Papornița Moșului
Rezultate 1 la 4 din 4

Subiect: another article on telomeres and longevity

  1. #1
    Administrator
    Data înscrierii
    10.10.2011
    Locație
    CRAIOVA
    Posturi
    78.785

    another article on telomeres and longevity

    On 8/4/2018 10:43 AM, Constantin Pandaru wrote:


    FYI ... your opinion counts!!
    .. who has proven the best, unrivaled solution??




    From:
    Date: August 3, 2018 at 8:53:20 AM CDT
    To: <
    Subject: another article on telomeres and longevity


    ..., and professional explanation :
    https://www.physiology.org/doi/10.11...rev.00024.2007
    www.popservice.ro
    www.papornitamosului.ro
    [U][COLOR=#800080][
    e-mail - adipop@popservice.ro
    ID Messenger: zalmoxa_adipop
    skype - adrianpop58
    http://www.sfatulbatranilor.ro/forum.php
    https://www.facebook.com/groups/611112328972709/ - REGIA DE RECONSTRUCTIE A ROMANIEI
    https:/https://www.facebook.com/grou...IZEGETUSA2050/ - Piciumanii doresc PACE
    https://www.facebook.com/groups/1086016084901078/ - Avangarda de Sacrificiu

  2. #2
    Administrator
    Data înscrierii
    10.10.2011
    Locație
    CRAIOVA
    Posturi
    78.785
    Telomeres, lifestyle, cancer, and aging

    Masood A. Shammas

    Author information ► Copyright and License information ► Disclaimer


    The publisher's final edited version of this article is available at Curr Opin Clin Nutr Metab Care
    See other articles in PMC that cite the published article.




    Abstract

    Purpose of review

    There has been growing evidence that lifestyle factors may affect the health and lifespan of an individual by affecting telomere length. The purpose of this review was to highlight the importance of telomeres in human health and aging and to summarize possible lifestyle factors that may affect health and longevity by altering the rate of telomere shortening.

    Recent findings

    Recent studies indicate that telomere length, which can be affected by various lifestyle factors, can affect the pace of aging and onset of age-associated diseases.

    Summary

    Telomere length shortens with age. Progressive shortening of telomeres leads to senescence, apoptosis, or oncogenic transformation of somatic cells, affecting the health and lifespan of an individual. Shorter telomeres have been associated with increased incidence of diseases and poor survival. The rate of telomere shortening can be either increased or decreased by specific lifestyle factors. Better choice of diet and activities has great potential to reduce the rate of telomere shortening or at least prevent excessive telomere attrition, leading to delayed onset of age-associated diseases and increased lifespan. This review highlights the role of telomeres in aging and describes the lifestyle factors which may affect telomeres, human health, and aging.


    Keywords: aging, cancer, lifestyle, oxidative stress, telomere

    Introduction

    Telomeres, the specific DNA–protein structures found at both ends of each chromosome, protect genome from nucleolytic degradation, unnecessary recombination, repair, and interchromosomal fusion. Telomeres therefore play a vital role in preserving the information in our genome. As a normal cellular process, a small portion of telomeric DNA is lost with each cell division. When telomere length reaches a critical limit, the cell undergoes senescence and/or apoptosis. Telomere length may therefore serve as a biological clock to determine the lifespan of a cell and an organism. Certain agents associated with specific lifestyles may expedite telomere shortening by inducing damage to DNA in general or more specifically at telomeres and may therefore affect health and lifespan of an individual. In this review we highlight the lifestyle factors that may adversely affect health and lifespan of an individual by accelerating telomere shortening and also those that can potentially protect telomeres and health of an individual.

    Structure and function of telomeres

    Telomeres, the DNA–protein complexes at chromosome ends (Fig. 1), protect genome from degradation and interchromosomal fusion. Telomeric DNA is associated with telomere-binding proteins and a loop structure mediated by TRF2 protects the ends of human chromosomes against exonucleolytic degradation [1], and may also prime telomeric DNA synthesis by a mechanism similar to ‘gap filling’ in homologous recombination [2]. As shown in Fig. 2, telomere shortening occurs at each DNA replication, and if continued leads to chromosomal degradation and cell death [3]. Telomerase activity, the ability to extend telomeres, is present in germline and certain hematopoietic cells, whereas somatic cells have low or undetectable levels of this activity and their telomeres undergo a progressive shortening with replication (Fig. 2). Telomerases are reactivated in most cancers and immortalized cells. However, a subset of cancer/immortalized cells lack telomerase activity and maintain telomere length by alternative mechanisms, probably involving genetic (homologous) recombination [4], which is elevated in most immortal/cancer cell lines [5]. We have found that telomerase physically interacts with recombinase family of proteins and inhibitors of homologous recombination reducing telomere length in telomerase positive Barrett’s adenocarcinoma cells (unpublished data from our laboratory). This suggests that recombinational repair is closely connected to telomere maintenance.
    Figure 1
    Telomeres, the DNA–protein structures which protect chromosomesOur chromosomes end with repeats of conserved ‘TTAGGG’ sequence. These sequences interact with specific proteins and attain a looped conformation which protects chromosomal DNA from degradation. The length of telomeric DNA shortens with each cell division and when it reaches below a critical limit, the cell undergoes replicative senescence or apoptotic cell death. The length of telomeric DNA determines the lifespan of a cell in culture.



    Figure 2
    Length of telomeric DNA is important for lifespan of a cell(a) Telomere length can be prevented from shortening by an enzyme Telomerase. Telomerase has a protein subunit (hTERT) and an RNA subunit (hTR). This enzyme is active in germline and stem cells and maintains their telomere length by adding ‘TTAGGG’ repeats to the ends of chromosomes. Therefore, telomeres do not shorten in these types of cells. (b) Telomerase is inactive in normal somatic cells. These cells, therefore, lose telomeres over time and when telomere length reaches below a critical limit, cells either senesce or die. (c) In the absence of appropriate signals for senescence or apoptotic death, continued cell division leads to severe telomere shortening and genomic instability. Although rare, but cells which survive this crisis, activate a telomere maintenance mechanism (either telomerase or homologous recombination-based ALT) and may become oncogenic. Therefore, most cancer cells have very short but stable telomeres. TA, telomere attrition; TL, telomere length.




    Telomere shortening, cancer, and aging

    Telomeres shorten with age and rate of telomere shortening may indicate the pace of aging.
    Telomere length decreases with age and may predict lifespan

    Normal diploid cells lose telomeres with each cell division and therefore have a limited lifespan in culture. Human liver tissues have been reported to lose 55 base pairs of telomeric DNA per year [6]. Rate of telomere shortening in rapidly renewing gastric mucosal cells is also similar to that observed for liver tissue. The expression of stathmin and EF-1a, the biomarkers for telomeric dysfunction and DNA damage in a cell, increases with age and age-related diseases in humans [7,8]. Telomere length negatively correlates with age whereas the expression of p16, which increases in aging cells, positively correlates with age [7,8].
    Accelerated telomere shortening in genetic disorder dyskeratosis congenital is associated with an early onset of several age-associated disorders and reduced lifespan. Telomerase activity, the ability to add telomeric repeats to the chromosome ends, is present in germline, hematopoietic, stem, and certain other rapidly renewing cells but extremely low or absent in most normal somatic cells. Transgenic induction of a telomerase gene in normal human cells extends their lifespan [9]. Cawthon et al. [10] showed that individuals with shorter telomeres had significantly poor survival due to higher mortality rate caused by heart and infectious diseases. Progressive shortening of telomeres leads to senescence, apoptotic cell death, or oncogenic transformation of somatic cells in various tissues. Telomere length, which can be affected by various lifestyle factors, may determine overall health, lifespan, and the rate at which an individual is aging [11•].

    Accelerated telomere shortening may increase the pace of aging

    As a normal cellular process, telomere length decreases with age [12,13]. Telomere length in humans seems to decrease at a rate of 24.8–27.7 base pairs per year [12,13]. Telomere length, shorter than the average telomere length for a specific age group, has been associated with increased incidence of age-related diseases and/or decreased lifespan in humans [10,14,15]. Telomere length is affected by a combination of factors including donor age [16], genetic, epigenetic make-up and environment [1720], social and economic status [21,22], exercise [21], body weight [12,23], and smoking [12,24]. Gender does not seem to have any significant effect on the rate of telomere loss [13]. When telomere length reaches below a critical limit, the cells undergo senescence and/or apoptosis [25,26].
    Certain lifestyle factors such as smoking, obesity, lack of exercise, and consumption of unhealthy diet can increase the pace of telomere shortening, leading to illness and/or premature death. Accelerated telomere shortening is associated with early onset of many age-associated health problems, including coronary heart disease [2729], heart failure [30], diabetes [31], increased cancer risk [32,33], and osteoporosis [34]. The individuals whose leukocyte telomeres are shorter than the corresponding average telomere length have three-fold higher risk to develop myocardial infarction [13]. Evaluation of telomere length in elders shows that the individuals with shorter telomeres have a much higher rate of mortality than those with longer telomeres [10]. Excessive or accelerated telomere shortening can affect health and lifespan at multiple levels. Shorter telomeres can also induce genomic instability [35,36] by mediating interchromosomal fusion and may contribute to telomere stabilization and development of cancer [36,37]. Consistently, telomerase activity in most cancer cells is elevated whereas telomere length is shorter, relative to corresponding control cells. We have shown that telomere length is shorter in cancer cell lines and primary cancer cells purified by laser capture microdissection [38,39]. However, inhibition of telomere maintenance mechanisms and continued telomere shortening induces senescence and/or apoptosis in immortal/cancer cells [3846].
    Several studies indicate that shorter telomeres are a risk factor for cancer. Individuals with shorter telomeres seem to have a greater risk for development of lung, bladder, renal cell, gastrointestinal, and head and neck cancers [32,33]. Certain individuals may also be born with shorter telomeres or may have genetic disorder leading to shorter telomeres. Such individuals are at a greater risk to develop premature coronary heart disease [13,28] and premature aging. Deficiency of telomerase RNA gene in a genetic disorder dyskeratosis congenita leads to shorter telomeres and is associated with premature graying, predisposition to cancer, vulnerability to infections, progressive bone marrow failure, and premature death in adults [47].


    Impact of smoking and obesity on telomeres and aging

    Smoking and obesity seem to have adverse effect on telomeres and aging.
    Smoking may expedite telomere shortening and process of aging

    Excessive telomere shortening can also lead to genomic instability [35,36] and tumorigenesis [36,37]. Consistently, the telomeres in most cancer cells are shorter relative to normal cells. Smoking is associated with accelerated telomere shortening [8]. The dosage of cigarette smoking is shown to negatively correlate with telomere length [8]. A dose-dependent increase in telomere shortening has been observed in blood cells of tobacco smokers [33,48]. A study conducted in white blood cells of women indicates that telomeric DNA is lost at an average rate of ‘25.7–27.7 base pairs’ per year and with daily smoking of each pack of cigarettes, an additional ‘5 base pairs’ is lost [12]. Therefore, the telomere attrition caused by smoking one pack of cigarettes a day for a period of 40 years is equivalent to 7.4 years of life [12]. Babizhayev et al. [11•] have proposed that telomere length can serve as a biomarker for evaluation of the oxidative damage caused by smoking and may also predict the rate at which an individual is aging. The authors also propose that oxidative damage leading to telomere shortening can be prevented by antioxidant therapy [11•]. In summary, the smoking increases oxidative stress, expedites telomere shortening, and may increase the pace of aging process.

    Obesity is associated with excessive telomere shortening

    Obesity is also associated with increased oxidative stress and DNA damage. Furukawa et al.[49] showed that the waist circumference and BMI significantly correlate with the elevated plasma and urinary levels of reactive oxygen species. Song et al. [8] have shown that BMI strongly correlates with biomarkers of DNA damage, independent of age. The obesity related increased oxidative stress is probably due to a deregulated production of adipocytokines. Obese KKAy mice display higher plasma levels of reactive oxygen species and lipid peroxidation, relative to control C57BL/6 mice [49]. The elevated levels of reactive oxygen species in obese mice were detected in white adipose tissue but not in other tissues, indicating that the oxidative stress detected in plasma could be attributed to oxidizing agents produced in the fat tissue. Moreover, the transcript levels and activities of antioxidant enzymes including catalase and dismutase were significantly lower in white adipose tissue of obese relative to control mice. The authors propose that a lack of antioxidant defense and elevated NADPH oxidase pathway in accrued fat probably led to increased oxidative stress in obese animals. Oxidative stress can induce DNA damage and may therefore expedite telomere shortening. Telomeres in obese women have been shown to be significantly shorter than those in lean women of the same age group [12]. The excessive loss of telomeres in obese individuals was calculated to be equivalent to 8.8 years of life, an effect which seems to be worse than smoking. Together these data indicate that obesity has a negative impact on telomeres and may unnecessarily expedite the process of aging.


    Impact of environment, nature of work, and stress on telomeres and aging

    Environment, nature of profession, and stress can also affect the rate of telomere shortening and health.
    Exposure to harmful agents and nature of profession may affect telomere shortening

    Hoxha et al. [50•] evaluated telomere length in the leukocytes derived from office workers and traffic police officers exposed to traffic pollution. Exposure to pollution was indicated by the levels of toluene and benzene. The investigators found that telomere length in traffic police officers was shorter within each age group, relative to telomere length in office workers. Similarly the lymphocytes of coke-oven workers, exposed to polycyclic aromatic hydrocarbons, had significantly shorter telomeres and increased evidence of DNA damage and genetic instability, relative to control subjects [51••]. Reduction in telomere length in these workers, although did not correlate with age and markers of DNA damage, significantly correlated with the number of years the workers were exposed to harmful agents. Telomere attrition has been associated with increased cancer risk [32,33] and coke-oven workers are at a greater risk to develop lung cancer. Telomere attrition in lymphocytes is also associated with aging [16]. Consistently, the reduced telomere length in the lymphocytes of coke-oven workers was also associated with hypomethylation of p53 promoter [51••], which may induce the expression of p53 [52], leading to inhibition of growth or induction of apoptosis [36]. Thus the exposure to genotoxic agents, which may induce damage to DNA in general or more extensively at telomeres, can increase cancer risk and pace of aging.

    Stress increases the pace of telomere shortening and aging

    The stress is associated with release of glucocorticoid hormones by the adrenal gland. These hormones have been shown to reduce the levels of antioxidant proteins [53] and may therefore cause increased oxidative damage to DNA [54] and accelerated telomere shortening [55]. Consistently, the women, exposed to stress in their daily life, had evidence of increased oxidative pressure, reduced telomerase activity, and shorter telomeres in peripheral blood mononuclear cells, relative to the women in the control group [56]. Importantly, the difference in telomere length in these two groups of women was equivalent to 10 years of life, indicating that the women under stress were at a risk for early onset of age-related health problems. Because telomere length may indicate an individual’s biological age, the stress would adversely affect health and longevity.


    Impact of diet, dietary restriction, and exercise on telomeres and aging

    What we eat and how much we eat can significantly affect our telomeres, health, and longevity.
    Impact of fiber, fat, and protein on telomeres

    Cassidy et al. [57••] studied the association of leukocyte telomere length with various lifestyle factors in a relatively large group of women. Telomere length positively correlated with dietary intake of fiber and negatively associated with waist circumference and dietary intake of polyunsaturated fatty acids, especially linoleic acid. Reduction in protein intake of food also seems to increase longevity. Reduction in the protein content of food by 40%, led to a 15% increase in the lifespan of rats. The rats subjected to a protein-restricted diet early in life displayed a long-term suppression of appetite, reduced growth rate, and increased lifespan [58,59], and the increased lifespan in such animals was associated with significantly longer telomeres in kidney [58]. Consistently, the highest life expectancy of Japanese is associated with low protein and high-carbohydrate intake in diet. The source of protein also seems to be an important factor as replacing casein with the soy protein in rats, is associated with delayed incidence of chronic nephropathies and increased lifespan.

    Dietary intake of antioxidants reduces the rate of telomere shortening

    A study by Farzaneh-Far et al. [60] indicates that a diet containing antioxidant omega-3 fatty acids is associated with reduced rate of telomere shortening, whereas a lack of these antioxidants correlates with increased rate of telomere attrition in study participants. The authors followed omega-3 fatty acid levels in blood and telomere length in these individuals over a period of 5 years and found an inverse correlation, indicating that antioxidants reduce the rate of telomere shortening. Similarly, the women who consumed a diet lacking antioxidants had shorter telomeres and a moderate risk for development of breast cancer, whereas the consumption of a diet rich in antioxidants such as vitamin E, vitamin C, and beta-carotene was associated with longer telomeres and lower risk of breast cancer [61]. Antioxidants can potentially protect telomeric DNA from oxidative damage caused by extrinsic and intrinsic DNA damaging agents.


    Dietary restriction reduces the pace of aging

    Dietary restriction or eating less has an extremely positive impact on health and longevity. Reducing food intake in animals leads to reduced growth rate [58,59], reduced oxidative burden and reduced damage to DNA [59], and therefore keeps the animals in a biologically younger state and can increase their lifespan by up to 66% [59]. It has been shown that dietary restriction in rodents delays the onset of age-associated diseases and increases the lifespan. Rats subjected to a protein-restricted diet early in life displayed a long-term suppression of appetite, reduced growth rate, and increased lifespan [58,59]. The increased lifespan in such animals was associated with significantly longer telomeres in kidney [58]. Because oxidative stress can substantially accelerate telomere shortening, the reduction in oxidative stress by dietary restriction is expected to preserve telomeres and other cellular components.

    Exercise may preserve telomeres and reduce the pace of aging

    Song et al. [8] have demonstrated that duration of exercise inversely correlates with biomarkers for damage to DNA and telomeres and with p16 expression, a biomarker for aging human cell. Exercise can reduce harmful fat and help mobilize waste products for faster elimination, leading to reduced oxidative stress and preservation of DNA and telomeres. Werner et al. [62•] showed that exercise was associated with elevated telomerase activity and suppression of several apoptosis proteins, including p53 and p16, in mice. Consistently, in humans the leukocytes derived from athletes had elevated telomerase activity and reduced telomere shortening, relative to nonathletes [62•]. Exercise seems to be associated with reduced oxidative stress and elevated expression of telomere stabilizing proteins and may therefore reduce the pace of aging and age-associated diseases.

    Conclusion

    Telomeres shorten with age and progressive telomere shortening leads to senescence and/or apoptosis. Shorter telomeres have also been implicated in genomic instability and oncogenesis. Older people with shorter telomeres have three and eight times increased risk to die from heart and infectious diseases, respectively. Rate of telomere shortening is therefore critical to an individual’s health and pace of aging. Smoking, exposure to pollution, a lack of physical activity, obesity, stress, and an unhealthy diet increase oxidative burden and the rate of telomere shortening. To preserve telomeres and reduce cancer risk and pace of aging, we may consider to eat less; include antioxidants, fiber, soy protein and healthy fats (derived from avocados, fish, and nuts) in our diet; and stay lean, active, healthy, and stress-free through regular exercise and meditation. Foods such as tuna, salmon, herring, mackerel, halibut, anchovies, cat-fish, grouper, flounder, flax seeds, chia seeds, sesame seeds, kiwi, black raspberries, lingonberry, green tea, broccoli, sprouts, red grapes, tomatoes, olive fruit, and other vitamin C-rich and E-rich foods are a good source of antioxidants. These combined with a Mediterranean type of diet containing fruits, and whole grains would help protect telomeres.

    Key points

    • Telomere length shortens with age.
    • Rate of telomere shortening may indicate the pace of aging.
    • Lifestyle factors such as smoking, lack of physical activity, obesity, stress, exposure to pollution, etc. can potentially increase the rate of telomere shortening, cancer risk, and pace of aging.
    • Dietary restriction, appropriate diet (high fiber, plenty of antioxidants, lean/low protein, adding soy protein to diet), and regular exercise can potentially reduce the rate of telomere shortening, disease risk, and pace of aging.




    Acknowledgments

    I would like to extend my gratitude to Dr Nikhil C. Munshi for his support. Research work conducted in my laboratory and some of the work discussed here is supported in part by National Institutes of Health grants ‘R01CA125711’ to MAS and ‘R01CA124929’ to NCM.


    References and recommended reading

    Papers of particular interest, published within the annual period of review, have been highlighted as:
    • of special interest
    •• of outstanding interest
    Additional references related to this topic can also be found in the Current World Literature section in this issue (pp. 101–102).
    1. Van Steensel B, Smogorzewska A, de Lange T. TRF2 protects human telomeres from end-to-end fusions. Cell. 1998;92:401–413. [PubMed]
    2. Griffith JD, Comeau L, Rosenfield S, et al. Mammalian telomeres end in a large duplex loop. Cell. 1999;97:503–514. [PubMed]
    3. Shin JS, Hong A, Solomon MJ, Lee CS. The role of telomeres and telomerase in the pathology of human cancer and aging. Pathology. 2006;38:103–113. [PubMed]
    4. Dunham MA, Neumann AA, Fasching CL, Reddel RR. Telomere maintenance by recombination in human cells. Nat Genet. 2000;26:447–450. [PubMed]
    5. Shammas MA, Shmookler Reis RJ, Koley H, et al. Dysfunctional homologous recombination mediates genomic instability and progression in myeloma. Blood. 2009;113:2290–2297.[PMC free article] [PubMed]
    6. Takubo K, Nakamura K, Izumiyama N, et al. Telomere shortening with aging in human liver. J Gerontol A Biol Sci Med Sci. 2000;55:B533–B536. [PubMed]
    7. Jiang H, Schiffer E, Song Z, et al. Proteins induced by telomere dysfunction and DNA damage represent biomarkers of human aging and disease. Proc Natl Acad Sci U S A. 2008;105:11299–11304. [PMC free article] [PubMed]
    8. Song Z, von Figura G, Liu Y, et al. Lifestyle impacts on the aging-associated expression of biomarkers of DNA damage and telomere dysfunction in human blood. Aging Cell. 2010;9:607–615. [PMC free article] [PubMed]
    9. Bodnar AG, Ouellette M, Frolkis M, et al. Extension of life-span by introduction of telomerase into normal human cells. Science. 1998;279:349–352. [PubMed]
    10. Cawthon RM, Smith KR, O’Brien E, et al. Association between telomere length in blood and mortality in people aged 60 years or older. Lancet. 2003;361:393–395. [PubMed]
    11•. Babizhayev MA, Savel?yeva EL, Moskvina SN, Yegorov YE. Telomere length is a biomarker of cumulative oxidative stress, biologic age, and an independent predictor of survival and therapeutic treatment requirement associated with smoking behavior. Am J Therapeutics. 2010 [Epub ahead of print]. This is an important paper indicating that telomere length can serve as a marker of overall oxidative stress may therefore be utilized as a diagnostic tool to assess the damage caused to DNA and telomeres in smokers and people exposed to similar environments/pollution, and so on. [PubMed]
    12. Valdes AM, Andrew T, Gardner JP, et al. Obesity, cigarette smoking, and telomere length in women. Lancet. 2005;366:662–664. [PubMed]
    13. Brouilette S, Singh RK, Thompson JR, et al. White cell telomere length and risk of premature myocardial infarction. Arterioscler Thromb Vasc Biol. 2003;23:842–846. [PubMed]
    14. Farzaneh-Far R, Cawthon RM, Na B, et al. Prognostic value of leukocyte telomere length in patients with stable coronary artery disease: data from the Heart and Soul Study. Arterioscler Thromb Vasc Biol. 2008;28:1379–1384. [PMC free article] [PubMed]
    15. Yang Z, Huang X, Jiang H, et al. Short telomeres and prognosis of hypertension in a Chinese population. Hypertension. 2009;53:639–645. [PMC free article] [PubMed]
    16. Frenck RW, Jr, Blackburn EH, Shannon KM. The rate of telomere sequence loss in human leukocytes varies with age. Proc Natl Acad Sci U S A. 1998;95:5607–5610. [PMC free article][PubMed]
    17. Steinert S, Shay JW, Wright WE. Modification of subtelomeric DNA. Mol Cell Biol. 2004;24:4571–4580. [PMC free article] [PubMed]
    18. Munoz P, Blanco R, Flores JM, Blasco MA. XPF nuclease-dependent telomere loss and increased DNA damage in mice overexpressing TRF2 result in premature aging and cancer. Nat Genet. 2005;37:1063–1071. [PubMed]
    19. Celli GB, de Lange T. DNA processing is not required for ATM mediated telomere damage response after TRF2 deletion. Nat Cell Biol. 2005;7:712–718. [PubMed]
    20. Benetti R, Garcia-Cao M, Blasco MA. Telomere length regulates the epigenetic status of mammalian telomeres and subtelomeres. Nat Genet. 2007;39:243–250. [PubMed]
    21. Cherkas LF, Hunkin JL, Kato BS, et al. The association between physical activity in leisure time and leukocyte telomere length. Arch Intern Med. 2008;168:154–158. [PubMed]
    22. Adams J, Martin-Ruiz C, Pearce MS, et al. No association between socioeconomic status and white blood cell telomere length. Aging Cell. 2007;6:125–128. [PubMed]
    23. Nordfjall K, Eliasson M, Stegmayr B, et al. Telomere length is associated with obesity parameters but with a gender difference. Obesity (Silver Spring) 2008;16:2682–2689. [PubMed]
    24. Nawrot TS, Staessen JA, Gardner JP, Aviv A. Telomere length and possible link to X chromosome. Lancet. 2004;363:507–510. [PubMed]
    25. Gong JG, Costanzo A, Yang HQ, et al. The tyrosine kinase c-Abl regulates p73 in apoptotic response to cisplatin-induced DNA damage. Nature. 1999;399:806–809. [PubMed]
    26. Stiewe T, Putzer BM. p73 in apoptosis. Apoptosis. 2001;6:447–452. [PubMed]
    27. Fitzpatrick AL, Kronmal RA, Gardner JP, et al. Leukocyte telomere length and cardiovascular disease in the cardiovascular health study. Am J Epidemiol. 2007;165:14–21.[PubMed]
    28. Brouilette SW, Moore JS, McMahon AD, et al. Telomere length, risk of coronary heart disease, and statin treatment in the West of Scotland Primary Prevention Study: a nested case-control study. Lancet. 2007;369:107–114. [PubMed]
    29. Zee RY, Michaud SE, Germer S, Ridker PM. Association of shorter mean telomere length with risk of incident myocardial infarction: a prospective, nested case-control approach. Clin Chim Acta. 2009;403:139–141. [PMC free article] [PubMed]
    30. Van der Harst P, van der Steege G, de Boer RA, et al. Telomere length of circulating leukocytes is decreased in patients with chronic heart failure. J Am Coll Cardiol. 2007;49:1459–1464. [PubMed]
    31. Sampson MJ, Winterbone MS, Hughes JC, et al. Monocyte telomere shortening and oxidative DNA damage in type 2 diabetes. Diabetes Care. 2006;29:283–289. [PubMed]
    32. Wu X, Amos CI, Zhu Y, et al. Telomere dysfunction: a potential cancer predisposition factor. J Natl Cancer Inst. 2003;95:1211–1218. [PubMed]
    33. McGrath M, Wong JY, Michaud D, et al. Telomere length, cigarette smoking, and bladder cancer risk in men and women. Cancer Epidemiol Biomarkers Prev. 2007;16:815–819.[PubMed]
    34. Valdes AM, Richards JB, Gardner JP, et al. Telomere length in leukocytes correlates with bone mineral density and is shorter in women with osteoporosis. Osteoporos Int. 2007;18:1203–1210. [PubMed]
    35. De Lange T. Telomere-related genome instability in cancer. Cold Spring Harb Symp Quant Biol. 2005;70:197–204. [PubMed]
    36. Chin L, Artandi SE, Shen Q, et al. p53 deficiency rescues the adverse effects of telomere loss and cooperates with telomere dysfunction to accelerate carcinogenesis. Cell. 1999;97:527–538. [PubMed]
    37. Meeker AK. Telomeres and telomerase in prostatic intraepithelial neoplasia and prostate cancer biology. Urol Oncol. 2006;24:122–130. [PubMed]
    38. Shammas MA, Qazi A, Batchu RB, et al. Telomere maintenance in LCM purified Barrett’s adenocarcinoma cells and impact of telomerase inhibition in vivo. Clin Cancer Res. 2008;14:4971–4980. [PMC free article] [PubMed]
    39. Shammas MA, Shmookler Reis RJ, Koley H, et al. Telomerase inhibition and cell growth arrest following porphyrin treatment of multiple myeloma cells. Mol Cancer Therapeutics. 2003;2:825–833. [PubMed]
    40. Shammas MA, Simmons CG, Corey D, Shmookler Reis RJ. Telomerase inhibition by peptide nucleic acids reverses ‘immortality’ of transformed human cells. Oncogene. 1999;18:6191–6200. [PubMed]
    41. Akiyama M, Hideshima T, Shammas MA, et al. Molecular sequelae of oligonucleotide N3′-P5′ phosphoramidate targeting telomerase RNA in human multiple myeloma cells. Cancer Research. 2003;63:6187–6194. [PubMed]
    42. Shammas MA, Koley H, Beer David G, et al. Growth arrest, apoptosis and telomere shortening of Barrett’s associated adenocarcinoma cells by a telomerase inhibitor. Gastroenterology. 2004;126:1337–1346. [PubMed]
    43. Shammas MA, Raney KD, Subramanian S, Shmookler Reis RJ. Telomere length, cell growth potential, and DNA integrity of human immortal cells are all compromised by peptide nucleic acids targeted to the telomere or telomerase. Exp Cell Res. 2004;295:204–214.
    44. Shammas MA, Shmookler Reis RJ, Koley H, et al. Telomerase inhibition and cell growth arrest following telomestatin treatment of multiple myeloma cells. Clin Cancer Res. 2004;10:770–776. [PubMed]
    45. Shammas MA, Koley H, Protopopov A, et al. Telomerase inhibition by siRNA causes senescence and apoptosis in Barrett’s adenocarcinoma cells: mechanism and therapeutic potential. Mol Cancer. 2005;4:24. [PMC free article] [PubMed]
    46. Shammas MA, Koley H, Protopopov A, et al. Telomerase inhibition, telomere shortening and apoptotic cell death in multiple myeloma cells following exposure to a novel and potent telomerase inhibitor (GRN163L), targeting RNA component of telomerase. Leukemia. 2008;22:1410–1418.
    47. Vulliamy T, Marrone A, Goldman F, et al. The RNA component of telomerase is mutated in autosomal dominant dyskeratosis congenita. Nature. 2001;413:432–435. [PubMed]
    48. Morlá M, Busquets X, Pons J, et al. Telomere shortening in smokers with and without COPD. Eur Respir J. 2006;27:525–528. [PubMed]
    49. Furukawa S, Fujita T, Shimabukuro M, et al. Increased oxidative stress in obesity and its impact on metabolic syndrome. J Clin Invest. 2004;114:1752–1761. [PMC free article][PubMed]
    50•. Hoxha M, Dioni L, Bonzini M, et al. Association between leukocyte telomere shortening and exposure to traffic pollution: a cross-sectional study on traffic officers and indoor office workers. Environ Health. 2009;8:41. This is an interesting paper which compares telomere length among office workers and police officers exposed to traffic pollution, indicating the possible impact of pollution on health and aging. [PMC free article] [PubMed]
    51••. Pavanello S, Pesatori AC, Dioni L, et al. Shorter telomere length in peripheral blood lymphocytes of workers exposed to polycyclic aromatic hydrocarbons. Carcinogenesis. 2010;31:216–221. This is an important paper which shows that telomere length in coke-oven workers, exposed to polycyclic aromatic hydrocarbons, is reduced relative to controls, and the reduction in telomeres correlates with the duration of exposure. [PMC free article] [PubMed]
    52. Esteller M. Epigenetics provides a new generation of oncogenes and tumour-suppressor genes. Br J Cancer. 2006;94:179–183. [PMC free article] [PubMed]
    53. Patel R, McIntosh L, McLaughlin J, et al. Disruptive effects of glucocorticoids on glutathione peroxidase biochemistry in hippocampal cultures. J Neurochem. 2002;82:118–125.[PubMed]
    54. Irie M, Asami S, Ikeda M, Kasai H. Depressive state relates to female oxidative DNA damage via neutrophil activation. Biochem Biophys Res Commun. 2003;311:1014–1018.[PubMed]
    55. Von Zglinicki T. Oxidative stress shortens telomeres. Trends Biochem Sci. 2002;27:339–344. [PubMed]
    56. Epel ES, Blackburn EH, Lin J, et al. Accelerated telomere shortening in response to life stress. Proc Natl Acad Sci U S A. 2004;101:17312–17315. [PMC free article] [PubMed]
    57••. Cassidy A, De Vivo I, Liu Y, et al. Associations between diet, lifestyle factors, and telomere length in women. Am J Clin Nutr. 2010;91:1273–1280. This is an important paper in which the relationship of diet and other lifestyle factors with telomeres has been studied in a large group of women. The paper describes the positive impact of fiber and negative effect of polyunsaturated fat on telomeres. This study did not find any association of telomere length with physical activity and smoking. [PMC free article] [PubMed]
    58. Jennings BJ, Ozanne SE, Dorling MW, Hales CN. Early growth determines longevity in male rats and may be related to telomere shortening in the kidney. FEBS Lett. 1999;448:4–8.[PubMed]
    59. Jennings BJ, Ozanne SE, Hales CN. Nutrition, oxidative damage, telomere shortening, and cellular senescence: individual or connected agents of aging? Mol Genet Metab. 2000;71:32–42.[PubMed]
    60. Farzaneh-Far R, Lin J, Epel ES, et al. Association with marine omega-2-fatty acid levels with telomeric aging in patients with coronary heart disease. AMA. 2010;303:250–257.[PMC free article] [PubMed]
    61. Gammon SJ, Terry MB, Wang Q, et al. Telomere length, oxidative damage, and antioxidant breast cancer risk. Int J Cancer. 2009;124:1637–1643. [PMC free article] [PubMed]
    62•. Werner C, Fürster T, Widmann T, et al. Physical exercise prevents cellular senescence in circulating leukocytes and in the vessel wall. Circulation. 2009;120:2438–2447. This paper demonstrates the positive impact of exercise on telomeres, in both the mice and humans.[PubMed]

    www.popservice.ro
    www.papornitamosului.ro
    [U][COLOR=#800080][
    e-mail - adipop@popservice.ro
    ID Messenger: zalmoxa_adipop
    skype - adrianpop58
    http://www.sfatulbatranilor.ro/forum.php
    https://www.facebook.com/groups/611112328972709/ - REGIA DE RECONSTRUCTIE A ROMANIEI
    https:/https://www.facebook.com/grou...IZEGETUSA2050/ - Piciumanii doresc PACE
    https://www.facebook.com/groups/1086016084901078/ - Avangarda de Sacrificiu

  3. #3
    Administrator
    Data înscrierii
    10.10.2011
    Locație
    CRAIOVA
    Posturi
    78.785
    www.popservice.ro
    www.papornitamosului.ro
    [U][COLOR=#800080][
    e-mail - adipop@popservice.ro
    ID Messenger: zalmoxa_adipop
    skype - adrianpop58
    http://www.sfatulbatranilor.ro/forum.php
    https://www.facebook.com/groups/611112328972709/ - REGIA DE RECONSTRUCTIE A ROMANIEI
    https:/https://www.facebook.com/grou...IZEGETUSA2050/ - Piciumanii doresc PACE
    https://www.facebook.com/groups/1086016084901078/ - Avangarda de Sacrificiu

  4. #4
    Administrator
    Data înscrierii
    10.10.2011
    Locație
    CRAIOVA
    Posturi
    78.785
    http://www.sfatulbatranilor.ro/threa...-and-longevity


    MULTUMESC !


    Sarut maini Doamnei !


    Cele bune,
    adi
    www.popservice.ro
    www.papornitamosului.ro
    [U][COLOR=#800080][
    e-mail - adipop@popservice.ro
    ID Messenger: zalmoxa_adipop
    skype - adrianpop58
    http://www.sfatulbatranilor.ro/forum.php
    https://www.facebook.com/groups/611112328972709/ - REGIA DE RECONSTRUCTIE A ROMANIEI
    https:/https://www.facebook.com/grou...IZEGETUSA2050/ - Piciumanii doresc PACE
    https://www.facebook.com/groups/1086016084901078/ - Avangarda de Sacrificiu

Informații subiect

Utilizatori care navighează în acest subiect

Momentan sunt 1 utilizatori care navighează în acest subiect. (0 membri și 1 vizitatori)

Marcaje

Marcaje

Permisiuni postare

  • Nu poți posta subiecte noi
  • Nu poți răspunde la subiecte
  • Nu poți adăuga atașamente
  • Nu poți edita posturile proprii
  •