Aging of the Vasculature

  • Chapter
  • First Online:
Cardiovascular Disease in the Elderly

Part of the book series: Contemporary Cardiology ((CONCARD))

  • 722 Accesses

Abstract

Vascular disease and its consequences remain the most common causes of mortality and significant lifelong disability in developed, and most develo**, countries. Age is the most potent risk factor for the responsible vascular changes and although these are in part dependent on the “quality of the arterial tissue” we inherit, their trajectory can be impacted by the environment our vessels are exposed to. This chapter will review the molecular, cellular, structural, and functional changes associated with aging, the impact of these changes on cardiovascular outcomes, and lifestyle and pharmacologic interventions designed to modify them.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Springer+ Basic
EUR 32.99 /Month
  • Get 10 units per month
  • Download Article/Chapter or Ebook
  • 1 Unit = 1 Article or 1 Chapter
  • Cancel anytime
Subscribe now
Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 84.99
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 109.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free ship** worldwide - see info
Hardcover Book
USD 159.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free ship** worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

Similar content being viewed by others

References

  1. Martin CF. Osler as clinician and teacher. Can Med Assoc J. 1920;10(Spec Issue):82–6.

    CAS  Google Scholar 

  2. Gimbrone MA Jr, Garcia-Cardena G. Endothelial cell dysfunction and the pathobiology of atherosclerosis. Circ Res. 2016;118(4):620–36.

    Article  CAS  Google Scholar 

  3. Hays AG, et al. Coronary vasomotor responses to isometric handgrip exercise are primarily mediated by nitric oxide: a noninvasive MRI test of coronary endothelial function. Am J Physiol Heart Circ Physiol. 2015;308(11):H1343–50.

    Article  CAS  Google Scholar 

  4. Jones SP, Bolli R. The ubiquitous role of nitric oxide in cardioprotection. J Mol Cell Cardiol. 2006;40(1):16–23.

    Article  CAS  Google Scholar 

  5. Virani SS, et al. Heart disease and stroke statistics-2020 update: a report from the American Heart Association. Circulation. 2020;141(9):e139–596.

    Article  Google Scholar 

  6. Vespa J, Medina L, Armstrong DM. Demographic turning points for the United States: population projections for 2020 to 2060. Current Population Reports. Washington, DC: Census Bureau; 2020. p. P25–1144.

    Google Scholar 

  7. Rizvi F, et al. Effects of aging on cardiac oxidative stress and transcriptional changes in pathways of reactive oxygen species generation and clearance. J Am Heart Assoc. 2021;10(16):e019948.

    Article  CAS  Google Scholar 

  8. Finkel T, Holbrook NJ. Oxidants, oxidative stress and the biology of ageing. Nature. 2000;408(6809):239–47.

    Article  CAS  Google Scholar 

  9. Csiszar A, et al. Aging-induced phenotypic changes and oxidative stress impair coronary arteriolar function. Circ Res. 2002;90(11):1159–66.

    Article  CAS  Google Scholar 

  10. Toth P, et al. Resveratrol treatment rescues neurovascular coupling in aged mice: role of improved cerebromicrovascular endothelial function and downregulation of NADPH oxidase. Am J Physiol Heart Circ Physiol. 2014;306(3):H299–308.

    Article  CAS  Google Scholar 

  11. Yang YM, et al. eNOS uncoupling and endothelial dysfunction in aged vessels. Am J Physiol Heart Circ Physiol. 2009;297(5):H1829–36.

    Article  CAS  Google Scholar 

  12. Donato AJ, et al. Direct evidence of endothelial oxidative stress with aging in humans: relation to impaired endothelium-dependent dilation and upregulation of nuclear factor-kappaB. Circ Res. 2007;100(11):1659–66.

    Article  CAS  Google Scholar 

  13. Csiszar A, et al. Inflammation and endothelial dysfunction during aging: role of NF-kappaB. J Appl Physiol (1985). 2008;105(4):1333–41.

    Article  CAS  Google Scholar 

  14. Wang M, et al. Matrix metalloproteinases promote arterial remodeling in aging, hypertension, and atherosclerosis. Hypertension. 2015;65(4):698–703.

    Article  CAS  Google Scholar 

  15. Ungvari Z, et al. Cerebral microhemorrhages: mechanisms, consequences, and prevention. Am J Physiol Heart Circ Physiol. 2017;312(6):H1128–43.

    Article  Google Scholar 

  16. Fleenor BS, et al. Superoxide signaling in perivascular adipose tissue promotes age-related artery stiffness. Aging Cell. 2014;13(3):576–8.

    Article  CAS  Google Scholar 

  17. Ungvari Z, Sonntag WE, Csiszar A. Mitochondria and aging in the vascular system. J Mol Med (Berl). 2010;88(10):1021–7.

    Article  CAS  Google Scholar 

  18. Ungvari Z, et al. Increased mitochondrial H2O2 production promotes endothelial NF-kappaB activation in aged rat arteries. Am J Physiol Heart Circ Physiol. 2007;293(1):H37–47.

    Article  CAS  Google Scholar 

  19. Ungvari Z, et al. Age-associated vascular oxidative stress, Nrf2 dysfunction, and NF-{kappa}B activation in the nonhuman primate Macaca mulatta. J Gerontol A Biol Sci Med Sci. 2011;66(8):866–75.

    Article  Google Scholar 

  20. Springo Z, et al. Aging exacerbates pressure-induced mitochondrial oxidative stress in mouse cerebral arteries. J Gerontol A Biol Sci Med Sci. 2015;70(11):1355–9.

    Article  CAS  Google Scholar 

  21. Gioscia-Ryan RA, et al. Mitochondria-targeted antioxidant (MitoQ) ameliorates age-related arterial endothelial dysfunction in mice. J Physiol. 2014;592(12):2549–61.

    Article  CAS  Google Scholar 

  22. Pearson KJ, et al. Resveratrol delays age-related deterioration and mimics transcriptional aspects of dietary restriction without extending life span. Cell Metab. 2008;8(2):157–68.

    Article  CAS  Google Scholar 

  23. Nissanka N, Moraes CT. Mitochondrial DNA damage and reactive oxygen species in neurodegenerative disease. FEBS Lett. 2018;592(5):728–42.

    Article  CAS  Google Scholar 

  24. Bratic A, Larsson NG. The role of mitochondria in aging. J Clin Invest. 2013;123(3):951–7.

    Article  CAS  Google Scholar 

  25. Yu E, et al. Mitochondrial DNA damage can promote atherosclerosis independently of reactive oxygen species through effects on smooth muscle cells and monocytes and correlates with higher-risk plaques in humans. Circulation. 2013;128(7):702–12.

    Article  CAS  Google Scholar 

  26. Sack MN, Finkel T. Mitochondrial metabolism, sirtuins, and aging. Cold Spring Harb Perspect Biol. 2012;4(12):a013102.

    Article  Google Scholar 

  27. Wang M, et al. Proinflammation: the key to arterial aging. Trends Endocrinol Metab. 2014;25(2):72–9.

    Article  CAS  Google Scholar 

  28. Kang C, et al. The DNA damage response induces inflammation and senescence by inhibiting autophagy of GATA4. Science. 2015;349(6255):aaa5612.

    Article  Google Scholar 

  29. Freund A, Patil CK, Campisi J. p38MAPK is a novel DNA damage response-independent regulator of the senescence-associated secretory phenotype. EMBO J. 2011;30(8):1536–48.

    Article  CAS  Google Scholar 

  30. Bailey-Downs LC, et al. Aging exacerbates obesity-induced oxidative stress and inflammation in perivascular adipose tissue in mice: a paracrine mechanism contributing to vascular redox dysregulation and inflammation. J Gerontol A Biol Sci Med Sci. 2013;68(7):780–92.

    Article  CAS  Google Scholar 

  31. Tucsek Z, et al. Aging exacerbates obesity-induced cerebromicrovascular rarefaction, neurovascular uncoupling, and cognitive decline in mice. J Gerontol A Biol Sci Med Sci. 2014;69(11):1339–52.

    Article  CAS  Google Scholar 

  32. Hasegawa Y, et al. Blockade of the nuclear factor-kappaB pathway in the endothelium prevents insulin resistance and prolongs life spans. Circulation. 2012;125(9):1122–33.

    Article  CAS  Google Scholar 

  33. Gano LB, et al. The SIRT1 activator SRT1720 reverses vascular endothelial dysfunction, excessive superoxide production, and inflammation with aging in mice. Am J Physiol Heart Circ Physiol. 2014;307(12):H1754–63.

    Article  CAS  Google Scholar 

  34. Karasawa T, Takahashi M. Role of NLRP3 inflammasomes in atherosclerosis. J Atheroscler Thromb. 2017;24(5):443–51.

    Article  CAS  Google Scholar 

  35. Song Y, et al. Aging enhances the basal production of IL-6 and CCL2 in vascular smooth muscle cells. Arterioscler Thromb Vasc Biol. 2012;32(1):103–9.

    Article  CAS  Google Scholar 

  36. Wang GC, et al. Cytomegalovirus infection and the risk of mortality and frailty in older women: a prospective observational cohort study. Am J Epidemiol. 2010;171(10):1144–52.

    Article  Google Scholar 

  37. Shah AV, Bennett MR. DNA damage-dependent mechanisms of ageing and disease in the macro- and microvasculature. Eur J Pharmacol. 2017;816:116–28.

    Article  CAS  Google Scholar 

  38. Durik M, et al. Nucleotide excision DNA repair is associated with age-related vascular dysfunction. Circulation. 2012;126(4):468–78.

    Article  CAS  Google Scholar 

  39. Matsumoto T, et al. Aging-associated vascular phenotype in mutant mice with low levels of BubR1. Stroke. 2007;38(3):1050–6.

    Article  CAS  Google Scholar 

  40. Liu J, et al. Roles of telomere biology in cell senescence, replicative and chronological ageing. Cells. 2019;8(1):54.

    Article  CAS  Google Scholar 

  41. Zhang W, et al. Epigenetic modifications in cardiovascular aging and diseases. Circ Res. 2018;123(7):773–86.

    Article  CAS  Google Scholar 

  42. Gensous N, et al. The impact of caloric restriction on the epigenetic signatures of aging. Int J Mol Sci. 2019;20(8):2022.

    Article  CAS  Google Scholar 

  43. Kida Y, Goligorsky MS. Sirtuins, cell senescence, and vascular aging. Can J Cardiol. 2016;32(5):634–41.

    Article  Google Scholar 

  44. de Lucia C, et al. microRNA in cardiovascular aging and age-related cardiovascular diseases. Front Med (Lausanne). 2017;4:74.

    Article  Google Scholar 

  45. Tarantini S, et al. IGF-1 deficiency in a critical period early in life influences the vascular aging phenotype in mice by altering miRNA-mediated post-transcriptional gene regulation: implications for the developmental origins of health and disease hypothesis. Age (Dordr). 2016;38(4):239–58.

    Article  CAS  Google Scholar 

  46. Lopez-Otin C, et al. The hallmarks of aging. Cell. 2013;153(6):1194–217.

    Article  CAS  Google Scholar 

  47. Campisi J, d’Adda di Fagagna F. Cellular senescence: when bad things happen to good cells. Nat Rev Mol Cell Biol. 2007;8(9):729–40.

    Article  CAS  Google Scholar 

  48. Erusalimsky JD. Vascular endothelial senescence: from mechanisms to pathophysiology. J Appl Physiol (1985). 2009;106(1):326–32.

    Article  CAS  Google Scholar 

  49. Childs BG, et al. Senescent intimal foam cells are deleterious at all stages of atherosclerosis. Science. 2016;354(6311):472–7.

    Article  CAS  Google Scholar 

  50. Ungvari Z, et al. Ionizing radiation promotes the acquisition of a senescence-associated secretory phenotype and impairs angiogenic capacity in cerebromicrovascular endothelial cells: role of increased DNA damage and decreased DNA repair capacity in microvascular radiosensitivity. J Gerontol A Biol Sci Med Sci. 2013;68(12):1443–57.

    Article  CAS  Google Scholar 

  51. Roos CM, et al. Chronic senolytic treatment alleviates established vasomotor dysfunction in aged or atherosclerotic mice. Aging Cell. 2016;15(5):973–7.

    Article  CAS  Google Scholar 

  52. Kumar S, Dietrich N, Kornfeld K. Angiotensin converting enzyme (ACE) inhibitor extends Caenorhabditis elegans life span. PLoS Genet. 2016;12(2):e1005866.

    Article  Google Scholar 

  53. Wang M, et al. Proinflammatory profile within the grossly normal aged human aortic wall. Hypertension. 2007;50(1):219–27.

    Article  CAS  Google Scholar 

  54. Wang M, et al. Angiotensin II activates matrix metalloproteinase type II and mimics age-associated carotid arterial remodeling in young rats. Am J Pathol. 2005;167(5):1429–42.

    Article  CAS  Google Scholar 

  55. Wang M, et al. Aging increases aortic MMP-2 activity and angiotensin II in nonhuman primates. Hypertension. 2003;41(6):1308–16.

    Article  CAS  Google Scholar 

  56. McCurley A, Jaffe IZ. Mineralocorticoid receptors in vascular function and disease. Mol Cell Endocrinol. 2012;350(2):256–65.

    Article  CAS  Google Scholar 

  57. Bonnans C, Chou J, Werb Z. Remodelling the extracellular matrix in development and disease. Nat Rev Mol Cell Biol. 2014;15(12):786–801.

    Article  CAS  Google Scholar 

  58. Toth P, et al. Aging exacerbates hypertension-induced cerebral microhemorrhages in mice: role of resveratrol treatment in vasoprotection. Aging Cell. 2015;14(3):400–8.

    Article  CAS  Google Scholar 

  59. Fleenor BS, et al. Arterial stiffening with ageing is associated with transforming growth factor-beta1-related changes in adventitial collagen: reversal by aerobic exercise. J Physiol. 2010;588(Pt 20):3971–82.

    Article  CAS  Google Scholar 

  60. Jacob MP. Extracellular matrix remodeling and matrix metalloproteinases in the vascular wall during aging and in pathological conditions. Biomed Pharmacother. 2003;57(5–6):195–202.

    Article  CAS  Google Scholar 

  61. Tarantini S, et al. Insulin-like growth factor 1 deficiency exacerbates hypertension-induced cerebral microhemorrhages in mice, mimicking the aging phenotype. Aging Cell. 2017;16(3):469–79.

    Article  CAS  Google Scholar 

  62. Keymel S, et al. Impaired endothelial progenitor cell function predicts age-dependent carotid intimal thickening. Basic Res Cardiol. 2008;103(6):582–6.

    Article  Google Scholar 

  63. Rauscher FM, et al. Aging, progenitor cell exhaustion, and atherosclerosis. Circulation. 2003;108(4):457–63.

    Article  Google Scholar 

  64. Zhu G, et al. Young environment reverses the declined activity of aged rat-derived endothelial progenitor cells: involvement of the phosphatidylinositol 3-kinase/Akt signaling pathway. Ann Vasc Surg. 2009;23(4):519–34.

    Article  Google Scholar 

  65. Chirinos JA, et al. Large-artery stiffness in health and disease: JACC state-of-the-art review. J Am Coll Cardiol. 2019;74(9):1237–63.

    Article  Google Scholar 

  66. Paneni F, et al. The aging cardiovascular system: understanding it at the cellular and clinical levels. J Am Coll Cardiol. 2017;69(15):1952–67.

    Article  Google Scholar 

  67. Angoff R, Mosarla RC, Tsao CW. Aortic stiffness: epidemiology, risk factors, and relevant biomarkers. Front Cardiovasc Med. 2021;8:709396.

    Article  Google Scholar 

  68. Fritze O, et al. Age-related changes in the elastic tissue of the human aorta. J Vasc Res. 2012;49(1):77–86.

    Article  Google Scholar 

  69. Zieman SJ, Melenovsky V, Kass DA. Mechanisms, pathophysiology, and therapy of arterial stiffness. Arterioscler Thromb Vasc Biol. 2005;25(5):932–43.

    Article  CAS  Google Scholar 

  70. Monk BA, George SJ. The effect of ageing on vascular smooth muscle cell behaviour—a mini-review. Gerontology. 2015;61(5):416–26.

    Article  CAS  Google Scholar 

  71. Pescatore LA, Gamarra LF, Liberman M. Multifaceted mechanisms of vascular calcification in aging. Arterioscler Thromb Vasc Biol. 2019;39(7):1307–16.

    Article  CAS  Google Scholar 

  72. Ungvari Z, et al. Mechanisms of vascular aging. Circ Res. 2018;123(7):849–67.

    Article  CAS  Google Scholar 

  73. Bramwell JC, Hill AV. The velocity of pulse wave in man. Proc R Soc Lond Ser B Biol Sci. 1922;93(652):298–306.

    Google Scholar 

  74. Reference Values for Arterial Stiffness Collaboration. Determinants of pulse wave velocity in healthy people and in the presence of cardiovascular risk factors: ‘establishing normal and reference values’. Eur Heart J. 2010;31(19):2338–50.

    Article  Google Scholar 

  75. Najjar SS, et al. Age and gender affect ventricular-vascular coupling during aerobic exercise. J Am Coll Cardiol. 2004;44(3):611–7.

    Article  Google Scholar 

  76. Redfield MM, et al. Age- and gender-related ventricular-vascular stiffening: a community-based study. Circulation. 2005;112(15):2254–62.

    Article  Google Scholar 

  77. De Tombe PP, et al. Ventricular stroke work and efficiency both remain nearly optimal despite altered vascular loading. Am J Phys. 1993;264(6 Pt 2):H1817–24.

    Google Scholar 

  78. Segers P, Stergiopulos N, Westerhof N. Relation of effective arterial elastance to arterial system properties. Am J Physiol Heart Circ Physiol. 2002;282(3):H1041–6.

    Article  CAS  Google Scholar 

  79. AlGhatrif M, et al. Longitudinal uncoupling of the heart and arteries with aging in a community-dwelling population. Geroscience. 2021;43(2):551–61.

    Article  Google Scholar 

  80. Vasan RS, et al. Arterial stiffness and long-term risk of health outcomes: the Framingham Heart Study. Hypertension. 2022;79(5):1045–56.

    Article  CAS  Google Scholar 

  81. Ben-Shlomo Y, et al. Aortic pulse wave velocity improves cardiovascular event prediction: an individual participant meta-analysis of prospective observational data from 17,635 subjects. J Am Coll Cardiol. 2014;63(7):636–46.

    Article  Google Scholar 

  82. Sutton-Tyrrell K, et al. Elevated aortic pulse wave velocity, a marker of arterial stiffness, predicts cardiovascular events in well-functioning older adults. Circulation. 2005;111(25):3384–90.

    Article  Google Scholar 

  83. Liao D, et al. Arterial stiffness and the development of hypertension. The ARIC study. Hypertension. 1999;34(2):201–6.

    Article  CAS  Google Scholar 

  84. Najjar SS, et al. Pulse wave velocity is an independent predictor of the longitudinal increase in systolic blood pressure and of incident hypertension in the Baltimore Longitudinal Study of Aging. J Am Coll Cardiol. 2008;51(14):1377–83.

    Article  Google Scholar 

  85. Franklin SS, et al. Does the relation of blood pressure to coronary heart disease risk change with aging? The Framingham Heart Study. Circulation. 2001;103(9):1245–9.

    Article  CAS  Google Scholar 

  86. Chae CU, et al. Increased pulse pressure and risk of heart failure in the elderly. JAMA. 1999;281(7):634–9.

    Article  CAS  Google Scholar 

  87. Haider AW, et al. Systolic blood pressure, diastolic blood pressure, and pulse pressure as predictors of risk for congestive heart failure in the Framingham Heart Study. Ann Intern Med. 2003;138(1):10–6.

    Article  Google Scholar 

  88. Omote K, Verbrugge FH, Borlaug BA. Heart failure with preserved ejection fraction: mechanisms and treatment strategies. Annu Rev Med. 2022;73:321–37.

    Article  Google Scholar 

  89. Reddy YNV, et al. Arterial stiffening with exercise in patients with heart failure and preserved ejection fraction. J Am Coll Cardiol. 2017;70(2):136–48.

    Article  Google Scholar 

  90. Shaikh AY, et al. Relations of arterial stiffness and brachial flow-mediated dilation with new-onset atrial fibrillation: the Framingham Heart Study. Hypertension. 2016;68(3):590–6.

    Article  CAS  Google Scholar 

  91. Pase MP, et al. Aortic stiffness and the risk of incident mild cognitive impairment and dementia. Stroke. 2016;47(9):2256–61.

    Article  Google Scholar 

  92. Cui C, et al. Aortic stiffness is associated with increased risk of incident dementia in older adults. J Alzheimers Dis. 2018;66(1):297–306.

    Article  CAS  Google Scholar 

  93. Bown CW, et al. Elevated aortic pulse wave velocity relates to longitudinal gray and white matter changes. Arterioscler Thromb Vasc Biol. 2021;41(12):3015–24.

    Article  CAS  Google Scholar 

  94. Yannoutsos A, et al. Clinical relevance of aortic stiffness in end-stage renal disease and diabetes: implication for hypertension management. J Hypertens. 2018;36(6):1237–46.

    Article  CAS  Google Scholar 

  95. Peralta CA, et al. Association of pulse pressure, arterial elasticity, and endothelial function with kidney function decline among adults with estimated GFR >60 mL/min/1.73 m(2): the Multi-Ethnic Study of Atherosclerosis (MESA). Am J Kidney Dis. 2012;59(1):41–9.

    Article  Google Scholar 

  96. Appel LJ, et al. A clinical trial of the effects of dietary patterns on blood pressure. DASH Collaborative Research Group. N Engl J Med. 1997;336(16):1117–24.

    Article  CAS  Google Scholar 

  97. Menotti A, Puddu PE. How the Seven Countries Study contributed to the definition and development of the Mediterranean diet concept: a 50-year journey. Nutr Metab Cardiovasc Dis. 2015;25(3):245–52.

    Article  CAS  Google Scholar 

  98. LaRocca TJ, Martens CR, Seals DR. Nutrition and other lifestyle influences on arterial aging. Ageing Res Rev. 2017;39:106–19.

    Article  Google Scholar 

  99. Mozaffarian D, Wu JHY. Flavonoids, dairy foods, and cardiovascular and metabolic health: a review of emerging biologic pathways. Circ Res. 2018;122(2):369–84.

    Article  CAS  Google Scholar 

  100. Wang M, et al. Calorie restriction curbs proinflammation that accompanies arterial aging, preserving a youthful phenotype. J Am Heart Assoc. 2018;7(18):e009112.

    Article  Google Scholar 

  101. Gates PE, et al. Dietary sodium restriction rapidly improves large elastic artery compliance in older adults with systolic hypertension. Hypertension. 2004;44(1):35–41.

    Article  CAS  Google Scholar 

  102. Figueroa A, et al. Effects of diet and/or low-intensity resistance exercise training on arterial stiffness, adiposity, and lean mass in obese postmenopausal women. Am J Hypertens. 2013;26(3):416–23.

    Article  CAS  Google Scholar 

  103. Dengo AL, et al. Arterial destiffening with weight loss in overweight and obese middle-aged and older adults. Hypertension. 2010;55(4):855–61.

    Article  CAS  Google Scholar 

  104. Santos-Parker JR, LaRocca TJ, Seals DR. Aerobic exercise and other healthy lifestyle factors that influence vascular aging. Adv Physiol Educ. 2014;38(4):296–307.

    Article  Google Scholar 

  105. Lopes S, et al. Exercise training reduces arterial stiffness in adults with hypertension: a systematic review and meta-analysis. J Hypertens. 2021;39(2):214–22.

    Article  CAS  Google Scholar 

  106. Vaitkevicius PV, et al. Effects of age and aerobic capacity on arterial stiffness in healthy adults. Circulation. 1993;88(4 Pt 1):1456–62.

    Article  CAS  Google Scholar 

  107. De Cesaris R, et al. Forearm arterial distensibility in patients with hypertension: comparative effects of long-term ACE inhibition and beta-blocking. Clin Pharmacol Ther. 1993;53(3):360–7.

    Article  Google Scholar 

  108. Dudenbostel T, Glasser SP. Effects of antihypertensive drugs on arterial stiffness. Cardiol Rev. 2012;20(5):259–63.

    Article  Google Scholar 

  109. Gurven M, Blackwell AD, Rodríguez DE, Stieglitz J, Kaplan H. Does blood pressure inevitably rise with age?: Longitudinal evidence among forager-horticulturalists. Hypertension. 2012 Jul;60(1):25-33. doi: https://doi.org/10.1161/HYPERTENSIONAHA.111.189100.. Epub 2012 May 21. PMID: 22700319; PMCID: PMC3392307.

  110. Tsao CW, Aday AW, Almarzooq ZI, Alonso A, Beaton AZ, Bittencourt MS, Boehme AK, Buxton AE, Carson AP, Commodore-Mensah Y, MSV E, Evenson KR, Eze-Nliam C, Ferguson JF, Generoso G, Ho JE, Kalani R, Khan SS, Kissela BM, Knutson KL, Levine DA, Lewis TT, Liu J, Loop MS, Ma J, Mussolino ME, Navaneethan SD, Perak AM, Poudel R, Rezk-Hanna M, Roth GA, Schroeder EB, Shah SH, Thacker EL, LB VW, Virani SS, Voecks JH, Wang NY, Yaffe K, Martin SS. Heart Disease and Stroke Statistics-2022 Update: A Report From the American Heart Association. Circulation. 2022;145(8):e153–639. https://doi.org/10.1161/CIR.0000000000001052. Epub 2022 Jan 26. Erratum in: Circulation. 2022 Sep 6;146(10):e141

  111. Avolio AP, Deng FQ, Li WQ, Luo YF, Huang ZD, **ng LF, O’Rourke MF. Effects of aging on arterial distensibility in populations with high and low prevalence of hypertension: comparison between urban and rural communities in China. Circulation. 1985 Feb;71(2):202-210. doi: https://doi.org/10.1161/01.cir.71.2.202.

  112. He J, Klag MJ, Whelton PK, Chen JY, Mo JP, Qian MC, Mo PS, He GQ. Migration, blood pressure pattern, and hypertension: the Yi Migrant Study. Am J Epidemiol. 1991;134(10):1085–101. https://doi.org/10.1093/oxfordjournals.aje.a116012.

  113. Halcox JP, et al. Prognostic value of coronary vascular endothelial dysfunction. Circulation. 2002;106(6):653–8.

    Article  Google Scholar 

  114. Houghton JL, et al. Effect of African-American race and hypertensive left ventricular hypertrophy on coronary vascular reactivity and endothelial function. Hypertension. 1997;29(3):706–14.

    Article  CAS  Google Scholar 

  115. Cosson E, et al. Impaired coronary endothelium-dependent vasodilation is associated with microalbuminuria in patients with type 2 diabetes and angiographically normal coronary arteries. Diabetes Care. 2006;29(1):107–12.

    Article  Google Scholar 

  116. Treasure CB, et al. Beneficial effects of cholesterol-lowering therapy on the coronary endothelium in patients with coronary artery disease. N Engl J Med. 1995;332(8):481–7.

    Article  CAS  Google Scholar 

  117. Schachinger V, Britten MB, Zeiher AM. Prognostic impact of coronary vasodilator dysfunction on adverse long-term outcome of coronary heart disease. Circulation. 2000;101(16):1899–906.

    Article  CAS  Google Scholar 

  118. Doucette JW, et al. Validation of a Doppler guide wire for intravascular measurement of coronary artery flow velocity. Circulation. 1992;85(5):1899–911.

    Article  CAS  Google Scholar 

  119. Zeiher AM, et al. Modulation of coronary vasomotor tone in humans. Progressive endothelial dysfunction with different early stages of coronary atherosclerosis. Circulation. 1991;83(2):391–401.

    Article  CAS  Google Scholar 

  120. Ludmer PL, et al. Paradoxical vasoconstriction induced by acetylcholine in atherosclerotic coronary arteries. N Engl J Med. 1986;315(17):1046–51.

    Article  CAS  Google Scholar 

  121. Niccoli G, Scalone G, Crea F. Coronary functional tests in the catheterization laboratory - pathophysiological and clinical relevance. Circ J. 2015;79(4):676–84.

    Article  CAS  Google Scholar 

  122. Randaccio P. A program for optimizing radiation beam parameters in radiotherapy (author’s transl). Radiol Med. 1979;65(10):741–6.

    CAS  Google Scholar 

  123. Nabel EG, et al. Dilation of normal and constriction of atherosclerotic coronary arteries caused by the cold pressor test. Circulation. 1988;77(1):43–52.

    Article  CAS  Google Scholar 

  124. Gordon JB, et al. Atherosclerosis influences the vasomotor response of epicardial coronary arteries to exercise. J Clin Invest. 1989;83(6):1946–52.

    Article  CAS  Google Scholar 

  125. Leucker TM, et al. Coronary endothelial dysfunction is associated with elevated serum PCSK9 levels in people with HIV independent of low-density lipoprotein cholesterol. J Am Heart Assoc. 2018;7(19):e009996.

    Article  Google Scholar 

  126. Beanlands RS, et al. Noninvasive quantification of regional myocardial flow reserve in patients with coronary atherosclerosis using nitrogen-13 ammonia positron emission tomography. Determination of extent of altered vascular reactivity. J Am Coll Cardiol. 1995;26(6):1465–75.

    Article  CAS  Google Scholar 

  127. Taqueti VR, Di Carli MF. Coronary microvascular disease pathogenic mechanisms and therapeutic options: JACC state-of-the-art review. J Am Coll Cardiol. 2018;72(21):2625–41.

    Article  Google Scholar 

  128. George RT, et al. Quantification of myocardial perfusion using dynamic 64-detector computed tomography. Investig Radiol. 2007;42(12):815–22.

    Article  Google Scholar 

  129. Corretti MC, et al. Guidelines for the ultrasound assessment of endothelial-dependent flow-mediated vasodilation of the brachial artery: a report of the International Brachial Artery Reactivity Task Force. J Am Coll Cardiol. 2002;39(2):257–65.

    Article  Google Scholar 

  130. Sara JD, et al. Prevalence of coronary microvascular dysfunction among patients with chest pain and nonobstructive coronary artery disease. JACC Cardiovasc Interv. 2015;8(11):1445–53.

    Article  Google Scholar 

  131. Hays AG, et al. Noninvasive visualization of coronary artery endothelial function in healthy subjects and in patients with coronary artery disease. J Am Coll Cardiol. 2010;56(20):1657–65.

    Article  Google Scholar 

  132. Hundley WG, et al. Assessment of coronary arterial flow and flow reserve in humans with magnetic resonance imaging. Circulation. 1996;93(8):1502–8.

    Article  CAS  Google Scholar 

  133. Leucker TM, et al. Evolocumab, a PCSK9-monoclonal antibody, rapidly reverses coronary artery endothelial dysfunction in people living with HIV and people with dyslipidemia. J Am Heart Assoc. 2020;9(14):e016263.

    Article  CAS  Google Scholar 

  134. Shufelt CL, et al. Cardiac magnetic resonance imaging myocardial perfusion reserve index assessment in women with microvascular coronary dysfunction and reference controls. Cardiovasc Diagn Ther. 2013;3(3):153–60.

    Google Scholar 

  135. Dorbala S, et al. Coronary microvascular dysfunction is related to abnormalities in myocardial structure and function in cardiac amyloidosis. JACC Heart Fail. 2014;2(4):358–67.

    Article  Google Scholar 

  136. Kelle S, et al. A bi-center cardiovascular magnetic resonance prognosis study focusing on dobutamine wall motion and late gadolinium enhancement in 3,138 consecutive patients. J Am Coll Cardiol. 2013;61(22):2310–2.

    Article  Google Scholar 

  137. Haluska B, et al. Automated edge-detection technique for measurement of brachial artery reactivity: a comparison of concordance with manual measurements. Ultrasound Med Biol. 2001;27(9):1285–9.

    Article  CAS  Google Scholar 

  138. Matsuzawa Y, et al. Prognostic value of flow-mediated vasodilation in brachial artery and fingertip artery for cardiovascular events: a systematic review and meta-analysis. J Am Heart Assoc. 2015;4(11):e002270.

    Article  Google Scholar 

  139. Allan RB, et al. A comparison of flow-mediated dilatation and peripheral artery tonometry for measurement of endothelial function in healthy individuals and patients with peripheral arterial disease. Eur J Vasc Endovasc Surg. 2013;45(3):263–9.

    Article  CAS  Google Scholar 

  140. Bonetti PO, et al. Noninvasive identification of patients with early coronary atherosclerosis by assessment of digital reactive hyperemia. J Am Coll Cardiol. 2004;44(11):2137–41.

    Article  Google Scholar 

  141. Iantorno M, et al. Simultaneous noninvasive assessment of systemic and coronary endothelial function. Circ Cardiovasc Imaging. 2016;9(3):e003954.

    Article  Google Scholar 

  142. Minhas AS, et al. Imaging assessment of endothelial function: an index of cardiovascular health. Front Cardiovasc Med. 2022;9:778762.

    Article  CAS  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Thorsten M. Leucker .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2023 The Author(s), under exclusive license to Springer Nature Switzerland AG

About this chapter

Check for updates. Verify currency and authenticity via CrossMark

Cite this chapter

Leucker, T.M., Goldenberg, J., Gerstenblith, G. (2023). Aging of the Vasculature. In: Leucker, T.M., Gerstenblith, G. (eds) Cardiovascular Disease in the Elderly. Contemporary Cardiology. Humana, Cham. https://doi.org/10.1007/978-3-031-16594-8_4

Download citation

  • DOI: https://doi.org/10.1007/978-3-031-16594-8_4

  • Published:

  • Publisher Name: Humana, Cham

  • Print ISBN: 978-3-031-16593-1

  • Online ISBN: 978-3-031-16594-8

  • eBook Packages: MedicineMedicine (R0)

Publish with us

Policies and ethics

Navigation