Research article - (2015)14, 239 - 245
Effect of Intensive Exercise in Early Adult Life on Telomere Length in Later Life in Men
Merja K. Laine1,2,, Johan G. Eriksson1,3-6, Urho M. Kujala7, Rahul Raj8, Jaakko Kaprio9-11, Heli M. Bäckmand8,12, Markku Peltonen3, Seppo Sarna9
1Department of General Practice and Primary Health Care, University of Helsinki, Helsinki, Finland
10Department of Mental Health and Substance Abuse Services, National Institute for Health and Welfare, Helsinki, Finland
11Institute for Molecular Medicine, University of Helsinki, Helsinki, Finland
12City of Vantaa, Health and Social Welfare Department, Finland
2Vantaa Health Center/Network of Academic Health Centers, University of Helsinki, Helsinki, Finland
3Division of Welfare and Health Promotion Department of Chronic Disease Prevention, Diabetes Prevention Unit, National Institute for Health and Welfare, Helsinki, Finland
4Folkhälsan Research Centre, Helsinki, Finland
5Unit of General Practice, Helsinki University Central Hospital, Helsinki, Finland
6Vaasa Central Hospital, Vaasa, Finland
7Department of Health Sciences, University of Jyväskylä, Jyväskylä, Finland
8Department of Neurosurgery, Helsinki University Central Hospital, Helsinki, Finland
9Department of Public Health, University of Helsinki, Helsinki, Finland

Merja K. Laine
✉ Department of General Practice and Primary Health Care, University of Helsinki, Tukholmankatu 8 B, 00290 Helsinki, Finland.
Email: merja.k.laine@helsinki.fi
Received: 26-11-2014 -- Accepted: 20-01-2015
Published (online): 01-06-2015

ABSTRACT

A career as an elite-class male athlete seems to improve metabolic heath in later life and is also associated with longer life expectancy. Telomere length is a biomarker of biological cellular ageing and could thus predict morbidity and mortality. The main aim of this study was to assess the association between vigorous elite-class physical activity during young adulthood on later life leukocyte telomere length (LTL). The study participants consist of former male Finnish elite athletes (n = 392) and their age-matched controls (n = 207). Relative telomere length was determined from peripheral blood leukocytes by quantitative real-time polymerase chain reaction. Volume of leisure-time physical activity (LTPA) was self-reported and expressed in metabolic equivalent hours. No significant difference in mean age-adjusted LTL in late life (p = 0.845) was observed when comparing former male elite athletes and their age-matched controls. Current volume of LTPA had no marked influence on mean age-adjusted LTL (p for trend 0.788). LTL was inversely associated with age (p = 0.004).Our study findings suggest that a former elite athlete career is not associated with LTL later in life.

Key words: Aging, athlete, DNA repeats, physical activity

Key Points
  • A career as an elite-class athlete is associated with improved metabolic health in late life and is associated with longer life expectancy.
  • A career as an elite-class athlete during young adulthood was not associated with leukocyte telomere length in later life.
  • Current volume of leisure-time physical activity did not influence telomere length in later life.
INTRODUCTION

Physical activity has a positive influence on health and general well-being and it has been associated with increased longevity, better physical functioning and self-rated heath in older age (Backmand et al., 2010; Cherkas et al., 2008; Kettunen et al., 2014; Warburton et al., 2006). A career as an elite-class athlete during young adulthood seems to improve metabolic heath and reduce coronary heart disease in later life (Kujala et al., 2013; Laine et al., 2014; Kujala et al., 2013; Sarna et al., 1997).

Telomeres consist of DNA repeats and associated proteins located at the ends of the chromosomes (Blackburn et al., 2006; de Lange, 2005). Telomeres play an important role in maintaining genomic stability and regulating cellular replicative capacity (Allsopp et al., 1992; Blackburn et al., 2006). Telomere length is heritable, and length declines with increasing age (Njajou et al., 2012; Nordfjall et al., 2005; Shammas, 2011). Especially in early life, the impact of inheritance on telomere length is strong, but it seems to diminish by age (Svenson et al., 2011). Shorter telomeres have been associated with increased incidence of several chronic non-communicable diseases and with shorter life span (Ludlow and Roth 2011; Shammas, 2011; Salpea and Humphries, 2010; Wong and Collins 2003). Several factors including smoking, obesity and an unhealthy diet, all conditions associated with an increase in oxidative stress and inflammation, have been linked with telomere shortening (Crous-Bou et al., 2014; Ornish et al., 2013; Shammas, 2011; Tiainen et al., 2012; Woo et al., 2010;). It is believed that telomere length could be a biomarker of biological cellular age and thus predicts morbidity and mortality (Bojesen, 2013; Woo et al., 2014; von Zglinicki and Martin-Ruiz, 2005).

Few studies have investigated the association between exercise training and leukocyte telomere length (LTL) with inconsistent results: positive, none and inverted U-shaped associations have been described (Du et al., 2012; Savela et al. 2013; Woo et al., 2008). Interestingly, a U-shaped relationship has been observed in both sedentary and extremely active individuals (Ludlow et al., 2013). Most commonly moderate levels of physical activity have been associated with longer LTL (Kim et al., 2012; Mirabello et al., 2009).

In Finland, a questionnaire-based study focusing upon former male elite athletes and their age -matched controls was initiated in 1985 (repeated 1995 and 2005). Twenty-three years later in 2008 clinical examinations including a physical examination and laboratory tests were performed.

We could not identify any previous studies that have evaluated the association of long-term influence of vigorous elite-class physical activity during young adulthood on later life LTL.

We hypothesized that the male former elite athletes having a history of vigorous physical activity during young adulthood have longer LTL than the controls later in life thus explaining the former athletes’ better metabolic health and longer life expectancy (Kettunen et al., 2014; Laine et al., 2014).

METHODS
Study subjects

A detailed description of the study design and participants has been published previously (Laine et al., 2014). Briefly, in Finland in the year 1985 a questionnaire-based study was initiated including former male elite athletes and their age- and area-matched healthy controls. Former elite male athletes consist of those who had represented Finland in major international competitions between 1920 and 1965. They were divided into three groups: endurance sports (long and middle distance running, cross country skiing), mixed sports (soccer, ice hockey, basketball, track and field: jumpers, sprinters, hurdlers, decathletes) and power sports (boxing, wrestling, weight lifting, track and field throwers). The division was made according to the type of training needed to achieve optimal results. In 2008, an invitation to a clinical study was sent to those alive, whom had answered at least once to the previous questionnaires sent in 1985, 1995 or 2001 (n = 1183). All together 599 men participated (392 former athletes and 207 controls) and they composed the present study cohort. Compared with the controls, the former athletes have longer life expectancy, lower cancer incidence, and better metabolic health (Laine et al., 2014; Kettunen et al., 2014; Sormunen et al., 2014).

The ethics committee of the Hospital District of Helsinki and Uusimaa approved the study, and all subjects have provided written informed consent.

The clinical examinations

Trained study nurses performed the physical examinations including assessment of height, weight and blood pressure (BP) as well as taking the blood samples.

Height was measured without shoes by a measuring tape against a wall to an accuracy of 0.1 cm. Weight was measured in light indoor clothing by a body composition device (InBody 3.0, Biospace, Seoul, Korea) to an accuracy of 0.1 kg. If the participant had a pacemaker (n = 14), weight was measured by a digital scale with the same accuracy. BMI was calculated as weight (kg) divided by height squared (m2). Plasma high sensitive C-reactive protein was measured by latex immunoturbidometric method (Sentinel Diagnostics, Milan, Italy).

Leukocyte telomere length measurement

Leukocyte telomere length was measured from DNA extracted from peripheral blood. We used a quantitative polymerase chain reaction (qPCR) -based method (Cawthon 2002), as described previously (Eerola et al., 2010; Kananen et al., 2010; Kao et al., 2008). We used β-hemoglobin (Hgb) as a single copy reference gene. Separate reactions for telomere and Hgb reaction were carried out in paired 384-well plates in which matched sample well positions were used. Ten nanograms of DNA were used for each reaction, performed in triplicate. Every plate included a 7-point standard curve, which was used to create a standard curve and to perform absolute quantification of each sample. Samples and standard dilutions were transferred into the plates using a DNA Hydra 96 robot and dried overnight at +37oC. Specific reaction mix for telomere reaction included 270 nM tel1b primer (5†²-CGGTTT(GTTTGG)5GTT-3†²) and 900 nM tel2b primer (5†²-GGCTTG(CCTTAC)5CCT-3†²), 150 nM ROX (Invitrogen), 0.2X SYBR Green I (Invitrogen), 5 mM DTT (Sigma-Aldrich), 1% DMSO (Sigma-Aldrich), 0.2 mM of each dNTP (Fermentas), and 1.25 U AmpliTaq Gold DNA polymerase (Applied Biosystems) in a total volume of 15 µl AmpliTaq Gold Buffer I. Hgb reaction mix included 300 nM Hgb1 primer (5†²-GCTTCTGACACAACTGTGTTCACTAGC-3†²) and Hgb2 primer (5†²-CACCAACTTCATCCACGTTCACC-3†²) in a total volume of 15 µl of iQ SyBrGreen supermix (BioRad). The cycling conditions for telomere amplification were: 10 minutes at 95 °C followed by 25 cycles at 95 °C for 15 s and 54°C for 2 min with signal acquisition. The cycling conditions for Hgb amplification were: 95 °C for 10 min followed by 35 cycles at 95 °C for 15 s, 58 °C for 20 s, 72 °C for 20 s with signal acquisition. Reactions were performed with CFX384 Real-Time PCR Detection System (Bio-Rad). Melt-curve analysis was carried out in the end of the run to ensure specific primer binding.

We used the Bio-Rad CFX Manager software to perform quality control, and samples with standard deviation of >0.5 between triplicates were omitted from the analysis. Five control samples analyzed on each plate were used for calculating the coefficient of variation, which was 7.14%. Thirteen participants had missing data of LTL.

Assessment of life-style factors

Information on smoking status, consumption of alcohol, educational attainment and marital status were self-reported and obtained from structured questionnaires. There were missing data from nine participants regarding smoking habits, for 32 participants on alcohol consumption, for four participants on educational attainment and for one participant about marital status. Participants were classified as smokers if they reported having smoked over 100 cigarettes in their lifetime and still smoked daily or almost daily at least one cigarette or quit smoking less than six months ago.

Assessment of leisure-time physical activity

Leisure-time physical activity (LTPA) was assessed by a questionnaire that included information about average intensity, duration and frequency of the activity during the previous three months. Intensity of LTPA was asked by the following question “Is your physical activity during leisure time about as tiring (intensive) on average as? 1 = walking (4 metabolic equivalent [MET]), 2 = walking and jogging alternately (6 MET), 3 = jogging (10 MET), 4 = running (13 MET)”. Duration of LTPA was asked by the question “What is the mean duration of your average physical exercise session? 1 = < 15 minutes, 2 = 15-29 minutes, 3 = 30-59 minutes, 4 = 1-2 hours, 5= > 2 hours”. Frequency of LTPA was asked by the question “How many times per month you do participate in physical exercise? 1 = < once a month, 2 = 1-2 times/month, 3 = 3-5 times/month, 4 = 6-10 times/month, 5 = 11-19 time/month, 6 = > 20 times/month”. For each of intensity category a metabolic equivalent value (MET-value, 1 MET = 3.5 ml O2/kg/min or 1 kcal/kg/h) was determined. The volume of LTPA was expressed in MET-hours, which was calculated by multiplying the intensity (MET), duration and frequency. This method has been validated against detailed physical activity interview (Waller et al., 2008). Fifteen participants had missing data of LTPA.

Statistical analyses

Data has been reported as means D standard deviations (SD). Percentage differences were tested using cross-tabulation and Chi-Square test. Means were compared by one-way ANOVA, and post hoc tests by Bonferroni correction. Correlation was assesses with the Pearson contingency coefficient for continuous variables and with the Lambda contingency coefficient for classified variables. Adjusting with other variables was done by general linear model. Statistical analyses were carried out using IBM SPSS version 21.0 (IBM Ltd, Armonk, New York, USA). P-values <0.05 were considered statistically significant.

RESULTS
Baseline characteristics

The former athletes were older (p = 0.024) and less likely to be smokers (p = 0.014) than the controls (Table 1). No significant difference was observed in BMI (p = 0.285), but age-adjusted fat free mass was significantly higher among the former athletes compared to the controls (p < 0.001, Table 1). The former athletes’ current volume of LTPA was significantly higher than that of the controls (p < 0.001, Table 1).

Leukocyte telomere length

Age was inversely associated with LTL (r = -0.119, p = 0.004), no significant associations were observed with the other assessed covariates: fat free mass (r = 0.028, p = 0.511), LTPA (r = 0.058, p = 0.164), smoking (r = 0.015, p = 0.936), alcohol intake (r = 0.005, p = 0.897), high sensitive C-reactive protein (r = -0.007, p=0.859), educational attainment (r = 0.018, p=0.453), and marital status (r = 0.026, p = 0.148). There were no significant differences in mean age-adjusted LTL between the athlete and control groups (p = 0.845, Table 1). Further adjusting for other covariates had only minimal influence on the results (Table 1). There were no significant differences in LTL between the different athlete groups (three group ANOVA p=1.000, data not shown). Participants’ current volume of LTPA was not associated with mean age-adjusted LTL (Figure 1; for LTPA categories the p for trend 0.788, Table 2).

DISCUSSION

Our findings of the present study suggest that a former career as an elite-class athlete during young adulthood did not influence mean age-adjusted LTL in later life. Neither did the participants’ current volume of LTPA show a significant association with mean age-adjusted LTL. However, as expected participants’ age was inversely associated with LTL.

Reports on the association between physical exercise and LTL have been inconsistent in previous studies (Ludlow et al., 2013). The former male athletes in our study had a history of vigorous exercise training during young adulthood. This is, however, a time period when only minor influence on LTL is expected by exercise training (LaRocca et al., 2010). In later life, current volume of LTPA seems to show no association with LTL supporting the results of a large Chinese study with participants from the same age-group (Woo et al., 2008). However, our results are contrary to a few studies with participants from the same age-group showing a positive association or an inverted U relationship with physical activity in later life or in midlife and LTL (Krauss et al., 2011; LaRocca et al., 2010; Ludlow et al., 2008; Savela et al., 2013).

The underlying mechanisms by which physical activity affects telomere length and the reasons for the different findings between studies are not clear. Oxidative stress, expression of telomere stabilizing proteins, growth- and stress-related hormones and their associated pathways have been suggested to play important roles (Carrero et al., 2008; Cherkas et al., 2008; Ludlow and Roth 2011; Shammas, 2011). Short-term exercise training has not been shown to be associated with LTL, but it modulates telomere-regulating proteins (Werner et al., 2008). Thus, it seems like long-term or more strenuous exercise training is required in order to influence LTL (LaRocca et al., 2010; Ludlow et al., 2013). Moreover, methods used for telomere length determination, cell types used for DNA extraction, the various ages of the individuals in the study cohorts, small study samples as well as variation in collection of exercise and physical activity data likely explain the discrepancies in results (Ludlow et al., 2013).

In most studies evaluating the association between physical exercise and telomere length the mode of exercise has been endurance or aerobic-type training giving a positive (Cherkas et al., 2008; Du et al., 2012; LaRocca et al., 2010), none (Mathur et al., 2013; Woo et al., 2008) or inverted U-shaped association (Collins et al., 2003; Savela et al., 2013). Little is known about the association between resistance training and telomere length. Probably, there is no association between long-term resistance training and telomere length, however negative associations between heavy-resistance training and telomere length has been described when telomeres are measured from skeletal muscles (Kadi et al., 2008).

Generally, oxidative stress and inflammation are both known to cause accelerated telomere shortening (von Zglinicki, 2002). With smoking, a significant dose-dependent relationship has been observed; each pack-year smoked was equivalent to an additional 5 base pairs of telomere length lost (Valdes et al., 2005). Obesity also increases oxidative stress and inflammation providing a link with accelerated telomere loss (Valdes et al., 2005; Shammas, 2011). Healthy dietary patterns like Mediterranean diet have been shown to be associated with longer LTL (Crous-Bou et al., 2014). Furthermore, depression, psychological stress, poor sleep quality and low educational attainment have all been linked with shorter LTL (Hartmann et al., 2010; Needham et al., 2013; Ornish et al., 2013; Prather et al., 2011).

We acknowledge some limitations to our study. LTL was measured only once and in this study in later life. Thus, with this cross-sectional study design is not possible to estimate the telomere attrition rate, which is thought to be better biomarker of biological cellular aging than once measured telomere length (Nilsson, 2014). Further our data is restricted to only survivors. Later life LTPA as well as other life-style factors were self-reported. Self-reported measures of LTPA have been reported to be both higher and lower than directly measured, but on average, the validity and reliability of questionnaires are better in groups than in individuals (Haskell, 2012; Prince et al., 2008; Wilson et al., 1986). Furthermore, information on long-term dietary habits of the participants was not available; this area should be focused upon in future research. Finally, all our study participants were men and of European ancestry which might influence the generalizability of the results.

CONCLUSION
In conclusion, we did not observe any significant differences between former male elite athletes and their matched controls in regard to LTL in late life. Neither did the participants’ current volume of LTPA associate with mean LTL.

ACKNOWLEDGEMENTS

The study was funded by the Ministry of Education and Culture, the Juho Vainio Foundation, the Finnish Heart Research Foundation, Paavo Nurmi Foundation, the Finnish Cultural Foundation, and by a grant from Medical Society of Finland, Finska Läkaresällskapet. We would like to thank National Institute for Health and Welfare, Department of Public Health, University of Helsinki, Sports & Exercise Medicine Department of Health Sciences, University of Jyväskylä, Paavo Nurmi Centre, Turku and ORTON Research Institute, Invalid Foundation, Helsinki for collaboration during the large epidemiological and clinical research program in the year 2008. We would like to thank Iiris Hovatta (Research Programs Unit - Molecular Neurology, Biomedicum Helsinki, University of Helsinki, Finland; Department of Biosciences, Viikki Biocenter, University of Helsinki, Finland; Mental Health and Substance Abuse Services, National Institute for Health and Welfare, Helsinki, Finland) and Laura Kananen (Research Programs Unit - Molecular Neurology, Biomedicum Helsinki, University of Helsinki, Finland) for measuring the leukocyte telomere length.

AUTHOR BIOGRAPHY
     
 
Merja K. Laine
 
Employment:Department of General Practice and Primary Care Health Care, University of Helsinki, Helsinki, Finland; Primary Health Care Center of Vantaa, Vantaa, Finland
 
Degree: MD
 
Research interests: Diabetes, obesity, prevention
  E-mail: merja.k.laine@helsinki.fi
   
   

     
 
Johan G. Eriksson
 
Employment:Depart. of General Practice and Primary Care Health Care, Univ. of Helsinki, Helsinki, Finland; Unit of General Practice, Helsinki Univ. General Hospital, Helsinki, Finland; Depart. of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Finland; Vaasa Central Hospital, Vaasa, Finland
 
Degree: MD, DMSc
 
Research interests: Type 2 diabetes, obesity, prevention, programming of health and disease
  E-mail: johan.eriksson@helsinki.fi
   
   

     
 
Urho M. Kujala
 
Employment:Professor of Sports and Exercise Medicine
 
Degree: MD, PhD
 
Research interests: Physical activity and health
  E-mail: urho.m.kujala@jyu.fi
   
   

     
 
Rahul Raj
 
Employment:Department of Neurosurgery, Helsinki University Central Hospital, Helsinki, Finland
 
Degree: MD, PhD
 
Research interests: Research interests: traumatic brain injury, subarachnoid hemorrhage, neurocritical care
  E-mail: rahul.raj@helsinki.fi
   
   

     
 
Jaakko Kaprio
 
Employment:Professor of Genetic Epidemiology
 
Degree: MD, PhD
 
Research interests: Research interests: Genetic epidemiology on non-communicable diseases and their behavioral risk factors
  E-mail: jaakko.kaprio@helsinki.fi
   
   

     
 
Heli M. Bäckmand
 
Employment:Health and Social Welfare Department, City of Vantaa, Finland
 
Degree: PhD
 
Research interests: Physical activity, public health, life-habits
  E-mail: heli.backmand@vantaa.fi
   
   

     
 
Markku Peltonen
 
Employment:National Institute for Health and Welfare
 
Degree: PhD
 
Research interests: Epidemiology and prevention of cardiovascular disease and diabetes
  E-mail: markku.peltonen@thl.fi
   
   

     
 
Seppo Sarna
 
Employment:Emeritus professor
 
Degree: PhD
 
Research interests: Biostatistics
  E-mail: seppo.sarna@helsinki.fi
   
   

REFERENCES
Allsopp R.C., Vaziri H., Patterson C., Goldstein S., Younglai E.V., Futcher A.B., Greider C.W., Harley C.B. (1992) Telomere length predicts replicative capacity of human fibroblasts. Proceedings of the National Academy of Sciences of the United States of America 89, 10114-10118.
Backmand H., Kujala U., Sarna S., Kaprio J. (2010) Former athletes’ health-related lifestyle behaviours and self-rated health in late adulthood. International Journal of Sports Medicine 31, 751-758.
Blackburn E.H., Greider C.W., Szostak J.W. (2006) Telomeres and telomerase: the path from maize, Tetrahymena and yeast to human cancer and aging. Nature Medicine 12, 1133-1138.
Bojesen S.E. (2013) Telomeres and human health. Journal of Internal Medicine 274, 399-413.
Carrero J.J., Shiels P.G., Stenvinkel P. (2008) Telomere biology alterations as a mortality risk factor in CKD. American Journal of Kidney Diseases: The Official Journal of the National Kidney Foundation 51, 1076-1077.
Cawthon R.M. (2002) Telomere measurement by quantitative PCR. Nucleic Acids Research 30, e47-.
Cherkas L.F., Hunkin J.L., Kato B.S., Richards J.B., Gardner J.P., Surdulescu G.L., Kimura M., Lu X., Spector T.D., Aviv A. (2008) The association between physical activity in leisure time and leukocyte telomere length. Archives of Internal Medicine 168, 154-158.
Collins M., Renault V., Grobler L.A., St Clair Gibson A., Lambert M.I., Wayne Derman E., Butler-Browne G.S., Noakes T.D., Mouly V. (2003) Athletes with exercise-associated fatigue have abnormally short muscle DNA telomeres. Medicine and Science in Sports and Exercise 35, 1524-1528.
Crous-Bou M., Fung T.T., Prescott J., Julin B., Du M., Sun Q., Rexrode K.M., Hu F.B., De Vivo I. (2014) Mediterranean diet and telomere length in Nurses’ Health Study: population based cohort study. British Medical Journal 349, g6674-.
de Lange T. (2005) Shelterin: the protein complex that shapes and safeguards human telomeres. Genes & Development 19, 2100-2110.
Du M., Prescott J., Kraft P., Han J., Giovannucci E., Hankinson S.E., De Vivo I. (2012) Physical activity, sedentary behavior, and leukocyte telomere length in women. American Journal of Epidemiology 175, 414-422.
Eerola J., Kananen L., Manninen K., Hellstrom O., Tienari P.J., Hovatta I. (2010) No evidence for shorter leukocyte telomere length in Parkinson’s disease patients. The Journals of Gerontology. Series A, Biological Sciences and Medical Sciences 65, 1181-1184.
Hartmann N., Boehner M., Groenen F., Kalb R. (2010) Telomere length of patients with major depression is shortened but independent from therapy and severity of the disease. Depression and Anxiety 27, 1111-1116.
Haskell W.L. (2012) Physical activity by self-report: a brief history and future issues. Journal of Physical Activity & Health 9, S5-10.
Kadi F., Ponsot E., Piehl-Aulin K., Mackey A., Kjaer M., Oskarsson E., Holm L. (2008) The effects of regular strength training on telomere length in human skeletal muscle. Medicine and Science in Sports and Exercise 40, 82-87.
Kananen L., Surakka I., Pirkola S., Suvisaari J., Lonnqvist J., Peltonen L., Ripatti S., Hovatta I. (2010) Childhood adversities are associated with shorter telomere length at adult age both in individuals with an anxiety disorder and controls. PloS One 5, e10826-.
Kao H.T., Cawthon R.M., Delisi L.E., Bertisch H.C., Ji F., Gordon D., Li P., Benedict M.M., Greenberg W.M., Porton B. (2008) Rapid telomere erosion in schizophrenia. Molecular Psychiatry 13, 118-119.
Kettunen J.A., Kujala U.M., Kaprio J., Backmand H., Peltonen M., Eriksson J.G., Sarna S. (2014) All-cause and disease-specific mortality among male, former elite athletes: an average 50-year follow-up. British Journal of Sports Medicine , -.
Kim J.H., Ko J.H., Lee D.C., Lim I., Bang H. (2012) Habitual physical exercise has beneficial effects on telomere length in postmenopausal women. Menopause (New York, N.Y.) 19, 1109-1115.
Krauss J., Farzaneh-Far R., Puterman E., Na B., Lin J., Epel E., Blackburn E., Whooley M.A. (2011) Physical fitness and telomere length in patients with coronary heart disease: findings from the Heart and Soul Study. PloS One 6, e26983-.
Kujala U.M., Makinen V.P., Heinonen I., Soininen P., Kangas A.J., Leskinen T.H., Rahkila P., Wurtz P., Kovanen V., Cheng S., Sipila S., Hirvensalo M., Telama R., Tammelin T., Savolainen M.J., Pouta A., O’Reilly P.F., Mantyselka P., Viikari J., Kahonen M., Lehtimaki T., Elliott P., Vanhala M.J., Raitakari O.T., Jarvelin M.R., Kaprio J., Kainulainen H., Ala-Korpela M. (2013) Long-term leisure-time physical activity and serum metabolome. Circulation 127, 340-348.
Laine M.K., Eriksson J.G., Kujala U.M., Wasenius N.S., Kaprio J., Backmand H.M., Peltonen M., Mertsalmi T.H., Sarna S. (2014) A former career as a male elite athlete-does it protect against type 2 diabetes in later life?. Diabetologia 57, 270-274.
LaRocca T.J., Seals D.R., Pierce G.L. (2010) Leukocyte telomere length is preserved with aging in endurance exercise-trained adults and related to maximal aerobic capacity. Mechanisms of Ageing and Development 131, 165-167.
Ludlow A.T., Ludlow L.W., Roth S.M. (2013) Do telomeres adapt to physiological stress? Exploring the effect of exercise on telomere length and telomere-related proteins. BioMed Research International 2013, 601368-.
Ludlow A.T., Roth S.M. (2011) Physical activity and telomere biology: exploring the link with aging-related disease prevention. Journal of Aging Research 2011, 790378-.
Ludlow A.T., Zimmerman J.B., Witkowski S., Hearn J.W., Hatfield B.D., Roth S.M. (2008) Relationship between physical activity level, telomere length, and telomerase activity. Medicine and Science in Sports and Exercise 40, 1764-1771.
Mathur S., Ardestani A., Parker B., Cappizzi J., Polk D., Thompson P.D. (2013) Telomere length and cardiorespiratory fitness in marathon runners. Journal of Investigative Medicine: the official publication of the American Federation for Clinical Research 61, 613-615.
Mirabello L., Huang W.Y., Wong J.Y., Chatterjee N., Reding D., Crawford E.D., De Vivo I., Hayes R.B., Savage S.A. (2009) The association between leukocyte telomere length and cigarette smoking, dietary and physical variables, and risk of prostate cancer. Aging Cell 8, 405-413.
Needham B.L., Adler N., Gregorich S., Rehkopf D., Lin J., Blackburn E.H., Epel E.S. (2013) Socioeconomic status, health behavior, and leukocyte telomere length in the National Health and Nutrition Examination Survey, 1999-2002. Social Science & Medicine 85, 1-8.
Nilsson P.M. (2014) Mediterranean diet and telomere length. British Medical Journal (Clinical Research Ed.) 349, g6843-.
Njajou O.T., Cawthon R.M., Blackburn E.H., Harris T.B., Li R., Sanders J.L., Newman A.B., Nalls M., Cummings S.R., Hsueh W.C. (2012) Shorter telomeres are associated with obesity and weight gain in the elderly. International Journal of Obesity (2005) 36, 1176-1179.
Nordfjall K., Larefalk A., Lindgren P., Holmberg D., Roos G. (2005) Telomere length and heredity: Indications of paternal inheritance. Proceedings of the National Academy of Sciences of the United States of America 102, 16374-16378.
Ornish D., Lin J., Chan J.M., Epel E., Kemp C., Weidner G., Marlin R., Frenda S.J., Magbanua M.J., Daubenmier J., Estay I., Hills N.K., Chainani-Wu N., Carroll P.R., Blackburn E.H. (2013) Effect of comprehensive lifestyle changes on telomerase activity and telomere length in men with biopsy-proven low-risk prostate cancer: 5-year follow-up of a descriptive pilot study. The Lancet. Oncology 14, 1112-1120.
Prather A.A., Puterman E., Lin J., O’Donovan A., Krauss J., Tomiyama A.J., Epel E.S., Blackburn E.H. (2011) Shorter leukocyte telomere length in midlife women with poor sleep quality. Journal of Aging Research 2011, 721390-.
Prince S.A., Adamo K.B., Hamel M.E., Hardt J., Connor Gorber S., Tremblay M. (2008) A comparison of direct versus self-report measures for assessing physical activity in adults: a systematic review. The International Journal of Behavioral Nutrition and Physical Activity 5, 56-5868-5-56.
Salpea K.D., Humphries S.E. (2010) Telomere length in atherosclerosis and diabetes. Atherosclerosis 209, 35-38.
Sarna S., Kaprio J., Kujala U.M., Koskenvuo M. (1997) Health status of former elite athletes. The Finnish experience. Aging (Milan, Italy) 9, 35-41.
Savela S., Saijonmaa O., Strandberg T.E., Koistinen P., Strandberg A.Y., Tilvis R.S., Pitkala K.H., Miettinen T.A., Fyhrquist F. (2013) Physical activity in midlife and telomere length measured in old age. Experimental Gerontology 48, 81-84.
Shammas M.A. (2011) Telomeres, lifestyle, cancer, and aging. Current Opinion in Clinical Nutrition and Metabolic Care 14, 28-34.
Sormunen J., Backmand H.M., Sarna S., Kujala U.M., Kaprio J., Dyba T., Pukkala E. (2014) Lifetime physical activity and cancer incidence--a cohort study of male former elite athletes in Finland. Journal of Science and Medicine in Sport 17, 479-484.
Svenson U., Nordfjall K., Baird D., Roger L., Osterman P., Hellenius M.L., Roos G. (2011) Blood cell telomere length is a dynamic feature. PloS One 6, e21485-.
Tiainen A.M., Mannisto S., Blomstedt P.A., Moltchanova E., Perala M.M., Kaartinen N.E., Kajantie E., Kananen L., Hovatta I., Eriksson J.G. (2012) Leukocyte telomere length and its relation to food and nutrient intake in an elderly population. European Journal of Clinical Nutrition 66, 1290-1294.
Valdes A.M., Andrew T., Gardner J.P., Kimura M., Oelsner E., Cherkas L.F., Aviv A., Spector T.D. (2005) Obesity, cigarette smoking, and telomere length in women. Lancet 366, 662-664.
von Zglinicki T. (2002) Oxidative stress shortens telomeres. Trends in Biochemical Sciences 27, 339-344.
von Zglinicki T., Martin-Ruiz C.M. (2005) Telomeres as biomarkers for ageing and age-related diseases. Current Molecular Medicine 5, 197-203.
Waller K., Kaprio J., Kujala U.M. (2008) Associations between long-term physical activity, waist circumference and weight gain: a 30-year longitudinal twin study. International Journal of Obesity (2005) 32, 353-361.
Warburton D.E., Nicol C.W., Bredin S.S. (2006) Health benefits of physical activity: the evidence. Canadian Medical Association Journal 174, 801-809.
Werner C., Hanhoun M., Widmann T., Kazakov A., Semenov A., Poss J., Bauersachs J., Thum T., Pfreundschuh M., Muller P., Haendeler J., Bohm M., Laufs U. (2008) Effects of physical exercise on myocardial telomere-regulating proteins, survival pathways, and apoptosis. Journal of the American College of Cardiology 52, 470-482.
Wilson P.W., Paffenbarger R.S., Morris J.N., Havlik R.J. (1986) Assessment methods for physical activity and physical fitness in population studies: report of a NHLBI workshop. American Heart Journal 111, 1177-1192.
Wong J.M., Collins K. (2003) Telomere maintenance and disease”. Lancet 362, 983-988.
Woo J., Suen E., Tang N.L.S. (2010) Telomeres and the ageing process. Reviews in Clinical Gerontology 20, 1-9.
Woo J., Tang N.L., Suen E., Leung J.C., Leung P.C. (2008) Telomeres and frailty. Mechanisms of Ageing and Development 129, 642-648.
Woo J., Yu R., Tang N., Leung J. (2014) Telomere length is associated with decline in grip strength in older persons aged 65 years and over. Age (Dordrecht, Netherlands) 36, 9711-014-9711-7.








Back
|
PDF
|
Share