Telomere, aging and age-related diseases - Springer Link

2 downloads 0 Views 204KB Size Report
Apr 3, 2013 - Thus, the loss of telomeres may lead to DNA damage. The relationship ... Many theories have been proposed to explain the aging process.
Aging Clin Exp Res (2013) 25:139–146 DOI 10.1007/s40520-013-0021-1

MINI REVIEW

Telomere, aging and age-related diseases Huanjiu Xi • Changyong Li • Fu Ren Hailong Zhang • Luping Zhang



Received: 11 July 2011 / Accepted: 30 March 2012 / Published online: 3 April 2013 Ó Springer International Publishing Switzerland 2013

Abstract Aging is an inevitable biological process that affects most living organisms. The process of aging is regulated at the level of the organism, as well as at the level of tissues and cells. Despite the enormous consequences associated with the aging process, relatively little systematic effort has been expended on the scientific understanding of this important life process. Many theories have been proposed to explain the aging process, the centerpiece of which is molecular damage. Located at the ends of eukaryotic chromosomes and synthesized by telomerase, telomeres maintain the stabilization of chromosomes. Thus, the loss of telomeres may lead to DNA damage. The relationship between cellular senescence and telomere shortening is well established. Furthermore, telomere attrition occurs with age, and is proposed to be a fundamental factor in the aging process. Here, we review the contemporary literatures to explore the current views on the correlation of telomere loss and telomerase action with aging and age-related diseases. Keywords Telomere  Telomerase  Aging  Cell senescence  Age-related diseases

Introduction Aging is characterized as degenerative changes progressively occurring in most cells, organisms and species. The nature of the aging process has been the subject of considerable speculation. Over the past four decades, there has been a concerted effort, at the molecular level, to understand better the changes that take place during aging. Observations from these studies have implicated aging as a genetically programed process where the total life span of an organism is remarkably constant and species specific. Many theories have been proposed to explain the aging process. Some researchers have suggested that telomeres play an important role in cellular senescence [1–3]. Indeed, of all the various theories proposed, the telomere hypothesis has become the recent focus of debate. Telomere length shortening has been recognized as not only a marker of biological aging but also reveals an important role in multiple aging-related diseases [2, 4–7]. The aim of this review is to summarize the relationships between telomere loss and human aging and age-related diseases.

Telomere and cell senescence H. Xi (&)  F. Ren  H. Zhang Institute of Anthropology, Liaoning Medical University, No.40, Section 3, Songpo Road, Jinzhou, Liaoning 121001, People’s Republic of China e-mail: [email protected] C. Li Department of Physiology, School of Basic Medical Sciences, Wuhan University, Hubei 430071, People’s Republic of China L. Zhang Department of Anatomy, Second Military Medical University, Shanghai 200433, People’s Republic of China

Telomere structure and the length regulation Telomeres, located at the ends of chromosomes in eukaryotes, are composed of TTAGGG repeats that are extended 9–15 kb, which can protect the chromosomes from degradation, fusion and recombination [8]. Normal telomeres do not trigger a DNA damage response. Extensive studies in unicellular organisms have shown that telomere function requires both specific DNA sequences and specific proteins [9]. The current telomere model

123

140

includes a telomere (T)-loop, and a displacement (D)-loop associated with telomere-binding proteins. As a protective cap, this structure can prevent the chromosome from being recognized as DNA damnification by DNA repair complexes, which would subsequently result in end-to-end fusion, recombination and degradation, and thus lead to chromosome instability and cell death [10]. The regulation of telomere length is a complex process, requiring the telomerase enzyme complex and numerous regulatory proteins. The shelterin complex consists of six proteins, TRF1, TRF2, RAP1, TIN2, TPP1 and POT1. It binds to both the duplex and ssDNA regions through specific protein–DNA interactions and shelters the telomere from a series of unwanted activities [11]. The two major telomerebinding proteins are telomeric repeat binding factor 1 (TRF1) and 2 (TRF2) [12]. They can function individually or interact with other binding proteins, such as POT1, Rap1, TIN2, hRap1, Ku86 and the Mre11/Rad50/Nbs1 DNA repair complex [13]. These binding proteins both play an important role in the regulation of telomere length. TRF1 is a negative regulator of telomere length, which depends on a feedback mechanism involving telomerase, whilst TRF2 is found in the double-strand T-loop and in the loop tail junction, and probably acts in the stabilization of the G-strand overhang at D-loop [12]. More recently, ataxia telangiectasia mutated (ATM), a PI-3 kinase essential for maintaining genomic stability, has been shown to regulate proteasome-dependent subnuclear localization of TRF1, which is important for telomere maintenance [14]. Many more proteins involved in the proper function of human telomeres are expected to be identified in the future. Telomerase is a ribonucleoprotein reverse transcriptase which stabilizes telomere length by adding TTAGGG repeats to the ends of the chromosomes. It consists of two main components, telomere RNA, which acts as template for telomere synthesis, and telomere reverse transcriptase, which catalyzes the elongation [15]. Whereas telomerase activity is low or undetectable in most normal somatic cells, it is detected at higher levels both in the germ cells and in the majority of cancer cells [2, 16], in the latter case, the activity of telomerase maintains the length of telomere and confers cancer cells high proliferation capacity. It has been shown that telomerase activity was associated with cell cycle deregulation in human cancer cells and telomerase activity was inversely associated with the level of cell differentiation [17]. Human cells must overcome two barriers to proliferate and achieve immortalization [8]. The first is mortality stage 1, which results in telomere shortening and cell proliferation arrest. Following the first stage, the telomeres continue to shorten and eventually reach the second proliferation barrier, mortality stage 2. Cells with extremely short telomeres then undergo apoptosis. Rarely, cells escape the

123

Aging Clin Exp Res (2013) 25:139–146

crisis and telomerase is activated causing immortalization to occur [18, 19]. Currently, little information is available on how germ cells allow telomerase-mediated telomere elongation which differs from somatic cells. Lanza et al. [20] have indicated that telomeres can also be elongated during development. Research in these areas will eventually elucidate the diversities in both gene and protein expression between the germ cells and somatic cells and the importance of telomere length regulation in the latter. However, the greatest threat to telomere function seems to be the gradual loss of telomere DNA in somatic cells with each round of cell division. The gradual shortening of telomeres is assumed to limit the capability of cells to proliferate, and eventually affect the health and lifespan of an individual [21, 22]. Recently, there has been growing evidence that lifestyle factors may affect human health and lifespan by affecting telomere length [2, 11, 23, 24]. Interestingly, telomerase activity was increased with improvements in nutrition and lifestyle in peripheral blood mononuclear cells [25]. These studies provide compelling reasons to conduct intervention studies to reduce early illness and extend years of healthy living in the future. Optimal diet and healthy lifestyle behaviors have 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. Telomere hypothesis of cell senescence Incomplete replication of the 30 end of telomere DNA is thought to be responsible for the replication-dependent telomere shortening. In most somatic cells, telomeres shorten with successive cell divisions. Therefore, normal somatic cells undergo a finite number of cell divisions, and the maximum number of divisions is known as ‘‘Hayflick limit’’ [26]. The mechanism by which cell division ceases due to telomere shortening is not yet fully understood, although three possible hypotheses have so far been proposed. The first hypothesis suggests that telomeres are characterized by heterochromatin. After the telomere length is shorted, it will attack the nearby DNA and will thus affect the expression of genes that are responsible for the regulation of growth. It has been found that gene transferation near telomere DNA has been restrained in low-level eukaryote. The second hypothesis assumes that the total loss of telomere DNA will produce a damaging signal, which will activate P53 and thus cause an inhibition in growth. The third hypothesis proposes that it is not the damaging signal but the shortened telomere itself that activates P53 and causes permanent restraint of growth. The key mediator of the response to dysfunctional telomere is P53 [16]. Recent

Aging Clin Exp Res (2013) 25:139–146

evidence indicates that at the senescence stage critically short telomeres lose the capping function and this can induce DNA damage, which is partially dependent on the activity of the PI3-kinase-like kinase ATM [27]. Although the precise mechanisms causing dysfunctional telomeres to trigger a DNA damage response are unclear, the telomere erosion that occurs during normal replication induces all of the hallmarks of a DNA damage response [28]. Mutation of ATM also attenuates apoptotic responses to telomere dysfunction caused by dominant negative expression of telomere-binding protein TRF2 [29]. Thus, the disruption of the native telomere structure triggers an active DNA damage response, which is largely indistinguishable from the response to ionizing radiation [1]. Telomere and its complex are involved in biological aging and disease processes. The evidence to support the assumption that telomere shortening is related to aging was put forward by Allsopp et al. [26]. They analyzed human fibroblasts from 31 donors and found relatively weak correlations between proliferative ability and donor age. However, there was a striking correlation between replicative capacity and initial telomere length. Their findings suggested that telomere length may be used as a biomarker of somatic cell aging in humans, which is consistent with the important role of telomere loss involving in mechanisms of aging. Wright et al. [30] subsequently used widowed nucleotide acid to dispose immortal human cells in order to extend telomere length and then integrate them with fatal cells. They found that these crossbreed cells tend to have better longevity than those immortal cells that have not been disposed by widowed nucleotide acid. In this case, Wright et al. provided direct evidence to support the hypothesis that length of telomere determines the multiplying capacity of human cells. An increasing body of evidence have demonstrated that the shortening of telomeres to a critical or threshold length acts as a signal for cell senescence [3, 26]. The replicative capacity was found to be directly proportional to the mean telomere length. More importantly, the variability in mean telomere length at senescence was markedly less than that at early passage. Although initial telomere length cannot account for all of the interclonal variability in replicative capacity, these observations supported the existence of a critical telomere length in senescing cells and an important role of telomere shortening in cell senescence [31]. Importantly, when telomerase was introduced into somatic cells, it extended the proliferative lifespan and prevents replicative senescence in human lens epithelial cells [32]. Moreover, studies have also shown that the replicative life span of various human cells can be prolonged by induced expression of the telomerase reverse transcriptase (hTERT) gene. Bodnar et al. [33] transfected

141

two telomerase-negative normal human cell types (retinal pigment epithelial cells and foreskin fibroblasts) with vectors encoding the human telomerase catalytic subunit. The telomerase-negative control clones exhibited telomere shortening and senescence, while the telomerase-expressing clones had elongated telomeres, divided vigorously, and showed reduced staining for beta-galactosidase, a biomarker for senescence. Notably, the telomeraseexpressing clones exhibited a normal karyotype and have already exceeded their normal life span by at least 20 doublings. Thus, this research establishes a causal relationship between telomere shortening and in vitro cellular senescence. New trends in telomerase manipulation show that hTERT can be used to immortalize a variety of differentiated human cell types including epithelial cells of renal proximal tubules [19, 34]. In humans, telomere length positively correlates with years of healthy life in health, aging, and body composition [35]. Telomere length could be considered as a biological parameter that intertwines replicative history and exposure to environmental stress [7]. In addition, there is a high variance in telomere length in humans. Telomere in female is longer than male and in African Americans than in White Americans [36]. Our own data have also demonstrated that telomeres shorten in human peripheral blood leukocytes in an age-dependent manner, and that telomere length was significantly longer in peripheral blood leukocytes from females than in those from males, specially, in two age groups (5–14 years old and 55–64 years old) [37]. Our findings suggest that estimation of human age according to telomere shortening in peripheral blood leukocytes is a novel method especially when there is no morphologic information in forensic practice [37]. In some types of premature senility syndrome, there is exceptional metabolization occurring with the normal somatic cell division. Vaziri et al. [38] have shown that Down syndrome patients showed a significantly higher rate of telomere loss with donor age compared with age-matched controls and they suggested that accelerated telomere loss is a biomarker of premature immunosenescence of Down syndrome patients and that it may play a role in this process.

Telomere and age-related diseases It is well established that telomere length and telomerase activity are important factors in the pathogenesis of human diseases [2, 6, 7, 39, 40]. Telomerase activation may prove to be useful in the treatment of chronic and degenerative diseases associated with telomere loss [41]. We summarize information on the age-related diseases below, which have been proposed to be related to telomeres.

123

142

Cardiovascular diseases Atherosclerosis is also an aging-related systemic disease. The studies in vitro and vivo showed a tight interplay between cell senescence and atherosclerosis [42]. Telomere length is a new marker of cardiovascular risk [6, 43, 44]. In the vascular endothelium, shorter telomeres are found in those areas of the arterial wall that are more susceptible to atherosclerosis because of higher haemodynamic stress. It is believed that this stress results in a more rapid cell turnover [45]. In addition, vascular endothelial cells and smooth muscle cells in the vicinity of atherosclerotic lesions often stain positive for the activity of b-galactosidase, which is a rather nonspecific marker for cellular senescence [46, 47]. Furthermore, the induction of telomere dysfunction in endothelial cells in vitro generates a senescent phenotype with an atherogenic protein expression profile [46]. Healthy individuals with shorter telomere were likely to develop hypertension, and hypertensive individuals with shorter telomere were more susceptible to develop atherosclerosis [48]. The animal experiments showed that endothelial progenitor cells from hypertensive patients and from spontaneous hypertensive rats exhibit reduced telomerase activity and accelerated senescence [49]. Heart failure is a frequent cause of death in the aging human population. To determine whether telomere shortening leads to a cardiac phenotype, Leri et al. studied heart function in the telomerase knockout mouse. They showed that telomere attrition is a key factor in the attenuation of cardiac myocyte proliferation, myocyte hypertrophy and heart failure. Telomere shortening with age might also contribute to cardiac failure in humans, opening the possibility for new therapies [50]. Chronic heart failure is characterized by increased myocyte apoptosis and telomere erosion [51]. The degree of telomere shortening is associated with the severity of disease [52]. In humans, the formation of myocytes from telomerase-positive cardiac stem cells appears to be necessary for cardiac homeostasis [53]. Endomyocardial tissue from heart failure patients has been shown to have higher levels of both apoptotic and apparently senescent cells, and the telomeres are shorter than their control counterparts [54, 55]. These reports demonstrate that the heart diseases are characterized by shorter telomeres, increased cellular senescence and cell death. Diabetes Type 2 diabetes is caused by a combination of peripheral insulin resistance and b-cell dysfunction. Zee and Salpea et al. [56, 57] showed that short telomere were significantly associated with type 2 diabetes, which could be partially attributed to the high oxidative stress in the patients with type 2 diabetes. Moreover, short telomeres are predictors of

123

Aging Clin Exp Res (2013) 25:139–146

progression of diabetic nephropathy [58] and of all-cause mortality in the patients with type 1 diabetes [59]. In a prospective follow-up study, Astrup et al. [59] found no differences in telomere length between patients with or without diabetic nephropathy. Young adult mice that are deficient in telomerase exhibit impaired glucose tolerance, which suggests short telomere can impair replicative capacity of pancreatic b-cell [60]. Telomeres also were shorter in pre-diabetes patients with impaired glucose tolerance than in healthy controls and in diabetic patients with atherosclerotic plaques than patients without atherosclerosis [61]. Information from the Framingham Heart Study suggests that subclinical presence of insulin resistance was associated with reduced telomere length [62]. The subjects with type 2 diabetes and with atherosclerotic plaques had shorter telomere than those without plaques [61]. Cancer The classical telomere hypothesis suggests that the telomere shortening provides a tumor suppressor mechanism to cease the growth of transformed cells. In normal somatic cells, the absence of telomerase can lead to telomere shortening and cell senescence. However, the majority of cancer cells exhibit a high telomerase activity [16]. The reappearance of telomerase activity is triggered by as yet unclear molecular mechanisms and enables cancer cells to maintain telomere length. Studies have revealed a dual role of telomere shortening in carcinogenesis. On the one hand, shortened telomeres induce chromosomal instability, which is the most important cause of cancer initiation during aging [27]. On the other hand, telomere stabilization is required for tumor progression [27, 63]. The initiation of chromosomal aberrations by telomere shortening might contribute to the increased cancer rate of cancer onset during aging. The necessity of telomere stabilization for tumor progression in most human cancers is achieved by the reactivation of telomerase [63]. However, reports of cancer formation in telomerase transgenic mice suggest that telomerase also exhibits tumor-promoting activity. Several observations on the presence of telomerase in non-transformed human cells [63] and the extra-telomeric function of telomerase [64] might provide some explanations for the tumor initiating activity of telomerase. Further understanding of the genetic alterations that cooperate with telomere dysfunction might help us to find new tumor marker genes and therapeutic targets to prevent and treat cancer in old people. Clinical data showed that telomere length of lymphocytes is shorter in patients with some cancers in the head, neck, breast, bladder, prostate, lung and kidney [65].

Aging Clin Exp Res (2013) 25:139–146

Telomere length undergoes shortening in early stage gastric carcinoma and lengthening in advanced gastric carcinoma. Additionally, telomere shortening may initiate the tumorigenesis of gastric carcinoma [66]. The ongoing basic research in this field helps to constantly define new targets and to increase the specificity, safety and efficacy of existing therapeutic approaches directed against telomeres and telomerase. Hahn et al. [67] have demonstrated that inhibition of telomerase limits the growth of human cancer cells. Therefore, therapies targeting telomerase with telomerase inhibitors revealed the potential for cancer therapy [68–70]. Recent research developments for each of the anti-telomerase approaches including antisense-oligonucleotides, hammerhead ribozymes, dominant negative hTERT, reverse-transcriptase inhibitors, immunotherapy, G-quadruplex stabilisers, gene therapy, small molecule inhibitors and RNA interference have been provided [71]. However, there are several limitations of anti-telomerase cancer therapy. For example, there is a lag phase between telomerase inhibition and the occurrence of senescence as a reversal of the immortal phenotype, and so their use will be probably restricted to enhance the efficiency of other anticancer agents or to prevent cancer relapse following standard therapies. Moreover, the role played by the telomerase-independent mechanism, which was reported to maintain telomere length in cell lines as well as in particular subset of tumors is still unclear. Uncoupling tumor suppression from aging represents undoubtedly one of the most daunting challenges for telomere and cellular senescence research [72]. Despite these problems, it is still exciting to discover that a promising therapeutic vaccine and a telomerase antagonist are now being employed in clinical and preclinical studies. Immune system diseases The immune system is a prime example of a highly dynamic cellular system, for which telomere maintenance is pivotal. Immune competence is strictly dependent on rapid expansions of clonal T- and B-cell populations, and telomere loss may contribute to defective immune responses in the elderly. Equally interestingly, accelerated T-cell aging combined with telomeric shortening may prompt an autoimmune response and thereby explain the increased susceptibility for chronic inflammatory diseases in the elderly [73]. Recent reports have suggested that telomere shortening is involved in the dramatic age-related alterations of the immune system, and this is considered one of the major factors affecting morbidity and mortality [74]. More recent data indicate that telomerase activity and telomere length are modified in various systemic immunemediated diseases and appear to be connected with premature immunosenescence [75].

143

The gradual decline with age in the capacity of the immune system to recognize and eliminate antigens is an important causal factor for many diseases in the elderly [40]. Although both the innate and adaptive immune responses are weakened in old people, it is the decline in T-cell action that proves to have the most injurious effect on the elderly [76]. T cells become clonally exhausted, which leads to a decline in the adaptability of the response of the immune system [77]. The senescence of T cells in vivo is well documented: as the number of population doublings in culture increases then there is a progressive decrease in the proportion of T cells that express CD28. This results in an inability of the T cells to proliferate [78] and a reduction in telomerase induction by CD28, as well as a resistance to apoptosis [79, 80]. In addition, research on bone marrow transplantation has provided important clues for the in vivo interactions of immunosenescence and telomere length and consequences [81]. The main body of clinical evidence that provides a role for telomere erosion in the decline of immunity comes from studies on premature aging syndromes such as ataxia telangiectasia and dyskeratosis congenital where dysfunctional telomeres are associated with a higher susceptibility to infection. In addition, it has been shown that individuals with short telomeres are eight times more likely to die of infectious diseases than those with long telomeres [82]. Dyskeratosis congenital Defective telomere function or mutations in the DNA repair system can induce some human disorders associated with shorter telomere length, such as dyskeratosis congenital (DC) [83]. Most direct evidence for telomere shortening during human aging comes from research on DC, which is a premature aging disease characterized by telomerase and telomere dysfunction [84, 85]. Autosomal dominant dyskeratosis congenita is associated with mutations in the RNA component of telomerase, while X-linked dyskeratosis congenita is due to mutations in the gene encoding dyskerin, a protein implicated in both telomerase function and ribosomal RNA processing [86]. Thus, it might be one of the first diseases where a telomerase activating gene therapy can be implemented. This approach might help to prolong the lifespan of DC patients and might also shed some light on the therapeutic potentials and risks involved in telomerase activation therapy.

A concluding remark Although an increased knowledge about the biomarker value of telomere length in human aging might significantly

123

144

Aging Clin Exp Res (2013) 25:139–146

improve our insight into the pathophysiology of human aging and aging-associated diseases, and yield a valuable risk factor, the true nature of the association remains unclear. Fewer studies have assessed the relationship between telomere length and age-sensitive measures of function that decline with normal aging in human population studies [87]. In addition, it is difficult to explain rationally some of the research results. For example, the ‘‘forced’’ expression of telomerase in cells with normallength telomeres has shown that telomerase promotes growth and survival in a manner that is uncoupled from telomere lengthening. This finding suggests that telomerase might play a fundamental role in tumor growth and survival, even at stages when telomeres are sufficiently long [88]. In addition, telomerase activity is low or undetectable in the endothelial and epithelial cells from the human cornea. These human corneal endothelial cells have long telomeres throughout life and their limited replicative ability does not appear to result from critically short telomere lengths [89]. As we know, the regulation of the process of aging is highly complex, involving a combination of telomere alterations, DNA damage, DNA methylation and cellular oxidation. Many theories have been proposed to explain the aging process. Telomeres have been proposed to play an important role in cellular senescence, and the telomere hypothesis, which is the focus of much debate and some controversy, will manifest an important theoretical and practical significance in anti-aging research. Indeed, the current findings have already inspired a number of potential therapeutic strategies that are based on telomerase and telomeres. Although the impact of these findings remains speculative, it is a hopeful thought that telomere elongation could possibly originate from an increased telomerase activity. Acknowledgments This work was supported by grants from the National Natural and Science Foundation of China (30830062, 30971529, 30840047). Conflict of interest

The authors disclose no conflicts.

References 1. Reaper PM, di Fagagna FD, Jackson SP (2004) Activation of the DNA damage response by telomere attrition—a passage to cellular senescence. Cell Cycle 3:543–546 2. Shammas MA (2011) Telomeres, lifestyle, cancer, and aging. Current Opinion Clin Nutr Metab Care 14:28–34 3. Takubo K, Aida J, Izumiyama-Shimomura N, Ishikawa N, Sawabe M, Kurabayashi R, Shiraishi H et al (2010) Changes of telomere length with aging. Geriatr Gerontol Int 10:S197–S206 4. Aubert G, Lansdorp PM (2008) Telomeres and aging. Physiol Rev 88:557–579

123

5. Wong LSM, van der Harst P, de Boer RA, Huzen J, van Gilst WH, van Veldhuisen DJ (2010) Aging, telomeres and heart failure. Heart Fail Rev 15:479–486 6. Huzen J, de Boer RA, van Veldhuisen DJ, van Gilst WH, van der Harst P (2010) The emerging role of telomere biology in cardiovascular disease. Frontiers Biosci Landmark 15:35–45 7. Oeseburg H, de Boer RA, van Gilst WH, van der Harst P (2010) Telomere biology in healthy aging and disease. Pflugers Arch 459:259–268 8. Lin KW, Yan J (2005) The telomere length dynamic and methods of its assessment. J Cell Mol Med 9:977–989 9. Zakian VA (1996) Structure, function, and replication of Saccharomyces cerevisiae telomeres. Annu Rev Genet 30:141–172 10. Griffith JD, Comeau L, Rosenfield S, Stansel RM, Bianchi A, Moss H, de Lange T (1999) Mammalian telomeres end in a large duplex loop. Cell 97:503–514 11. Stewart JA, Chaiken MF, Wang F, Price CM (2012) Maintaining the end: roles of telomere proteins in end-protection, telomere replication and length regulation. Mutat Res 730:12–19 12. Broccoli D, Smogorzewska A, Chong L, de Lange T (1997) Human telomeres contain two distinct Myb-related proteins, TRF1 and TRF2. Nat Genet 17:231–235 13. Kim SH, Kaminker P, Campisi J (1999) TIN2, a new regulator of telomere length in human cells. Nat Genet 23:405–412 14. McKerlie MA, Lin S, Zhu XD (2012) ATM regulates proteasome-dependent subnuclear localization of TRF1, which is important for telomere maintenance. Nucleic Acids Res 40:3975– 3989 15. Mason M, Schuller A, Skordalakes E (2011) Telomerase structure function. Curr Opin Struct Biol 21:92–100 16. Artandi SE, DePinho RA (2010) Telomeres and telomerase in cancer. Carcinogenesis 31:9–18 17. Landberg G, Nielsen NH, Nilsson P, Emdin SO, Cajander J, Roos G (1997) Telomerase activity is associated with cell cycle deregulation in human breast cancer. Cancer Res 57:549–554 18. Halvorsen TL, Leibowitz G, Levine F (1999) Telomerase activity is sufficient to allow transformed cells to escape from crisis. Mol Cell Biol 19:1864–1870 19. Shay JW, Wright WE (2005) Senescence and immortalization: role of telomeres and telomerase. Carcinogenesis 26:867–874 20. Lanza RP, Cibelli JB, Blackwell C, Cristofalo VJ, Francis MK, Baerlocher GM, Mak J et al (2000) Extension of cell life-span and telomere length in animals cloned from senescent somatic cells. Science 288:665–669 21. Lansdorp PM (2000) Repair of telomeric DNA prior to replicative senescence. Mech Ageing Dev 118:23–34 22. Martinez P, Blasco MA (2011) Telomeric and extra-telomeric roles for telomerase and the telomere-binding proteins. Nat Rev Cancer 11:161–176 23. Diaz VA, Mainous AG 3rd, Everett CJ, Schoepf UJ, Codd V, Samani NJ (2010) Effect of healthy lifestyle behaviors on the association between leukocyte telomere length and coronary artery calcium. Am J Cardiol 106:659–663 24. Cassidy A, De Vivo I, Liu Y, Han J, Prescott J, Hunter DJ, Rimm EB (2010) Associations between diet, lifestyle factors, and telomere length in women. Am J Clin Nutr 91:1273–1280 25. Ornish D, Lin J, Daubenmier J, Weidner G, Epel E, Kemp C, Magbanua MJ et al (2008) Increased telomerase activity and comprehensive lifestyle changes: a pilot study. Lancet Oncol 9:1048–1057 26. Allsopp RC, Vaziri H, Patterson C, Goldstein S, Younglai EV, Futcher AB, Greider CW et al (1992) Telomere Length Predicts Replicative Capacity of Human Fibroblasts. Proc Natl Acad Sci USA 89:10114–10118 27. Greenberg RA (2005) Telomeres, crisis and cancer. Curr Mol Med 5:213–218

Aging Clin Exp Res (2013) 25:139–146 28. di Fagagna FD, Reaper PM, Clay-Farrace L, Fiegler H, Carr P, von Zglinicki T, Saretzki G et al (2003) A DNA damage checkpoint response in telomere-initiated senescence. Nature 426:194–198 29. Karlseder J, Broccoli D, Dai YM, Hardy S, de Lange T (1999) p53- and ATM-dependent apoptosis induced by telomeres lacking TRF2. Science 283:1321–1325 30. Wright WE, Brasiskyte D, Piatyszek MA, Shay JW (1996) Experimental elongation of telomeres extends the lifespan of immortal x normal cell hybrids. EMBO J 15:1734–1741 31. Allsopp RC, Harley CB (1995) Evidence for a critical telomere length in senescent human fibroblasts. Exp Cell Res 219:130–136 32. Huang XQ, Wang J, Liu JP, Feng H, Liu WB, Yan Q, Liu Y et al (2005) hTERT extends proliferative lifespan and prevents oxidative stress-induced apoptosis in human lens epithelial cells. Invest Ophthalmol Vis Sci 46:2503–2513 33. Bodnar AG, Ouellette M, Frolkis M, Holt SE, Chiu CP, Morin GB, Harley CB et al (1998) Extension of life-span by introduction of telomerase into normal human cells. Science 279:349–352 34. Wieser M, Stadler G, Jennings P, Streubel B, Pfaller W, Ambros P, Riedl C et al (2008) hTERT alone immortalizes epithelial cells of renal proximal tubules without changing their functional characteristics. Am J Physiol Renal Physiol 295:F1365–F1375 35. Njajou OT, Hsueh WC, Blackburn EH, Newman AB, Wu SH, Li RL, Simonsick EM et al (2009) Association between telomere length, specific causes of death, and years of healthy life in health, aging, and body composition, a population-based cohort study. J Gerontol Ser A Biol Sci Med Sci 64:860–864 36. Hunt SC, Chen W, Gardner JP, Kimura M, Srinivasan SR, Eckfeldt JH, Berenson GS et al (2008) Leukocyte telomeres are longer in African Americans than in whites: the National Heart, Lung, and Blood Institute Family Heart Study and the Bogalusa Heart Study. Aging Cell 7:451–458 37. Ren F, Li CY, Xi HJ, Wen YF, Huang KQ (2009) Estimation of human age according to telomere shortening in peripheral blood leukocytes of Tibetan. Am J Forensic Med Pathol 30:252–255 38. Vaziri H, Schachter F, Uchida I, Wei L, Zhu XM, Effros R, Cohen D et al (1993) Loss of telomeric DNA during aging of normal and trisomy-21 human-lymphocytes. Am J Hum Genet 52:661–667 39. Andrews NP, Fujii H, Goronzy JJ, Weyand CM (2010) Telomeres and immunological diseases of aging. Gerontology 56:390–403 40. von Zglinicki T, Martin-Ruiz CM (2005) Telomeres as biomarkers for ageing and age-related diseases. Curr Mol Med 5: 197–203 41. Harley CB (2005) Telomerase therapeutics for degenerative diseases. Curr Mol Med 5:205–211 42. Asselbergs FW, van der Harst P, Jessurun GAJ, Tio RA, van Gilst WH (2005) Clinical impact of vasomotor function assessment and the role of ACE-inhibitors and statins. Vascul Pharmacol 42: 125–140 43. Mainous AG, Diaz VA (2010) Telomere length as a risk marker for cardiovascular disease: the next big thing? Expert Rev Mol Diagn 10:969–971 44. De Meyer T, Rietzschel ER, De Buyzere ML, Van Criekinge W, Bekaert S (2011) Telomere length and cardiovascular aging: the means to the ends? Ageing Res Rev 10:297–303 45. Chang E, Harley CB (1995) Telomere length and replicative aging in human vascular tissues. Proc Natl Acad Sci USA 92: 11190–11194 46. Minamino T, Miyauchi H, Yoshida T, Ishida Y, Yoshida H, Komuro I (2002) Endothelial cell senescence in human atherosclerosis—role of telomere in endothelial dysfunction. Circulation 105:1541–1544 47. Fenton M, Barker S, Kurz DJ, Erusalimsky JD (2001) Cellular senescence after single and repeated balloon catheter denudations

145

48.

49.

50.

51.

52.

53.

54.

55.

56.

57.

58.

59.

60.

61.

62.

63. 64. 65.

66.

of rabbit carotid arteries. Arterioscler Thromb Vasc Biol 21: 220–226 Yang ZW, Huang X, Jiang H, Zhang YR, Liu HX, Qin C, Eisner GM et al (2009) Short telomeres and prognosis of hypertension in a chinese population. Hypertension 53:639–695 Imanishi T, Moriwaki C, Hano T, Nishio I (2005) Endothelial progenitor cell senescence is accelerated in both experimental hypertensive rats and patients with essential hypertension. J Hypertens 23:1831–1837 Leri A, Franco S, Zacheo A, Barlucchi L, Chimenti S, Limana F, Nadal-Ginard B et al (2003) Ablation of telomerase and telomere loss leads to cardiac dilatation and heart failure associated with p53 upregulation. EMBO J 22:131–139 Wong LSM, de Boer RA, Samani NJ, van Veldhuisen DJ, Harst PD (2008) Telomere biology in heart failure. Eur J Heart Fail 10:1049–1056 van der Harst P, van der Steege G, de Boer RA, Voors AA, Hall AS, Mulder MJ, van Gilst WH et al (2007) Telomere length of circulating leukocytes is decreased in patients with chronic heart failure. J Am Coll Cardiol 49:1459–1464 Urbanek K, Quaini F, Tasca G, Torella D, Castaldo C, NadalGinard B, Leri A et al (2003) Intense myocyte formation from cardiac stem cells in human cardiac hypertrophy. Proc Natl Acad Sci USA 100:10440–10445 Oh H, Wang SC, Prahash A, Sano M, Moravec CS, Taffett GE, Michael LH et al (2003) Telomere attrition and Chk2 activation in human heart failure. Proc Natl Acad Sci USA 100:5378–5383 Chimenti C, Kajstura J, Torella D, Urbanek K, Heleniak H, Colussi C, Di Meglio F et al (2003) Senescence and death of primitive cells and myocytes lead to premature cardiac aging and heart failure. Circ Res 93:604–613 Zee RYL, Castonguay AJ, Barton NS, Germer S, Martin M (2010) Mean leukocyte telomere length shortening and type 2 diabetes mellitus: a case-control study. Transl Res 155:166–169 Salpea KD, Talmud PJ, Cooper JA, Maubaret CG, Stephens JW, Abelak K, Humphries SE (2010) Association of telomere length with type 2 diabetes, oxidative stress and UCP2 gene variation. Atherosclerosis 209:42–50 Fyhrquist F, Tiitu A, Saijonmaa O, Forsblom C, Groop PH (2010) Telomere length and progression of diabetic nephropathy in patients with type 1 diabetes. J Intern Med 267:278–286 Astrup AS, Tarnow L, Jorsal A, Lajer M, Nzietchueng R, Benetos A, Rossing P et al (2010) Telomere length predicts allcause mortality in patients with type 1 diabetes. Diabetologia 53:45–48 Kuhlow D, Florian S, von Figura G, Weimer S, Schulz N, Petzke KJ, Zarse K et al (2010) Telomerase deficiency impairs glucose metabolism and insulin secretion. Aging (US) 2:650–658 Adaikalakoteswari A, Balasubramanyam M, Ravikumar R, Deepa R, Mohan V (2007) Association of telomere shortening with impaired glucose tolerance and diabetic macroangiopathy. Atherosclerosis 195:83–89 Demissie S, Levy D, Benjamin EJ, Cupples LA, Gardner JP, Herbert A, Kimura M et al (2006) Insulin resistance, oxidative stress, hypertension, and leukocyte telomere length in men from the Framingham Heart Study. Aging Cell 5:325–330 Hahn WC (2003) Role of telomeres and telomerase in the pathogenesis of human cancer. J Clin Oncol 21:2034–2043 Chung HK, Cheong C, Song J, Lee HW (2005) Extratelomeric functions of telomerase. Curr Mol Med 5:233–241 Wu XF, Amos CI, Zhu Y, Zhao H, Grossman BH, Shay JW, Luo S et al (2003) Telomere dysfunction: a potential cancer predisposition factor. J Natl Cancer Inst 95:1211–1218 Mu YC, Zhang Q, Mei LH, Liu XS, Yang WL, Yu JR (2011) Telomere shortening occurs early during gastrocarcinogenesis. Med Oncol. doi:10.1007/s12032-011-9866-3

123

146 67. Hahn WC, Stewart SA, Brooks MW, York SG, Eaton E, Kurachi A, Beijersbergen RL et al (1999) Inhibition of telomerase limits the growth of human cancer cells. Nat Med 5:1164–1170 68. Guittat L, Alberti P, Mergny JL, Riou JF, Teulade-Fichou MP, Mailliet P (2001) Telomerase inhibitors: possible consequences in cancer therapy. M S-Med Sci 17:1076–1081 69. Perry PJ, Arnold JRP, Jenkins TC (2001) Telomerase inhibitors for the treatment of cancer: the current perspective. Expert Opin Investig Drugs 10:2141–2156 70. Hashizume R, Gupta N (2010) Telomerase inhibitors for the treatment of brain tumors and the potential of intranasal delivery. Curr Opin Mol Ther 12:168–175 71. Agrawal A, Dang S, Gabrani R (2012) Recent patents on antitelomerase cancer therapy. Recent Pat Anticancer Drug Discov 7:102–117 72. Malavolta M, Mocchegiani E, Bertoni-Freddari C (2004) New trends in biomedical aging research. Gerontology 50:420–424 73. Goronzy JJ, Fujii H, Weyand CM (2006) Telomeres, immune aging and autoimmunity. Exp Gerontol 41:246–251 74. Effros RB (2003) Genetic alterations in the ageing immune system: impact on infection and cancer. Mech Ageing Dev 124:71–77 75. Georgin-Lavialle S, Aouba A, Mouthon L, Londono-Vallejo JA, Lepelletier Y, Gabet AS, Hermine O (2010) The telomere/telomerase system in autoimmune and systemic immune-mediated diseases. Autoimmun Rev 9:646–651 76. Pawelec G, Akbar A, Caruso C, Effros R, Grubeck-Loebenstein B, Wikby A (2004) Is immunosenescence infectious? Trends Immunol 25:406–410 77. Akbar AN, Beverley PCL, Salmon M (2004) Opinion: will telomere erosion lead to a loss of T-cell memory? Nat Rev Immunol 4:737–743 78. Effros RB, Boucher N, Porter V, Zhu XM, Spaulding C, Walford RL, Kronenberg M et al (1994) Decline in Cd28(?) T-cells in centenarians and in long-term t-cell cultures—a possible cause

123

Aging Clin Exp Res (2013) 25:139–146

79.

80.

81.

82.

83.

84.

85.

86. 87.

88. 89.

for both in vivo and in vitro immunosenescence. Exp Gerontol 29:601–609 Weng NP, Levine BL, June CH, Hodes RJ (1996) Regulated expression of telomerase activity in human T lymphocyte development and activation. J Exp Med 183:2471–2479 Spaulding C, Guo W, Effros RB (1999) Resistance to apoptosis in human CD8? T cells that reach replicative senescence after multiple rounds of antigen-specific proliferation. Exp Gerontol 34:633–644 Weng NP (2001) Interplay between telomere length and telomerase in human leukocyte differentiation and aging. J Leukoc Biol 70:861–867 Cawthon RM, Smith KR, O’Brien E, Sivatchenko A, Kerber RA (2003) Association between telomere length in blood and mortality in people aged 60 years or older. Lancet 361:393–395 Mitchell JR, Wood E, Collins K (1999) A telomerase component is defective in the human disease dyskeratosis congenita. Nature 402:551–555 Mason PJ, Wilson DB, Bessler M (2005) Dyskeratosis congenita—a disease of dysfunctional telomere maintenance. Curr Mol Med 5:159–170 Walne AJ, Marrone A, Dokal I (2005) Dyskeratosis congenita: a disorder of defective telomere maintenance? Int J Hematol 82:184–189 Bessler M, Wilson DB, Mason PJ (2004) Dyskeratosis congenita and telomerase. Curr Opin Pediatr 16:23–28 Mather KA, Jorm AF, Parslow RA, Christensen H (2011) Is telomere length a biomarker of aging? A review. J Gerontol Ser A Biol Sci Med Sci 66:202–213 Cong YS, Shay JW (2008) Actions of human telomerase beyond telomeres. Cell Res 18:725–732 Egan CA, Savre-Train I, Shay JW, Wilson SE, Bourne WM (1998) Analysis of telomere lengths in human corneal endothelial cells from donors of different ages. Invest Ophthalmol Vis Sci 39:648–653