1. Chien KR, Karsenty G. Longevity and lineages: toward the integrative biology of degenerative diseases in heart, muscle, and bone. Cell. 2005; 120:533–544.
4. Triposkiadis F, Xanthopoulos A, Butler J. Cardiovascular aging and heart failure: JACC review topic of the week. J Am Coll Cardiol. 2019; 74:804–813.
5. Yoshii M, Horinaka S, Yabe A, Mori Y, Nakano S, Ishimura K, et al. The relationship between coronary artery disease and parameters of arteriosclerosis in patients with ischemic heart disease. J Hypertens. 2006; 24:214–215.
7. Minamino T, Miyauchi H, Yoshida T, Ishida Y, Yoshida H, Komuro I. Endothelial cell senescence in human atherosclerosis: role of telomere in endothelial dysfunction. Circulation. 2002; 105:1541–1544.
8. Shimizu I, Yoshida Y, Suda M, Minamino T. DNA damage response and metabolic disease. Cell Metab. 2014; 20:967–977.
11. Sano M, Minamino T, Toko H, Miyauchi H, Orimo M, Qin Y, et al. p53-induced inhibition of Hif-1 causes cardiac dysfunction during pressure overload. Nature. 2007; 446:444–448.
12. Shimizu I, Yoshida Y, Katsuno T, Tateno K, Okada S, Moriya J, et al. p53-induced adipose tissue inflammation is critically involved in the development of insulin resistance in heart failure. Cell Metab. 2012; 15:787.

13. Shimizu I, Yoshida Y, Moriya J, Nojima A, Uemura A, Kobayashi Y, et al. Semaphorin3E-induced inflammation contributes to insulin resistance in dietary obesity. Cell Metab. 2013; 18:491–504.
15. Yokoyama M, Shimizu I, Nagasawa A, Yoshida Y, Katsuumi G, Wakasugi T, et al. p53 plays a crucial role in endothelial dysfunction associated with hyperglycemia and ischemia. J Mol Cell Cardiol. 2019; 129:105–117.
16. Yoshida Y, Shimizu I, Katsuumi G, Jiao S, Suda M, Hayashi Y, et al. p53-induced inflammation exacerbates cardiac dysfunction during pressure overload. J Mol Cell Cardiol. 2015; 85:183–198.
17. Hayflick L, Moorhead PS. The serial cultivation of human diploid cell strains. Exp Cell Res. 1961; 25:585–621.
18. Shimizu I, Minamino T. Cellular senescence in cardiac diseases. J Cardiol. 2019; 74:313–319.
19. Minamino T, Orimo M, Shimizu I, Kunieda T, Yokoyama M, Ito T, et al. A crucial role for adipose tissue p53 in the regulation of insulin resistance. Nat Med. 2009; 15:1082–1087.
21. Anderson R, Lagnado A, Maggiorani D, Walaszczyk A, Dookun E, Chapman J, et al. Length-independent telomere damage drives post-mitotic cardiomyocyte senescence. EMBO J. 2019; 38:e100492.

24. Chang J, Wang Y, Shao L, Laberge RM, Demaria M, Campisi J, et al. Clearance of senescent cells by ABT263 rejuvenates aged hematopoietic stem cells in mice. Nat Med. 2016; 22:78–83.
25. Cafueri G, Parodi F, Pistorio A, Bertolotto M, Ventura F, Gambini C, et al. Endothelial and smooth muscle cells from abdominal aortic aneurysm have increased oxidative stress and telomere attrition. PLoS One. 2012; 7:e35312.

26. Marchand A, Atassi F, Gaaya A, Leprince P, Le Feuvre C, Soubrier F, et al. The Wnt/beta-catenin pathway is activated during advanced arterial aging in humans. Aging Cell. 2011; 10:220–232.
27. Morgan RG, Ives SJ, Lesniewski LA, Cawthon RM, Andtbacka RH, Noyes RD, et al. Age-related telomere uncapping is associated with cellular senescence and inflammation independent of telomere shortening in human arteries. Am J Physiol Heart Circ Physiol. 2013; 305:H251–H258.

28. Lin JR, Shen WL, Yan C, Gao PJ. Downregulation of dynamin-related protein 1 contributes to impaired autophagic flux and angiogenic function in senescent endothelial cells. Arterioscler Thromb Vasc Biol. 2015; 35:1413–1422.
29. Melk A, Schmidt BM, Takeuchi O, Sawitzki B, Rayner DC, Halloran PF. Expression of p16INK4a and other cell cycle regulator and senescence associated genes in aging human kidney. Kidney Int. 2004; 65:510–520.
30. Rajapakse AG, Yepuri G, Carvas JM, Stein S, Matter CM, Scerri I, et al. Hyperactive S6K1 mediates oxidative stress and endothelial dysfunction in aging: inhibition by resveratrol. PLoS One. 2011; 6:e19237.

32. Telomeres Mendelian Randomization Collaboration. Haycock PC, Burgess S, Nounu A, Zheng J, Okoli GN, et al. Association between telomere length and risk of cancer and non-neoplastic diseases: a mendelian randomization study. JAMA Oncol. 2017; 3:636–651.
33. Riches K, Clark E, Helliwell RJ, Angelini TG, Hemmings KE, Bailey MA, et al. Progressive development of aberrant smooth muscle cell phenotype in abdominal aortic aneurysm disease. J Vasc Res. 2018; 55:35–46.
34. Imai SI, Guarente L. It takes two to tango: NAD
+ and sirtuins in aging/longevity control. NPJ Aging Mech Dis. 2016; 2:16017.
36. Bordone L, Guarente L. Calorie restriction, SIRT1 and metabolism: understanding longevity. Nat Rev Mol Cell Biol. 2005; 6:298–305.
37. Guarente L, Picard F. Calorie restriction--the SIR2 connection. Cell. 2005; 120:473–482.
38. Liu Y, Wang TT, Zhang R, Fu WY, Wang X, Wang F, et al. Calorie restriction protects against experimental abdominal aortic aneurysms in mice. J Exp Med. 2016; 213:2473–2488.

39. Gao P, Zhang H, Zhang Q, Fang X, Wu H, Wang M, et al. Caloric restriction exacerbates angiotensin II-induced abdominal aortic aneurysm in the absence of p53. Hypertension. 2019; 73:547–560.
40. Zhang WM, Liu Y, Li TT, Piao CM, Liu O, Liu JL, et al. Sustained activation of ADP/P2ry12 signaling induces SMC senescence contributing to thoracic aortic aneurysm/dissection. J Mol Cell Cardiol. 2016; 99:76–86.
41. Satoh M, Ishikawa Y, Takahashi Y, Itoh T, Minami Y, Nakamura M. Association between oxidative DNA damage and telomere shortening in circulating endothelial progenitor cells obtained from metabolic syndrome patients with coronary artery disease. Atherosclerosis. 2008; 198:347–353.
42. Hammadah M, Al Mheid I, Wilmot K, Ramadan R, Abdelhadi N, Alkhoder A, et al. Telomere shortening, regenerative capacity, and cardiovascular outcomes. Circ Res. 2017; 120:1130–1138.
44. Haycock PC, Heydon EE, Kaptoge S, Butterworth AS, Thompson A, Willeit P. Leucocyte telomere length and risk of cardiovascular disease: systematic review and meta-analysis. BMJ. 2014; 349:g4227.

47. Nasu T, Satoh M, Takahashi Y, Takahashi K, Hitomi S, Morino Y, et al. Expression of miR-23a induces leukocyte telomere shortening and is associated with poor clinical outcomes in patients with coronary artery disease. Eur Heart J. 2017; 38:ehx501.P838.

51. Latorre E, Pilling LC, Lee BP, Bandinelli S, Melzer D, Ferrucci L, et al. The
VEGFA156b isoform is dysregulated in senescent endothelial cells and may be associated with prevalent and incident coronary heart disease. Clin Sci (Lond). 2018; 132:313–325.
52. Guirguis-Blake JM, Evans CV, Redmond N, Lin JS. Screening for peripheral artery disease using the ankle-brachial index: updated evidence report and systematic review for the US preventive services task force. JAMA. 2018; 320:184–196.
53. Oberdier MT, Morrell CH, Lakatta EG, Ferrucci L, AlGhatrif M. Subclinical longitudinal change in ankle-brachial index with aging in a community-dwelling population is associated with central arterial stiffening. J Am Heart Assoc. 2019; 8:e011650.

54. Olinic DM, Spinu M, Olinic M, Homorodean C, Tataru DA, Liew A, et al. Epidemiology of peripheral artery disease in Europe: VAS educational paper. Int Angiol. 2018; 37:327–334.
56. Paradis S, Charles AL, Georg I, Goupilleau F, Meyer A, Kindo M, et al. Aging exacerbates ischemia-reperfusion-induced mitochondrial respiration impairment in skeletal muscle. Antioxidants (Basel). 2019; 8:E168.

57. Kokubun T, Saitoh SI, Miura S, Ishida T, Takeishi Y. Telomerase plays a pivotal role in collateral growth under ischemia by suppressing age-induced oxidative stress, expression of p53, and pro-apoptotic proteins. Int Heart J. 2019; 60:736–745.
58. Sosińska-Zawierucha P, Maćkowiak B, Staniszewski R, Sumińska-Jasińska K, Maj M, Krasiński Z, et al. Sulodexide slows down the senescence of aortic endothelial cells exposed to serum from patients with peripheral artery diseases. Cell Physiol Biochem. 2018; 45:2225–2232.
59. Hansson GK, Libby P. The immune response in atherosclerosis: a double-edged sword. Nat Rev Immunol. 2006; 6:508–519.
60. Libby P, Ridker PM, Hansson GK. Progress and challenges in translating the biology of atherosclerosis. Nature. 2011; 473:317–325.
61. Matthews C, Gorenne I, Scott S, Figg N, Kirkpatrick P, Ritchie A, et al. Vascular smooth muscle cells undergo telomere-based senescence in human atherosclerosis: effects of telomerase and oxidative stress. Circ Res. 2006; 99:156–164.

62. Faggiotto A, Ross R, Harker L. Studies of hypercholesterolemia in the nonhuman primate. I. Changes that lead to fatty streak formation. Arteriosclerosis. 1984; 4:323–340.
63. Wang YC, Lee AS, Lu LS, Ke LY, Chen WY, Dong JW, et al. Human electronegative LDL induces mitochondrial dysfunction and premature senescence of vascular cells in vivo
. Aging Cell. 2018; 17:e12792.
64. de Lange T. Protection of mammalian telomeres. Oncogene. 2002; 21:532–540.
65. Wang J, Uryga AK, Reinhold J, Figg N, Baker L, Finigan A, et al. Vascular smooth muscle cell senescence promotes atherosclerosis and features of plaque vulnerability. Circulation. 2015; 132:1909–1919.
66. Reaux A, Fournie-Zaluski MC, Llorens-Cortes C. Angiotensin III: a central regulator of vasopressin release and blood pressure. Trends Endocrinol Metab. 2001; 12:157–162.
67. Kunieda T, Minamino T, Nishi J, Tateno K, Oyama T, Katsuno T, et al. Angiotensin II induces premature senescence of vascular smooth muscle cells and accelerates the development of atherosclerosis via a p21-dependent pathway. Circulation. 2006; 114:953–960.
68. Miao SB, Xie XL, Yin YJ, Zhao LL, Zhang F, Shu YN, et al. Accumulation of smooth muscle 22α protein accelerates senescence of vascular smooth muscle cells via stabilization of p53
in vitro and
in vivo
. Arterioscler Thromb Vasc Biol. 2017; 37:1849–1859.
69. Gardner SE, Humphry M, Bennett MR, Clarke MCH. Senescent vascular smooth muscle cells drive inflammation through an interleukin-1α-dependent senescence-associated secretory phenotype. Atherosclerosis. 2016; 244:E5.

71. Yoshino J, Baur JA, Imai SI. NAD+ intermediates: the biology and therapeutic potential of NMN and NR. Cell Metab. 2018; 27:513–528.
72. Watson A, Nong Z, Yin H, O'Neil C, Fox S, Balint B, et al. Nicotinamide phosphoribosyltransferase in smooth muscle cells maintains genome integrity, resists aortic medial degeneration, and is suppressed in human thoracic aortic aneurysm disease. Circ Res. 2017; 120:1889–1902.
74. Torella D, Leosco D, Indolfi C, Curcio A, Coppola C, Ellison GM, et al. Aging exacerbates negative remodeling and impairs endothelial regeneration after balloon injury. Am J Physiol Heart Circ Physiol. 2004; 287:H2850–H2860.

75. Yokoyama M, Okada S, Nakagomi A, Moriya J, Shimizu I, Nojima A, et al. Inhibition of endothelial p53 improves metabolic abnormalities related to dietary obesity. Cell Reports. 2014; 7:1691–1703.
76. Warboys CM, de Luca A, Amini N, Luong L, Duckles H, Hsiao S, et al. Disturbed flow promotes endothelial senescence via a p53-dependent pathway. Arterioscler Thromb Vasc Biol. 2014; 34:985–995.
77. Steinberg D. Clinical trials of antioxidants in atherosclerosis: are we doing the right thing? Lancet. 1995; 346:36–38.
78. Hofmann P, Sommer J, Theodorou K, Kirchhof L, Fischer A, Li Y, et al. Long non-coding RNA H19 regulates endothelial cell aging via inhibition of STAT3 signalling. Cardiovasc Res. 2019; 115:230–242.
79. Furuuchi R, Shimizu I, Yoshida Y, Hayashi Y, Ikegami R, Suda M, et al. Boysenberry polyphenol inhibits endothelial dysfunction and improves vascular health. PLoS One. 2018; 13:e0202051.

80. Tsuboi T, Maeda M, Hayashi T. Administration of L-arginine plus L-citrulline or L-citrulline alone successfully retarded endothelial senescence. PLoS One. 2018; 13:e0192252.

81. Sakai C, Ishida M, Ohba H, Yamashita H, Uchida H, Yoshizumi M, et al. Fish oil omega-3 polyunsaturated fatty acids attenuate oxidative stress-induced DNA damage in vascular endothelial cells. PLoS One. 2017; 12:e0187934.

82. Lesniewski LA, Seals DR, Walker AE, Henson GD, Blimline MW, Trott DW, et al. Dietary rapamycin supplementation reverses age-related vascular dysfunction and oxidative stress, while modulating nutrient-sensing, cell cycle, and senescence pathways. Aging Cell. 2017; 16:17–26.
83. Sjøberg KA, Frøsig C, Kjøbsted R, Sylow L, Kleinert M, Betik AC, et al. Exercise increases human skeletal muscle insulin sensitivity via coordinated increases in microvascular perfusion and molecular signaling. Diabetes. 2017; 66:1501–1510.
84. Ji LL. Redox signaling in skeletal muscle: role of aging and exercise. Adv Physiol Educ. 2015; 39:352–359.
86. Reid MB, Durham WJ. Generation of reactive oxygen and nitrogen species in contracting skeletal muscle: potential impact on aging. Ann N Y Acad Sci. 2002; 959:108–116.
88. Rossman MJ, Kaplon RE, Hill SD, McNamara MN, Santos-Parker JR, Pierce GL, et al. Endothelial cell senescence with aging in healthy humans: prevention by habitual exercise and relation to vascular endothelial function. Am J Physiol Heart Circ Physiol. 2017; 313:H890–H895.

89. Calvert PA, Liew TV, Gorenne I, Clarke M, Costopoulos C, Obaid DR, et al. Leukocyte telomere length is associated with high-risk plaques on virtual histology intravascular ultrasound and increased proinflammatory activity. Arterioscler Thromb Vasc Biol. 2011; 31:2157–2164.
90. Cawthon RM, Smith KR, O'Brien E, Sivatchenko A, Kerber RA. Association between telomere length in blood and mortality in people aged 60 years or older. Lancet. 2003; 361:393–395.
95. Lewis-McDougall FC, Ruchaya PJ, Domenjo-Vila E, Shin Teoh T, Prata L, Cottle BJ, et al. Aged-senescent cells contribute to impaired heart regeneration. Aging Cell. 2019; 18:e12931.

96. Walaszczyk A, Dookun E, Redgrave R, Tual-Chalot S, Victorelli S, Spyridopoulos I, et al. Pharmacological clearance of senescent cells improves survival and recovery in aged mice following acute myocardial infarction. Aging Cell. 2019; 18:e12945.

97. Hartmann JT, Haap M, Kopp HG, Lipp HP. Tyrosine kinase inhibitors - a review on pharmacology, metabolism and side effects. Curr Drug Metab. 2009; 10:470–481.
98. Wilson WH, O'Connor OA, Czuczman MS, LaCasce AS, Gerecitano JF, Leonard JP, et al. Navitoclax, a targeted high-affinity inhibitor of BCL-2, in lymphoid malignancies: a phase 1 dose-escalation study of safety, pharmacokinetics, pharmacodynamics, and antitumour activity. Lancet Oncol. 2010; 11:1149–1159.