Clinical Impact and Mechanisms of Nonatherosclerotic Vascular Aging: The New Kid to Be Blocked

Canadian Journal of Cardiology - Tập 39 - Trang 1839-1858 - 2023
Soroush Mohammadi Jouabadi1,2, Ehsan Ataei Ataabadi1, Keivan Golshiri1, Daniel Bos2,3, Bruno H.C. Stricker2, A.H. Jan Danser1, Francesco Mattace-Raso4, Anton J.M. Roks1
1Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands
2Department of Epidemiology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
3Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands
4Division of Geriatric Medicine, Department of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands

Tài liệu tham khảo

Mozaffarian, 2015, Heart disease and stroke statistics—2015 update: a report from the American Heart Association, Circulation, 131, e29 Knuuti, 2020, 2019 ESC guidelines for the diagnosis and management of chronic coronary syndromes, Eur Heart J, 41, 407, 10.1093/eurheartj/ehz425 Kyhl, 2021, Conductance artery stiffness impairs atrio-ventriculo-arterial coupling before manifestation of arterial hypertension or left ventricular hypertrophic remodelling, Sci Rep, 11, 10.1038/s41598-021-93614-w O’Rourke, 1968, Pressure wave transmission along the human aorta. Changes with age and in arterial degenerative disease, Circ Res, 23, 567, 10.1161/01.RES.23.4.567 Kucharska-Newton, 2019, Determinants of vascular age: an epidemiological perspective, Clin Chem, 65, 108, 10.1373/clinchem.2018.287623 Benetos, 2002, Determinants of accelerated progression of arterial stiffness in normotensive subjects and in treated hypertensive subjects over a 6-year period, Circulation, 105, 1202, 10.1161/hc1002.105135 Van Popele, 2001, Association between arterial stiffness and atherosclerosis: the Rotterdam Study, Stroke, 32, 454, 10.1161/01.STR.32.2.454 Roman, 1992, Parallel cardiac and vascular adaptation in hypertension, Circulation, 86, 1909, 10.1161/01.CIR.86.6.1909 Safar, 2006, Metabolic syndrome and age-related progression of aortic stiffness, J Am Coll Cardiol, 47, 72, 10.1016/j.jacc.2005.08.052 Mitchell, 2004, Changes in arterial stiffness and wave reflection with advancing age in healthy men and women: the Framingham Heart Study, Hypertension, 43, 1239, 10.1161/01.HYP.0000128420.01881.aa 2010, Determinants of pulse wave velocity in healthy people and in the presence of cardiovascular risk factors: “establishing normal and reference values.”, Eur Heart J, 31, 2338, 10.1093/eurheartj/ehq165 Laucyte-Cibulskiene, 2022, Clusters of risk factors in metabolic syndrome and their influence on central blood pressure in a global study, Sci Rep, 12, 10.1038/s41598-022-18094-y Avolio, 1985, Effects of aging on arterial distensibility in populations with high and low prevalence of hypertension: comparison between urban and rural communities in China, Circulation, 71, 202, 10.1161/01.CIR.71.2.202 Cavalcante, 2011, Aortic stiffness: current understanding and future directions, J Am Coll Cardiol, 57, 1511, 10.1016/j.jacc.2010.12.017 Verwoert, 2014, Arterial stiffness and hypertension in a large population of untreated individuals: the Rotterdam Study, J Hypertens, 32, 1606, 10.1097/HJH.0000000000000237 Mattace-Raso, 2006, Arterial stiffness and risk of coronary heart disease and stroke: the Rotterdam Study, Circulation, 113, 657, 10.1161/CIRCULATIONAHA.105.555235 Sedaghat, 2015, Arterial stiffness and decline in kidney function, Clin J Am Soc Nephrol, 10, 2190, 10.2215/CJN.03000315 Van Sloten, 2015, Carotid stiffness is associated with incident stroke: a systematic review and individual participant data meta-analysis, J Am Coll Cardiol, 66, 2116, 10.1016/j.jacc.2015.08.888 Poels, 2012, Arterial stiffness and cerebral small vessel disease: the Rotterdam Scan Study, Stroke, 43, 2637, 10.1161/STROKEAHA.111.642264 Scuteri, 2007, Arterial stiffness as an independent predictor of longitudinal changes in cognitive function in the older individual, J Hypertens, 25, 1035, 10.1097/HJH.0b013e3280895b55 Selwaness, 2014, Arterial stiffness is associated with carotid intraplaque hemorrhage in the general population: the Rotterdam study, Arterioscler Thromb Vasc Biol, 34, 927, 10.1161/ATVBAHA.113.302603 Loutzenhiser, 2002, Renal myogenic response: kinetic attributes and physiological role, Circ Res, 90, 1316, 10.1161/01.RES.0000024262.11534.18 Safar, 2004, Arterial stiffness and kidney function, Hypertension, 43, 163, 10.1161/01.HYP.0000114571.75762.b0 Li, 2016, Bone mineral density is negatively associated with arterial stiffness in men with hypertension, J Clin Hypertens, 18, 1106, 10.1111/jch.12848 Kim, 2011, Skeletal muscle mass to visceral fat area ratio is associated with metabolic syndrome and arterial stiffness: the Korean Sarcopenic Obesity Study (KSOS), Diabetes Res Clin Pract, 93, 285, 10.1016/j.diabres.2011.06.013 Kohara, 2017, Muscle mass decline, arterial stiffness, white matter hyperintensity, and cognitive impairment: Japan Shimanami Health Promoting Program study, J Cachexia Sarcopenia Muscle, 8, 557, 10.1002/jcsm.12195 Waller, 1989, Anatomy, histology, and pathology of the major epicardial coronary arteries relevant to echocardiographic imaging techniques, J Am Soc Echocardiogr, 2, 232, 10.1016/S0894-7317(89)80084-7 Vos, 2016, Predominance of nonatherosclerotic internal elastic lamina calcification in the intracranial internal carotid artery, Stroke, 47, 221, 10.1161/STROKEAHA.115.011196 Amann, 2008, Media calcification and intima calcification are distinct entities in chronic kidney disease, Clin J Am Soc Nephrol, 3, 1599, 10.2215/CJN.02120508 Kockelkoren, 2017, Computed tomographic distinction of intimal and medial calcification in the intracranial internal carotid artery, PLoS One, 12, 10.1371/journal.pone.0168360 Psychogios, 2022, European Stroke Organisation guidelines on treatment of patients with intracranial atherosclerotic disease, Eur Stroke J, 7, III, 10.1177/23969873221099715 van den Beukel, 2022, Morphological subtypes of intracranial internal carotid artery arteriosclerosis and the risk of stroke, Stroke, 53, 1339, 10.1161/STROKEAHA.121.036213 Compagne, 2018, Intracranial carotid artery calcification and effect of endovascular stroke treatment, Stroke, 49, 2961, 10.1161/STROKEAHA.118.022400 Luijten, 2021, Intracranial carotid artery calcification subtype and collaterals in patients undergoing endovascular thrombectomy, Atherosclerosis, 337, 1, 10.1016/j.atherosclerosis.2021.10.005 Steppan, 2011, Vascular stiffness and increased pulse pressure in the aging cardiovascular system, Cardiol Res Pract, 2011, 10.4061/2011/263585 Ataei Ataabadi, 2020, Nitric oxide–cGMP signaling in hypertension: current and future options for pharmacotherapy, Hypertension, 76, 1055, 10.1161/HYPERTENSIONAHA.120.15856 Lacolley, 2018, Smooth muscle cell and arterial aging: basic and clinical aspects, Cardiovasc Res, 114, 513, 10.1093/cvr/cvy009 López-Otín, 2013, The hallmarks of aging, Cell, 153, 1194, 10.1016/j.cell.2013.05.039 López-Otín, 2023, Hallmarks of aging: an expanding universe, Cell, 186, 243, 10.1016/j.cell.2022.11.001 Schumacher, 2021, The central role of DNA damage in the ageing process, Nature, 592, 695, 10.1038/s41586-021-03307-7 Gyenis, 2023, Genome-wide RNA polymerase stalling shapes the transcriptome during aging, Nat Genet, 55, 268, 10.1038/s41588-022-01279-6 Vougioukalaki, 2022, Different responses to DNA damage determine ageing differences between organs, Aging Cell, 21, 10.1111/acel.13562 Wu, 2014, Genomic instability and vascular aging: a focus on nucleotide excision repair, Trends Cardiovasc Med, 24, 61, 10.1016/j.tcm.2013.06.005 Bautista-Niño, 2016, DNA damage: a main determinant of vascular aging, Int J Mol Sci, 17, 748, 10.3390/ijms17050748 Piekarowicz, 2019, Hutchinson-Gilford progeria syndrome—current status and prospects for gene therapy treatment, Cells, 8, 88, 10.3390/cells8020088 Olive, 2010, Cardiovascular pathology in Hutchinson-Gilford progeria: correlation with the vascular pathology of aging, Arterioscler Thromb Vasc Biol, 30, 2301, 10.1161/ATVBAHA.110.209460 Stehbens, 1999, Histological and ultrastructural features of atherosclerosis in progeria, Cardiovasc Pathol, 8, 29, 10.1016/S1054-8807(98)00023-4 Durik, 2012, Nucleotide excision DNA repair is associated with age-related vascular dysfunction, Circulation, 126, 468, 10.1161/CIRCULATIONAHA.112.104380 Samani, 2008, Chromosome 9p21 and cardiovascular disease: the story unfolds, Circ Cardiovasc Genet, 1, 81, 10.1161/CIRCGENETICS.108.832527 Benetos, 2001, Telomere length as an indicator of biological aging: the gender effect and relation with pulse pressure and pulse wave velocity, Hypertension, 37, 381, 10.1161/01.HYP.37.2.381 Panayiotou, 2010, Leukocyte telomere length is associated with measures of subclinical atherosclerosis, Atherosclerosis, 211, 176, 10.1016/j.atherosclerosis.2010.01.037 Burnett-Hartman, 2012, Telomere-associated polymorphisms correlate with cardiovascular disease mortality in Caucasian women: the Cardiovascular Health Study, Mech Ageing Dev, 133, 275, 10.1016/j.mad.2012.03.002 Khalid, 2019, Epigenetic modifications associated with pathophysiological effects of lead exposure, J Environ Sci Health C, 37, 235 Scherrer, 2012, Systemic and pulmonary vascular dysfunction in children conceived by assisted reproductive technologies, Circulation, 125, 1890, 10.1161/CIRCULATIONAHA.111.071183 Ding, 2018, Epigenetic regulation of vascular aging and age-related vascular diseases, Adv Exp Med Biol, 1086, 55, 10.1007/978-981-13-1117-8_4 Herman, 2021, Epigenetic dysregulation in cardiovascular aging and disease, J Cardiovasc Aging, 1, 10 Lin, 2021, Targeting epigenetic mechanisms in vascular aging, Front Cardiovasc Med, 8 Kumar, 2013, Histone and DNA methylation-mediated epigenetic downregulation of endothelial Kruppel-like factor 2 by low-density lipoprotein cholesterol, Arterioscler Thromb Vasc Biol, 33, 1936, 10.1161/ATVBAHA.113.301765 Wei, 2018, SMAD7 methylation as a novel marker in atherosclerosis, Biochem Biophys Res Commun, 496, 700, 10.1016/j.bbrc.2018.01.121 Ma, 2018, Homocysteine-induced proliferation of vascular smooth muscle cells occurs via PTEN hypermethylation and is mitigated by resveratrol, Mol Med Rep, 17, 5312 Xu, 2019, Aberrant MFN2 transcription facilitates homocysteine-induced VSMCs proliferation via the increased binding of c-Myc to DNMT1 in atherosclerosis, J Cell Mol Med, 23, 4611, 10.1111/jcmm.14341 Challen, 2012, Dnmt3a is essential for hematopoietic stem cell differentiation, Nat Genet, 44, 23, 10.1038/ng.1009 Zaina, 2014, DNA methylation map of human atherosclerosis, Circ Cardiovasc Genet, 7, 692, 10.1161/CIRCGENETICS.113.000441 Aavik, 2019, DNA methylation processes in atheosclerotic plaque, Atherosclerosis, 281, 168, 10.1016/j.atherosclerosis.2018.12.006 Seungyoon, 2018, Mechanisms orchestrating mitochondrial dynamics for energy homeostasis, J Mol Biol, 430, 3922, 10.1016/j.jmb.2018.07.027 Soubannier, 2009, Positioning mitochondrial plasticity within cellular signaling cascades, Biochim Biophys Acta Mol Cell Res, 1793, 154, 10.1016/j.bbamcr.2008.07.008 Simon, 2000, Role of reactive oxygen species (ROS) in apoptosis induction, Apoptosis, 5, 415, 10.1023/A:1009616228304 Rambold, 2011, Mechanisms of mitochondria and autophagy crosstalk, Cell Cycle, 10, 4032, 10.4161/cc.10.23.18384 Youn, 2021, Mitochondrial dysfunction associated with autophagy and mitophagy in cerebrospinal fluid cells of patients with delayed cerebral ischemia following subarachnoid hemorrhage, Sci Rep, 11, 10.1038/s41598-021-96092-2 Foote, 2018, Restoring mitochondrial DNA copy number preserves mitochondrial function and delays vascular aging in mice, Aging Cell, 17, 10.1111/acel.12773 Yu, 2017, Mitochondrial respiration is reduced in atherosclerosis, promoting necrotic core formation and reducing relative fibrous cap thickness, Arterioscler Thromb Vasc Biol, 37, 2322, 10.1161/ATVBAHA.117.310042 Rossman, 2020, Targeting mitochondrial fitness as a strategy for healthy vascular aging, Clin Sci, 134, 1491, 10.1042/CS20190559 Rossman, 2018, Chronic supplementation with a mitochondrial antioxidant (MitoQ) improves vascular function in healthy older adults, Hypertension, 71, 1056, 10.1161/HYPERTENSIONAHA.117.10787 Park, 2020, Vasodilatory and vascular mitochondrial respiratory function with advancing age: evidence of a free radically mediated link in the human vasculature, Am J Physiol Regul Integr Comp Physiol, 318, R701, 10.1152/ajpregu.00268.2019 Chan, 2006, Mitochondrial uncoupling protein-4 regulates calcium homeostasis and sensitivity to store depletion-induced apoptosis in neural cells, J Biol Chem, 281, 37391, 10.1074/jbc.M605552200 Duchen, 2004, Roles of mitochondria in health and disease, Diabetes, 53, S96, 10.2337/diabetes.53.2007.S96 Tyrrell, 2020, Age-associated mitochondrial dysfunction accelerates atherogenesis, Circ Res, 126, 298, 10.1161/CIRCRESAHA.119.315644 Schleicher, 2007, Oxidative stress, AGE, and atherosclerosis, Kidney Int Suppl, S17, 10.1038/sj.ki.5002382 Ungvari, 2010, Mitochondria and aging in the vascular system, J Mol Med (Berl), 88, 1021, 10.1007/s00109-010-0667-5 Sell, 2012, Molecular basis of arterial stiffening: role of glycation—a mini-review, Gerontology, 58, 227, 10.1159/000334668 Jüttner, 2022, Pharmacological developments in antihypertensive treatment through nitric oxide–cGMP modulation, Adv Pharmacol, 94, 57, 10.1016/bs.apha.2022.01.001 Scalera, 2006, Effect of L-arginine on asymmetric dimethylarginine (ADMA) or homocysteine-accelerated endothelial cell aging, Biochem Biophys Res Commun, 345, 1075, 10.1016/j.bbrc.2006.05.015 Scalera, 2004, Endogenous nitric oxide synthesis inhibitor asymmetric dimethyl l-arginine accelerates endothelial cell senescence, Arterioscler Thromb Vasc Biol, 24, 1816, 10.1161/01.ATV.0000141843.77133.fc Chen, 1999, AMP-activated protein kinase phosphorylation of endothelial NO synthase, FEBS Lett, 443, 285, 10.1016/S0014-5793(98)01705-0 Fleming, 2001, Phosphorylation of Thr495 regulates Ca2+/calmodulin-dependent endothelial nitric oxide synthase activity, Circ Res, 88, e68, 10.1161/hh1101.092677 Yoon, 2010, Alterations in the activity and expression of endothelial NO synthase in aged human endothelial cells, Mech Ageing Dev, 131, 119, 10.1016/j.mad.2009.12.010 Liu, 2012, Ginsenoside Rb1 reverses H2O2-induced senescence in human umbilical endothelial cells: involvement of eNOS pathway, J Cardiovasc Pharmacol, 59, 222, 10.1097/FJC.0b013e31823c1d34 Jo-Watanabe, 2014, Glyoxalase I reduces glycative and oxidative stress and prevents age-related endothelial dysfunction through modulation of endothelial nitric oxide synthase phosphorylation, Aging Cell, 13, 519, 10.1111/acel.12204 Ataei Ataabadi, 2021, Vascular ageing features caused by selective DNA damage in smooth muscle cell, Oxid Med Cell Longev, 2021, 10.1155/2021/2308317 Wu, 2017, Dietary restriction but not angiotensin II type 1 receptor blockade improves DNA damage-related vasodilator dysfunction in rapidly aging Ercc1Δ/− mice, Clin Sci (Lond), 131, 1941, 10.1042/CS20170026 Uijl, 2022, Conventional vasopressor and vasopressor-sparing strategies to counteract the blood pressure–lowering effect of small interfering RNA targeting angiotensinogen, J Am Heart Assoc, 11 Uijl, 2021, No evidence for brain renin-angiotensin system activation during DOCA-salt hypertension, Clin Sci (Lond), 135, 259, 10.1042/CS20201239 Cruz-López, 2021, Perivascular adipose tissue in vascular function: does locally synthesized angiotensinogen play a role?, J Cardiovasc Pharmacol, 78, S53, 10.1097/FJC.0000000000001027 Sun, 2020, Megalin: a novel endocytic receptor for prorenin and renin, Hypertension, 75, 1242, 10.1161/HYPERTENSIONAHA.120.14845 Batenburg, 2004, Angiotensin II type 2 receptor–mediated vasodilation in human coronary microarteries, Circulation, 109, 2296, 10.1161/01.CIR.0000128696.12245.57 Verdonk, 2012, Compound 21 induces vasorelaxation via an endothelium– and angiotensin II type 2 receptor–independent mechanism, Hypertension, 60, 722, 10.1161/HYPERTENSIONAHA.112.196022 Verdonk, 2015, Association studies suggest a key role for endothelin-1 in the pathogenesis of preeclampsia and the accompanying renin-angiotensin-aldosterone system suppression, Hypertension, 65, 1316, 10.1161/HYPERTENSIONAHA.115.05267 Moltzer, 2010, Effects of angiotensin metabolites in the coronary vascular bed of the spontaneously hypertensive rat: loss of angiotensin II type 2 receptor–mediated vasodilation, Hypertension, 55, 516, 10.1161/HYPERTENSIONAHA.109.145037 Ungvari, 2018, Mechanisms of vascular aging, Circ Res, 123, 849, 10.1161/CIRCRESAHA.118.311378 Yoon, 2016, Age-associated changes in the vascular renin-angiotensin system in mice, Oxid Med Cell Longev, 2016, 10.1155/2016/6731093 Liaudet, 2009, Role of peroxynitrite in the redox regulation of cell signal transduction pathways, Front Biosci (Landmark Ed), 14, 4809, 10.2741/3569 Boulanger, 1998, Neuronal nitric oxide synthase is expressed in rat vascular smooth muscle cells: activation by angiotensin II in hypertension, Circ Res, 83, 1271, 10.1161/01.RES.83.12.1271 Kawano, 2000, Angiotensin II has multiple profibrotic effects in human cardiac fibroblasts, Circulation, 101, 1130, 10.1161/01.CIR.101.10.1130 Thomas, 2006, Inflammation in cardiovascular disease and regulation of the actin cytoskeleton in inflammatory cells: the actin cytoskeleton as a target, Cardiovasc Hematol Agents Med Chem, 4, 165, 10.2174/187152506776369926 Minamino, 2003, Ras induces vascular smooth muscle cell senescence and inflammation in human atherosclerosis, Circulation, 108, 2264, 10.1161/01.CIR.0000093274.82929.22 Cheng, 2020, The roles of signaling in cytoskeletal changes, random movement, direction-sensing and polarization of eukaryotic cells, Cells, 9, 10.3390/cells9061437 Huveneers, 2015, Between Rho(k) and a hard place: the relation between vessel wall stiffness, endothelial contractility, and cardiovascular disease, Circ Res, 116, 895, 10.1161/CIRCRESAHA.116.305720 Mu, 2020, Cytoskeleton stiffness regulates cellular senescence and innate immune response in Hutchinson-Gilford progeria syndrome, Aging Cell, 19, 10.1111/acel.13152 Ivanov, 2021, Novel functions of the septin cytoskeleton: shaping up tissue inflammation and fibrosis, Am J Pathol, 191, 40, 10.1016/j.ajpath.2020.09.007 Campisi, 2013, Aging, cellular senescence, and cancer, Annu Rev Physiol, 75, 685, 10.1146/annurev-physiol-030212-183653 Niedernhofer, 2018, Nuclear genomic instability and aging, Annu Rev Biochem, 87, 295, 10.1146/annurev-biochem-062917-012239 Hernandez-Segura, 2018, Hallmarks of cellular senescence, Trends Cell Biol, 28, 436, 10.1016/j.tcb.2018.02.001 Childs, 2014, Senescence and apoptosis: dueling or complementary cell fates?, EMBO Rep, 15, 1139, 10.15252/embr.201439245 Itahana, 2013, Colorimetric detection of senescence-associated β galactosidase, Methods Mol Biol, 965, 143, 10.1007/978-1-62703-239-1_8 Lee, 2006, Senescence-associated beta-galactosidase is lysosomal beta-galactosidase, Aging Cell, 5, 187, 10.1111/j.1474-9726.2006.00199.x van Deursen, 2014, The role of senescent cells in ageing, Nature, 509, 439, 10.1038/nature13193 Baker, 2016, Naturally occurring p16(Ink4a)-positive cells shorten healthy lifespan, Nature, 530, 184, 10.1038/nature16932 Kumari, 2021, Mechanisms of cellular senescence: cell cycle arrest and senescence associated secretory phenotype, Front Cell Dev Biol, 9, 10.3389/fcell.2021.645593 Saleh, 2019, Tumor cell escape from therapy-induced senescence as a model of disease recurrence after dormancy, Cancer Res, 79, 1044, 10.1158/0008-5472.CAN-18-3437 Lapasset, 2011, Rejuvenating senescent and centenarian human cells by reprogramming through the pluripotent state, Genes Dev, 25, 2248, 10.1101/gad.173922.111 Kim, 2006, Upregulation of chicken p15INK4b at senescence and in the developing brain, J Cell Sci, 119, 2435, 10.1242/jcs.02989 Thanapaul, 2021, An insight into aging, senescence, and their impacts on wound healing, Adv Geriatr Med Res, 3 Andrade, 2022, Role of senescent cells in cutaneous wound healing, Biology (Basel), 11 Huang, 2022, Cellular senescence: the good, the bad and the unknown, Nat Rev Nephrol, 18, 611, 10.1038/s41581-022-00601-z Freund, 2010, Inflammatory networks during cellular senescence: causes and consequences, Trends Mol Med, 16, 238, 10.1016/j.molmed.2010.03.003 Franceschi, 2000, Inflamm-aging. An evolutionary perspective on immunosenescence, Ann N Y Acad Sci, 908, 244, 10.1111/j.1749-6632.2000.tb06651.x Youm, 2013, Canonical Nlrp3 inflammasome links systemic low-grade inflammation to functional decline in aging, Cell Metab, 18, 519, 10.1016/j.cmet.2013.09.010 Chen, 2016, Age-associated sirtuin 1 reduction in vascular smooth muscle links vascular senescence and inflammation to abdominal aortic aneurysm, Circ Res, 119, 1076, 10.1161/CIRCRESAHA.116.308895 Ungvari, 2007, Increased mitochondrial H2O2 production promotes endothelial NF-κB activation in aged rat arteries, Am J Physiol Heart Circ Physiol, 293, H37, 10.1152/ajpheart.01346.2006 Guo, 2019, Endothelial SIRT1 prevents age-induced impairment of vasodilator responses by enhancing the expression and activity of soluble guanylyl cyclase in smooth muscle cells, Cardiovasc Res, 115, 678, 10.1093/cvr/cvy212 Wang, 2011, SIRT1 and AMPK in regulating mammalian senescence: a critical review and a working model, FEBS Lett, 585, 986, 10.1016/j.febslet.2010.11.047 Ren, 2009, Role of interleukin-1β during pain and inflammation, Brain Res Rev, 60, 57, 10.1016/j.brainresrev.2008.12.020 Abbate, 2020, Interleukin-1 blockade inhibits the acute inflammatory response in patients with ST-segment-elevation myocardial infarction, J Am Heart Assoc, 9 Reiss, 2017, Interleukin-6 in atherosclerosis: atherogenic or atheroprotective?, Clin Lipidol, 12, 14 Markousis-Mavrogenis, 2019, The clinical significance of interleukin-6 in heart failure: results from the BIOSTAT-CHF study, Eur J Heart Fail, 21, 965, 10.1002/ejhf.1482 Esteve, 2007, Serum interleukin-6 correlates with endothelial dysfunction in healthy men independently of insulin sensitivity, Diabetes Care, 30, 939, 10.2337/dc06-1793 Sprague, 2009, Inflammatory cytokines in vascular dysfunction and vascular disease, Biochem Pharmacol, 78, 539, 10.1016/j.bcp.2009.04.029 Donato, 2012, TNF-α impairs endothelial function in adipose tissue resistance arteries of mice with diet-induced obesity, Am J Physiol Heart Circ Physiol, 303, H672, 10.1152/ajpheart.00271.2012 Goetze, 1999, TNF-α–induced migration of vascular smooth muscle cells is MAPK dependent, Hypertension, 33, 183, 10.1161/01.HYP.33.1.183 Davis, 2012, TNF-α–mediated proliferation of vascular smooth muscle cells involves Raf-1–mediated inactivation of Rb and transcription of E2F1-regulated genes, Cell Cycle, 11, 109, 10.4161/cc.11.1.18473 Mistriotis, 2017, Vascular aging: molecular mechanisms and potential treatments for vascular rejuvenation, Ageing Res Rev, 37, 94, 10.1016/j.arr.2017.05.006 Hasegawa, 2012, Blockade of the nuclear factor-κB pathway in the endothelium prevents insulin resistance and prolongs life spans, Circulation, 125, 1122, 10.1161/CIRCULATIONAHA.111.054346 Griendling, 2000, NAD(P)H oxidase: role in cardiovascular biology and disease, Circ Res, 86, 494, 10.1161/01.RES.86.5.494 García-Martín, 2021, EHP-101 alleviates angiotensin II–induced fibrosis and inflammation in mice, Biomed Pharmacother, 142, 10.1016/j.biopha.2021.112007 Kirkland, 2020, Senolytic drugs: from discovery to translation, J Intern Med, 288, 518, 10.1111/joim.13141 Ungvari, 2020, Mechanisms of vascular aging, a geroscience perspective: JACC focus seminar, J Am Coll Cardiol, 75, 931, 10.1016/j.jacc.2019.11.061 Rabinovitch, 2023, Are senolytic agents guilty of overkill or inappropriate age discrimination?, Circulation, 147, 667, 10.1161/CIRCULATIONAHA.122.060247 Kennedy, 2014, Geroscience: linking aging to chronic disease, Cell, 159, 709, 10.1016/j.cell.2014.10.039 Cai, 2022, Induction of accelerated aging in a mouse model, Cells, 11, 1418, 10.3390/cells11091418 Withaar, 2021, Heart failure with preserved ejection fraction in humans and mice: embracing clinical complexity in mouse models, Eur Heart J, 42, 4420, 10.1093/eurheartj/ehab389 Barros, 2021, Vascular aging in rodent models: contrasting mechanisms driving the female and male vascular senescence, Front Aging, 2, 10.3389/fragi.2021.727604 Newcomer, 2005, Heterogeneous vasodilator responses of human limbs: influence of age and habitual endurance training, Am J Physiol Heart Circ Physiol, 289, H308, 10.1152/ajpheart.01151.2004 Weeda, 1997, Disruption of mouse ERCC1 results in a novel repair syndrome with growth failure, nuclear abnormalities and senescence, Curr Biol, 7, 427, 10.1016/S0960-9822(06)00190-4 Vermeij, 2016, Genome integrity in aging: human syndromes, mouse models, and therapeutic options, Annu Rev Pharmacol Toxicol, 56, 427, 10.1146/annurev-pharmtox-010814-124316 Vermeij, 2016, Restricted diet delays accelerated ageing and genomic stress in DNA-repair–deficient mice, Nature, 537, 427, 10.1038/nature19329 Sims, 2023, DNA damage–induced stalling of transcription drives aging through gene expression imbalance, DNA Repair (Amst), 125, 10.1016/j.dnarep.2023.103483 Bautista-Niño, 2020, Local endothelial DNA repair deficiency causes aging-resembling endothelial-specific dysfunction, Clin Sci (Lond), 134, 727, 10.1042/CS20190124 de Boer, 2023, DNA repair in cardiomyocytes is critical for maintaining cardiac function in mice, Aging Cell, 22, 10.1111/acel.13768 Yousefzadeh, 2020, Tissue specificity of senescent cell accumulation during physiologic and accelerated aging of mice, Aging Cell, 19, 10.1111/acel.13094 Nakano-Kurimoto, 2009, Replicative senescence of vascular smooth muscle cells enhances the calcification through initiating the osteoblastic transition, Am J Physiol Heart Circ Physiol, 297, H1673, 10.1152/ajpheart.00455.2009 Liu, 2013, Prelamin A accelerates vascular calcification via activation of the DNA damage response and senescence-associated secretory phenotype in vascular smooth muscle cells, Circ Res, 112, e99, 10.1161/CIRCRESAHA.111.300543 Gomez-Sanchez, 2020, Vascular aging and its relationship with lifestyles and other risk factors in the general Spanish population: Early Vascular Ageing Study, J Hypertens, 38, 1110, 10.1097/HJH.0000000000002373 Lessiani, 2016, Arterial stiffness and sedentary lifestyle: role of oxidative stress, Vascul Pharmacol, 79, 1, 10.1016/j.vph.2015.05.017 Karimi, 2016, Effects of combined healthy lifestyle factors on functional vascular aging: the Rotterdam Study, J Hypertens, 34, 853, 10.1097/HJH.0000000000000861 Ghebre, 2016, Vascular aging: implications for cardiovascular disease and therapy, Transl Med (Sunnyvale), 6, 183, 10.4172/2161-1025.1000183 Rubio-Ruiz, 2014, Non-steroidal anti-inflammatory drugs attenuate the vascular responses in aging metabolic syndrome rats, Acta Pharmacol Sin, 35, 1364, 10.1038/aps.2014.67 Lesniewski, 2011, Salicylate treatment improves age-associated vascular endothelial dysfunction: potential role of nuclear factor κB and forkhead box O phosphorylation, J Gerontol A Biol Sci Med Sci, 66, 409, 10.1093/gerona/glq233 Pierce, 2009, Nuclear factor-κB activation contributes to vascular endothelial dysfunction via oxidative stress in overweight/obese middle-aged and older humans, Circulation, 119, 1284, 10.1161/CIRCULATIONAHA.108.804294 Nowak, 2017, IL-1 inhibition and vascular function in CKD, J Am Soc Nephrol, 28, 971, 10.1681/ASN.2016040453 Vallejo, 2014, The interleukin-1 receptor antagonist anakinra improves endothelial dysfunction in streptozotocin-induced diabetic rats, Cardiovasc Diabetol, 13, 158, 10.1186/s12933-014-0158-z Ikonomidis, 2008, Inhibition of interleukin-1 by anakinra improves vascular and left ventricular function in patients with rheumatoid arthritis, Circulation, 117, 2662, 10.1161/CIRCULATIONAHA.107.731877 Ikonomidis, 2019, Differential effects of inhibition of interleukin 1 and 6 on myocardial, coronary and vascular function, Clin Res Cardiol, 108, 1093, 10.1007/s00392-019-01443-9 Kume, 2011, Tocilizumab monotherapy reduces arterial stiffness as effectively as etanercept or adalimumab monotherapy in rheumatoid arthritis: an open-label randomized controlled trial, J Rheumatol, 38, 2169, 10.3899/jrheum.110340 Bosello, 2008, TNF-α blockade induces a reversible but transient effect on endothelial dysfunction in patients with long-standing severe rheumatoid arthritis, Clin Rheumatol, 27, 833, 10.1007/s10067-007-0803-y Lee, 2017, Interaction of IL-6 and TNF-α contributes to endothelial dysfunction in type 2 diabetic mouse hearts, PLoS One, 12 Elmazoglu, 2021, S-Allylcysteine inhibits chondrocyte inflammation to reduce human osteoarthritis via targeting RAGE, TLR4, JNK, and Nrf2 signaling: comparison with colchicine, Biochem Cell Biol, 99, 645, 10.1139/bcb-2021-0004 Kajikawa, 2019, Effect of short-term colchicine treatment on endothelial function in patients with coronary artery disease, Int J Cardiol, 281, 35, 10.1016/j.ijcard.2019.01.054 Toprover, 2020, Initiating guideline-concordant gout treatment improves arterial endothelial function and reduces intercritical inflammation: a prospective observational study, Arthritis Res Ther, 22, 169, 10.1186/s13075-020-02260-6 Nidorf, 2019, The effect of low-dose colchicine in patients with stable coronary artery disease: the LoDoCo2 trial rationale, design, and baseline characteristics, Am Heart J, 218, 46, 10.1016/j.ahj.2019.09.011 Tardif, 2019, Efficacy and safety of low-dose colchicine after myocardial infarction, N Engl J Med, 381, 2497, 10.1056/NEJMoa1912388 Cimmino, 2021, Colchicine inhibits the prothrombotic effects of oxLDL in human endothelial cells, Vascul Pharmacol, 137, 10.1016/j.vph.2020.106822 Zhang, 2022, Therapeutic potential of colchicine in cardiovascular medicine: a pharmacological review, Acta Pharmacol Sin, 43, 2173, 10.1038/s41401-021-00835-w Yilmaz, 2021, The efficacy of colchicine on carotid intima-media thickness: a prospective comparative study, J Stroke Cerebrovasc Dis, 30, 10.1016/j.jstrokecerebrovasdis.2020.105580 Ozalper, 2017, Evaluation of endothelial dysfunction in patients with familial Mediterranean fever: the relationship between the levels of asymmetric dimethylarginine and endocan with carotid intima-media thickness and endothelium-dependent vasodilation, Clin Rheumatol, 36, 2071, 10.1007/s10067-016-3532-2 Cameron, 2016, Anti-inflammatory effects of metformin irrespective of diabetes status, Circ Res, 119, 652, 10.1161/CIRCRESAHA.116.308445 Rena, 2017, The mechanisms of action of metformin, Diabetologia, 60, 1577, 10.1007/s00125-017-4342-z Man, 2019, The role of sirtuin1 in regulating endothelial function, arterial remodeling and vascular aging, Front Physiol, 10, 1173, 10.3389/fphys.2019.01173 Mengozzi, 2022, Targeting SIRT1 rescues age- and obesity-induced microvascular dysfunction in ex vivo human vessels, Circ Res, 131, 476, 10.1161/CIRCRESAHA.122.320888 de Picciotto, 2016, Nicotinamide mononucleotide supplementation reverses vascular dysfunction and oxidative stress with aging in mice, Aging Cell, 15, 522, 10.1111/acel.12461 Kiss, 2020, Nicotinamide mononucleotide (NMN) supplementation promotes neurovascular rejuvenation in aged mice: transcriptional footprint of SIRT1 activation, mitochondrial protection, anti-inflammatory, and anti-apoptotic effects, Geroscience, 42, 527, 10.1007/s11357-020-00165-5 Tarantini, 2019, Treatment with the poly(ADP-ribose) polymerase inhibitor PJ-34 improves cerebromicrovascular endothelial function, neurovascular coupling responses and cognitive performance in aged mice, supporting the NAD+ depletion hypothesis of neurovascular aging, Geroscience, 41, 533, 10.1007/s11357-019-00101-2 Abdellatif, 2021, Nicotinamide for the treatment of heart failure with preserved ejection fraction, Sci Transl Med, 13, 10.1126/scitranslmed.abd7064 Martens, 2018, Chronic nicotinamide riboside supplementation is well-tolerated and elevates NAD+ in healthy middle-aged and older adults, Nat Commun, 9, 1286, 10.1038/s41467-018-03421-7 Katayoshi, 2023, Nicotinamide adenine dinucleotide metabolism and arterial stiffness after long-term nicotinamide mononucleotide supplementation: a randomized, double-blind, placebo-controlled trial, Sci Rep, 13, 2786, 10.1038/s41598-023-29787-3 Freeberg, 2022, Nicotinamide riboside supplementation for treating elevated systolic blood pressure and arterial stiffness in midlife and older adults, Front Cardiovasc Med, 9, 10.3389/fcvm.2022.881703 Gasek, 2021, Strategies for targeting senescent cells in human disease, Nat Aging, 1, 870, 10.1038/s43587-021-00121-8 Zhang, 2023, Targeting cellular senescence with senotherapeutics: senolytics and senomorphics, FEBS J, 290, 1362, 10.1111/febs.16350 Hartman, 2020, BCL-w: apoptotic and non-apoptotic role in health and disease, Cell Death Dis, 11, 260, 10.1038/s41419-020-2417-0 Robbins, 2021, Senolytic drugs: reducing senescent cell viability to extend health span, Annu Rev Pharmacol Toxicol, 61, 779, 10.1146/annurev-pharmtox-050120-105018 Mijit, 2020, Role of p53 in the regulation of cellular senescence, Biomolecules, 10, 10.3390/biom10030420 Huang, 2021, Senolytic peptide FOXO4-DRI selectively removes senescent cells from in vitro expanded human chondrocytes, Front Bioeng Biotechnol, 9, 10.3389/fbioe.2021.677576 Malaise, 2017, Senolytic treatments applied to osteoarthritis: a step towards the end of orthopedic surgery, AME Medical Journal, 2, 161, 10.21037/amj.2017.10.01 An, 2017, USP7 inhibitor P5091 inhibits Wnt signaling and colorectal tumor growth, Biochem Pharmacol, 131, 29, 10.1016/j.bcp.2017.02.011 Zhang, 2022, Cellular senescence: a key therapeutic target in aging and diseases, J Clin Invest, 132 Baar, 2017, Targeted apoptosis of senescent cells restores tissue homeostasis in response to chemotoxicity and aging, Cell, 169, 132, 10.1016/j.cell.2017.02.031 Demaria, 2014, An essential role for senescent cells in optimal wound healing through secretion of PDGF-AA, Dev Cell, 31, 722, 10.1016/j.devcel.2014.11.012 Yosef, 2016, Directed elimination of senescent cells by inhibition of BCL-W and BCL-XL, Nat Commun, 7, 10.1038/ncomms11190 Böger, 2004, Asymmetric dimethylarginine, an endogenous inhibitor of nitric oxide synthase, explains the “l-arginine paradox” and acts as a novel cardiovascular risk factor, J Nutr, 134, 2842S Bode-Böger, 2005, Asymmetric dimethylarginine (ADMA) accelerates cell senescence, Vasc Med, 10, S65, 10.1177/1358836X0501000110 Scalera, 2010, Nitric oxide–asymmetric dimethylarginine system in endothelial cell senescence, 483 Cimmino, 2023, Colchicine in athero-thrombosis: molecular mechanisms and clinical evidence, Int J Mol Sci, 24 Zheng, 2015, Atorvastatin protects endothelium by decreasing asymmetric dimethylarginine in dyslipidemia rats, Lipids Health Dis, 14, 41, 10.1186/s12944-015-0041-2 Chaib, 2022, Cellular senescence and senolytics: the path to the clinic, Nat Med, 28, 1556, 10.1038/s41591-022-01923-y Cai, 2022, Celecoxib, beyond anti-inflammation, alleviates tendon-derived stem cell senescence in degenerative rotator cuff tendinopathy, Am J Sports Med, 50, 2488, 10.1177/03635465221098133 Ota, 2010, Induction of endothelial nitric oxide synthase, SIRT1, and catalase by statins inhibits endothelial senescence through the Akt pathway, Arterioscler Thromb Vasc Biol, 30, 2205, 10.1161/ATVBAHA.110.210500 Liu, 2015, Simvastatin suppresses breast cancer cell proliferation induced by senescent cells, Sci Rep, 5, 10.1038/srep17895 Moiseeva, 2013, Metformin inhibits the senescence-associated secretory phenotype by interfering with IKK/NF-κB activation, Aging Cell, 12, 489, 10.1111/acel.12075 Avogaro, 2013, The endothelium abridges insulin resistance to premature aging, J Am Heart Assoc, 2 Tilstra, 2012, NF-κB inhibition delays DNA damage–induced senescence and aging in mice, J Clin Invest, 122, 2601, 10.1172/JCI45785 Childs, 2016, Senescent intimal foam cells are deleterious at all stages of atherosclerosis, Science, 354, 472, 10.1126/science.aaf6659 Roos, 2016, Chronic senolytic treatment alleviates established vasomotor dysfunction in aged or atherosclerotic mice, Aging Cell, 15, 973, 10.1111/acel.12458 Raffaele, 2022, The costs and benefits of senotherapeutics for human health, Lancet Healthy Longev, 3, e67, 10.1016/S2666-7568(21)00300-7 An, 2020, Inhibition of 3-phosphoinositide–dependent protein kinase 1 (PDK1) can revert cellular senescence in human dermal fibroblasts, Proc Natl Acad Sci U S A, 117, 31535, 10.1073/pnas.1920338117 Golshiri, 2020, The importance of the nitric oxide–cGMP pathway in age-related cardiovascular disease: focus on phosphodiesterase-1 and soluble guanylate cyclase, Basic Clin Pharmacol Toxicol, 127, 67, 10.1111/bcpt.13319 Golshiri, 2020, Chronic sildenafil treatment improves vasomotor function in a mouse model of accelerated aging, Int J Mol Sci, 21, 4667, 10.3390/ijms21134667 Golshiri, 2021, Selective phosphodiesterase 1 inhibition ameliorates vascular function, reduces inflammatory response, and lowers blood pressure in aging animals, J Pharmacol Exp Ther, 378, 173, 10.1124/jpet.121.000628 Golshiri, 2021, The effects of acute and chronic selective phosphodiesterase 1 inhibition on smooth muscle cell–associated aging features, Front Pharmacol, 12 Ataei Ataabadi, 2022, Soluble guanylate cyclase activator BAY 54-6544 improves vasomotor function and survival in an accelerated ageing mouse model, Aging Cell, 21, 10.1111/acel.13683 Hirano, 2015, Discovery of GSK2795039, a novel small molecule NADPH oxidase 2 inhibitor, Antioxid Redox Signal, 23, 358, 10.1089/ars.2014.6202 Snell, 2016, Repurposing FDA-approved drugs for anti-aging therapies, Biogerontology, 17, 907, 10.1007/s10522-016-9660-x Born, 2023, Eliminating senescent cells can promote pulmonary hypertension development and progression, Circulation, 147, 650, 10.1161/CIRCULATIONAHA.122.058794 Nidorf, 2020, Colchicine in patients with chronic coronary disease, N Engl J Med, 383, 1838, 10.1056/NEJMoa2021372