homeostasis. Proceedings of the National Academy of Sciences of the United States of America. 2010;107(22):10268-10273
  63. 63. Li L, Zhang HN, Chen HZ, Gao P, Zhu LH, Li HL, Lv X, Zhang QJ, Zhang R, Wang Z. SIRT1 acts as a modulator of neointima formation following vascular injury in mice. Circulation Research. 2011;108(10):1180-1189
  64. 64. Thompson AM, Wagner R, Rzucidlo EM. Age-related loss of SirT1 expression results in dysregulated human vascular smooth muscle cell function. American Journal of Physiology-Heart and Circulatory Physiology. 2014;307(4):H533-H541
  65. 65. Gorenne I, Kumar S, Gray K, Figg N, Yu H, Mercer J, Bennett M. Vascular smooth muscle cell sirtuin 1 protects against DNA damage and inhibits atherosclerosis. Circulation. 2013;127(3):386-396
  66. 66. Xia J, Wu X, Yang Y, Zhao Y, Fang M, Xie W, Wang H, Xu Y. SIRT1 deacetylates RFX5 and antagonizes repression of collagen type I (COL1A2) transcription in smooth muscle cells. Biochemical and Biophysical Research Communications. 2012;428(2):264-270
  67. 67. Kim YH, Bae JU, Lee SJ, Park SY, Kim CD. SIRT1 attenuates PAF-induced MMP-2 production via down-regulation of PAF receptor expression in vascular smooth muscle cells. Vascular Pharmacology. 2015;72:35-42
  68. 68. Xu J, Li L, Yun HF, Han YS. MiR-138 promotes smooth muscle cells proliferation and migration in db/db mice through down-regulation of SIRT1. Biochemical and Biophysical Research Communications. 2015;463(4):1159-1164
  69. 69. Li L, Gao P, Zhang H, Chen H, Zheng W, Lv X, Xu T, Wei Y, Liu D, Liang C. SIRT1 inhibits angiotensin II-induced vascular smooth muscle cell hypertrophy. Acta Biochimica Et Biophysica Sinica. 2010;43(2):103-109
  70. 70. Gao P, Xu TT, Lu J, Li L, Xu J, Hao DL, Chen HZ, Liu DP. Overexpression of SIRT1 in vascular smooth muscle cells attenuates angiotensin II-induced vascular remodeling and hypertension in mice. Journal of Molecular Medicine. 2014;92(4):347-357
  71. 71. Badi I, Burba I, Ruggeri C, Zeni F, Bertolotti M, Scopece A, Pompilio G, Raucci A. MicroRNA-34a induces vascular smooth muscle cells senescence by SIRT1 downregulation and promotes the expression of age-associated pro-inflammatory secretory factors. The Journals of Gerontology. Series A, Biological Sciences and Medical Sciences. 2014;70(11):1304-1311
  72. 72. Kopp P. Resveratrol, a phytoestrogen found in red wine. A possible explanation for the conundrum of the ‘French paradox’? European Journal of Endocrinology. 1998;138(6):619-620
  73. 73. Catalgol B, Batirel S, Taga Y, Ozer NK. Resveratrol: French paradox revisited. Frontiers in Pharmacology. 2012;3(141):1-18
  74. 74. Xu Q, Xia P, Li X, Wang W, Liu Z, Gao X. Tetramethylpyrazine ameliorates high glucose-induced endothelial dysfunction by increasing mitochondrial biogenesis. PLoS One. 2014;9(2):e88243
  75. 75. Zhang W, Huang Q, Zeng Z, Wu J, Zhang Y, Chen Z. Sirt1 inhibits oxidative stress in vascular endothelial cells. Oxidative Medicine and Cellular Longevity. 2017;2017:7543973
  76. 76. Hung CH, Chan SH, Chu PM, Tsai KL. Quercetin is a potent anti-atherosclerotic compound by activation of SIRT1 signaling under oxLDL stimulation. Molecular Nutrition & Food Research. 2015;59(10):1905-1917
  77. 77. Milne JC, Lambert PD, Schenk S, Carney DP, Smith JJ, Gagne DJ, Jin L, Boss O, Perni RB, Vu CB, Bemis JE, Xie R, Disch JS, Ng PY, Nunes JJ, Lynch AV, Yang H, Galonek H, Israelian K, Choy W, Iffland A, Lavu S, Medvedik O, Sinclair DA, Olefsky JM, Jirousek MR, Elliott PJ, Westphal CH. Small molecule activators of SIRT1 as therapeutics for the treatment of type 2 diabetes. Nature. 2007;450(7170):712-716
  78. 78. Marampon F, Gravina GL, Festuccia C, Popov VM, Colapietro EA, Sanita P, Musio D, De Felice F, Lenzi A, Jannini EA, Di Cesare E, Tombolini V. Vitamin D protects endothelial cells from irradiation-induced senescence and apoptosis by modulating MAPK/SirT1 axis. Journal of Endocrinological Investigation. 2016;39(4):411-422
  79. 79. Villalba JM, Alcaín FJ. Sirtuin activators and inhibitors. BioFactors. 2012;38(5):349-359
  80. 80. Alcendor RR, Gao S, Zhai P, Zablocki D, Holle E, Yu X, Tian B, Wagner T, Vatner SF, Sadoshima J. Sirt1 regulates aging and resistance to oxidative stress in the heart. Circulation Research. 2007;100(10):1512-1121

Written By

Yumeng Guo and Yu Wang

Submitted: 08 May 2017 Reviewed: 08 December 2017 Published: 22 February 2018