You are on page 1of 9

Journals of Gerontology: Biological Sciences

cite as: J Gerontol A Biol Sci Med Sci, 2017, Vol. 00, No. 00, 1–9
doi:10.1093/gerona/glx082
Advance Access publication May 16, 2017

Special Issue: Caloric Restriction and Restrictive Diets: Interventions that Target the
Biology of Aging: Review in Depth

Telomeres, Nutrition, and Longevity: Can We Really


Navigate Our Aging?
Nikolina Škrobot  Vidaček,1 Lucia  Nanić,1 Sanda  Ravlić,1 Mary  Sopta,1 Marko  Gerić,2
Goran Gajski,2 Vera Garaj-Vrhovac,2 and Ivica Rubelj1
Department of Molecular Biology, Ruđer Bošković Institute, Zagreb, Croatia. 2Mutagenesis Unit, Institute for Medical Research and
1

Occupational Health, Zagreb, Croatia.

Address correspondence to: Ivica Rubelj, PhD, Laboratory for Molecular and Cellular Biology, Division of Molecular Biology, RBI, Bijenička cesta
54, 10 000 Zagreb, Croatia. E-mail: rubelj@irb.hr

Received: November 28, 2016; Editorial Decision Date: April 20, 2017

Decision Editor: Rozalyn Anderson, PhD

Abstract
Telomeres are dynamic chromosome-end structures that serve as guardians of genome stability. They are known to be one of the major determinants
of aging and longevity in higher mammals. Studies have demonstrated a direct correlation between telomere length and life expectancy, stress, DNA
damage, and onset of aging-related diseases. This review discusses the most important factors that influence our telomeres. Various genetic and
environmental factors such as diet, physical activity, obesity, and stress are known to influence health and longevity as well as telomere dynamics.
Individuals currently have the opportunity to modulate the dynamics of their aging and health span, monitor these processes, and even make future
projections by following their telomere dynamics. As telomeres react to positive as well as negative health factors, we should be able to directly
influence our telomere metabolism, slow their deterioration, and diminish our aging and perhaps extend our life and health span.

Keywords: Telomeres—Human aging—Longevity—Dietary restriction

In recent years, evidence has accumulated revealing the molecular well. In various population studies, telomere length is measured
mechanisms of aging. Among the primary hallmarks of aging that in peripheral leukocytes that correlates well with telomere length
have been described thus far (1), telomere length has garnered much in other body tissues (11). Average telomere length has been used
attention in the past two decades. Telomeres are specialized struc- as a reliable biomarker for prediction of health span and longevity
tures found at the ends of chromosomes and in addition to being both in humans and animals, with the exception of the oldest old
the primary gatekeepers of genome stability, they play major role in subpopulation (12). Therefore, besides measuring average telomere
senescence and aging (2). Telomeres are dynamic structures in that length, it is important to follow changes in telomere length during
they shorten with each cell division (3). Since even a single too short time as well as proportion of short telomeres in overall sample (13).
telomere is detected as DNA damage which induces permanent cell Several methodologies have been routinely used for telomere length
cycle arrest, sooner or later all cycling somatic cells will permanently measurement, including Q-PCR which measures the average length
stop dividing and enter senescence, whether in vitro or in vivo (4). of bulk telomeres, while Southern blotting and Q-PNA-FISH-based
Indeed, numerous studies point to the accumulation of senescent cells methods measure telomere length distributions. The latter methods
as the main cause of aging and aging-related diseases observed in vari- provide a measure of the shortest telomere fraction which is more
ous organisms (5–7). The strongest evidence for a telomere ­controlled relevant for cell survival and as such is a better predictive factor for
mechanism of aging comes from experiments where ectopic expres- morbidity and mortality in humans (13).
sion of telomerase in normal cells or transgenic animals prevents Of particular interest is the fact that environmental and lifestyle
senescence and postpones or even reverses aging (8–10). factors influence telomere metabolism and their effects on health
In addition to measuring telomere length at the cellular level, and longevity (14,15). Therefore, in this review, we explore the rela-
telomere shortening can be followed at the population level as tionship between telomeres, genome integrity, aging, and lifestyle in
© The Author 2017. Published by Oxford University Press on behalf of The Gerontological Society of America. All rights reserved.
1
For permissions, please e-mail: journals.permissions@oup.com.
2 Journals of Gerontology: BIOLOGICAL SCIENCES, 2017, Vol. 00, No. 00

light of recent findings in primates (16,17) and humans (14,15) that


open up new perspectives on how to combat aging and aging-related
diseases. Telomere molecular metabolism underlies the observa-
tions of the U.S. Centers for Diseases Control and Prevention (CDC,
2009) and the World Health Organization (WHO, 2005) that aging-
related diseases, such as cancer, type 2-diabetes, and coronary heart
disease, could be reduced between 40% and 80% just by changing
one’s lifestyle. This means primarily implementing an antiaging diet
and moderate but regular physical activity.

Free Radical Effects on Telomeres


The biology of genome stability and telomeres includes many inter-
actions between these two factors. Short and dysfunctional telom-
eres are the initiating point for cell senescence, cell death, and DNA
instability (18). The effect of aging on the level of DNA damage in
human cells has been assessed in many studies (19,20). Over time,
genome damage accumulates probably due to lower DNA repair
capacity, lower chromosome segregation, and cell cycle checkpoint
efficacy (21). Unhealthy factors, which include endogenous geno-
toxins, inadequate nutrition, and other unfavourable lifestyle fac-
tors, are also responsible for both an increase in baseline genome
damage and accelerated telomere shortening. Most of these fac-
Figure 1.  Influence of endogenous and exogenous factors on aging. Aging is
tors have an indirect mechanism of action through free radical a plastic phenomenon that can be either positively or negatively influenced
formation and oxidative stress. This points to a conclusion that by various endogenous or exogenous factors. Accumulating evidence points
since reactive oxygen species (ROS) are predominantly responsible to telomeres and mitochondria as the main factors in cellular aging control
for age-related damage at the cellular and tissue levels (22,23), a that consequently dictate the aging process at the organismal level. Recently,
reduction in calorie intake would lead to a reduction in energy these two aspects have been connected in that critically short telomeres
trigger the DNA damage response which results in an increase in p53 protein.
metabolism and ROS production. Consequently, this would result
In addition to activating mechanisms of cell cycle arrest through activation
in diminished cell and tissue damage. Indeed, ROS strongly influ- of p21, increasing levels of p53 also results in the repression of PGC1α and
ences replicative senescence and aging through accelerated tel- β, the main promotors of mitochondrial biogenesis, causing compromised
omere shortening caused primarily by accumulation of nicks in the mitochondrial function. Dysfunctional mitochondria release even more
G-rich strand (24). reactive oxygen species (ROS) into the cytoplasm causing further damage
Under physiological conditions, mitochondria are the main to the cell and accelerating the onset of senescence. External factors such as
irradiation, pollution, environmental stress as well as unhealthy habits like
source of ROS and it has been demonstrated that senescence of
smoking, alcohol consumption, unhealthy food, lack of exercise, obesity, etc.
human cells is related to dysfunctional mitochondria and shorter tel- can contribute to increased ROS production and other negative effects of
omeres (25) (Figure 1). Overproduction of ROS results in oxidative aging. On the other hand, cellular defence mechanisms like ROS scavenging
stress which can lead to oxidation of biomolecules including lipids, enzymes, antioxidants such as vitamins, flavonoids and carotenoids can
proteins, and DNA over time. Such loss of DNA structure or its diminish ROS production and preserve our telomeres, reduce mitochondrial
information content typically cannot be replaced (26). Obviously, damage and extend our health span. Positive lifestyle habits such as regular
moderate exercise, relaxation and healthy diet can similarly improve our
there is a valid reason why chronic oxidative stress is perceived as
aging profile.
one of the leading causes of many metabolic and neurodegenerative
diseases and chronic inflammation. Inflammation causes accelerated
white blood cell turnover and additional telomere attrition resulting metabolism linked to some of the beneficial effects of calorie restric-
in accelerated aging (27,28). tion (1).
Furthermore, mitochondrial and telomere mechanisms of aging Also, mitochondria can contribute to aging independently of
have been united and a direct molecular connection between telomere ROS, through numerous types of mitochondrial dysfunctions like
attrition and mitochondrial dysfunction has been established (29). It impaired mitochondrial biogenesis, decreased mitophagy and fission
has been shown that p53 activation induced by telomere shortening, fusion ratios, mutations and deletions in mtDNA or perturbations
in addition to halting the cell cycle, downregulates the expression of on electron transport chain. All this can cause reduced efficiency of
PGC1α and β which leads to reduction of mitochondrial biogenesis mitochondrial bioenergetics (1).
and function. Dysfunctional mitochondria demonstrate metabolic Nevertheless, it should be emphasized that ROS are normal
changes, such that they exhibit defective ATP generation, increased intermediaries of biochemical reactions which serve as important
ROS production, and decreased levels of ROS-detoxifying enzymes physiological regulators of intracellular signaling pathways (30).
which cause additional damage both to mitochondrial and genomic Depending on the circumstances, they can be either harmful or ben-
DNA, including telomeres (29) (Figure 1). eficial to living systems (31). Beneficial effects of ROS occur at low/
Besides p53 protein, sirtuin family of NAD-dependent protein moderate concentrations and involve physiological roles in cellular
deacetylases and ADP-ribosyltransferases plays an important role responses to anoxia, defense against infectious agents, or induction
in control of mitochondrial function. SIRT1 modulates mitochon- of a mitogenic response (30). In contrast, a significant increase in
drial biogenesis through deacetylation and activation of PGC-1α oxidative stress induces premature senescence in many human cell
while SIRT3 deacetylates mitochondrial enzymes involved in energy types (32). Thus, the delicate balance between the beneficial and
Journals of Gerontology: BIOLOGICAL SCIENCES, 2017, Vol. 00, No. 00 3

harmful effects of free radicals is a very important aspect of human aging-related pathologies so it has yet to be established whether
aging (33) as proposed in the gradual ROS response hypothesis (23). resveratrol can slow the aging process by lowering the incidence of
chronic diseases in humans. To achieve this goal, it is necessary to
establish the optimal dosage of resveratrol administration because of
Nutrients and Food Groups That Protect its very low bioavailability in humans. Furthermore, resveratrol and
Telomeres its metabolites accumulate in human cells in a tissue-specific manner
As mentioned earlier, oxidative stress has a great impact on telomere which greatly depends on the dosage. Different dietary factors, for
shortening. It is expected that an excess of oxidants can be offset example, high-fat meals consumed by some individuals may influ-
with antioxidants, substances capable of donating electrons, and ence plasma resveratrol concentrations which may differ consider-
neutralizing free radicals in the cell (34). Therefore, experiments ably within population (46).
were performed examining the effect of the most common antioxi-
dant compounds found in food such as vitamins, minerals, polyphe- Fats
nols, and omega-3 fatty acids in prevention of telomere shortening Omega-3 fatty acids have been recognized over the last decade as
and possibly slowing down the aging process (35) (Figure 1). important molecules for well-being and in particular helpful for the
cardiovascular system. It has been shown that there is a negative cor-
Vitamins and Minerals relation between the blood level of marine omega-3 fatty acids (doc-
Although in vitro experiments have shown positive results with osahexaenoic and eicosapentaenoic acids) in patients with coronary
antioxidants regarding telomere metabolism (36), similar results heart disease and the rate of their telomere attrition over a 5-year
with in vivo experiments are considered more relevant because period (47). A recent randomized controlled 4 months trial showed
more complex biological factors might influence telomere shorten- that it is not omega-3 itself, but rather the ratio between omega-
ing. Vitamins and minerals have particularly garnered the spotlight 3(n-3) and omega-6 (n-6) fatty acids that is important, as telomere
recently because of their widespread use among the general popula- length increases with decreasing n-6:n-3 plasma ratios regarding
tion and the potential for increasing commercial value. Vitamins C, baseline telomere length. Telomerase activity was unchanged during
D, E, folate, and β-carotene and the minerals zinc and magnesium the study in these individuals but there was a significant negative cor-
have shown positive effects in protection against oxidative stress and relation between telomere length and biomarkers of oxidative stress
inflammation which leads to telomere protection and are positively and inflammation, two factors that affect telomere shortening (48).
associated with telomere length in humans (37). So, the individuals with higher endogenous n-6:n-3 polyunsaturated
fatty acid ratios would have greater benefits from simple nutritional
intervention in the form of omega-3 supplementation.
Naturally Occurring Polyphenols
Polyphenols are naturally occurring chemicals that have also shown
positive effects on telomere length and organismal aging. Study con- Food Groups
ducted on a Belgian population demonstrated antioxidant properties Based on the premise that higher intakes of fresh fruits, vegetables,
of theaflavins, polyphenols found in green and black tea, as well legumes, fish, poultry, and whole grains lower markers of inflamma-
as its negative association with biomarkers of inflammation (38). tion (49), it can be concluded that antioxidant-rich food may have a
Consequently, polyphenols may have a positive influence on tel- positive influence on telomere length. Indeed, consumption of seeds,
omere length. However, in western society’s tea drinking is gener- nuts, legumes, seaweeds, and coffee was associated with longer tel-
ally associated with a healthier lifestyle which could have influenced, omeres (50). In addition, dietary fiber intake, specifically from cereals
these observed levels of inflammation markers compared to the gen- and whole grains, has a positive effect on telomere length in women
eral population. Elderly Chinese who are habitual tea drinkers have (51). An interesting finding is that colonocyte telomere shortening
longer telomeres which correspond to an average increase of 5 years in rats fed with red or white meat can be attenuated by the inclu-
in life span as compared to their counterparts who do not drink tea sion of resistant starch in the diet suggesting a protective effect of
as frequently (39). This association was observed only in men but dietary fiber (52). Furthermore, intake of processed meats was nega-
not in women. A  particular weakness of this study is that it does tively linked to telomere length in a multiethnic study (53) but not
not include young or middle-aged male subjects and as both studies among African American and Hispanic population (50). Because of
are cross-sectional in nature a strict causal relationship cannot be their high fat and protein content, heat-generated glycotoxins are
established. produced that increase cell-oxidant stress and promote inflamma-
Another significant naturally occurring polyphenol related to tel- tion (54) which has been associated with inflammatory diseases like
omere length is resveratrol which activates SIRT1, an intracellular type-2 diabetes and atherosclerosis (55). There have been conflict-
regulatory protein that regulates important metabolic and physio- ing reports regarding fruit and vegetable consumption and telomere
logical processes (40). It is commonly found in the skin of red grapes length. However, there is at least some evidence that they can have a
and has antioxidant and anti-inflammatory properties as well as a positive influence on telomere length and further studies are needed
positive effect on general health in mammals (40,41). Resveratrol to confirm these results (50,56).
delays senescence at the cellular level, increases telomere length Accumulation of information on the positive effects of various
and telomerase activity in rodents but does not extend life span in food compounds on molecular mechanisms that dictate the aging
healthy rodents or in a rodent model of premature aging (42,43). process and resistance to developing diseases open up the opportu-
However, synthetic small molecules, sirtuin agonists SRT2104 and nity to make better choices towards healthier nutritional habits that
SRT1720, extend both mean and maximal life span of mice fed a will prolong our health and life span. To benefit the most from all of
standard diet (44,45). In humans, resveratrol has been reported the aforementioned positive effects, the best sources of antioxidants
to decrease oxidative stress and attenuate inflammation, reduce are from food rather than dietary supplements due to the synergistic
the risk of cardiovascular diseases and diabetes (46). These are all effect of a variety of compounds found in the original plant product.
4 Journals of Gerontology: BIOLOGICAL SCIENCES, 2017, Vol. 00, No. 00

In addition, the exact dosage and side effects for dietary supplements feeding (68) have also been used as alternative models for impacting
have yet to be established. Accordingly, it has been shown that some telomere length and health span. Most of these interventions have
compounds, like β-carotene, are positively related to lung cancer positive effects on overall health and longevity in model organisms
incidence in smokers (57). (68,69). It has been shown that short-term dietary restriction has
multiple benefits in mammals including modulation of inflammatory
responses (70), reduction in cell senescence, and decreases in oxida-
Dietary Patterns with Effect on Telomere tive stress markers in the small intestinal epithelium and liver in mice
Length and Aging (71). Recent work suggests that a fasting-mimicking diet (FMD)
An association between telomere length and nutritional status can consisting of cycles of short-term FMD followed by a standard ad
be seen since birth because exclusive breastfeeding in the first 4–6 libitum diet can produce health benefits in people and extend the
weeks of life is associated with longer telomeres in Latino preschool life span of mice (72). The aforementioned evidence suggests that
children at 4 and 5 years of age (58). Later in life various diets show beneficial effects on health span and life span can be achieved by less
beneficial effects on human health. Among those, reduced calorie invasive dietary interventions, rather than CR, which do not require
intake has raised much attention both among scientists and the gen- long lasting or overall reduction in calorie intake.
eral public because it is the only nonpharmacological intervention While a positive effect of CR on telomere length and telomerase
known to date that slows down the aging process and increases both activity in rodents has been well documented (73), there have been
average and maximal life span in rodents, fish, fruit flies, worms, and very few reports regarding the association of energy intake with
yeast (59,60). telomere length in humans. A  cross-sectional Multiethnic Study
of Atherosclerosis that encompasses men and women showed a
Calorie Restriction nonsignificant negative correlation of energy intake with telomere
A longitudinal calorie restriction (CR) studies in rhesus monkeys length (53). The absence of a correlation between energy intake
(Macaca mulatta) implied that CR can prolong life span in longer- and telomere length was also found in the Nurses’ Health Study
lived species (61). These investigations raised the hope that CR (51) and in elderly Chinese men (39). In contrast, research con-
might be effective in humans. However, contradictory results were ducted on young adults in Jerusalem showed a significant nega-
later published which indicated that, while some health benefits were tive correlation of dietary energy and macronutrient intake with
observed, CR had no effect on longevity in these monkeys (62). Both both baseline and follow-up telomere length in men only (74). It
studies demonstrated that CR in tested animals delayed the onset of is important to emphasize that the test subjects in this study were
several aging associated pathologies such as diabetes, cancer, cardio- younger (age 30–43) than in other reports. Recently an article was
vascular diseases, and brain atrophy (61,62). Another study showed published describing the connection of plasma irisin levels and
that CR does not significantly affect telomere length in the skin and telomere length. Irisin is a hormone released from skeletal mus-
muscle of rhesus monkeys (63). Possible differences in the life span cles after exercise which may induce CR-like effects by increas-
of CR animals can be explained by differences in dietary composi- ing energy cost from adipose tissue. Using telomere length as a
tion, vitamin and mineral supplementation, husbandry, and genetic marker of aging, this study showed a significant positive correla-
background. It was shown that all calories are not the same, and tion between plasma irisin levels and telomere length (75).
significantly it is the quality of the source of calories that matters
(62). Meanwhile, the effect of CR on humans has been poorly stud- Mediterranean Diet
ied. The great number of centenarians and the high average life span In the last 15  years, a lot of attention has focused on the
of Okinawa women who consumed 15%–20% less calories than Mediterranean diet, proven to be one of the healthiest diets in the
mainland Japanese throughout their lifetime may be living proof world (50,76). It respects all the principles of the healthy antiaging
that moderate CR in combination with a well-balanced diet can diet mentioned earlier as it is based on the intake of seasonal fruits,
have significant effect on human life span (64). CR in the Biosphere vegetables, nuts and seeds, whole grains, olive oil, fish and low fat
2 experiment due to unexpected food insufficiency showed that meat, and dairy products as well as moderate intake of alcohol. As
healthy nonobese humans on a low-calorie, nutrient-dense diet a result, the Mediterranean diet has been linked to low morbidity,
show physiologic, hematologic, hormonal, and biochemical alterna- lower occurrence of some chronic, especially cardiovascular diseases
tions consistent with caloric restricted rodents and primates (65). and consequently greater longevity (50,76). The Mediterranean diet
Following the first 6  months of the first randomized human trial, lowers the level of oxidative stress markers and inflammation due
it was shown that CR had positive physiological and psychologi- to a high abundance of antioxidant compounds such as omega-3
cal effects including improvement of several markers of aging (64). and resveratrol (48,77). Importantly, it has a direct positive effect
This was confirmed by a later study where the effects of CR on dis- on telomere length (48,78). Additional confirmation of a positive
ease risk and human survival suggested potential benefits for aging- influence of the Mediterranean diet on telomere length came from a
related outcomes (66). Although the effect of CR intervention on life Nurses’ Health Study which is one of the largest research studies into
span in humans remains to be determined unequivocally, evidence the risk factors for major chronic diseases in women. Adherence to a
thus far suggests that it can be a successful step to prolonged health Mediterranean diet managed to preserve a telomere length that cor-
span and healthy aging. responded to 4.5 years of aging which is comparable to the effects
Without strong proof that long-term CR indeed prolongs human of smoking (4.6 years) and physical activity (4.4 years) on the tel-
life and the fact that it is very hard for most individuals to practice omere shortening rate (79). The Mediterranean diet also stimulates
calorie restriction in an overfeeding environment, later studies have telomerase activity in peripheral blood mononuclear cells (50). If
focused on compounds, like resveratrol that mimic calorie restriction combined with moderate exercise, this diet demonstrates elevated
metabolism (67). The effects of various forms of dietary restrictions, long-term improvement in endothelial microvascular and cardiores-
including normocaloric diets with planned deficiencies (in particu- piratory functions, important for both better health and increased
lar macronutrients: proteins or carbohydrates), and time-restricted life expectancy (80).
Journals of Gerontology: BIOLOGICAL SCIENCES, 2017, Vol. 00, No. 00 5

Vegetarian Diets dose-dependent relation with smoking was established and smoking


Vegetarians, including vegetarian subgroups, which differ in their one pack of cigarettes a day over a 40-year period was equivalent
degree of avoidance of meat consumption and based on a degree to an additional 18% of telomere length lost (eg, 7,4 years of life)
of restriction of other animal products (milk, eggs, etc.), have also compared with the rate in the overall sample (89). The connection
been studied for effects on health span and telomere length. Even between passive smoking or smoking cessation and telomere length
though it is perceived as very healthy by the general public, observed has not been investigated adequately. It was shown that prenatal
consequences of a vegetarian diet are higher intake of antioxidants tobacco exposure greatly affects telomere length in children leading
and lower fat intake but at the same time deficiencies in vitamins to a significant health impact (93). Further, longer telomeres were
B, D, iron, and calcium were also shown (81). Over time, conflict- indicated among former smokers but this phenomenon is limited to
ing results were reported regarding general health, antioxidant sta- a 3–16-year period after smoking cessation. It should be emphasized
tus, and DNA damage observed in vegetarians (82) and so far, no that this finding only implied an association rather than causation.
clear conclusions about beneficial effects can be drawn from these Clearly, further investigations on biological pathways underlying
studies without a thorough multibiomarker meta-analysis. As far these results are needed (92).
as telomeres are concerned, there is very little data. One case–con-
trol study conducted on an Indian subjects with coronary artery Exercise
disease showed trend toward longer telomeres in vegetarians com- Correlation between physical activity, sedentary behavior, and tel-
pared with subjects on a mixed diet without a statistical significance omere length has been extensively studied. Physical activity is a
(83). Additional research is needed specifically designed to test this factor that is positively associated with greater leukocyte telomere
correlation. length among healthy individuals and twin volunteers while seden-
tary behavior is negatively correlated with telomere length (88). In
addition, research in patients with coronary heart disease and post-
Behevioral and Psychological Factors That menopausal women with stage I-III breast cancer indicates a strong
Affect Telomeres and Aging association between physical fitness and telomere length (88). On the
In recent years, a growing body of literature has emerged showing other hand, there are some studies that show no significant associa-
a connection between telomere length and various lifestyle factors. tions between physical activity and telomere length (88). It seems
Thus, beside genetics, smoking, obesity, lack of physical activity, and likely that there is no clear answer yet at this point. After adjustment
sleep, stress or depression can increase the rate of telomere short- for other patient characteristics, including markers of cardiac disease
ening that directly correlates to increased incidence of cancer and severity and physical inactivity, participants with low exercise capac-
mortality rate. In contrast, positive correlation of telomere length ity had 94% greater odds of having short telomere length than those
with healthy lifestyle is well-documented, suggesting a great impact with high exercise capacity. A recent 30-year long study showed that
of these factors on telomere maintenance and consequently overall people who practiced moderate physical activity in midlife had longer
health status (84). telomeres in old age than participants who had low or high physi-
cal activity (88). These results suggest a dose-related link between
Obesity and Smoking intensity of physical exercise, telomere length, and baseline DNA
Smoking and obesity affect an increasing number of people world- damage where regular moderate physical activity is most beneficial.
wide. There are opposite results regarding obesity related pheno- In addition, different forms of physical activity should be performed
types and telomere length correlation. Multiple studies have shown at certain ages to obtain the positive effect on telomere maintenance.
that high body mass index (BMI) correlates with short telomere It seems likely that in older adults, physical capacity like the ability
length, DNA damage, and cancer incidence (85). Individuals with to perform activities of daily living, sit-to-stand performance, and
higher total and abdominal adiposity have shorter telomeres sug- walking distance, rather than exercise, may have a greater impact on
gesting that obesity may accelerate the aging process (86), and recip- telomere length (88). In contrast, at younger age, intensity of exercise
rocally shorter telomere length may be a risk factor for increased rather than duration of physical activity may have a beneficial effect
adiposity (87). It should be emphasized that negative effect of obe- on telomeres (88). Further research is needed to find the optimal
sity-related phenotypes on telomere length may be more evident in exercise conditions for telomere length maintenance.
women than men (88). Valdes and colleagues conducted a scientific
study in which participants were 45 monozygotic and 516 dizygotic Alcohol Consumption
twin pairs of Caucasian women aged 18–76 years. The telomeres of Consumption of alcohol is a custom in many societies in the world
obese women were 240 bp shorter than those of their slender coun- and its ambiguous impact on human health is extensively studied
terparts (89). In contrast, some other studies did not find a signifi- over the years. In a long-term study over three decades, a negative
cant association between shorter telomere length and obesity (87). relationship of even minor alcohol consumption in midlife and tel-
Recently, a systematic review was published assessing the effects of omere length in old age is described in the Helsinki Businessman
obesity on telomere length. Sixty-three original studies with a total study (50). Differences in telomere length between nonalcohol users
of 119,439 subjects that satisfied strict inclusion criteria showed that and the group with highest alcohol consumption rate could be rep-
despite weak to moderate associations between obesity and telomere resented as a 10-year difference in biological age. In contrast, the
length, there was a trend toward an negative correlation between Copenhagen City Heart Study shows no association between alco-
those two (90). hol consumption and telomere length measured after 10 years (50).
Tobacco smoke produces free radicals which cause damage to While Nordic populations usually drink spirits, Mediterranean pop-
lipids, proteins, DNA, and other endogenous cellular structures ulations characteristically have a regular intake of red wine and beer,
including telomeres and increase various inflammation biomarkers both polyphenol-rich beverages. One study explored the association
(91). A  negative correlation between active smoking and telomere between telomere length and alcohol consumption in an Italian pop-
length has been indicated in many studies (85,92). In addition, a ulation but no effects on telomere length were detected (50).
6 Journals of Gerontology: BIOLOGICAL SCIENCES, 2017, Vol. 00, No. 00

Mental Health causality but rather are indicative of an association (15). We should


It is well known that better mental health greatly contributes to emphasize that there was no clinical evidence of prostate cancer pro-
physical health as well. Despite some contradictory reports, there are gression in these subjects during the test period. Still, one should
a large number of results that support a connection between shorter keep in mind that lifestyle changes cannot be substituted for conven-
telomere length and chronically high levels of perceived stress, tional treatment in patients with more aggressive forms of cancer.
especially in caregivers, maternal stress during pregnancy, and the A recent study that followed 231 healthy women over a short,
number of childhood adversities (11). There is also a positive cor- 1-year period showed that for every major life stress event like ill-
relation between accelerated telomere shortening and psychological ness, death of a family member or friend, financial, employment or
disorders like depression and high phobic anxiety (11). Also, sleep marital problems that occurred during that time, there was a sig-
deprivation (less than 5 hours) shortens telomeres by 6% compared nificantly greater decline in telomere length. However, these impacts
with subjects sleeping 7 hours or more (11). were ameliorated by healthy behaviors that included self-reported
physical activity, typical food consumption, and sleep quality (94). It
It should be stated that most of these studies are observational
seems likely that a combination of healthy behaviors has a stronger
and there were conflicting results for each of the above-mentioned
influence on telomere length than each factor alone (94). The weak-
factors. These inconsistencies are likely due to the biological diversi-
ness of this study is that results are limited to postmenopausal and
ties in the subjects such as genetic polymorphisms, ethnicity, age, and
nonsmoking primarily Caucasian women that are highly educated,
sex differences. Furthermore, DNA extraction methods, study par-
healthy, and have lower levels of stress than the national average.
ticipant sampling, and methodological differences are a contributing
factor for these discrepancies. Despite these limitations, the studies
reviewed provide insight into the complex connection between tel- Meditation
omere length and various lifestyle factors. In addition to lifestyle and diet, it is well known that better mental
health greatly contributes to physical health as well. There are a few
studies that directly link meditation and positive psychological change
Can Dietary and Lifestyle Interventions with telomerase activity showing that mental health is an adjustable
Improve Telomere Stability And Modulate the factor associated with telomere length. In a pilot study of yogic medi-
Aging Process? tation for depressed dementia caregivers, 23 subjects in the meditation
Lifestyle group were randomized to practice Kirtan Kriya and 16 subjects in
the relaxation group listened to relaxation music for 12 minutes per
With respect to the accumulating evidence that telomere length
day for eight weeks. Telomerase activity in peripheral blood mononu-
can be modulated by a variety of lifestyle factors, a longitudinal
clear cells was measured pre- and postintervention. The meditation
study has been conducted in order to closely examine the direct
group showed a 43% increase in telomerase activity, accompanied
influence of comprehensive lifestyle changes on telomerase activ-
by improved mental and cognitive functioning and lower levels of
ity and telomere length (14). Comprehensive lifestyle changes
depressive symptoms compared with a relaxation control group. This
imply a specific diet accompanied by stress management, mod-
increase in telomerase activity can be associated with a lower level
erate aerobic exercise, and group support sessions. In the pilot
of stress-induced cellular aging (95). In the study by Jacobs and col-
study that lasted for 3  months, 24 men with biopsy-diagnosed
leagues, 60 men and women matched for age, sex, body mass index,
low risk prostate cancer who chose active surveillance rather than
and prior meditation experience were randomly divided into two
conventional treatments were provided precooked meals based on
groups. Retreat participants meditated for 6 hours daily for 3 months
food low on fat and refined carbohydrates, with plenty of fruits,
and the other half served as a wait-list control group A  3-month
vegetables, unrefined grains, and legumes supplemented with soy,
retreat of intensive meditation training resulted in 30% higher telom-
fish oil, vitamins E and C, and selenium. Additionally, they were
erase activity in retreat participants as compared with a control group
asked to conduct moderate exercise in the form of walking 30
(96). Weak points in this study are the absence of a baseline measure
minutes/d, 6  days/wk. Stress management included gentle yoga
of telomerase activity, relatively small sample size, and the control
based stretching, breathing, meditation, and relaxation tech-
condition that did not match the experimental condition such that the
niques for 1 hour/d, 6  days/wk and psychological support ses-
control group was not spared from the stresses of everyday life (96).
sions once a week.
However, a recent meta-analytic review based on 190 participants in
The most important result of this study is that strictly follow-
four initial randomized control trials indicates a positive correlation
ing lifestyle recommendations for only 3  months was sufficient to
between mindfulness meditation and telomerase activity (97).
increase telomerase activity in peripheral blood mononuclear cells
(PBMC) of test subjects by almost 30%. This increase in telomerase
activity was correlated with improvements of risk factors for car- Antiaging Intervention Comes of Age
diovascular diseases as well as a decrease in the aging biomarker C A more direct and so far the only serious attempt to combat aging
reactive protein (CRP) value (14). Ornish and colleagues conducted through telomerase activation and telomere elongation is the use of
a 5-year follow-up study in order to determine whether the long- TA-65, a dietary supplement isolated from the Chinese traditional
term comprehensive lifestyle changes affect telomere length as well. medicine herb Astragalus membranaceus (98,99). This approach
Over time assessment of gross telomere shortening showed a reduc- opens up the possibility of health and life extension, or even reju-
tion such that the relative telomere length decreased in only ~30% of venation, as has been demonstrated in transgenic animals (8–10).
subjects in lifestyle intervention group compared to 64% of subjects Although some expression of telomerase in normal somatic cells
in the control group. It is interesting that a dose–response relation in culture was detected, the use of TA-65 did not show significant
has been established between the lifestyle change adherence score effects on human subjects during a 1-year trial (98,99). However,
and degree of change in telomere length. The weakness of this study while there was no or just a slight effect on average telomere length,
is its relatively small sample group; so, these results do not address more important reduction in the proportion of the shortest telomere
Journals of Gerontology: BIOLOGICAL SCIENCES, 2017, Vol. 00, No. 00 7

fraction (>4  kb or bottom 20%) was demonstrated in most sub- for biobehavioral research. Nurs Res. 2014;63:36–50. doi:10.1097/
jects. Since these trials were performed over a relatively short period NNR.0000000000000009
of time, it will take years before some positive effects of TA-65 on 12. Bär C, Blasco MA. Telomeres and telomerase as therapeutic targets to pre-
vent and treat age-related diseases. F1000Research. 2016;5. doi:10.12688/
health and longevity become more obvious.
f1000research.7020.1
13. Vera E, Blasco MA. Beyond average: potential for measurement of

short telomeres. Aging (Albany NY). 2012;4:379–392. doi:10.18632/
Conclusions
aging.100462
We live in an exciting era in which the research field of aging and 14. Ornish D, Lin J, Daubenmier J, et al. Increased telomerase activity and com-
senescence is quickly growing because the mechanisms of cell senes- prehensive lifestyle changes: a pilot study. Lancet Oncol. 2008;9:1048–
cence and aging have been largely elucidated. Telomere shorten- 1057. doi:10.1016/S1470-2045(08)70234-1
ing has been established as one of the main causes of cellular and 15. Ornish D, Lin J, Chan JM, et al. Effect of comprehensive lifestyle changes
organismal aging in higher mammals (100). Aging is a “disease” that on telomerase activity and telomere length in men with biopsy-proven low-
risk prostate cancer: 5-year follow-up of a descriptive pilot study. Lancet
affects the whole population without exception so finding a “cure”
Oncol. 2013;14:1112–1120. doi:10.1016/S1470-2045(13)70366-8
is the next logical step. But before we discover an effective cure, we
16. Gardner JP, Kimura M, Chai W, et al. Telomere dynamics in macaques and
have the opportunity to modulate our aging through comprehen-
humans. J Gerontol A Biol Sci Med Sci. 2007;62:367–374. doi:10.1093/
sive lifestyle changes. These findings may seem like common sense gerona/62.4.367
but they are based on hard scientific evidence about factors that are 17. Herbig U, Ferreira M, Condel L, Carey D, Sedivy JM. Cellular senescence
both beneficial or harmful for our health and aging. Armed with this in aging primates. Science. 2006;311:1257. doi:10.1126/science.1122446
knowledge, we can now monitor the aging process by measuring our 18. Kim Sh S, Kaminker P, Campisi J. Telomeres, aging and cancer: in

telomere status through commercially available services and respond search of a happy ending. Oncogene. 2002;21:503–511. doi:10.1038/sj/
accordingly. Thus, for the first time we really can navigate our aging! onc/1205077
19. Bautista-Niño PK, Portilla-Fernandez E, Vaughan DE, Danser AHJ, Roks
AJM. DNA damage: a main determinant of vascular aging. Int J Mol Sci.
Funding 2016;17. doi:10.3390/ijms17050748
20. Fenech M, Bonassi S. The effect of age, gender, diet and lifestyle on DNA
This work was supported by Zaklada Adris.
damage measured using micronucleus frequency in human peripheral
blood lymphocytes. Mutagenesis. 2011;26:43–49. doi:10.1093/mutage/
geq050
Conflict of Interest 21. Moskalev AA, Shaposhnikov MV, Plyusnina EN, et al. The role of DNA
The authors declare no conflict of interest. damage and repair in aging through the prism of Koch-like criteria. Ageing
Res Rev. 2013;12:661–684. doi:10.1016/j.arr.2012.02.001
22. De la Fuente M. Effects of antioxidants on immune system ageing. Eur J
References
Clin Nutr. 2002;56(suppl 3):S5–8. doi:10.1038/sj.ejcn.1601476
1. López-Otín C, Blasco MA, Partridge L, Serrano M, Kroemer G. 23. Hekimi S, Lapointe J, Wen Y. Taking a “good” look at free radicals in
The hallmarks of aging. Cell. 2013;153:1194–1217. doi:10.1016/ the aging process. Trends Cell Biol. 2011;21:569–576. doi:10.1016/
j.cell.2013.05.039 j.tcb.2011.06.008
2. Karlseder J, Smogorzewska A, de Lange T. Senescence induced by 24. von Zglinicki T, Pilger R, Sitte N. Accumulation of single-strand breaks is
altered telomere state, not telomere loss. Science. 2002;295:2446–2449. the major cause of telomere shortening in human fibroblasts. Free Radic
doi:10.1126/science.1069523 Biol Med. 2000;28:64–74. doi:10.1016/S0891-5849(99)00207-5
3. Harley CB, Futcher AB, Greider CW. Telomeres shorten during ageing of 25. Passos JF, Saretzki G, Ahmed S, et al. Mitochondrial dysfunction accounts
human fibroblasts. Nature. 1990;345:458–460. doi:10.1038/345458a0 for the stochastic heterogeneity in telomere-dependent senescence. PLoS
4. Blasco MA. Telomeres and human disease: ageing, cancer and beyond. Nat Biol. 2007;5:e110. doi:10.1371/journal.pbio.0050110
Rev Genet. 2005;6:611–622. doi:10.1038/nrg1656 26. Lieber MR, Karanjawala ZE. Ageing, repetitive genomes and DNA dam-
5. Baker DJ, Wijshake T, Tchkonia T, et al. Clearance of p16Ink4a-positive age. Nat Rev Mol Cell Biol. 2004;5:69–75. doi:10.1038/nrm1281
senescent cells delays ageing-associated disorders. Nature. 2011;479:232– 27. Salpea KD, Talmud PJ, Cooper JA, et al. Association of telomere length with
236. doi:10.1038/nature10600 type 2 diabetes, oxidative stress and UCP2 gene variation. Atherosclerosis.
6. Sugimoto M, Yamashita R, Ueda M. Telomere length of the skin in 2010;209:42–50. doi:10.1016/j.atherosclerosis.2009.09.070
association with chronological aging and photoaging. J Dermatol Sci. 28. Khan S, Chuturgoon AA, Naidoo DP. Telomeres and atherosclerosis.

2006;43:43–47. doi:10.1016/j.jdermsci.2006.02.004 Cardiovasc J Afr. 2012;23:563–571. doi:10.5830/CVJA-2012-056
7. Kim S, Jiang JC, Kirchman PA, Rubelj I, Helm EG, Jazwinski SM. 29. Sahin E, Colla S, Liesa M, et al. Telomere dysfunction induces metabolic
Cellular and Molecular Aging. In: Balducci L, Ershler WB, Lyman GH, and mitochondrial compromise. Nature. 2011;470:359–365. doi:10.1038/
eds. Comprehensive Geriatric Oncology. 2nd ed. Amsterdam: Harwood nature09787
Academic; 1998:123–55. 30. Finkel T. Signal transduction by reactive oxygen species. J Cell Biol.

8. Bernardes de Jesus B, Vera E, Schneeberger K, et  al. Telomerase gene 2011;194:7–15. doi:10.1083/jcb.201102095
therapy in adult and old mice delays aging and increases longevity with- 31. Valko M, Rhodes CJ, Moncol J, Izakovic M, Mazur M. Free radicals,
out increasing cancer. EMBO Mol Med. 2012;4:691–704. doi:10.1002/ metals and antioxidants in oxidative stress-induced cancer. Chem Biol
emmm.201200245 Interact. 2006;160:1–40. doi:10.1016/j.cbi.2005.12.009
9. Jaskelioff M, Muller FL, Paik JH, et  al. Telomerase reactivation 32. Colavitti R, Finkel T. Reactive oxygen species as mediators of cellular senes-
reverses tissue degeneration in aged telomerase-deficient mice. Nature. cence. IUBMB Life. 2005;57:277–281. doi:10.1080/15216540500091890
2011;469:102–106. doi:10.1038/nature09603 33. Pham-Huy LA, He H, Pham-Huy C. Free radicals, antioxidants in disease
10. Bodnar AG, Ouellette M, Frolkis M, et al. Extension of life-span by intro- and health. Int J Biomed Sci. 2008;4:89–96.
duction of telomerase into normal human cells. Science. 1998;279:349– 34. Sies H. Oxidative stress: oxidants and antioxidants. Exp Physiol.

352. doi:10.1126/science.279.5349.349 1997;82:291–295. doi:10.1113/expphysiol.1997.sp004024
11. Starkweather AR, Alhaeeri AA, Montpetit A, et  al. An integrative
35.
García-Calzón S, Moleres A, Martínez-González MA, Martínez
review of factors associated with telomere length and implications JA, Zalba G, Marti A; GENOI members. Dietary total antioxidant
8 Journals of Gerontology: BIOLOGICAL SCIENCES, 2017, Vol. 00, No. 00

capacity is associated with leukocyte telomere length in a children and 54. Vlassara H, Cai W, Crandall J, et al. Inflammatory mediators are induced
adolescent population. Clin Nutr. 2015;34:694–699. doi:10.1016/ by dietary glycotoxins, a major risk factor for diabetic angiopathy. Proc
j.clnu.2014.07.015 Natl Acad Sci USA. 2002;99:15596–15601. doi:10.1073/pnas.242407999
36. Yokoo S, Furumoto K, Hiyama E, Miwa N. Slow-down of age-dependent 55. Schulze MB, Hoffmann K, Manson JE, et  al. Dietary pattern, inflam-
telomere shortening is executed in human skin keratinocytes by hormesis- mation, and incidence of type 2 diabetes in women. Am J Clin Nutr.
like-effects of trace hydrogen peroxide or by anti-oxidative effects of pro- 2005;82:675–84; quiz 714.
vitamin C in common concurrently with reduction of intracellular oxida- 56. Rafie N, Golpour Hamedani S, Barak F, Safavi SM, Miraghajani M.

tive stress. J Cell Biochem. 2004;93:588–597. doi:10.1002/jcb.20208 Dietary patterns, food groups and telomere length: a systematic review
37. Paul L. Diet, nutrition and telomere length. J Nutr Biochem. 2011;22:895– of current studies. Eur J Clin Nutr. 2017;71:151–158. doi:10.1038/
901. doi:10.1016/j.jnutbio.2010.12.001 ejcn.2016.149
38. De Bacquer D, Clays E, Delanghe J, De Backer G. Epidemiological evi- 57. Sommer A, Vyas KS. A global clinical view on vitamin A and carotenoids.
dence for an association between habitual tea consumption and markers of Am J Clin Nutr. 2012;96:1204S–1206S. doi:10.3945/ajcn.112.034868
chronic inflammation. Atherosclerosis. 2006;189:428–435. doi:10.1016/ 58. Wojcicki JM, Heyman MB, Elwan D, Lin J, Blackburn E, Epel E. Early
j.atherosclerosis.2005.12.028 exclusive breastfeeding is associated with longer telomeres in Latino
39. Chan R, Woo J, Suen E, Leung J, Tang N. Chinese tea consumption is preschool children. Am J Clin Nutr. 2016;104:397–405. doi:10.3945/
associated with longer telomere length in elderly Chinese men. Br J Nutr. ajcn.115.115428
2010;103:107–113. doi:10.1017/S0007114509991383 59. Heilbronn LK, Ravussin E. Calorie restriction and aging: review of

40. Baur JA, Pearson KJ, Price NL, et  al. Resveratrol improves health and the literature and implications for studies in humans. Am J Clin Nutr.
survival of mice on a high-calorie diet. Nature. 2006;444:337–342. 2003;78:361–369.
doi:10.1038/nature05354 60. McCay CM, Crowell MF, Maynard LA. The effect of retarded growth
41. Vitseva O, Varghese S, Chakrabarti S, Folts JD, Freedman JE. Grape seed upon the length of life span and upon the ultimate body size one figure.
and skin extracts inhibit platelet function and release of reactive oxygen J Nutr. 1935;10:63–79.
intermediates. J Cardiovasc Pharmacol. 2005;46:445–451. 61. Colman RJ, Anderson RM, Johnson SC, et  al. Caloric restriction delays
42. da Luz PL, Tanaka L, Brum PC, et al. Red wine and equivalent oral phar- disease onset and mortality in rhesus monkeys. Science. 2009;325:201–
macological doses of resveratrol delay vascular aging but do not extend 204. doi:10.1126/science.1173635
life span in rats. Atherosclerosis. 2012;224:136–142. doi:10.1016/j. 62. Mattison JA, Roth GS, Beasley TM, et  al. Impact of caloric restriction
atherosclerosis.2012.06.007 on health and survival in rhesus monkeys from the NIA study. Nature.
43. Labbé A, Garand C, Cogger VC, et al. Resveratrol improves insulin resist- 2012;489:318–321. doi:10.1038/nature11432
ance hyperglycemia and hepatosteatosis but not hypertriglyceridemia, 63. Smith DL Jr, Mattison JA, Desmond RA, et al. Telomere dynamics in rhe-
inflammation, and life span in a mouse model for Werner syndrome. sus monkeys: no apparent effect of caloric restriction. J Gerontol A Biol
J  Gerontol A  Biol Sci Med Sci. 2011;66:264–278. doi:10.1093/gerona/ Sci Med Sci. 2011;66:1163–1168. doi:10.1093/gerona/glr136
glq184 64. Redman LM, Ravussin E. Caloric restriction in humans: impact on physi-
44. Mercken EM, Mitchell SJ, Martin-Montalvo A, et  al. SRT2104 extends ological, psychological, and behavioral outcomes. Antioxid Redox Signal.
survival of male mice on a standard diet and preserves bone and muscle 2011;14:275–287. doi:10.1089/ars.2010.3253
mass. Aging Cell. 2014;13:787–796. doi:10.1111/acel.12220 65. Walford RL, Mock D, Verdery R, MacCallum T. Calorie restriction in
45. Mitchell SJ, Martin-Montalvo A, Mercken EM, et al. The SIRT1 activator biosphere 2: alterations in physiologic, hematologic, hormonal, and bio-
SRT1720 extends lifespan and improves health of mice fed a standard diet. chemical parameters in humans restricted for a 2-year period. J Gerontol
Cell Rep. 2014;6:836–843. doi:10.1016/j.celrep.2014.01.031 A Biol Sci Med Sci. 2002;57:B211–B224. doi:10.1093/gerona/57.6.B211
46. Smoliga JM, Baur JA, Hausenblas HA. Resveratrol and health–a
66. Ravussin E, Redman LM, Rochon J, et  al.; CALERIE Study Group. A
comprehensive review of human clinical trials. Mol Nutr Food Res. 2-year randomized controlled trial of human caloric restriction: feasibility
2011;55:1129–1141. doi:10.1002/mnfr.201100143 and effects on predictors of health span and longevity. J Gerontol A Biol
47. Farzaneh-Far R, Lin J, Epel ES, Harris WS, Blackburn EH, Whooley MA. Sci Med Sci. 2015;70:1097–1104. doi:10.1093/gerona/glv057
Association of marine omega-3 fatty acid levels with telomeric aging 67. Lam YY, Peterson CM, Ravussin E. Resveratrol vs. calorie restriction:
in patients with coronary heart disease. JAMA. 2010;303:250–257. data from rodents to humans. Exp Gerontol. 2013;48:1018–1024.
doi:10.1001/jama.2009.2008 doi:10.1016/j.exger.2013.04.005
48. Kiecolt-Glaser JK, Epel ES, Belury MA, et al. Omega-3 fatty acids, oxida- 68. Lee C, Longo VD. Fasting vs dietary restriction in cellular protection
tive stress, and leukocyte telomere length: a randomized controlled trial. and cancer treatment: from model organisms to patients. Oncogene.
Brain Behav Immun. 2013;28:16–24. doi:10.1016/j.bbi.2012.09.004 2011;30:3305–3316. doi:10.1038/onc.2011.91
49. Lopez-Garcia E, Schulze MB, Fung TT, et al. Major dietary patterns are 69. Lee C, Longo V. Dietary restriction with and without caloric restric-

related to plasma concentrations of markers of inflammation and endothe- tion for healthy aging. F1000Research. 2016;5. doi:10.12688/
lial dysfunction. Am J Clin Nutr. 2004;80:1029–1035. f1000research.7136.1
50. Freitas-Simoes TM, Ros E, Sala-Vila A. Nutrients, foods, dietary patterns 70. Robertson LT, Mitchell JR. Benefits of short-term dietary restric-

and telomere length: update of epidemiological studies and randomized tri- tion in mammals. Exp Gerontol. 2013;48:1043–1048. doi:10.1016/
als. Metabolism. 2016;65:406–415. doi:10.1016/j.metabol.2015.11.004 j.exger.2013.01.009
51. Cassidy A, De Vivo I, Liu Y, et al. Associations between diet, lifestyle fac- 71. Wang C, Maddick M, Miwa S, et al. Adult-onset, short-term dietary restric-
tors, and telomere length in women. Am J Clin Nutr. 2010;91:1273–1280. tion reduces cell senescence in mice. Aging (Albany NY). 2010;2:555–566.
doi:10.3945/ajcn.2009.28947 doi:10.18632/aging.100196
52. O’Callaghan NJ, Toden S, Bird AR, Topping DL, Fenech M, Conlon MA. 72. Brandhorst S, Choi IY, Wei M, et  al. A periodic diet that mimics fast-
Colonocyte telomere shortening is greater with dietary red meat than ing promotes multi-system regeneration, enhanced cognitive perfor-
white meat and is attenuated by resistant starch. Clin Nutr. 2012;31: mance, and healthspan. Cell Metab. 2015;22:86–99. doi:10.1016/
60–64. doi:10.1016/j.clnu.2011.09.003 j.cmet.2015.05.012
53. Nettleton JA, Diez-Roux A, Jenny NS, Fitzpatrick AL, Jacobs DR
73. Vera E, Bernardes de Jesus B, Foronda M, Flores JM, Blasco MA.

Jr. Dietary patterns, food groups, and telomere length in the Multi- Telomerase reverse transcriptase synergizes with calorie restric-
Ethnic Study of Atherosclerosis (MESA). Am J Clin Nutr. 2008;88: tion to increase health span and extend mouse longevity. PLoS One.
1405–1412. 2013;8:e53760. doi:10.1371/journal.pone.0053760
Journals of Gerontology: BIOLOGICAL SCIENCES, 2017, Vol. 00, No. 00 9

74. Kark JD, Goldberger N, Kimura M, Sinnreich R, Aviv A. Energy intake 88. Denham J, O’Brien BJ, Charchar FJ. Telomere length maintenance and car-
and leukocyte telomere length in young adults. Am J Clin Nutr. 2012;95: dio-metabolic disease prevention through exercise training. Sports Med.
479–487. doi:10.3945/ajcn.111.024521 2016;46:1213–1237. doi:10.1007/s40279-016-0482-4
75. Rana KS, Arif M, Hill EJ, et  al. Plasma irisin levels predict telomere
89. Valdes AM, Andrew T, Gardner JP, et al. Obesity, cigarette smoking, and
length in healthy adults. Age (Dordr). 2014;36:995–1001. doi:10.1007/ telomere length in women. Lancet. 2005;366:662–664. doi:10.1016/
s11357-014-9620-9 S0140-6736(05)66630-5
76. Trichopoulou A. Traditional Mediterranean diet and longevity in the
90. Mundstock E, Sarria EE, Zatti H, et  al. Effect of obesity on telomere
elderly: a review. Public Health Nutr. 2004;7:943–947. doi:10.1079/ length: systematic review and meta-analysis. Obesity (Silver Spring).
PHN2004558 2015;23:2165–2174. doi:10.1002/oby.21183
77. Marín C, Yubero-Serrano EM, López-Miranda J, Pérez-Jiménez F.
91. Babizhayev MA, Yegorov YE. Smoking and health: association between
Endothelial aging associated with oxidative stress can be modulated telomere length and factors impacting on human disease, quality of
by a healthy mediterranean diet. Int J Mol Sci. 2013;14:8869–8889. life and life span in a large population-based cohort under the effect
doi:10.3390/ijms14058869 of smoking duration. Fundam Clin Pharmacol. 2011;25:425–442.
78. García-Calzón S, Gea A, Razquin C, et al. Longitudinal association of telomere doi:10.1111/j.1472-8206.2010.00866.x
length and obesity indices in an intervention study with a Mediterranean diet: 92. Wulaningsih W, Serrano FE, Utarini A, Matsuguchi T, Watkins J; PILAR
the PREDIMED-NAVARRA trial. Int J Obes 2005. 2014;38:177–182. doi: Research Network. Smoking, second-hand smoke exposure and smok-
10.1038/ijo.2013.68 ing cessation in relation to leukocyte telomere length and mortality.
79. Crous-Bou M, Fung TT, Prescott J, et al. Mediterranean diet and telomere Oncotarget. 2016;7:60419–60431. doi:10.18632/oncotarget.11051
length in Nurses’ Health Study: population based cohort study. BMJ. 93. Ip P, Chung BH, Ho FK, et  al. Prenatal tobacco exposure shortens

2014;349:g6674. doi:10.1136/bmj.g6674 telomere length in children. Nicotine Tob Res. 2017;19:111–118.
80. Klonizakis M, Alkhatib A, Middleton G. Long-term effects of an exer- doi:10.1093/ntr/ntw139
cise and Mediterranean diet intervention in the vascular function of an 94. Puterman E, Lin J, Krauss J, Blackburn EH, Epel ES. Determinants of
older, healthy population. Microvasc Res. 2014;95:103–107. doi:10.1016/ telomere attrition over 1 year in healthy older women: stress and health
j.mvr.2014.07.015 behaviors matter. Mol Psychiatry. 2015;20:529–535. doi:10.1038/
81. Gaby A. A review of the fundamentals of diet. Glob Adv Health Med mp.2014.70
Improv Healthc Outcomes Worldw. 2013;2:58–63. doi:10.7453/gahmj. 95. Lavretsky H, Epel ES, Siddarth P, et al. A pilot study of yogic medita-
2013.2.1.010 tion for family dementia caregivers with depressive symptoms: effects
82. Burkert NT, Muckenhuber J, Großschädl F, Rásky E, Freidl W. Nutrition on mental health, cognition, and telomerase activity. Int J Geriatr
and health - the association between eating behavior and various Psychiatry. 2013;28:57–65. doi:10.1002/gps.3790
health parameters: a matched sample study. PLoS One. 2014;9:e88278. 96. Jacobs TL, Epel ES, Lin J, et  al. Intensive meditation train-

doi:10.1371/journal.pone.0088278 ing, immune cell telomerase activity, and psychological media-
83. Mukherjee M, Brouilette S, Stevens S, Shetty KR, Samani NJ. Association tors. Psychoneuroendocrinology. 2011;36:664–681. doi:10.1016/j.
of shorter telomeres with coronary artery disease in Indian subjects. Heart. psyneuen.2010.09.010
2009;95:669–673. doi:10.1136/hrt.2008.150250 97. Schutte NS, Malouff JM. A meta-analytic review of the effects of mind-
84. Sun Q, Shi L, Prescott J, et  al. Healthy lifestyle and leukocyte telomere fulness meditation on telomerase activity. Psychoneuroendocrinology.
length in U.S.  women. PloS One. 2012;7:e38374. doi:10.1371/journal. 2014;42:45–48. doi:10.1016/j.psyneuen.2013.12.017
pone.0038374 98. Harley CB, Liu W, Blasco M, et al. A natural product telomerase acti-
85. Huzen J, Wong LS, van Veldhuisen DJ, et  al. Telomere length loss due vator as part of a health maintenance program. Rejuvenation Res.
to smoking and metabolic traits. J Intern Med. 2014;275:155–163. 2011;14:45–56. doi:10.1089/rej.2010.1085
doi:10.1111/joim.12149 99. Salvador L, Singaravelu G, Harley CB, Flom P, Suram A, Raffaele JM. A
86. Lee M, Martin H, Firpo MA, Demerath EW. Inverse association between natural product telomerase activator lengthens telomeres in humans: a
adiposity and telomere length: the Fels Longitudinal Study. Am J Hum randomized, double blind, and placebo controlled study. Rejuvenation
Biol. 2011;23:100–106. doi:10.1002/ajhb.21109 Res. 2016;19:478–484. doi:10.1089/rej.2015.1793
87. Njajou OT, Cawthon RM, Blackburn EH, et  al. Shorter telomeres are 100. Vidacek NS, Cukusić A, Ivanković M, et al. Abrupt telomere shorten-
associated with obesity and weight gain in the elderly. Int J Obes (Lond). ing in normal human fibroblasts. Exp Gerontol. 2010;45:235–242.
2012;36:1176–1179. doi:10.1038/ijo.2011.196 doi:10.1016/j.exger.2010.01.009

You might also like