A framework of biomarkers for vascular aging: a consensus statement by the Aging Biomarker Consortium

Aging Biomarker Consortium; Le Zhang, Jun Guo, Yuehong Liu, Shimin Sun, Baohua Liu, Qi Yang, Jun Tao, Xiao-Li Tian, Jun Pu, Huashan Hong, Miao Wang, Hou-Zao Chen, Jie Ren, Xiaoming Wang, Zhen Liang, Yuan Wang, Kai Huang, Weiqi Zhang, Jing Qu, Zhenyu Ju, Guang-Hui Liu, Gang Pei, Jian Li, Cuntai Zhang

PDF(505 KB)
PDF(505 KB)
Life Medicine ›› 2023, Vol. 2 ›› Issue (4) : 3. DOI: 10.1093/lifemedi/lnad033
Forum

A framework of biomarkers for vascular aging: a consensus statement by the Aging Biomarker Consortium

Author information +
History +

Abstract

Aging of the vasculature, which is integral to the functioning of literally all human organs, serves as a fundamental physiological basis for age-related alterations as well as a shared etiological mechanism for various chronic diseases prevalent in the elderly population. China, home to the world’s largest aging population, faces an escalating challenge in addressing the prevention and management of these age-related conditions. To meet this challenge, the Aging Biomarker Consortium of China has developed an expert consensus on biomarkers of vascular aging (VA) by synthesizing literature and insights from scientists and clinicians. This consensus provides a comprehensive assessment of biomarkers associated with VA and presents a systemic framework to classify them into three dimensions: functional, structural, and humoral. Within each dimension, the expert panel recommends the most clinically relevant VA biomarkers. For the functional domain, biomarkers reflecting vascular stiffness and endothelial function are high-lighted. The structural dimension encompasses metrics for vascular structure, microvascular structure, and distribution. Additionally, proinflammatory factors are emphasized as biomarkers with the humoral dimension. The aim of this expert consensus is to establish a foundation for assessing the extent of VA and conducting research related to VA, with the ultimate goal of improving the vascular health of the elderly in China and globally.

Cite this article

Download citation ▾
Aging Biomarker Consortium; Le Zhang, Jun Guo, Yuehong Liu, Shimin Sun, Baohua Liu, Qi Yang, Jun Tao, Xiao-Li Tian, Jun Pu, Huashan Hong, Miao Wang, Hou-Zao Chen, Jie Ren, Xiaoming Wang, Zhen Liang, Yuan Wang, Kai Huang, Weiqi Zhang, Jing Qu, Zhenyu Ju, Guang-Hui Liu, Gang Pei, Jian Li, Cuntai Zhang. A framework of biomarkers for vascular aging: a consensus statement by the Aging Biomarker Consortium. Life Medicine, 2023, 2(4): 3 https://doi.org/10.1093/lifemedi/lnad033

References

[1]
Augustin HG, Koh GY. Organotypic vasculature: from descriptive heterogeneity to functional pathophysiology. Science 2017;357:eaal2379.
CrossRef Google scholar
[2]
Zhang C, Tao J; Cardiovascular Group, Society of Geriatrics, Chinese Medical Association. Expert consensus on clinical assessment and intervention of vascular aging in China (2018). Aging Med 2018;1:228–37.
CrossRef Google scholar
[3]
Ungvari Z, Tarantini S, Sorond F, et al. Mechanisms of vascular aging, a geroscience perspective: JACC focus seminar. J Am Coll Cardiol 2020;75:931–41.
CrossRef Google scholar
[4]
Bao H, Cao J, Chen M, et al; Aging Biomarker Consortium. Biomarkers of aging. Sci China Life Sci 2023;66:893–1066.
CrossRef Google scholar
[5]
National Bureau of Statistics of China. Main Data of the Seventh National Population Census. 2021.
[6]
Cai Y, Song W, Li J, et al. The landscape of aging. Sci China Life Sci 2022;65:2354–454.
CrossRef Google scholar
[7]
Aging Biomarker Consortium, Jia Y-J, Wang J et al. A framework of biomarkers for brain aging: a consensus statement by the Aging Biomarker Consortium. Life Med 2023;2.
CrossRef Google scholar
[8]
Ren J, Song M, Zhang W, et al. The Aging Biomarker Consortium represents a new era for aging research in China. Nat Med 2023;29:2162–65.
CrossRef Google scholar
[9]
Sousa-Uva M, Head SJ, Thielmann M, et al. Methodology manual for European Association for Cardio-Thoracic Surgery (EACTS) clinical guidelines. Eur J Cardio-thoracic Surg: Off J Eur Assoc Cardiothoracic Surg 2015;48:809–16.
CrossRef Google scholar
[10]
Zhang W, Zhang S, Yan P, et al. A single-cell transcriptomic landscape of primate arterial aging. Nat Commun 2020;11:2202.
CrossRef Google scholar
[11]
Ma S, Chi X, Cai Y, et al. Decoding aging hallmarks at the single-cell Level. Ann Rev Biomed Data Sci 2023;6:129–52.
CrossRef Google scholar
[12]
Van Bortel LM, Laurent S, Boutouyrie P, et al.; Artery Society. Expert consensus document on the measurement of aortic stiffness in daily practice using carotid-femoral pulse wave velocity. J Hypertens 2012;30:445–8.
CrossRef Google scholar
[13]
Townsend RR, Wilkinson IB, Schiffrin EL, et al.; American Heart Association Council on Hypertension. Recommendations for improving and standardizing vascular research on arterial stiffness: a scientific statement from the American Heart Association. Hypertension 2015;66:698–722.
CrossRef Google scholar
[14]
Vaitkevicius PV, Fleg JL, Engel JH, et al. Effects of age and aerobic capacity on arterial stiffness in healthy adults. Circulation 1993;88:1456–62.
CrossRef Google scholar
[15]
Avolio AP, Deng FQ, Li WQ, et al. 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;71:202–10.
CrossRef Google scholar
[16]
Avolio AP, Kuznetsova T, Heyndrickx GR, et al. Arterial flow, pulse pressure and pulse wave velocity in men and women at various ages. Adv Exp Med Biol 2018;1065:153–68.
CrossRef Google scholar
[17]
Kim OY, Paik JK, Lee JY, et al. Follow-ups of metabolic, inflammatory and oxidative stress markers, and brachial-ankle pulse wave velocity in middle-aged subjects without metabolic syndrome. Clin Exp Hypertens 2013;35:382–8.
CrossRef Google scholar
[18]
Ishida A, Fujisawa M, Del Saz EG, et al. Arterial stiffness, not systolic blood pressure, increases with age in native Papuan populations. Hypertens Res: Off J Jpn Soc Hypertens 2018;41:539–46.
CrossRef Google scholar
[19]
Mitchell GF, Hwang S-J, Vasan RS, et al. Arterial stiffness and cardiovascular events: the Framingham Heart Study. Circulation 2010;121:505–11.
CrossRef Google scholar
[20]
Laurent S, Boutouyrie P, Asmar R, et al. Aortic stiffness is an independent predictor of all-cause and cardiovascular mortality in hypertensive patients. Hypertension 2001;37:1236–41.
CrossRef Google scholar
[21]
Voicehovska JG, Bormane E, Grigane A, et al. Association of arterial stiffness with chronic kidney disease progression and mortality. Heart Lung Circulat 2021;30:1694–701
CrossRef Google scholar
[22]
Pandey A, Khan H, Newman AB, et al. Arterial stiffness and risk of overall heart failure, heart failure with preserved ejection fraction, and heart failure with reduced ejection fraction: the health ABC study (health, aging, and body composition). Hypertension 2017;69:267–74.
CrossRef Google scholar
[23]
Vlachopoulos C, Aznaouridis K, Stefanadis C. Prediction of cardiovascular events and all-cause mortality with arterial stiffness: a systematic review and meta-analysis. J Am Coll Cardiol 2010;55:1318–27.
CrossRef Google scholar
[24]
Poulter NR, Prabhakaran D, Caulfield M. Hypertension. Lancet 2015;386:801–12.
CrossRef Google scholar
[25]
Humphrey JD, Harrison DG, Figueroa CA, et al. Central artery stiffness in hypertension and aging: a problem with cause and consequence. Circ Res 2016;118:379–81.
CrossRef Google scholar
[26]
Bianchini E, Lonnebakken MT, Wohlfahrt P, et al. Magnetic resonance imaging and computed tomography for the noninvasive assessment of arterial aging: a review by the VascAgeNet COST action. J Am Heart Assoc 2023;12:e027414.
CrossRef Google scholar
[27]
Azarine A, Garçon P, Stansal A, et al. Four-dimensional flow MRI: principles and cardiovascular applications. Radiographics: Rev Publ Radiol Soc North America, Inc 2019;39:632–48.
CrossRef Google scholar
[28]
Dyverfeldt P, Bissell M, Barker AJ, et al. 4D flow cardiovascular magnetic resonance consensus statement. J Cardiovasc Magnet Reson: Off J Soc Cardiovasc Magnet Reson 2015;17:72.
CrossRef Google scholar
[29]
Bissell MM, Raimondi F, Ait Ali L, et al. 4D Flow cardiovascular magnetic resonance consensus statement: 2023 update. J Cardiovasc Magnet Reson: Off J Soc Cardiovasc Magnet Reson 2023;25:40.
CrossRef Google scholar
[30]
Ohyama Y, Ambale-Venkatesh B, Noda C, et al. Aortic arch pulse wave velocity assessed by magnetic resonance imaging as a predictor of incident cardiovascular events: the MESA (Multi-Ethnic Study of Atherosclerosis). Hypertension 2017;70:524–30.
CrossRef Google scholar
[31]
Dyverfeldt P, Ebbers T, Lanne T. Pulse wave velocity with 4D flow MRI: systematic differences and age-related regional vascular stiffness. Magn Reson Imaging 2014;32:1266–71.
CrossRef Google scholar
[32]
Hickson SS, Butlin M, Graves M, et al. The relationship of age with regional aortic stiffness and diameter. JACC Cardiovasc Imaging 2010;3:1247–55.
CrossRef Google scholar
[33]
Saiki A, Ohira M, Yamaguchi T, et al. New horizons of arterial stiffness developed using Cardio-Ankle Cascular Index (CAVI). J Atheroscler Thromb 2020;27:732–48.
CrossRef Google scholar
[34]
Saiki A, Sato Y, Watanabe R, et al. The role of a novel arterial stiffness parameter, Cardio-Ankle Vascular Index (CAVI), as a surrogate marker for cardiovascular diseases. J Atheroscler Thromb 2016;23:155–68.
CrossRef Google scholar
[35]
Park SY, Chin SO, Rhee SY, et al. Cardio-ankle vascular index as a surrogate marker of early atherosclerotic cardiovascular disease in Koreans with type 2 diabetes mellitus. Diabetes Metab J 2018;42:285–95.
CrossRef Google scholar
[36]
McDermott MM, Criqui MH. Ankle-brachial index screening and improving peripheral artery disease detection and outcomes. JAMA 2018;320:143–5.
CrossRef Google scholar
[37]
Curry SJ, Krist AH, Owens DK, et al.; US Preventive Services Task Force. Screening for peripheral artery disease and cardiovascular disease risk assessment with the ankle-brachial index: US preventive services task force recommendation statement. JAMA 2018;320:177–83.
CrossRef Google scholar
[38]
Fowkes FGR, Murray GD, Butcher I, et al.; Ankle Brachial Index Collaboration. Ankle brachial index combined with Framingham Risk Score to predict cardiovascular events and mortality: a metaanalysis. JAMA 2008;300:197–208.
CrossRef Google scholar
[39]
Perlstein TS, Creager MA. The ankle-brachial index as a biomarker of cardiovascular risk: it’s not just about the legs. Circulation 2009;120:2033–5.
CrossRef Google scholar
[40]
Manzano L, Mostaza JM, Suarez C, et al.; Merito II Study Group. Prognostic value of the ankle-brachial index in elderly patients with a stable chronic cardiovascular event. J Thrombosis Haemostasis: JTH 2010;8:1176–84.
CrossRef Google scholar
[41]
Qian Z, Zhang Y, Yang N, et al. Close association between lifestyle and circulating FGF21 levels: a systematic review and meta-analysis. Front Endocrinol 2022;13:984828.
CrossRef Google scholar
[42]
Matsushima H, Hosomi N, Hara N, et al. Ability of the Ankle Brachial Index and brachial-ankle pulse wave velocity to predict the 3-month outcome in patients with non-cardioembolic stroke. J Atheroscler Thromb 2017;24:1167–73.
CrossRef Google scholar
[43]
Banerjee D, Plange-Rhule J, Chitalia N, et al. Pulse pressure relationships with demographics and kidney function in Ashanti, Ghana. Int J Hypertens 2018;2018:7864564.
CrossRef Google scholar
[44]
Tang KS, Medeiros ED, Shah AD. Wide pulse pressure: a clinical review. J Clin Hypertens 2020;22:1960–7.
CrossRef Google scholar
[45]
Franklin SS, Wong ND. Pulse pressure: how valuable as a diagnostic and therapeutic tool? J Am Coll Cardiol 2016;67:404–6.
CrossRef Google scholar
[46]
Franklin SS, Khan SA, Wong ND, et al. Is pulse pressure useful in predicting risk for coronary heart Disease? The Framingham heart study. Circulation 1999;100:354–60.
CrossRef Google scholar
[47]
Haider AW, Larson MG, Franklin SS, et al.; Framingham Heart Study. 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;138:10–6.
CrossRef Google scholar
[48]
Greve SV, Blicher MK, Kruger R, et al. Estimated carotid-femoral pulse wave velocity has similar predictive value as measured carotidfemoral pulse wave velocity. J Hypertens 2016;34:1279–89.
CrossRef Google scholar
[49]
Vlachopoulos C, Terentes-Printzios D, Laurent S, et al. Association of estimated pulse wave velocity with survival: a secondary analysis of SPRINT. JAMA Network Open 2019;2:e1912831.
CrossRef Google scholar
[50]
Pan F-S, Yu L, Luo J, et al. Carotid artery stiffness assessment by ultrafast ultrasound imaging: feasibility and potential influencing factors. J Ultrasound Med: Off J Am Instit Ultrasound Med 2018;37:2759–67.
CrossRef Google scholar
[51]
Yin L-X, Ma C-Y, Wang S, et al.; Study Investigators. Reference values of carotid ultrafast pulse-wave velocity: a prospective, multicenter, population-based study. J Am Soc Echocardiogr: Off Publ Am Soc Echocardiogr 2021;34:629–41
CrossRef Google scholar
[52]
Rasouli R, Baranger J, Slorach C, et al. Local arterial stiffness assessment: comparison of pulse wave velocity assessed by ultrafast ultrasound imaging versus the Bramwell–Hill equation. J Am Soc Echocardiogr: Off Publ Am Soc Echocardiogr 2022;35:1185–8.
CrossRef Google scholar
[53]
Zhu Z-Q, Chen L-S, Wang H, et al. Carotid stiffness and atherosclerotic risk: non-invasive quantification with ultrafast ultrasound pulse wave velocity. Eur Radiol 2019;29:1507–17.
CrossRef Google scholar
[54]
Mirault T, Pernot M, Frank M, et al. Carotid stiffness change over the cardiac cycle by ultrafast ultrasound imaging in healthy volunteers and vascular Ehlers-Danlos syndrome. J Hypertens 2015;33:1890–6; discussion 1896.
CrossRef Google scholar
[55]
Laurent S, Cockcroft J, Van Bortel L, et al.; European Network for Non-invasive Investigation of Large Arteries. Expert consensus document on arterial stiffness: methodological issues and clinical applications. Eur Heart J 2006;27:2588–605.
CrossRef Google scholar
[56]
Ohyama Y, Redheuil A, Kachenoura N, et al. Imaging insights on the Aorta in aging. Circ Cardiovasc Imaging 2018;11:e005617.
CrossRef Google scholar
[57]
Redheuil A, Yu WC, Wu CO, et al. Reduced ascending aortic strain and distensibility: earliest manifestations of vascular aging in humans. Hypertension 2010;55:319–26.
CrossRef Google scholar
[58]
Redheuil A, Wu CO, Kachenoura N, et al. Proximal aortic distensibility is an independent predictor of all-cause mortality and incident CV events: the MESA study. J Am Coll Cardiol 2014;64:2619–29.
CrossRef Google scholar
[59]
Tian X-L, Yang Li, Li Y. Endothelial cell senescence and age-related vascular diseases. J Genet Genomics 2014;41:485–95.
CrossRef Google scholar
[60]
Ungvari Z, Tarantini S, Kiss T, et al. Endothelial dysfunction and angiogenesis impairment in the ageing vasculature. Nat Rev Cardiol 2018;15:555–65.
CrossRef Google scholar
[61]
Zhu L, Xu C, Huo X, et al. The cyclooxygenase-1/mPGES-1/endothelial prostaglandin EP4 receptor pathway constrains myocardial ischemia-reperfusion injury. Nat Commun 2019;10:1888.
CrossRef Google scholar
[62]
Zhu L, Zhang Y, Guo Z, et al. Cardiovascular biology of prostanoids and drug discovery. Arterioscler Thromb Vasc Biol 2020;40:1454–63.
CrossRef Google scholar
[63]
Zhang Y, Steinmetz-Späh J, Idborg H, et al. Microsomal prostaglandin E synthase-1 inhibition prevents adverse cardiac remodelling after myocardial infarction in mice. Br J Pharmacol 2023;180:1981–98.
CrossRef Google scholar
[64]
Libby P, Buring JE, Badimon L, et al. Atherosclerosis. Nat Rev Dis Primers 2019;5:56.
CrossRef Google scholar
[65]
Bloom SI, Islam MT, Lesniewski LA, et al. Mechanisms and consequences of endothelial cell senescence. Nat Rev Cardiol 2023;20:38–51.
CrossRef Google scholar
[66]
Thijssen DHJ, Bruno RM, van Mil ACCM, et al. Expert consensus and evidence-based recommendations for the assessment of flow-mediated dilation in humans. Eur Heart J 2019;40:2534–47.
CrossRef Google scholar
[67]
Celermajer DS, Sorensen KE, Gooch VM, et al. Non-invasive detection of endothelial dysfunction in children and adults at risk of atherosclerosis. Lancet 1992;340:1111–5.
CrossRef Google scholar
[68]
Skaug E-A, Aspenes ST, Oldervoll L, et al. Age and gender differences of endothelial function in 4739 healthy adults: the HUNT3 Fitness Study. Eur J Prevent Cardiol 2013;20:531–40.
CrossRef Google scholar
[69]
Matsuzawa Y, Kwon T-G, Lennon RJ, 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:e002270.
CrossRef Google scholar
[70]
Kuvin JT, Patel AR, Sliney KA, et al. Assessment of peripheral vascular endothelial function with finger arterial pulse wave amplitude. Am Heart J 2003;146:168–74.
CrossRef Google scholar
[71]
Norimatsu K, Gondo K, Kusumoto T, et al. Association between lipid profile and endothelial dysfunction as assessed by the reactive hyperemia index. Clin Exp Hypertens 2021;43:125–30.
CrossRef Google scholar
[72]
Heffernan KS, Patvardhan EA, Kapur NK, et al. Peripheral augmentation index as a biomarker of vascular aging: an invasive hemodynamics approach. Eur J Appl Physiol 2012;112:2871–9.
CrossRef Google scholar
[73]
Bonetti PO, Pumper GM, Higano ST, et al. Noninvasive identification of patients with early coronary atherosclerosis by assessment of digital reactive hyperemia. J Am Coll Cardiol 2004;44:2137–41.
CrossRef Google scholar
[74]
Babcock MC, DuBose LE, Witten TL, et al. Assessment of macrovascular and microvascular function in aging males. J Appl Physiol 2021;130:96–103.
CrossRef Google scholar
[75]
Hamburg NM, Palmisano J, Larson MG, et al. Relation of brachial and digital measures of vascular function in the community: the Framingham heart study. Hypertension 2011;57:390–6.
CrossRef Google scholar
[76]
Heidari B, Lerman A, Lalia AZ, et al. Effect of metformin on microvascular endothelial function in polycystic ovary syndrome. Mayo Clin Proc 2019;94:2455–66.
CrossRef Google scholar
[77]
Reeson P, Choi K, Brown CE. VEGF signaling regulates the fate of obstructed capillaries in mouse cortex. Elife 2018;7:e33670.
CrossRef Google scholar
[78]
Alexandrou ME, Gkaliagkousi E, Loutradis C, et al. Haemodialysis and peritoneal dialysis patients have severely impaired postocclusive skin forearm vasodilatory response assessed with laser speckle contrast imaging. Clin Kidney J 2021;14:1419–27.
CrossRef Google scholar
[79]
Luo J, Yan Z, Guo S, et al. Recent advances in atherosclerotic disease screening using pervasive healthcare. IEEE Rev Biomed Eng 2022;15:293–308.
CrossRef Google scholar
[80]
Tao J, Jin Y-f, Yang Z, et al. Reduced arterial elasticity is associated with endothelial dysfunction in persons of advancing age: comparative study of noninvasive pulse wave analysis and laser Doppler blood flow measurement. Am J Hypertens 2004;17:654–9.
CrossRef Google scholar
[81]
Kotecha T, Martinez-Naharro A, Boldrini M, et al. Automated pixel-wise quantitative myocardial perfusion mapping by CMR to detect obstructive coronary artery disease and coronary microvascular dysfunction: validation against invasive coronary physiology. JACC Cardiovasc Imaging 2019;12:1958–69.
CrossRef Google scholar
[82]
Castle-Kirszbaum M, Parkin WG, Goldschlager T, et al. Cardiac output and cerebral blood flow: a systematic review of cardio-cerebral coupling. J Neurosurg Anesthesiol 2022;34:352–63.
CrossRef Google scholar
[83]
Roberts GS, Peret A, Jonaitis EM, et al. Normative cerebral hemodynamics in middle-aged and older adults using 4d flow MRI: initial analysis of vascular aging. Radiology 2023;307:e222685.
CrossRef Google scholar
[84]
Chang TY, Liu HL, Lee TH, et al. Change in cerebral perfusion after carotid angioplasty with stenting is related to cerebral vasoreactivity: a study using dynamic susceptibility-weighted contrast-enhanced MR imaging and functional MR imaging with a breath-holding paradigm. AJNR Am J Neuroradiol 2009;30:1330–6.
CrossRef Google scholar
[85]
Haller S, Zaharchuk G, Thomas DL, et al. Arterial spin labeling perfusion of the brain: emerging clinical applications. Radiology 2016;281:337–56.
CrossRef Google scholar
[86]
Li Y, Ying Y, Yao T, et al. Decreased water exchange rate across blood-brain barrier in hereditary cerebral small vessel disease. Brain 2023;146:3079–87.
CrossRef Google scholar
[87]
Kaczmarz S, Gottler J, Zimmer C, et al. Characterizing white matter fiber orientation effects on multi-parametric quantitative BOLD assessment of oxygen extraction fraction. J Cereb Blood Flow Metab 2020;40:760–74.
CrossRef Google scholar
[88]
Uwano I, Kudo K, Sato R, et al. Noninvasive assessment of oxygen extraction fraction in chronic ischemia using quantitative susceptibility mapping at 7 Tesla. Stroke 2017;48:2136–41.
CrossRef Google scholar
[89]
Stein JH, Korcarz CE, Hurst RT, et al. Use of carotid ultrasound to identify subclinical vascular disease and evaluate cardiovascular disease risk: a consensus statement from the American Society of Echocardiography Carotid Intima-Media Thickness Task Force. Endorsed by the Society for Vascular Medicine. J Am Soc Echocardiogr: Off Publ Am Soc Echocardiogr 2008;21:93–111.
CrossRef Google scholar
[90]
Homma S, Hirose N, Ishida H, et al. Carotid plaque and intima-media thickness assessed by b-mode ultrasonography in subjects ranging from young adults to centenarians. Stroke 2001;32:830–5.
CrossRef Google scholar
[91]
Polak JF, Pencina MJ, Pencina KM, et al. Carotid-wall intima-media thickness and cardiovascular events. N Engl J Med 2011;365:213–21.
CrossRef Google scholar
[92]
Zhao XX, Liu J, Zhao H, et al. The effect of cardiovascular risk factors on the carotid intima-media thickness in an old-aged cohort with hypertension: a longitudinal evolution with 4-year follow-up of a random clinical trial. Clini Exp Hypertens 2019;41:49–57.
CrossRef Google scholar
[93]
Huang Q, Liu Z, Wei M, et al. The atherogenic index of plasma and carotid atherosclerosis in a community population: a populationbased cohort study in China. Cardiovasc Diabetol 2023;22:125.
CrossRef Google scholar
[94]
Petrie JR, Chaturvedi N, Ford I, et al; REMOVAL Study Group. Cardiovascular and metabolic effects of metformin in patients with type 1 diabetes (REMOVAL): a double-blind, randomised, placebocontrolled trial. Lancet. Diabet Endocrinol 2017;5:597–609.
[95]
Blaha MJ, DeFilippis AP. Multi-Ethnic Study of Atherosclerosis (MESA): JACC Focus Seminar 5/8. J Am Coll Cardiol 2021;77:3195–216.
CrossRef Google scholar
[96]
Kong Q, Zhang Z, Yang Q, et al. 7T TOF-MRA shows modulated orifices of lenticulostriate arteries associated with atherosclerotic plaques in patients with lacunar infarcts. Eur J Radiol 2019;118:271–6.
CrossRef Google scholar
[97]
Zhang Z, Fan Z, Kong Q, et al. Visualization of the lenticulostriate arteries at 3T using black-blood T1-weighted intracranial vessel wall imaging: comparison with 7T TOF-MRA. Eur Radiol 2019;29:1452–9.
CrossRef Google scholar
[98]
Liu CY, Chen D, Bluemke DA, et al. Evolution of aortic wall thickness and stiffness with atherosclerosis: long-term follow up from the multi-ethnic study of atherosclerosis. Hypertension 2015;65:1015–9.
CrossRef Google scholar
[99]
Song JW, Pavlou A, Xiao J, et al. Vessel wall magnetic resonance imaging biomarkers of symptomatic intracranial atherosclerosis: a meta-analysis. Stroke 2021;52:193–202.
CrossRef Google scholar
[100]
Yang Q, Deng Z, Bi X, et al. Whole-brain vessel wall MRI: a parameter tune-up solution to improve the scan efficiency of three-dimensional variable flip-angle turbo spin-echo. J Magn Reson Imaging 2017;46:751–7.
CrossRef Google scholar
[101]
Wu F, Song H, Ma Q, et al; WISP Investigators. Hyperintense plaque on intracranial vessel wall magnetic resonance imaging as a predictor of artery-to-artery embolic infarction. Stroke 2018;49:905–11.
CrossRef Google scholar
[102]
Li Y, Choi WJ, Wei W, et al. Aging-associated changes in cerebral vasculature and blood flow as determined by quantitative optical coherence tomography angiography. Neurobiol Aging 2018;70:148–59.
CrossRef Google scholar
[103]
Lanzer P, Hannan FM, Lanzer JD, et al. Medial arterial calcification: JACC state-of-the-art review. J Am Coll Cardiol 2021;78:1145–65.
CrossRef Google scholar
[104]
Otsuka F, Sakakura K, Yahagi K, et al. Has our understanding of calcification in human coronary atherosclerosis progressed? Arterioscler Thromb Vasc Biol 2014;34:724–36.
CrossRef Google scholar
[105]
Lakatta EG, Levy D. Arterial and cardiac aging: major shareholders in cardiovascular disease enterprises: Part II: the aging heart in health: links to heart disease. Circulation 2003;107:346–54.
CrossRef Google scholar
[106]
Laclaustra M, Casasnovas JA, Fernández-Ortiz A, et al. Femoral and carotid subclinical atherosclerosis association with risk factors and coronary calcium: the AWHS study. J Am Coll Cardiol 2016;67:1263–74.
CrossRef Google scholar
[107]
Golub IS, Termeie OG, Kristo S, et al. Major global coronary artery calcium guidelines. JACC Cardiovasc Imaging 2023;16:98–117.
CrossRef Google scholar
[108]
Miname MH, Bittencourt MS, Pereira AC, et al. Vascular age derived from coronary artery calcium score on the risk stratification of individuals with heterozygous familial hypercholesterolaemia. Eur Heart J Cardiovasc Imaging 2020;21:251–7.
CrossRef Google scholar
[109]
Megale A, Wolosker N, Kalil V, et al. Calcium score predicts mortality after revascularization in critical limb ischemia. J Endovasc Ther 2022;29:438–43.
CrossRef Google scholar
[110]
Wang C, Zhang Y, Du J, et al. Quantitative susceptibility mapping for characterization of intraplaque hemorrhage and calcification in carotid atherosclerotic disease. J Magn Reson Imaging 2020;52:534–41.
CrossRef Google scholar
[111]
Wang Y, Osborne MT, Tung B, et al. Imaging cardiovascular calcification. J Am Heart Assoc 2018;7:e008564.
CrossRef Google scholar
[112]
Wang J, Börnert P, Zhao H, et al. Simultaneous noncontrast angiography and intraplaque hemorrhage (SNAP) imaging for carotid atherosclerotic disease evaluation. Magn Reson Med 2013;69:337–45.
CrossRef Google scholar
[113]
Li D, Qiao H, Han Y, et al. Histological validation of simultaneous non-contrast angiography and intraplaque hemorrhage imaging (SNAP) for characterizing carotid intraplaque hemorrhage. Eur Radiol 2021;31:3106–15.
CrossRef Google scholar
[114]
Gupta DK, Khandker N, Stacy K, et al. Utility of combining a simulation-based method with a lecture-based method for fundoscopy training in neurology residency. JAMA Neurol 2017;74:1223–7.
CrossRef Google scholar
[115]
Eppley SE, Mansberger SL, Ramanathan S, et al. Characteristics associated with adherence to annual dilated eye examinations among US patients with diagnosed diabetes. Ophthalmology 2019;126:1492–9.
CrossRef Google scholar
[116]
Song A, Lusk JB, Roh K-M, et al. Practice patterns of fundoscopic examination for diabetic retinopathy screening in primary care. JAMA Network Open 2022;5:e2218753.
CrossRef Google scholar
[117]
DellaCroce JT, Vitale AT. Hypertension and the eye. Curr Opin Ophthalmol 2008;19:493–8.
CrossRef Google scholar
[118]
Antonopoulos AS, Siasos G, Oikonomou E, et al. Arterial stiffness and microvascular disease in type 2 diabetes. Eur J Clin Invest 2021;51:e13380.
CrossRef Google scholar
[119]
Lovshin JA, Bjornstad P, Lovblom LE, et al. Atherosclerosis and microvascular complications: results from the Canadian study of longevity in type 1 diabetes. Diabetes Care 2018;41:2570–8.
CrossRef Google scholar
[120]
Ahadi S, Wilson KA, Babenko B, et al. Longitudinal fundus imaging and its genome-wide association analysis provide evidence for a human retinal aging clock. ELife 2023;12:e82364.
CrossRef Google scholar
[121]
Zhu Z, Shi D, Guankai P, et al. Retinal age gap as a predictive biomarker for mortality risk. Br J Ophthalmol 2023;107:547–54.
CrossRef Google scholar
[122]
Winkelmann JA, Eid A, Spicer G, et al. Spectral contrast optical coherence tomography angiography enables single-scan vessel imaging. Light Sci Appl 2019;8:7.
CrossRef Google scholar
[123]
Bullitt E, Zeng D, Mortamet B, et al. The effects of healthy aging on intracerebral blood vessels visualized by magnetic resonance angiography. Neurobiol Aging 2010;31:290–300.
CrossRef Google scholar
[124]
Li C, Rusinek H, Chen J, et al. Reduced white matter venous density on MRI is associated with neurodegeneration and cognitive impairment in the elderly. Front Aging Neurosci 2022;14:972282.
CrossRef Google scholar
[125]
Yang D, McCrann DJ, Nguyen H, et al. Increased polyploidy in aortic vascular smooth muscle cells during aging is marked by cellular senescence. Aging Cell 2007;6:257–60.
CrossRef Google scholar
[126]
Hansson GK, Libby P. The immune response in atherosclerosis: a double-edged sword. Nat Rev Immunol 2006;6:508–19
CrossRef Google scholar
[127]
Wolf D, Ley K. Immunity and inflammation in atherosclerosis. Circ Res 2019;124:315–27.
CrossRef Google scholar
[128]
Balistreri CR, Pisano C, Bertoldo F, et al. Red blood cell distribution width, vascular aging biomarkers, and endothelial progenitor cells for predicting vascular aging and diagnosing/prognosing age-related degenerative arterial diseases. Rejuvenation Res 2019;22:399–408.
CrossRef Google scholar
[129]
Buffa S, Borzì D, Chiarelli R, et al. Biomarkers for vascular ageing in aorta tissues and blood samples. Exp Gerontol 2019;128:110741.
CrossRef Google scholar
[130]
Li X, Li C, Zhang W, et al. Inflammation and aging: signaling pathways and intervention therapies. Signal Transduct Target Ther 2023;8:239.
CrossRef Google scholar
[131]
Ungvari Z, Csiszar A. The emerging role of IGF-1 deficiency in cardiovascular aging: recent advances. J Gerontol A Biol Sci Med Sci 2012;67:599–610.
CrossRef Google scholar
[132]
Higashi Y, Sukhanov S, Anwar A, et al. Aging, atherosclerosis, and IGF-1. J Gerontol A Biol Sci Med Sci 2012;67:626–39.
CrossRef Google scholar
[133]
Sherlala RA, Kammerer CM, Kuipers AL, et al. Relationship between serum IGF-1 and BMI differs by age. J Gerontol A Biol Sci Med Sci 2021;76:1303–8.
CrossRef Google scholar
[134]
Toth L, Czigler A, Hegedus E, et al. Age-related decline in circulating IGF-1 associates with impaired neurovascular coupling responses in older adults. GeroScience 2022;44:2771–83.
CrossRef Google scholar
[135]
Li J, Xiong M, Fu X-H, et al. Determining a multimodal aging clock in a cohort of Chinese women. Med 2023;4:825–48.
CrossRef Google scholar
[136]
Fulop GA, Ramirez-Perez FI, Kiss T, et al. IGF-1 deficiency promotes pathological remodeling of cerebral arteries: a potential mechanism contributing to the pathogenesis of intracerebral hemorrhages in aging. J Gerontol A Biol Sci Med Sci 2019;74:446–54.
CrossRef Google scholar
[137]
Sanders JL, Guo W, O’Meara ES, et al. Trajectories of IGF-I predict mortality in older adults: the Cardiovascular Health Study. J Gerontol A Biol Sci Med Sci 2018;73:953–9.
CrossRef Google scholar
[138]
Ameri P, Canepa M, Fabbi P, et al. Vitamin D modulates the association of circulating insulin-like growth factor-1 with carotid artery intima-media thickness. Atherosclerosis 2014;236:418–25.
CrossRef Google scholar
[139]
Fisher FM, Maratos-Flier E. Understanding the physiology of FGF21. Annu Rev Physiol 2016;78:223–41.
CrossRef Google scholar
[140]
Sorokina AG, Efimenko AY, Grigorieva OA, et al. Correlations between vessel stiffness and biomarkers of senescent cell in elderly patients. Kardiologiia 2022;62:15–22.
CrossRef Google scholar
[141]
Yang N, Zhang Y, Huang Y, et al. FGF21 at physiological concentrations regulates vascular endothelial cell function through multiple pathways. Biochim Biophys Acta Mol Basis Dis 2022;1868:166558.
CrossRef Google scholar
[142]
Zhang Y, Yan J, Yang N, et al. High-level serum fibroblast growth factor 21 concentration is closely associated with an increased risk of cardiovascular diseases: a systematic review and meta-analysis. Front Cardiovasc Med 2021;8:705273.
CrossRef Google scholar
[143]
Shen Y, Zhang X, Xu Y, et al. Serum FGF21 is associated with future cardiovascular events in patients with coronary artery disease. Cardiology 2018;139:212–8.
CrossRef Google scholar
[144]
Shen Y, Zhang X, Pan X, et al. Contribution of serum FGF21 level to the identification of left ventricular systolic dysfunction and cardiac death. Cardiovasc Diabetol 2017;16:106.
CrossRef Google scholar
[145]
Luo M, Yan D, Liang X, et al. Association between plasma fibulin-1 and brachial-ankle pulse wave velocity in arterial stiffness. Front Cardiovasc Med 2022;9:837490.
CrossRef Google scholar
[146]
Yasmin, Maskari RA, McEniery CM et al. The matrix proteins aggrecan and fibulin-1 play a key role in determining aortic stiffness. Sci Rep 2018;8:8550.
CrossRef Google scholar
[147]
Liu X, Liu Z, Wu Z, et al. Resurrection of endogenous retroviruses during aging reinforces senescence. Cell 2023;186:287–304.e26.
CrossRef Google scholar
[148]
Xie W, Ke Y, You Q, et al. Single-cell RNA sequencing and assay for transposase-accessible chromatin using sequencing reveals cellular and molecular dynamics of aortic aging in mice. Arterioscler Thromb Vasc Biol 2022;42:156–71.
CrossRef Google scholar
[149]
Weng N-P, Akbar AN, Goronzy J. CD28(-) T cells: their role in the age-associated decline of immune function. Trends Immunol 2009;30:306–12.
CrossRef Google scholar
[150]
Carrasco E, Gómez de Las Heras MM, Gabandé-Rodríguez E, et al. The role of T cells in age-related diseases. Nat Rev Immunol 2022;22:97–111.
CrossRef Google scholar
[151]
Rodriguez IJ, Lalinde Ruiz N, Llano León M, et al. Immunosenescence study of T cells: a systematic review. Front Immunol 2020;11:604591.
CrossRef Google scholar
[152]
Yang JX, Pan YY, Wang XX, et al. Endothelial progenitor cells in age-related vascular remodeling. Cell Transplant 2018;27:786–95.
CrossRef Google scholar
[153]
Xia W-H, Li J, Su C, et al. Physical exercise attenuates age-associated reduction in endothelium-reparative capacity of endothelial progenitor cells by increasing CXCR4/JAK-2 signaling in healthy men. Aging Cell 2012;11:111–9.
CrossRef Google scholar
[154]
Xia WH, Yang Z, Xu SY, et al. Age-related decline in reendothelialization capacity of human endothelial progenitor cells is restored by shear stress. Hypertension 2012;59:1225–31.
CrossRef Google scholar
[155]
Hill JM, Zalos G, Halcox JP, et al. Circulating endothelial progenitor cells, vascular function, and cardiovascular risk. N Engl J Med 2003;348:593–600.
CrossRef Google scholar
[156]
Jie KE, Goossens MH, van Oostrom O, et al. Circulating endothelial progenitor cell levels are higher during childhood than in adult life. Atherosclerosis 2009;202:345–7.
CrossRef Google scholar
[157]
Tao J, Wang Y, Yang Z, et al. Circulating endothelial progenitor cell deficiency contributes to impaired arterial elasticity in persons of advancing age. J Hum Hypertens 2006;20:490–5.
CrossRef Google scholar
[158]
Redondo S, Hristov M, Gordillo-Moscoso AA, et al. High-reproducible flow cytometric endothelial progenitor cell determination in human peripheral blood as CD34+/CD144+/CD3– lymphocyte subpopulation. J Immunol Methods 2008;335:21–7.
CrossRef Google scholar
[159]
Alique M, Ramirez-Carracedo R, Bodega G, et al. Senescent microvesicles: a novel advance in molecular mechanisms of atherosclerotic calcification. Int J Mol Sci 2018;19:2003.
CrossRef Google scholar
[160]
Wang J-M, Huang Y-J, Wang Y, et al. Increased circulating CD31+/ CD42- microparticles are associated with impaired systemic artery elasticity in healthy subjects. Am J Hypertens 2007;20:957–64.
CrossRef Google scholar
[161]
Grone M, Sansone R, Hoffken P, et al. Cocoa flavanols improve endothelial functional integrity in healthy young and elderly subjects. J Agric Food Chem 2020;68:1871–6.
CrossRef Google scholar
[162]
Amabile N, Cheng S, Renard JM, et al. Association of circulating endothelial microparticles with cardiometabolic risk factors in the Framingham Heart Study. Eur Heart J 2014;35:2972–9.
CrossRef Google scholar
[163]
Xu H, Li S, Liu YS. Roles and mechanisms of DNA methylation in vascular aging and related diseases. Front Cell Dev Biol 2021;9:699374.
CrossRef Google scholar
[164]
Baccarelli A, Tarantini L, Wright RO, et al. Repetitive element DNA methylation and circulating endothelial and inflammation markers in the VA normative aging study. Epigenetics 2010;5:222–8.
CrossRef Google scholar
[165]
Bakshi C, Vijayvergiya R, Dhawan V. Aberrant DNA methylation of M1-macrophage genes in coronary artery disease. Sci Rep 2019;9:1429.
CrossRef Google scholar
[166]
Martinez-Iglesias O, Carrera I, Carril JC, et al. DNA methylation in neurodegenerative and cerebrovascular disorders. Int J Mol Sci 2020;21:2220.
CrossRef Google scholar
[167]
Horvath S, Raj K. DNA methylation-based biomarkers and the epigenetic clock theory of ageing. Nat Rev Genet 2018;19:371–84.
CrossRef Google scholar
[168]
Duan R, Fu Q, Sun Y, et al. Epigenetic clock: a promising biomarker and practical tool in aging. Ageing Res Rev 2022;81:101743.
CrossRef Google scholar
[169]
Marioni RE, Shah S, McRae AF, et al. DNA methylation age of blood predicts all-cause mortality in later life. Genome Biol 2015;16:25.
CrossRef Google scholar
[170]
Horvath S. DNA methylation age of human tissues and cell types. Genome Biol 2013;14:R115.
CrossRef Google scholar
[171]
Hannum G, Guinney J, Zhao L, et al. Genome-wide methylation profiles reveal quantitative views of human aging rates. Mol Cell 2013;49:359–67.
CrossRef Google scholar
[172]
Levine ME, Lu AT, Quach A, et al. An epigenetic biomarker of aging for lifespan and healthspan. Aging 2018;10:573–91.
CrossRef Google scholar
[173]
Krištić J, Vučković F, Menni C, et al. Glycans are a novel biomarker of chronological and biological ages. J Gerontol A Biol Sci Med Sci 2014;69:779–89.
CrossRef Google scholar
[174]
Cao Q, Wu J, Wang X, et al. Noncoding RNAs in vascular aging. Oxid Med Cell Longev 2020;2020:7914957.
CrossRef Google scholar
[175]
Raucci A, Macri F, Castiglione S, et al. MicroRNA-34a: the bad guy in age-related vascular diseases. Cell Mol Life Sci 2021;78:7355–78.
CrossRef Google scholar
[176]
Zhao Y, Zhang Y, Zhang L, et al. The potential markers of circulating microRNAs and long non-coding RNAs in Alzheimer’s disease. Aging Dis 2019;10:1293–301.
CrossRef Google scholar
[177]
van den Berg MMJ, Krauskopf J, Ramaekers JG, et al. Circulating microRNAs as potential biomarkers for psychiatric and neurodegenerative disorders. Prog Neurobiol 2020;185:101732.
CrossRef Google scholar
[178]
Fulzele S, Mendhe B, Khayrullin A, et al. Muscle-derived miR-34a increases with age in circulating extracellular vesicles and induces senescence of bone marrow stem cells. Aging 2019;11:1791–803.
CrossRef Google scholar
[179]
Pang J, Xiong H, Yang H, et al. Circulating miR-34a levels correlate with age-related hearing loss in mice and humans. Exp Gerontol 2016;76:58–67.
CrossRef Google scholar
[180]
Du S, Ling H, Guo Z, et al. Roles of exosomal miRNA in vascular aging. Pharmacol Res 2021;165:105278.
CrossRef Google scholar
[181]
Toyama K, Igase M, Spin JM, et al. Exosome miR-501-3p elevation contributes to progression of vascular stiffness. Circ Rep 2021;3:170–7.
CrossRef Google scholar
[182]
Cheng Y-F, Gu X-J, Yang T-M, et al. Signature of miRNAs derived from the circulating exosomes of patients with amyotrophic lateral sclerosis. Front Aging Neurosci 2023;15:1106497.
CrossRef Google scholar
[183]
Panyard DJ, Yu B, Snyder MP. The metabolomics of human aging: advances, challenges, and opportunities. Sci Adv 2022;8:eadd6155.
CrossRef Google scholar
[184]
Bar N, Korem T, Weissbrod O, et al.; IMI DIRECT consortium. A reference map of potential determinants for the human serum metabolome. Nature 2020;588:135–40.
CrossRef Google scholar
[185]
Buergel T, Steinfeldt J, Ruyoga G, et al. Metabolomic profiles predict individual multidisease outcomes. Nat Med 2022;28:2309–20.
CrossRef Google scholar
[186]
Talmor-Barkan Y, Bar N, Shaul AA, et al. Metabolomic and microbiome profiling reveals personalized risk factors for coronary artery disease. Nat Med 2022;28:295–302.
CrossRef Google scholar
[187]
Collino S, Montoliu I, Martin F-PJ, et al. Metabolic signatures of extreme longevity in northern Italian centenarians reveal a complex remodeling of lipids, amino acids, and gut microbiota metabolism. PLoS One 2013;8:e56564.
CrossRef Google scholar
[188]
Dunn WB, Lin W, Broadhurst D, et al. Molecular phenotyping of a UK population: defining the human serum metabolome. Metabol: Off J Metabol Soc 2015;11:9–26.
CrossRef Google scholar
[189]
Zeng Y, Feng Q, Hesketh T, et al. Survival, disabilities in activities of daily living, and physical and cognitive functioning among the oldest-old in China: a cohort study. Lancet 2017;389:1619–29.
CrossRef Google scholar
[190]
Chak CM, Lacruz ME, Adam J, et al. Ageing investigation using two-time-point metabolomics data from KORA and CARLA studies. Metabolites 2019;9:44.
CrossRef Google scholar
[191]
Johnson LC, Parker K, Aguirre BF, et al. The plasma metabolome as a predictor of biological aging in humans. GeroScience 2019;41:895–906.
CrossRef Google scholar
[192]
Bunning BJ, Contrepois K, Lee-McMullen B, et al. Global metabolic profiling to model biological processes of aging in twins. Aging Cell 2020;19:e13073.
CrossRef Google scholar
[193]
Li H, Ren M, Li Q. 1H NMR-based metabolomics reveals the intrinsic interaction of age, plasma signature metabolites, and nutrient intake in the longevity population in Guangxi, China. Nutrients 2022;14:2539.
CrossRef Google scholar
[194]
Ke Y, Li D, Zhao M, et al. Gut flora-dependent metabolite trimethylamine-N-oxide accelerates endothelial cell senescence and vascular aging through oxidative stress. Free Radical Biol Med 2018;116:88–100.
CrossRef Google scholar
[195]
Choi RH, Tatum SM, Symons JD, et al. Ceramides and other sphingolipids as drivers of cardiovascular disease. Nat Rev Cardiol 2021;18:701–11.
CrossRef Google scholar
[196]
Yin W, Li F, Tan X, et al. Plasma ceramides and cardiovascular events in hypertensive patients at high cardiovascular risk. Am J Hypertens 2021;34:1209–16.
CrossRef Google scholar
[197]
Laaksonen R, Ekroos K, Sysi-Aho M, et al. Plasma ceramides predict cardiovascular death in patients with stable coronary artery disease and acute coronary syndromes beyond LDL-cholesterol. Eur Heart J 2016;37:1967–76.
CrossRef Google scholar
[198]
Liu Z, Li W, Geng L, et al. Cross-species metabolomic analysis identifies uridine as a potent regeneration promoting factor. Cell Discovery 2022;8:6.
CrossRef Google scholar
[199]
Shi D, Tan Q, Ruan J, et al. Aging-related markers in rat urine revealed by dynamic metabolic profiling using machine learning. Aging 2021;13:14322–41.
CrossRef Google scholar
[200]
Li X, Gao Y. Potential urinary aging markers of 20-month-old rats. PeerJ 2016;4:e2058.
CrossRef Google scholar
[201]
Teruya T, Goga H, Yanagida M. Aging markers in human urine: a comprehensive, non-targeted LC–MS study. FASEB BioAdv 2020;2:720–33.
CrossRef Google scholar
[202]
Shi F, Nie B, Gan W, et al. Oxidative damage of DNA, RNA and their metabolites in leukocytes, plasma and urine of Macaca mulatta: 8-oxoguanosine in urine is a useful marker for aging. Free Radic Res 2012;46:1093–8.
CrossRef Google scholar
[203]
Yao S-M, Zheng P-P, He W, et al. Urinary 8-OxoGsn as a potential indicator of mild cognitive impairment in frail patients with cardiovascular disease. Front Aging Neurosci 2021;13:672548.
CrossRef Google scholar
[204]
Andersson C, Johnson AD, Benjamin EJ, et al. 70-year legacy of the Framingham Heart Study. Nat Rev Cardiol 2019;16:687–98.
CrossRef Google scholar
[205]
Lam CSP, Xanthakis V, Sullivan LM, et al. Aortic root remodeling over the adult life course: longitudinal data from the Framingham Heart Study. Circulation 2010;122:884–90.
CrossRef Google scholar
[206]
Cooper LL, O’Donnell A, Beiser AS, et al. Association of aortic stiffness and pressure pulsatility with global amyloid-β and regional tau burden among Framingham Heart Study participants without dementia. JAMA Neurol 2022;79:710–9.
CrossRef Google scholar
[207]
Gong J, Wang G, Wang Y, et al. Nowcasting and forecasting the care needs of the older population in China: analysis of data from the China Health and Retirement Longitudinal Study (CHARLS). Lancet Public Health 2022;7:e1005–13.
CrossRef Google scholar
[208]
Wu Y, Tao Z, Qiao Y, et al. Prevalence and characteristics of somatic symptom disorder in the elderly in a community-based population: a large-scale cross-sectional study in China. BMC Psychiat 2022;22:257.
CrossRef Google scholar
[209]
Alimujiang A, Wiensch A, Boss J, et al. Association between life purpose and mortality among US adults older than 50 years. JAMA Network Open 2019;2:e194270.
CrossRef Google scholar
[210]
Börsch-Supan A, Brandt M, Hunkler C, et al; SHARE Central Coordination Team. Data resource profile: the Survey of Health, Ageing and Retirement in Europe (SHARE). Int J Epidemiol 2013;42:992–1001.
CrossRef Google scholar
[211]
Veronese N, Noale M, Sinclair A, et al. Risk of progression to diabetes and mortality in older people with prediabetes: the English longitudinal study on ageing. Age Ageing 2022;51:afab222.
CrossRef Google scholar
[212]
Kim GR, Sun J, Han M, et al. Evaluation of the directional relationship between handgrip strength and cognitive function: the Korean Longitudinal Study of Ageing. Age Ageing 2019;48:426–32.
CrossRef Google scholar
[213]
Wong R, Michaels-Obregon A, Palloni A. Cohort profile: the Mexican Health and Aging Study (MHAS). Int J Epidemiol 2017;46:e2.
CrossRef Google scholar
[214]
Perianayagam A, Prina M, Selvamani Y, et al. Sub-national patterns and correlates of depression among adults aged 45 years and older: findings from wave 1 of the Longitudinal Ageing Study in India. Lancet Psychiat 2022;9:645–59.
CrossRef Google scholar
[215]
Santamaría-Ulloa C, Chinnock A, Montero-López M. Association between obesity and mortality in the Costa Rican elderly: a cohort study. BMC Public Health 2022;22:1007.
CrossRef Google scholar
[216]
Li Z, Zhang W, Duan Y, et al. Biological age models based on a healthy Han Chinese population. Arch Gerontol Geriatr 2023;107:104905.
CrossRef Google scholar
[217]
Lu AT, Quach A, Wilson JG, et al. DNA methylation GrimAge strongly predicts lifespan and healthspan. Aging 2019;11:303–27.
CrossRef Google scholar
[218]
Nilsson Wadström B, Fatehali A-AH, Engström G, et al. A vascular aging index as independent predictor of cardiovascular events and total mortality in an elderly urban population. Angiology 2019;70:929–37.
CrossRef Google scholar
[219]
Singla A, Chobufo MD, Meesum A, et al. Prevalence and outcomes of low ankle brachial index by atherosclerotic cardiovascular disease risk level: insights from the National Health and Nutrition Examination Survey (NHANES). Am J Med Sci 2023;365:121–9.
CrossRef Google scholar
[220]
Ciardullo S, Bianconi E, Cannistraci R, et al. Peripheral artery disease and all-cause and cardiovascular mortality in patients with NAFLD. J Endocrinol Invest 2022;45:1547–53.
CrossRef Google scholar
[221]
Li Z, Zhang W, Duan Y, et al. Progress in biological age research. Front Public Health 2023;11:1074274.
CrossRef Google scholar
[222]
Lloyd-Jones DM, Allen NB, Anderson CAM, et al; American Heart Association. Life’s essential 8: updating and enhancing the American Heart Association’s Construct of Cardiovascular Health: a Presidential Advisory From the American Heart Association. Circulation 2022;146:e18–43.
CrossRef Google scholar

RIGHTS & PERMISSIONS

2023 The Author(s) 2023. Published by Oxford University Press on behalf of Higher Education Press.
AI Summary AI Mindmap
PDF(505 KB)

Accesses

Citations

Detail

Sections
Recommended

/