Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 1971 2023-02-25 17:30:57 |
2 format Meta information modification 1971 2023-02-27 04:34:13 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Sagris, M.; Theofilis, P.; Antonopoulos, A.S.; Tsioufis, K.; Tousoulis, D. Telomere Length and Coronary Artery Disease. Encyclopedia. Available online: https://encyclopedia.pub/entry/41666 (accessed on 27 July 2024).
Sagris M, Theofilis P, Antonopoulos AS, Tsioufis K, Tousoulis D. Telomere Length and Coronary Artery Disease. Encyclopedia. Available at: https://encyclopedia.pub/entry/41666. Accessed July 27, 2024.
Sagris, Marios, Panagiotis Theofilis, Alexios S. Antonopoulos, Konstantinos Tsioufis, Dimitris Tousoulis. "Telomere Length and Coronary Artery Disease" Encyclopedia, https://encyclopedia.pub/entry/41666 (accessed July 27, 2024).
Sagris, M., Theofilis, P., Antonopoulos, A.S., Tsioufis, K., & Tousoulis, D. (2023, February 25). Telomere Length and Coronary Artery Disease. In Encyclopedia. https://encyclopedia.pub/entry/41666
Sagris, Marios, et al. "Telomere Length and Coronary Artery Disease." Encyclopedia. Web. 25 February, 2023.
Telomere Length and Coronary Artery Disease
Edit

Coronary artery disease (CAD) is a multifactorial disease with a high prevalence, particularly in developing countries. The investigation of telomeres as a potential tool for the early detection of the atherosclerotic disease seems to be a promising method. Telomeres are repetitive DNA sequences located at the extremities of chromosomes that maintain genetic stability. Telomere length (TL) has been associated with several human disorders and diseases while its attrition rate varies significantly in the population. The rate of TL shortening ranges between 20 and 50 bp and is affected by factors such as the end-replication phenomenon, oxidative stress, and other DNA-damaging agents.

telomere length LTL telomerase

1. Telomere Length

Every person is born with a specific telomere length (TL) that ranges between 5 to 15 kb, which is affected as the years go by [1]. The rate of TL shortening hovers at 20–50 bp while it is dependent on several factors such as the end-replication phenomenon, oxidative stress, and other DNA-damaging agents [2][3][4][5]. According to the above-mentioned loop formation hypothesis, telomeric regions can form loops having a minimum length of about several thousand nucleotides, which can be used by the cell to quickly detect DNA breaks in this area [6]. By the end-replication phenomenon, a small telomeric DNA fragment is lost in every cell division due to the inability of transcription of the free 3′-chain. So naturally, telomeres reach a critical length and to such an extent that no loop can be formed. Telomere shortening is thought to be the cause of the restricted number of divisions in most human cells. Hayflick was the first to describe this occurrence and this phenomenon was later called the “Hayflick Limit” [7]. A DNA damage signal is received by the cell at this time, and telomeres lose their protective role. This critical shortening of TL leads the cell into senescence, and causative cell death, which is regulated by inner biochemical and pro-inflammatory changes via the transition of the cell into a senescence-associated secretory phenotype (SASP) [8][9]. The DNA-damage signal becomes permanent, leading to activation of cyclin-dependent inhibitor pathways, including either the p53/p21Cip or p16Ink4a/Rb while transcription factors such as nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), CCAAT/enhancer-binding protein (C/EBP), and tumor protein p53 are controlling the procession [4][5][9]. Although TL varies along the tissue types due to the altered proliferation rates, a correlation has been observed between TL in different tissues and peripheral blood leucocytes [1][2][3][10][11].

2. LTL and Atherosclerosis

Telomeres, called the “biological clock” of cells, are a recognized marker of cell senescence. They are widely affected by a variety of intrinsic and environmental factors via the upregulation of oxidative stress levels. It is a condition in which increased levels of reactive oxygen species (ROS), such as superoxide anions, hydrogen peroxide, and hydroxyl radicals, are present due to a biological imbalance [12]. Dysfunctional mitochondria and immune cells are responsible for the main expression of ROS . The high guanine content of telomeres makes telomeres an easy target for ROS, leading to guanine oxidation. These point mutations and single or double DNA breaks affect telomeres’ function and cell proliferation [13][14]. Consequently, oxidative stress leads to TL shortening which is closely associated with tissue age-related decline regardless of the telomerase function [15]. When a critical TL is reached, apoptotic mechanisms and molecular paths such as p53, MAP kinase (mitogen-activated protein kinase), and transcription factor kappa B are activated, leading to cell senescence [16]. Cells with SASP secrete a variety of pro- and inflammatory cytokines well known for their atherosclerotic effect [17]. SASP cells have been identified in vasculature regions with atherosclerotic plaques as well as in cardiomyocytes in biopsies of patients with heart failure [18][19][20]. In both conditions, the SASP phenotype was followed by significant short TL [18]. As a result, factors that induce oxidative stress and telomere shortening can lead to a vicious cycle that promotes a state of chronic inflammation, which causes vascular endothelial dysfunction and contributes to the development of atherosclerotic plaques.
Atherosclerosis is a multifactorial condition whose progression is affected by a variety of cardiovascular risk factors [21]. Recently, short LTL has been positively associated with cardiovascular risk factors such as high BMI, waist circumference, high levels of blood C-reactive protein, low levels of HDL (high-density lipoprotein), high levels of cholesterol and triglycerides, as well as insulin resistance and blood pressure [22][23][24]. In another study, although no association was observed between classical cardiovascular risk factors and short LTL, when the results were adjusted to smokers a strong statistical significance was detected [25]. Benetos et al. previously verified the link between hypertension and shorter telomeres [26]. In the same regard, Morgan et al. discovered that the exposed telomere ends led to arterial cell senescence in individuals with hypertension [27]. Finally, Haycock et al. concluded that patients with diabetes mellitus presented shorter LTL than their healthy counterparts [28]. Short telomeres have been linked to increased arterial stiffness, preclinical atherosclerosis, and poor diabetes management. This might be owing to the harmful consequences of persistent hyperglycemia and the accumulation of advanced glycation end products (AGEs) [29].
Atherosclerosis is characterized by the formation of plaques in the vessel wall. These emerge as a result of complex pathophysiological pathways involving pro- and anti-inflammatory cytokines [30][31]. Given the elevated cardiovascular risk in people with short LTL, it would be rational to assume that there is a connection between short LTL and subclinical atherosclerosis. Since the investigation of potential indicators of atherosclerosis progression constitutes a huge challenge worldwide, several studies are trying to detect the exact role of LTL in this context. Until now, the landscape has been quite hazy with the results from a number of studies being inconsistent. In 2016, a large study including 1459 middle-aged adults found no statistically significant link between shortened LTL and subclinical atherosclerosis [32][33]. Thus, TL does not seem to have a prognostic value in individuals without clinical signs of disease. Indeed, Nzietchueng and Nguyen et al. showed a significantly shorter TL in aortic cells with atherosclerotic lesions as well as in vasculature regions with low elasticity [34][35]. A possible explanation is that local vascular alterations affect TL as a result of oxidative stress. In the Framingham study, internal carotid artery intima-media thickness was linked with short LTL among obese males (BMI > 30 kg/m2) but not in the whole cohort. No association was observed between short LTL and common carotid artery intima-media thickness or carotid artery stenosis [36]. Trying to connect LTL with potential modifiable behavioral factors, Bountziouka et al. analyzed 422.797 patients from the UK Biobank. Although lifestyle changes appear to be quantitatively related with LTL, the magnitude of these effects is insufficient to appreciably affect the connection between LTL and various diseases or life expectancy [37]. In another study, Schellnegger et al. highlighted the detrimental role of sedentary life, not only in cardiovascular risk induction but also in LTL attrition. More specifically, a regular basis aerobic physical activity at a moderate to high level tends to help LTL preservation, while it is still unclear as to the optimal type and duration of exercising [38]. LTL was also not a significant predictor of intima-media thickness or plaque formation in the Asklepios study [32]. However, Panayiotou et al. showed an inverse association between LTL and common carotid artery intima-media thickness [39]. In the same aspect, the Strong Heart Study examined 2819 Americans without known cardiovascular risk factors for a follow-up period of 5.5 years to assess the predictive role of LTL in the occurrence and progression of carotid atherosclerosis. The shortest LTL had a 49% and 61% greater incidence of plaque formation and plaque development, respectively, than the longest LTL [40]. Finally, a strong association of carotid atherosclerosis with short LTL has been established in hypertensive patients, highlighting the unfavorable impact of hypertension on TL [41].

3. LTL and Cardiovascular Disease

Since observational studies illustrated that attrition of LTL is related to mortality, physicians are trying to investigate its role in cardiovascular disease (CVD), which is the leading cause of death worldwide [42]. The Hutchinson–Guilford Progeria Syndrome constitutes a striking example of age-related LTL shortening, in which the majority of the patients die from a myocardial infarction or stroke in their teenage years [43]. Premature senescence of fibroblasts as well as rapid TL shortening was observed in the cell cultures of these patients [43]. As such, there was a rationale for further exploration of LTL’s role in CVD, especially since studies showed that shorter LTL is associated with higher mortality rates. Indeed, in the Bruneck and LURIC studies, patients with a lower relative LTL presented higher death incidences in a 10-year follow-up [42][44][45]. In the same vein, multiple studies reported an increase in all-cause mortality risk, ranging from 17% to 66%, when patients with the longest telomeres were compared to subjects with the shortest telomeres [42][44][45][46][47]. However, some studies could not confirm the hypothesis of the association between LTL and CVD or mortality. This might be partially explained by the fact that some of these studies focused on low-risk populations with a modest number of events [48][49][50].
Several studies also investigated the association of LTL shortening with the presentation and progression of CAD. In two studies conducted in 2010 and 2021, physicians found that LTL in patients with stable CAD was significantly shorter than in healthy individuals of the same age (1.13 ± 0.52 CU in patients with CAD vs. 1.52 ± 0.81 CU in healthy individuals) which was further extended to sex analysis. It was interesting that men presented with shorter LTL than women, which could partially be related to the effect of estrogen, but this is an observation that remains hazy [45][51]. Trying to investigate the genetic background of premature CAD onset, Tian et al. compared Chinese patients with healthy individuals. It was shown that patients with premature CAD presented shorter LTL and higher circulating levels of oxidative stress components [52]. In another study, men with arterial hypertension, CAD, and early vascular aging (defined as arterial hypertension or CAD debut at young age—before 45 years, increased vascular wall stiffness according to the cardio-ankle vascular index), the LTL was significantly shorter than in men with arterial hypertension and CAD but without early vascular aging [53]. Haycock et al. conducted a meta-analysis on 43,000 individuals, including over 8000 patients with cardiovascular disease. It was demonstrated that, regardless of other risk factors, people with a shorter LTL were more likely to develop CAD [28]. In another meta-analysis published in 2020, shorter LTL was shown to be strongly connected to CAD severity, with Asians presenting the shortest LTL after ethnicity adjustment [54]. However, regarding the association of function stages of stable angina I–III (according to the Canadian Cardiovascular Society classifications) and LTL, no significant relation was observed [42][53].
One of the major manifestations of CAD is myocardial infarction (MI), and the development of biomarkers for early prognosis is referred to as mandatory [55][56][57][58][59]. LTL has been investigated as a potential biomarker with controversial results. A significantly shorter LTL has been observed in individuals with MI compared to healthy individuals even after adjustment for sex, body mass, and age [55]. Similarly, a study from the United Kingdom in 2017 demonstrated that in patients suffering from MI, LTL may be a useful prognostic biomarker for cardiovascular outcomes after the event, regardless of age. More particularly, MI patients with short LTL (defined as less than 0.96 CU in the study) presented significantly higher rates of all-cause and cardiovascular mortality within the first year after the event [60][61]. On the other hand, Russo et al. found no significant association between LTL and MI occurrence in young Italians [62]. Neither could Chan et al. confirm the hypothesis, as no statistically significant association was observed between relative LTL and adverse MI outcomes (death, recurrent MI, unplanned percutaneous coronary intervention revascularization, stroke, significant bleeding) in elderly Chinese patients, one year after their percutaneous coronary intervention [63]. Finally, while there was initially a connection between LTL and MI incidence in Czech women, the significance was lost after adjusting for major cardiovascular risk factors [61]. LTL has also been explored as a potential biomarker for the prognostication of stroke. The vast majority of the trials, which included over 37,000 and 25,000 people, respectively, could not establish a link between stroke risk and LTL [64][65].

References

  1. Boniewska-Bernacka, E.; Panczyszyn, A.; Klinger, M. Telomeres and telomerase in risk assessment of cardiovascular diseases. Exp. Cell Res. 2020, 397, 112361.
  2. Fitzpatrick, A.L.; Kronmal, R.A.; Kimura, M.; Gardner, J.P.; Psaty, B.M.; Jenny, N.S.; Tracy, R.P.; Hardikar, S.; Aviv, A. Leukocyte telomere length and mortality in the Cardiovascular Health Study. J. Gerontol. A Biol. Sci. Med. Sci. 2011, 66, 421–429.
  3. Friedrich, U.; Griese, E.; Schwab, M.; Fritz, P.; Thon, K.; Klotz, U. Telomere length in different tissues of elderly patients. Mech. Ageing Dev. 2000, 119, 89–99.
  4. Hayakawa, T.; Iwai, M.; Aoki, S.; Takimoto, K.; Maruyama, M.; Maruyama, W.; Motoyama, N. SIRT1 suppresses the senescence-associated secretory phenotype through epigenetic gene regulation. PLoS ONE 2015, 10, e0116480.
  5. Leon, K.E.; Buj, R.; Lesko, E.; Dahl, E.S.; Chen, C.W.; Tangudu, N.K.; Imamura-Kawasawa, Y.; Kossenkov, A.V.; Hobbs, R.P.; Aird, K.M. DOT1L modulates the senescence-associated secretory phenotype through epigenetic regulation of IL1A. J. Cell Biol. 2021, 220, e202008101.
  6. Yegorov, Y.E.; Chernov, D.N.; Akimov, S.S.; Akhmalisheva, A.K.; Smirnova, Y.B.; Shinkarev, D.B.; Semenova, I.V.; Yegorova, I.N.; Zelenin, A.V. Blockade of telomerase function by nucleoside analogs. Biochemistry 1997, 62, 1296–1305.
  7. Shay, J.W.; Wright, W.E. Hayflick, his limit, and cellular ageing. Nat. Rev. Mol. Cell Biol. 2000, 1, 72–76.
  8. Lopes-Paciencia, S.; Saint-Germain, E.; Rowell, M.C.; Ruiz, A.F.; Kalegari, P.; Ferbeyre, G. The senescence-associated secretory phenotype and its regulation. Cytokine 2019, 117, 15–22.
  9. Gao, L.; Zheng, W.G.; Wu, X.K.; Du, G.H.; Qin, X.M. Baicalein Delays H2O2-Induced Astrocytic Senescence through Inhibition of Senescence-Associated Secretory Phenotype (SASP), Suppression of JAK2/STAT1/NF-kappaB Pathway, and Regulation of Leucine Metabolism. ACS Chem. Neurosci. 2021, 12, 2320–2335.
  10. Yeh, J.K.; Wang, C.Y. Telomeres and Telomerase in Cardiovascular Diseases. Genes 2016, 7, 58.
  11. Dlouha, D.; Maluskova, J.; Kralova Lesna, I.; Lanska, V.; Hubacek, J.A. Comparison of the relative telomere length measured in leukocytes and eleven different human tissues. Physiol. Res. 2014, 63, S343–S350.
  12. Barnes, R.P.; Fouquerel, E.; Opresko, P.L. The impact of oxidative DNA damage and stress on telomere homeostasis. Mech. Ageing Dev. 2019, 177, 37–45.
  13. Correia-Melo, C.; Hewitt, G.; Passos, J.F. Telomeres, oxidative stress and inflammatory factors: Partners in cellular senescence? Longev. Healthspan 2014, 3, 1.
  14. Panth, N.; Paudel, K.R.; Parajuli, K. Reactive Oxygen Species: A Key Hallmark of Cardiovascular Disease. Adv. Med. 2016, 2016, 9152732.
  15. Grahame, T.J.; Schlesinger, R.B. Oxidative stress-induced telomeric erosion as a mechanism underlying airborne particulate matter-related cardiovascular disease. Part. Fibre Toxicol. 2012, 9, 21.
  16. Sack, M.N.; Fyhrquist, F.Y.; Saijonmaa, O.J.; Fuster, V.; Kovacic, J.C. Basic Biology of Oxidative Stress and the Cardiovascular System: Part 1 of a 3-Part Series. J. Am. Coll. Cardiol. 2017, 70, 196–211.
  17. Aviv, H.; Khan, M.Y.; Skurnick, J.; Okuda, K.; Kimura, M.; Gardner, J.; Priolo, L.; Aviv, A. Age dependent aneuploidy and telomere length of the human vascular endothelium. Atherosclerosis 2001, 159, 281–287.
  18. Sharifi-Sanjani, M.; Oyster, N.M.; Tichy, E.D.; Bedi, K.C., Jr.; Harel, O.; Margulies, K.B.; Mourkioti, F. Cardiomyocyte-Specific Telomere Shortening is a Distinct Signature of Heart Failure in Humans. J. Am. Heart Assoc. 2017, 6, e005086.
  19. Theofilis, P.; Sagris, M.; Oikonomou, E.; Antonopoulos, A.S.; Siasos, G.; Tsioufis, K.; Tousoulis, D. The impact of SGLT2 inhibitors on inflammation: A systematic review and meta-analysis of studies in rodents. Int. Immunopharmacol. 2022, 111, 109080.
  20. Theofilis, P.; Sagris, M.; Oikonomou, E.; Antonopoulos, A.S.; Siasos, G.; Tsioufis, K.; Tousoulis, D. Pleiotropic effects of SGLT2 inhibitors and heart failure outcomes. Diabetes Res. Clin. Pract. 2022, 188, 109927.
  21. Sagris, M.; Theofilis, P.; Antonopoulos, A.S.; Oikonomou, E.; Paschaliori, C.; Galiatsatos, N.; Tsioufis, K.; Tousoulis, D. Inflammation in Coronary Microvascular Dysfunction. Int. J. Mol. Sci. 2021, 22, 13471.
  22. Karimi, B.; Yunesian, M.; Nabizadeh, R.; Mehdipour, P. Serum Level of Total Lipids and Telomere Length in the Male Population: A Cross-Sectional Study. Am. J. Mens. Health 2019, 13, 1557988319842973.
  23. Boccardi, V.; Esposito, A.; Rizzo, M.R.; Marfella, R.; Barbieri, M.; Paolisso, G. Mediterranean diet, telomere maintenance and health status among elderly. PLoS ONE 2013, 8, e62781.
  24. Werner, C.M.; Hecksteden, A.; Morsch, A.; Zundler, J.; Wegmann, M.; Kratzsch, J.; Thiery, J.; Hohl, M.; Bittenbring, J.T.; Neumann, F.; et al. Differential effects of endurance, interval, and resistance training on telomerase activity and telomere length in a randomized, controlled study. Eur. Heart J. 2019, 40, 34–46.
  25. Koriath, M.; Muller, C.; Pfeiffer, N.; Nickels, S.; Beutel, M.; Schmidtmann, I.; Rapp, S.; Munzel, T.; Westermann, D.; Karakas, M.; et al. Relative Telomere Length and Cardiovascular Risk Factors. Biomolecules 2019, 9, 192.
  26. Benetos, A.; Toupance, S.; Gautier, S.; Labat, C.; Kimura, M.; Rossi, P.M.; Settembre, N.; Hubert, J.; Frimat, L.; Bertrand, B.; et al. Short Leukocyte Telomere Length Precedes Clinical Expression of Atherosclerosis: The Blood-and-Muscle Model. Circ. Res. 2018, 122, 616–623.
  27. Morgan, R.G.; Ives, S.J.; Walker, A.E.; Cawthon, R.M.; Andtbacka, R.H.; Noyes, D.; Lesniewski, L.A.; Richardson, R.S.; Donato, A.J. Role of arterial telomere dysfunction in hypertension: Relative contributions of telomere shortening and telomere uncapping. J. Hypertens. 2014, 32, 1293–1299.
  28. Haycock, P.C.; Heydon, E.E.; Kaptoge, S.; Butterworth, A.S.; Thompson, A.; Willeit, P. Leucocyte telomere length and risk of cardiovascular disease: Systematic review and meta-analysis. BMJ 2014, 349, g4227.
  29. Dudinskaya, E.N.; Tkacheva, O.N.; Shestakova, M.V.; Brailova, N.V.; Strazhesko, I.D.; Akasheva, D.U.; Isaykina, O.Y.; Sharashkina, N.V.; Kashtanova, D.A.; Boytsov, S.A. Short telomere length is associated with arterial aging in patients with type 2 diabetes mellitus. Endocr. Connect. 2015, 4, 136–143.
  30. Sagris, M.; Theofilis, P.; Antonopoulos, A.S.; Tsioufis, C.; Oikonomou, E.; Antoniades, C.; Crea, F.; Kaski, J.C.; Tousoulis, D. Inflammatory Mechanisms in COVID-19 and Atherosclerosis: Current Pharmaceutical Perspectives. Int. J. Mol. Sci. 2021, 22, 6607.
  31. Bennett, M.R.; Sinha, S.; Owens, G.K. Vascular Smooth Muscle Cells in Atherosclerosis. Circ. Res. 2016, 118, 692–702.
  32. De Meyer, T.; Rietzschel, E.R.; De Buyzere, M.L.; Langlois, M.R.; De Bacquer, D.; Segers, P.; Van Damme, P.; De Backer, G.G.; Van Oostveldt, P.; Van Criekinge, W.; et al. Systemic telomere length and preclinical atherosclerosis: The Asklepios Study. Eur. Heart J. 2009, 30, 3074–3081.
  33. Fernandez-Alvira, J.M.; Fuster, V.; Dorado, B.; Soberon, N.; Flores, I.; Gallardo, M.; Pocock, S.; Blasco, M.A.; Andres, V. Short Telomere Load, Telomere Length, and Subclinical Atherosclerosis: The PESA Study. J. Am. Coll. Cardiol. 2016, 67, 2467–2476.
  34. Nguyen, M.T.; Vryer, R.; Ranganathan, S.; Lycett, K.; Grobler, A.; Dwyer, T.; Juonala, M.; Saffery, R.; Burgner, D.; Wake, M. Telomere Length and Vascular Phenotypes in a Population-Based Cohort of Children and Midlife Adults. J. Am. Heart Assoc. 2019, 8, e012707.
  35. Nzietchueng, R.; Elfarra, M.; Nloga, J.; Labat, C.; Carteaux, J.P.; Maureira, P.; Lacolley, P.; Villemot, J.P.; Benetos, A. Telomere length in vascular tissues from patients with atherosclerotic disease. J. Nutr. Health Aging 2011, 15, 153–156.
  36. O’Donnell, C.J.; Demissie, S.; Kimura, M.; Levy, D.; Gardner, J.P.; White, C.; D’Agostino, R.B.; Wolf, P.A.; Polak, J.; Cupples, L.A.; et al. Leukocyte telomere length and carotid artery intimal medial thickness: The Framingham Heart Study. Arterioscler. Thromb. Vasc. Biol. 2008, 28, 1165–1171.
  37. Bountziouka, V.; Musicha, C.; Allara, E.; Kaptoge, S.; Wang, Q.; Angelantonio, E.D.; Butterworth, A.S.; Thompson, J.R.; Danesh, J.N.; Wood, A.M.; et al. Modifiable traits, healthy behaviours, and leukocyte telomere length: A population-based study in UK Biobank. Lancet Healthy Longev. 2022, 3, e321–e331.
  38. Schellnegger, M.; Lin, A.C.; Hammer, N.; Kamolz, L.P. Physical Activity on Telomere Length as a Biomarker for Aging: A Systematic Review. Sports Med. Open 2022, 8, 111.
  39. Panayiotou, A.G.; Nicolaides, A.N.; Griffin, M.; Tyllis, T.; Georgiou, N.; Bond, D.; Martin, R.M.; Hoppensteadt, D.; Fareed, J.; Humphries, S.E. Leukocyte telomere length is associated with measures of subclinical atherosclerosis. Atherosclerosis 2010, 211, 176–181.
  40. Chen, S.; Lin, J.; Matsuguchi, T.; Blackburn, E.; Yeh, F.; Best, L.G.; Devereux, R.B.; Lee, E.T.; Howard, B.V.; Roman, M.J.; et al. Short leukocyte telomere length predicts incidence and progression of carotid atherosclerosis in American Indians: The Strong Heart Family Study. Aging 2014, 6, 414–427.
  41. Benetos, A.; Gardner, J.P.; Zureik, M.; Labat, C.; Xiaobin, L.; Adamopoulos, C.; Temmar, M.; Bean, K.E.; Thomas, F.; Aviv, A. Short telomeres are associated with increased carotid atherosclerosis in hypertensive subjects. Hypertension 2004, 43, 182–185.
  42. Pusceddu, I.; Kleber, M.; Delgado, G.; Herrmann, W.; Marz, W.; Herrmann, M. Telomere length and mortality in the Ludwigshafen Risk and Cardiovascular Health study. PLoS ONE 2018, 13, e0198373.
  43. Ahmed, M.S.; Ikram, S.; Bibi, N.; Mir, A. Hutchinson-Gilford Progeria Syndrome: A Premature Aging Disease. Mol. Neurobiol. 2018, 55, 4417–4427.
  44. Pusceddu, I.; Herrmann, W.; Kleber, M.E.; Scharnagl, H.; Marz, W.; Herrmann, M. Telomere length, vitamin B12 and mortality in persons undergoing coronary angiography: The Ludwigshafen risk and cardiovascular health study. Aging 2019, 11, 7083–7097.
  45. Willeit, P.; Willeit, J.; Brandstatter, A.; Ehrlenbach, S.; Mayr, A.; Gasperi, A.; Weger, S.; Oberhollenzer, F.; Reindl, M.; Kronenberg, F.; et al. Cellular aging reflected by leukocyte telomere length predicts advanced atherosclerosis and cardiovascular disease risk. Arterioscler. Thromb. Vasc. Biol. 2010, 30, 1649–1656.
  46. Mons, U.; Muezzinler, A.; Schottker, B.; Dieffenbach, A.K.; Butterbach, K.; Schick, M.; Peasey, A.; De Vivo, I.; Trichopoulou, A.; Boffetta, P.; et al. Leukocyte Telomere Length and All-Cause, Cardiovascular Disease, and Cancer Mortality: Results from Individual-Participant-Data Meta-Analysis of 2 Large Prospective Cohort Studies. Am. J. Epidemiol. 2017, 185, 1317–1326.
  47. Wang, Q.; Zhan, Y.; Pedersen, N.L.; Fang, F.; Hagg, S. Telomere Length and All-Cause Mortality: A Meta-analysis. Ageing Res. Rev. 2018, 48, 11–20.
  48. Bischoff, C.; Petersen, H.C.; Graakjaer, J.; Andersen-Ranberg, K.; Vaupel, J.W.; Bohr, V.A.; Kolvraa, S.; Christensen, K. No association between telomere length and survival among the elderly and oldest old. Epidemiology 2006, 17, 190–194.
  49. Needham, B.L.; Rehkopf, D.; Adler, N.; Gregorich, S.; Lin, J.; Blackburn, E.H.; Epel, E.S. Leukocyte telomere length and mortality in the National Health and Nutrition Examination Survey, 1999-2002. Epidemiology 2015, 26, 528–535.
  50. Svensson, J.; Karlsson, M.K.; Ljunggren, O.; Tivesten, A.; Mellstrom, D.; Moverare-Skrtic, S. Leukocyte telomere length is not associated with mortality in older men. Exp. Gerontol. 2014, 57, 6–12.
  51. Starnino, L.; Dupuis, G.; Busque, L.; Bourgoin, V.; Dube, M.P.; Busseuil, D.; D’Antono, B. The associations of hostility and defensiveness with telomere length are influenced by sex and health status. Biol. Sex Differ. 2021, 12, 2.
  52. Tian, R.; Zhang, L.N.; Zhang, T.T.; Pang, H.Y.; Chen, L.F.; Shen, Z.J.; Liu, Z.; Fang, Q.; Zhang, S.Y. Association Between Oxidative Stress and Peripheral Leukocyte Telomere Length in Patients with Premature Coronary Artery Disease. Med. Sci. Monit. 2017, 23, 4382–4390.
  53. Bruno, R.M.; Nilsson, P.M.; Engstrom, G.; Wadstrom, B.N.; Empana, J.P.; Boutouyrie, P.; Laurent, S. Early and Supernormal Vascular Aging: Clinical Characteristics and Association With Incident Cardiovascular Events. Hypertension 2020, 76, 1616–1624.
  54. Xu, X.; Hu, H.; Lin, Y.; Huang, F.; Ji, H.; Li, Y.; Lin, S.; Chen, X.; Duan, S. Differences in Leukocyte Telomere Length between Coronary Heart Disease and Normal Population: A Multipopulation Meta-Analysis. Biomed Res. Int. 2019, 2019, 5046867.
  55. Capodanno, D.; Alfonso, F.; Levine, G.N.; Valgimigli, M.; Angiolillo, D.J. ACC/AHA Versus ESC Guidelines on Dual Antiplatelet Therapy: JACC Guideline Comparison. J. Am. Coll. Cardiol. 2018, 72, 2915–2931.
  56. Giannaki, A.; Sagris, M.; Toskas, P.; Antonopoulos, A.S.; Oikonomou, E.; Theofilis, P.; Lazaros, G.; Tousoulis, D. The Effect of Stress Management in Patients Post-acute Myocardial Infarction. Hellenic J. Cardiol. 2022, S1109–9666.
  57. Diavati, S.; Sagris, M.; Terentes-Printzios, D.; Vlachopoulos, C. Anticoagulation Treatment in Venous Thromboembolism: Options and Optimal Duration. Curr. Pharm. Des. 2022, 28, 296–305.
  58. Sagris, M.; Antonopoulos, A.S.; Theofilis, P.; Oikonomou, E.; Siasos, G.; Tsalamandris, S.; Antoniades, C.; Brilakis, E.S.; Kaski, J.C.; Tousoulis, D. Risk factors profile of young and older patients with myocardial infarction. Cardiovasc. Res. 2022, 118, 2281–2292.
  59. Sagris, M.; Antonopoulos, A.S.; Simantiris, S.; Oikonomou, E.; Siasos, G.; Tsioufis, K.; Tousoulis, D. Pericoronary fat attenuation index-a new imaging biomarker and its diagnostic and prognostic utility: A systematic review and meta-analysis. Eur. Heart J. Cardiovasc. Imaging 2022, 23, e526–e536.
  60. Margaritis, M.; Sanna, F.; Lazaros, G.; Akoumianakis, I.; Patel, S.; Antonopoulos, A.S.; Duke, C.; Herdman, L.; Psarros, C.; Oikonomou, E.K.; et al. Predictive value of telomere length on outcome following acute myocardial infarction: Evidence for contrasting effects of vascular vs. blood oxidative stress. Eur. Heart J. 2017, 38, 3094–3104.
  61. Dlouha, D.; Pitha, J.; Mesanyova, J.; Mrazkova, J.; Fellnerova, A.; Stanek, V.; Lanska, V.; Hubacek, J.A. Genetic variants within telomere-associated genes, leukocyte telomere length and the risk of acute coronary syndrome in Czech women. Clin. Chim. Acta 2016, 454, 62–65.
  62. Russo, A.; Palumbo, L.; Fornengo, C.; Di Gaetano, C.; Ricceri, F.; Guarrera, S.; Critelli, R.; Anselmino, M.; Piazza, A.; Gaita, F.; et al. Telomere length variation in juvenile acute myocardial infarction. PLoS ONE 2012, 7, e49206.
  63. Chan, D.; Martin-Ruiz, C.; Saretzki, G.; Neely, D.; Qiu, W.; Kunadian, V. The association of telomere length and telomerase activity with adverse outcomes in older patients with non-ST-elevation acute coronary syndrome. PLoS ONE 2020, 15, e0227616.
  64. Cao, W.; Li, X.; Zhang, X.; Zhang, J.; Sun, Q.; Xu, X.; Sun, M.; Tian, Q.; Li, Q.; Wang, H.; et al. No Causal Effect of Telomere Length on Ischemic Stroke and Its Subtypes: A Mendelian Randomization Study. Cells 2019, 8, 159.
  65. Jin, X.; Pan, B.; Dang, X.; Wu, H.; Xu, D. Relationship between short telomere length and stroke: A meta-analysis. Medicine 2018, 97, e12489.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , ,
View Times: 416
Revisions: 2 times (View History)
Update Date: 27 Feb 2023
1000/1000
Video Production Service