Calorie restriction

Calorie restriction, or caloric restriction, or energy restriction, is a dietary regimen that reduces calorie intake without incurring malnutrition or a reduction in essential nutrients. "Reduce" can be defined relative to the subject's previous intake before intentionally restricting calories, or relative to an average person of similar body type. Commonly consumed food components containing calories are carbohydrates, proteins and fat.

In preliminary research, some non-human species on calorie restriction diets without malnutrition may exhibit slowing of the biological aging process, resulting in an increase in both median and maximum lifespan,[1][2] but this effect is not universal.[3] In humans, the long-term health effects of moderate caloric restriction with sufficient nutrients are unknown.[4]

Health effects

Risks of malnutrition

The term "calorie restriction" as used in gerontology refers to dietary regimens that reduce calorie intake without incurring malnutrition.[4] If a restricted diet is not designed to include essential nutrients, malnutrition may result in serious deleterious effects, as shown in the Minnesota Starvation Experiment.[5] This study was conducted during World War II on a group of lean men, who restricted their calorie intake by 45%[6] for 6 months and composed roughly 77%[7] of their diet with carbohydrates.[5] As expected, this malnutrition resulted in many positive metabolic adaptations (e.g. decreased body fat, blood pressure, improved lipid profile, low serum T3 concentration, and decreased resting heart rate and whole-body resting energy expenditure), but also caused a wide range of negative effects, such as anemia, edema, muscle wasting, weakness, neurological deficits, dizziness, irritability, lethargy, and depression.[8]

Musculoskeletal losses

Short-term studies in humans report a loss of muscle mass and strength and reduced bone mineral density.[9] However, whether or not the reduction in bone mineral density actually harms bone health is unclear.[10] In a study in premenopausal women, bone mineral density after weight loss was higher when normalized for body weight;[10] reduced bone mineral density is also observed in humans undergoing long-term calorie restriction with adequate nutrition, but no fractures have been reported[11] and the reduction in bone mineral density was not associated with deleterious changes in bone microarchitecture.[10]

The authors of a 2007 review of the caloric restriction literature warned that "[i]t is possible that even moderate calorie restriction may be harmful in specific patient populations, such as lean persons who have minimal amounts of body fat."[12]

Lower-than-normal body mass index, high mortality

Caloric restriction diets typically lead to reduced body weight, yet reduced weight can come from other causes and is not in itself necessarily healthy. In some studies, low body weight has been associated with increased mortality, particularly in late middle-aged or elderly subjects. Low body weight in the elderly can be caused by pathological conditions associated with aging and predisposing to higher mortality (such as cancer, chronic obstructive pulmonary disorder, or depression) or of the cachexia (wasting syndrome) and sarcopenia (loss of muscle mass, structure, and function).[13] One study linked a body mass index lower than 18 in women with increased mortality from noncancer, non−cardiovascular disease causes.[14] The authors attempted to adjust for confounding factors (cigarette smoking, failure to exclude pre-existing disease); others argued that the adjustments were inadequate.[15]

"epidemiologists from the ACS (American Cancer Society), American Heart Association, Harvard School of Public Health, and other organizations raised specific methodologic questions about the recent Centers for Disease Control and Prevention study and presented analyses of other data sets. The main concern ... is that it did not adequately account for weight loss from serious illnesses such as cancer and heart disease ... [and] failed to account adequately for the effect of smoking on weight ... As a result, the Flegal study underestimated the risks from obesity and overestimated the risks of leanness."[16]

Such epidemiological studies of body weight are not about caloric restriction as used in anti-aging studies; they are not about caloric intake to begin with, as body weight is influenced by many factors other than energy intake, Moreover, "the quality of the diets consumed by the low-body mass index individuals are difficult to assess, and may lack nutrients important to longevity."[4] Typical low-calorie diets rarely provide the high nutrient intakes that are a necessary feature of an anti-aging calorie restriction diet.[17][18][19] As well, "The lower-weight individuals in the studies are not a caloric restriction because their caloric intake reflects their individual ad libitum set-points and not a reduction from that set-point."[4]

Triggering binge eating

In those who have a binge-eating disorder, calorie restriction can precipitate an episode of binge eating, but it does not seem to pose any such risk otherwise.[20]

Young or pregnant

Long-term caloric restriction at a level sufficient for slowing the aging process is generally not recommended in children, adolescents, and young adults (under the age of approximately 21), because this type of diet may interfere with natural physical growth, as has been observed in laboratory animals. In addition, mental development and physical changes to the brain take place in late adolescence and early adulthood that could be negatively affected by severe caloric restriction.[21] Pregnant women and women trying to become pregnant are advised not to practice calorie restriction, because low BMI may result in ovulatory dysfunction (infertility), and underweight mothers are more prone to preterm delivery.[21]

Miscellaneous concerns

It has also been noted that people losing weight on such diets risk developing cold sensitivity, menstrual irregularities, infertility, and hormonal changes.[22]

Mechanisms

Even though there has been research on caloric restriction for over 70 years, the mechanism by which caloric restriction works is not well understood.[2] Some explanations include reduced core body temperature,[23] reduced cellular divisions, lower metabolic rates and hormesis, according to one review of laboratory research.[24]

Temperature

Caloric restriction lowers the core body temperature, a phenomenon believed to be an adaptive response to reduce energy expenditure when nutrients availability is limited. Lowering the temperature may prolong the lifespan of cold blooded animals. Mice, which are warm blooded, have been engineered to have a reduced core body temperature which increased the lifespan independently of calorie restriction.[23]

Hormesis

Some research has pointed toward hormesis as an explanation for the benefits of caloric restriction, representing beneficial actions linked to a low-intensity biological stressor such as reduced calorie intake.[24] As a potential role for caloric restriction, the diet imposes a low-intensity biological stress on the organism, eliciting a defensive response that may help protect it against the disorders of aging.[25] In other words, caloric restriction places the organism in a defensive state so that it can survive adversity, resulting in improved health and longer life.[24] This switch to a defensive state may be controlled by longevity genes (see below).[26]

Evolution

It has been recently argued that during years of famine, it may be evolutionarily desirable for an organism to avoid reproduction and to up-regulate protective and repair enzyme mechanisms to ensure that it is fit for reproduction in future years. This argument seems to be supported by recent work studying hormones. Prolonged severe CR lowers total serum and free testosterone while increasing sex hormone binding globulin concentrations in humans; these effects are independent of adiposity.[27]

Lowering of the concentration of insulin and substances related to insulin, such as insulin-like growth factor 1 and growth hormone, has been shown to up-regulate autophagy, the repair mechanism of the cell.[28] A related hypothesis suggests that caloric restriction works by decreasing insulin levels and thereby up-regulating autophagy,[28][29] but caloric restriction affects many other health indicators, and it is still undecided whether insulin is the main concern.[30] Calorie restriction has been shown to increase DHEA in primates, but it has not been shown to increase DHEA in post-pubescent primates.[31][32] The extent to which these findings apply to humans is still under investigation.

Chromatin and PHA-4

Evidence suggests that the biological effects of caloric restriction are closely related to chromatin function.[33] One study determined that the gene PHA-4 is responsible for the longevity behind calorie restriction in roundworms, "with similar results expected in humans".[34]

Sirtuin-mediated mechanism

Preliminary research indicates that sirtuins are activated by fasting and serve as "energy sensors" during metabolism.[35] Sirtuins, specifically Sir2 (found in yeast) have been implicated in the aging of yeast,[36] and are a class of highly conserved, NAD+-dependent histone deacetylase enzymes.[37] Sir2 homologs have been identified in a wide range of organisms from bacteria to humans.[36][38] Yeast has 3 SIR genes (SIR2, SIR3, and SIR4) that are responsible for silencing mating type loci, telomeres, and rDNA.[39] Although all three genes are required for the silencing of mating type loci and telomeres, only SIR2 has been implicated in the silencing of rDNA.[40] In addition, SIR2 related genes also regulate formation of some specialized survival forms, such as spores in yeast[41] and daher larvae in C. elegans.[42] A study done by Kaeberlein et al. (1999) in yeast found that deletions of Sir2 decreased lifespan, and additional copies increased lifespan.[38]

In many calorie restriction studies, it is believed that Sir2 mediates the longevity effects from calorie restriction for several reasons. First, it was found that in yeast without SIR2, calorie restriction did not impart longevity in yeast;[43] second, in Sir2 mutants an abundance of extra-chromosomal ribosomal DNA circles (that typically limit lifespan) has been observed, and that mitigation of these circles restore regular life span, but still are resistant to calorie restriction-mediated longevity;[44][45] third, that caloric restriction increases the activity of Sir2 in vivo.[46] Although Sir2 has been implicated in calorie restriction-mediated longevity, the method by which Sir2 is regulated under caloric restriction is still debated.

Work on the mechanisms of caloric restriction has given hope to the synthesizing of future drugs to increase the human lifespan by simulating the effects of calorie restriction. In particular, the large number of genes and pathways reported to regulate the actions of caloric restriction in model organisms represent attractive targets for developing drugs that mimic the benefits of CR without its side effects.[47][48][49]

Sir2, or "silent information regulator 2", is a sirtuin, discovered in baker's yeast cells, that is hypothesized to suppress DNA instability.[50] In mammals, Sir2 is known as SIRT1. One proponent of the view that the gene Sir2 may underlie the effect of calorie restriction in mammals by protecting cells from dying under stress.[51] It is suggested that a low-calorie diet that requires less Nicotinamide adenine dinucleotide to metabolize may allow SIRT1 to be more active in its life-extending processes, possibly by a mechanism of SIRT1 releasing fat from storage cells.[52]

Sirtuins

Attempts are being made to develop drugs that act as CR mimetics, and much of that work has focused on a class of proteins called sirtuins.[53] Resveratrol has been reported to activate SIRT1 and extend the lifespan of yeast,[54] nematode worms, fruit flies,[55] vertebrate fish,[56] and mice consuming a high-caloric diet.[57] However, resveratrol does not extend life span in normal mice[58] and the effect of resveratrol on lifespan in nematodes and fruit flies has been disputed.[59]

There are studies that indicate that resveratrol may not function through SIRT1[60][61] but may work through other targets.[62][63] A clinical trial of the resveratrol formulation SRT501 was suspended.[64]

History

Benedict is known for the Harris–Benedict equation used to measure metabolic rate.

Ancient medicine, the province of Hippocrates and Galen after him, taught that the very fat were destined to die suddenly more often than the slender.[65] Around 1000 A.D. Avicenna taught the elderly to eat less than when they were young.[65] Around 1500 because his health was failing due to gluttony, the Venetian nobleman Luigi Cornaro adopted a calorie restricted diet at age 35 and went on to live to be 102 years old. His very successful book Discorsi della vita sobria[66] described his regimen, restricting himself to 350 g (12 oz) of food daily (including bread, egg yolk, meat, and soup) and 410 ml (14 US fl oz) of wine.[67][65]

In 1919 after observing starvation in Central Europe during World War I, Francis Benedict and his colleagues published Human Vitality and Efficiency Under Prolonged Restricted Diet[68] based on their experiment with 10% calorie reduction on male college students at the Carnegie Institution for Science.[69] Reduced rations had turned out to be "not necessarily cataclysmic." Faced with some evidence for what was unknown at the time but today is called metabolic adaptation, Benedict wanted to find the science behind what appeared to be an adjustment in metabolic rate when food intake was below energy expenditure.[69]

Hoping to learn how to refeed the people who had starved during World War II, between 1944 and 1945, 36 healthy conscientious objectors participated in the Minnesota Starvation Experiment, published in 1950 as The Biology of Human Starvation by lead investigator Ancel Keys and colleagues.[70][8] Because the men were receiving 40% CR and subject to malnutrition this study was not one of calorie restriction per se.[nb 1]

EA Vallejo published a study of approximately 35% CR in the Spanish language in 1957, testing CR without malnutrition in nonobese elderly persons.[72][73] About 30% CR for six months was achieved accidentally in the Biosphere 2 experiment during the 1990s.[72]

Research

Humans

Studies have been conducted to examine the effects of calorie restriction with adequate intake of nutrients in humans; however, long-term effects are unknown.[4] One objection to calorie restriction in humans is a claim that the physiological mechanisms determining longevity are complex, and that the effect would be small to negligible.[74] Effects of calorie restriction in humans over multiple years or decades may be small in comparison to conventional medical and public health interventions, but have not yet been clearly determined.[4][75]

Biomarkers for cardiovascular risk

A review of the effects of calorie restriction on the aging heart and vasculature concluded that "Data from animal and human studies indicate that [beyond the effects of "implementation of healthier diets and regular exercise"], more drastic interventions, i.e., calorie restriction with adequate nutrition (CRAN), may have additional beneficial effects on several metabolic and molecular factors that are modulating cardiovascular aging itself (e.g., cardiac and arterial stiffness and heart rate variability)."[76] Studies of long-term practitioners of rigorous calorie restriction show that their risk factors for atherosclerosis are substantially improved in a manner consistent with experimental studies in rodent models of atherosclerosis and nonhuman primates.[76] Risk factors such as c-reactive protein; serum triglycerides, low-density lipoprotein, high-density lipoprotein; blood pressure; and fasting blood sugar, are substantially more favorable than persons consuming usual Western diets and comparable or better than long-term endurance exercisers.[76] Similar effects were also seen during a "natural experiment" in Biosphere 2,[76] and effects on blood pressure, cholesterol level, and resting heart rate were seen in subjects in the “Minnesota Starvation Experiment” during World War II.[8] Cardiac "diastolic function was better in subjects who practiced strict calorie restriction for 3–15 years than that in healthy age- and sex-matched control subjects ... calorie restriction subjects had less ventricular stiffness and less viscous loss of diastolic recoil, both of which would be consistent with less myocardial fibrosis.[76] "These effects, in combination with other benefits of calorie restriction, such as protection against obesity, diabetes, hypertension, and cancer, suggest that CR may have a major beneficial effect on health span, life span, and quality of life in humans."[76]

Primates

In a 2017 collaborative report on rhesus monkeys by scientists of the US National Institute on Aging and the University of Wisconsin, caloric restriction in the presence of adequate nutrition was effective in delaying the effects of aging.[31][77] Older age of onset, female sex, lower body weight and fat mass, reduced food intake, diet quality, and lower fasting blood glucose levels were factors associated with fewer disorders of aging and with improved survival rates.[31][77] Specifically, reduced food intake was beneficial in adult and older primates, but not in younger monkeys.[31] The study indicated that caloric restriction provided health benefits with fewer age-related disorders in elderly monkeys and, because rhesus monkeys are genetically similar to humans, the benefits and mechanisms of caloric restriction may apply to human health during aging.[31]

Rodents

It has been known since the 1930s that reducing the number of calories fed to laboratory rodents increases their life spans. The life extension varies for each species, but on average there was a 30–40% increase in life span in both mice and rats.[30] In late adulthood, acute CR partially or completely reverses age-related alterations of liver, brain and heart proteins, and mice placed on CR at 19 months of age show an increases in life span.[78]

Yeast

Fungi models are very easy to manipulate, and many crucial steps toward the understanding of aging have been made with them. Many studies were undertaken on budding yeast and fission yeast to analyze the cellular mechanisms behind increased longevity due to calorie restriction. First, calorie restriction is often called dietary restriction because the same effects on life span can be achieved by only changing the nutrient quality without changing the number of calories. Data from Guarente and others showed that genetic manipulations in nutrient-signaling pathways could mimic the effects of dietary restriction. In some cases, dietary restriction requires mitochondrial respiration to increase longevity (chronological aging), and in some other cases not (replicative aging). Nutrient sensing in yeast controls stress defense, mitochondrial functions, Sir2, and others. These functions are all known to regulate aging. Genes involved in these mechanisms are TOR, PKA, SCH9, MSN2/4, RIM15, and SIR2.[79][80][81][82][83] Importantly, yeast responses to CR can be modulated by genetic background. Therefore, while some strains respond to calorie restriction with increased lifespan, in others calorie restriction shortens it [84]

Activity levels

Calorie restriction preserves muscle tissue in nonhuman primates[85][86] and rodents.[87][88] Mechanisms include reduced muscle cell apoptosis and inflammation;[87] protection against[88] or adaptation to[85] age-related mitochondrial abnormalities; and preserved muscle stem cell function.[89] Muscle tissue grows when stimulated, so it has been suggested that the calorie-restricted test animals exercised more than their companions on higher calories, perhaps because animals enter a foraging state during calorie restriction. However, studies show that overall activity levels are no higher in calorie restriction than ad libitum animals in youth.[90] Laboratory rodents placed on a calorie restriction diet tend to exhibit increased activity levels (particularly when provided with exercise equipment) at feeding time. Monkeys undergoing calorie restriction also appear more restless immediately before and after meals.[91]

Behavior

Observations in some accounts of animals undergoing calorie restriction have noted an increase in stereotyped behaviors.[91] For example, monkeys on calorie restriction have demonstrated an increase in licking, sucking, and rocking behavior.[92] A calorie restriction regimen may also lead to increased aggressive behavior in animals.[91]

Obese controls

It has sometimes been suggested that the lives of calorie-restricted animals are only extended relative to control animals whose lives are artificially shortened by weight-gain from unnatural ad libitum feeding in the laboratory.[93] However, studies designed to test this hypothesis suggest that reduced fat mass is not a major contributor to the longevity effects of calorie restriction.[4]

See also

Notes

  1. Vitousek et al. write in 2004, "The relevance of the classic Minnesota study of human CR (Keys et al., 1950) is specifically disavowed (e.g. Heilbronn & Ravussin, 2003; Walford, 2000; Weindruch & Walford, 1988), on the grounds that substandard nutrition must have been responsible for the depression, irritability, social withdrawal, asexuality, fatigue and food preoccupation that subjects experienced."[71]

References

  1. Omodei, D; Fontana, L (Jun 6, 2011). "Calorie restriction and prevention of age-associated chronic disease". FEBS Lett. 585 (11): 1537–42. doi:10.1016/j.febslet.2011.03.015. PMC 3439843. PMID 21402069. Retrieved 28 November 2015.
  2. 1 2 Anderson, R. M.; Shanmuganayagam, D.; Weindruch, R. (2009). "Caloric Restriction and Aging: Studies in Mice and Monkeys". Toxicologic Pathology. 37 (1): 47–51. doi:10.1177/0192623308329476. PMC 3734859. PMID 19075044.
  3. "Can We Prevent Aging?". National Institute on Aging, US National Institutes of Health, Bethesda, MD. 29 July 2016. Archived from the original on 26 September 2016.
  4. 1 2 3 4 5 6 7 Spindler, Stephen R. (2010). "Biological Effects of Calorie Restriction: Implications for Modification of Human Aging". The Future of Aging. pp. 367–438. doi:10.1007/978-90-481-3999-6_12. ISBN 978-90-481-3998-9.
  5. 1 2 Keys A, Brozek J, Henschels A & Mickelsen O & Taylor H. The Biology of Human Starvation, 1950. University of Minnesota Press, Minneapolis
  6. Keys A 1950, p. 114.
  7. Keys A 1950, pp. 1213–1214.
  8. 1 2 3 Kalm, Leah M.; Semba, Richard D. (June 1, 2005). "They Starved So That Others Be Better Fed: Remembering Ancel Keys and the Minnesota Experiment". J. Nutr. 135 (6): 1347–1352. doi:10.1093/jn/135.6.1347. PMID 15930436.
  9. Morley, John E; Chahla, Elie; Alkaade, Saad (2010). "Antiaging, longevity and calorie restriction". Current Opinion in Clinical Nutrition and Metabolic Care. 13 (1): 40–5. doi:10.1097/MCO.0b013e3283331384. PMID 19851100.
  10. 1 2 3 Gower BA, Casazza K (October–December 2013). "Divergent Effects of Obesity on Bone Health". Journal of Clinical Densitometry. 16 (4): 450–454. doi:10.1016/j.jocd.2013.08.010. PMC 5321047. PMID 24063845.
  11. Bonewald LF, Kiel DP, Clemens TL, Esser K, Orwoll ES, O'Keefe RJ, Fielding RA (September 2013). "Forum on bone and skeletal muscle interactions: Summary of the proceedings of an ASBMR workshop". Journal of Bone and Mineral Research. 28 (9): 1857–1865. doi:10.1002/jbmr.1980. PMC 3749267. PMID 23671010.
  12. Fontana, L.; Klein, S. (2007). "Aging, Adiposity, and Calorie Restriction". JAMA. 297 (9): 986–94. doi:10.1001/jama.297.9.986. PMID 17341713.
  13. Hu, Frank (2008). "Interpreting Epidemiologic Evidence and Causal Inference in Obesity Research". In Frank B. Hu. Obesity Epidemiology. New York, NY: Oxford University Press. pp. 38–52. ISBN 978-0-19-531291-1. Retrieved 2011-02-20.
  14. Flegal, K. M.; Graubard, B. I.; Williamson, D. F.; Gail, M. H. (2007). "Cause-Specific Excess Deaths Associated With Underweight, Overweight, and Obesity". JAMA. 298 (17): 2028–37. doi:10.1001/jama.298.17.2028. PMID 17986696.
  15. Holzman, Donald (2005-05-27). "Panel Suggests Methodology Flawed of Recent CDC Obesity Study". Medscape Medical News. Retrieved 2011-02-21.
  16. "Researchers weigh risks due to overweight". CA: A Cancer Journal for Clinicians. 55 (5): 268–9. 2005. doi:10.3322/canjclin.55.5.268.
  17. St. Jeor, S. T.; Howard, B. V.; Prewitt, T. E.; Bovee, V.; Bazzarre, T.; Eckel, R. H.; Nutrition Committee Of The Council On Nutrition (2001). "Dietary Protein and Weight Reduction: A Statement for Healthcare Professionals From the Nutrition Committee of the Council on Nutrition, Physical Activity, and Metabolism of the American Heart Association". Circulation. 104 (15): 1869–74. doi:10.1161/hc4001.096152. PMID 11591629.
  18. De Souza, RJ; Swain, JF; Appel, LJ; Sacks, FM (2008). "Alternatives for macronutrient intake and chronic disease: a comparison of the OmniHeart diets with popular diets and with dietary recommendations". The American Journal of Clinical Nutrition. 88 (1): 1–11. doi:10.1093/ajcn/88.1.1. PMC 2674146. PMID 18614716.
  19. Ma, Y; Pagoto, S; Griffith, J; Merriam, P; Ockene, I; Hafner, A; Olendzki, B (2007). "A Dietary Quality Comparison of Popular Weight-Loss Plans". Journal of the American Dietetic Association. 107 (10): 1786–91. doi:10.1016/j.jada.2007.07.013. PMC 2040023. PMID 17904938.
  20. Binge-Eating Disorder: Clinical Foundations and Treatment (1 ed.). The Guilford Press. 2007. p. 15. ISBN 978-1-59385-594-9. It can be concluded that caloric restriction does not appear to be associated with the development of binge eating in individuals who have never reported problems with binge eating.
  21. 1 2 "Risks". Archived from the original on 2010-09-19. Retrieved 2010-07-28.
  22. Marzetti, E.; Wohlgemuth, S. E.; Anton, S. D.; Bernabei, R; Carter, C. S.; Leeuwenburgh, C (2012-10-19). "Cellular mechanisms of cardioprotection by calorie restriction: state of the science and future perspectives". Clin. Geriatr. Med. 25 (4): 715–32, ix. doi:10.1016/j.cger.2009.07.002. PMC 2786899. PMID 19944269.
  23. 1 2 Conti, B; Sanchez-Alavez, M; Winsky-Sommerer, R; Morale, MC; Lucero, J; Brownell, S; Fabre, V; Huitron-Resendiz, S; Henriksen, S; Zorrilla, EP; de Lecea, L; Bartfai, T (3 November 2006). "Transgenic mice with a reduced core body temperature have an increased life span". Science. 314 (5800): 825–8. Bibcode:2006Sci...314..825C. doi:10.1126/science.1132191. PMID 17082459.
  24. 1 2 3 Nikolai, Sibylle; Pallauf, Kathrin; Huebbe, Patricia; Rimbach, Gerald (22 September 2015). "Energy restriction and potential energy restriction mimetics". Nutrition Research Reviews. 28 (2): 1–21. doi:10.1017/S0954422415000062. PMID 26391585. Retrieved 8 November 2015.
  25. Martins, I; Galluzzi, L; Kroemer, G (2011). "Hormesis, cell death and aging". Aging. 3 (9): 821–8. doi:10.18632/aging.100380. PMC 3227447. PMID 21931183.
  26. Mattson, M (2008). "Dietary Factors, Hormesis and Health". Ageing Research Reviews. 7 (1): 43–8. doi:10.1016/j.arr.2007.08.004. PMC 2253665. PMID 17913594.
  27. Cangemi, Roberto; Friedmann, Alberto J.; Holloszy, John O.; Fontana, Luigi (2010). "Long-term effects of calorie restriction on serum sex-hormone concentrations in men". Aging Cell. 9 (2): 236–42. doi:10.1111/j.1474-9726.2010.00553.x. PMC 3569090. PMID 20096034.
  28. 1 2 Bergamini, E; Cavallini, G; Donati, A; Gori, Z (2003). "The anti-ageing effects of caloric restriction may involve stimulation of macroautophagy and lysosomal degradation, and can be intensified pharmacologically". Biomedecine & Pharmacotherapy. 57 (5–6): 203–8. doi:10.1016/S0753-3322(03)00048-9.
  29. Cuervo, Ana Maria; Bergamini, Ettore; Brunk, Ulf T; Dröge, Wulf; Ffrench, Martine; Terman, Alexei (2005). "Autophagy and Aging: the Importance of Maintaining "Clean" Cells". Autophagy. 1 (3): 131–40. doi:10.4161/auto.1.3.2017. PMID 16874025.
  30. 1 2 Mattson MP (2005). "Energy intake, meal frequency, and health: a neurobiological perspective". Annu. Rev. Nutr. (Review). 25: 237–60. doi:10.1146/annurev.nutr.25.050304.092526. PMID 16011467.
  31. 1 2 3 4 5 Mattison, Julie A; Colman, Ricki J; Beasley, T Mark; Allison, David B; Kemnitz, Joseph W; Roth, George S; Ingram, Donald K; Weindruch, Richard; De Cabo, Rafael; Anderson, Rozalyn M (2017). "Caloric restriction improves health and survival of rhesus monkeys". Nature Communications. 8: 14063. Bibcode:2017NatCo...814063M. doi:10.1038/ncomms14063. PMC 5247583. PMID 28094793.
  32. Urbanski, H F.; Downs, J L; Garyfallou, V T; Mattison, J A; Lane, M A; Roth, G S; Ingram, D K (2004). "Effect of Caloric Restriction on the 24-Hour Plasma DHEAS and Cortisol Profiles of Young and Old Male Rhesus Macaques". Annals of the New York Academy of Sciences. 1019: 443–7. doi:10.1196/annals.1297.081. PMID 15247063.
  33. Vaquero, A.; Reinberg, D. (2009). "Calorie restriction and the exercise of chromatin". Genes & Development. 23 (16): 1849–69. doi:10.1101/gad.1807009. PMC 2725938. PMID 19608767.
  34. "The gene for longevity, if you're a worm". ABC News. 2007. Retrieved 2007-05-03.
  35. Chang, H. C; Guarente, L (2013). "SIRT1 and other sirtuins in Metabolism". Trends in Endocrinology and Metabolism. 25 (3): 138–145. doi:10.1016/j.tem.2013.12.001. hdl:1721.1/104067. PMC 3943707. PMID 24388149.
  36. 1 2 Guarente, L. (2007). "Sirtuins in aging and disease". Cold Spring Harbor Symposia on Quantitative Biology. 72: 483–488. doi:10.1101/sqb.2007.72.024. ISSN 0091-7451. PMID 18419308.
  37. Lin, Su-Ju; Ford, Ethan; Haigis, Marcia; Liszt, Greg; Guarente, Leonard (2004-01-01). "Calorie restriction extends yeast life span by lowering the level of NADH". Genes & Development. 18 (1): 12–16. doi:10.1101/gad.1164804. ISSN 0890-9369. PMC 314267. PMID 14724176.
  38. 1 2 Kaeberlein M.; McVey M.; Guarente L. (1999). "The SIR2/3/4 complex and SIR2 alone promote longevity in Saccharomyces cerevisiae by two different mechanisms. '". Genes & Development. 13 (19): 2570–2580. doi:10.1101/gad.13.19.2570. PMC 317077. PMID 10521401.
  39. Guarente, Leonard (2000). "Sir2 links chromatin silencing, metabolism, and aging". Genes & Development. 14 (9): 1021–1026. doi:10.1101/gad.14.9.1021 (inactive 2018-08-27). ISSN 0890-9369. PMID 10809662.
  40. Smith, J. S.; Boeke, J. D. (1997-01-15). "An unusual form of transcriptional silencing in yeast ribosomal DNA". Genes & Development. 11 (2): 241–254. doi:10.1101/gad.11.2.241. ISSN 0890-9369. PMID 9009206.
  41. Margolskee, Jeanne P. (1988-03-01). "The sporulation capable (sca) mutation of Saccharomyces cerevisiae is an allele of the SIR2 gene". Molecular and General Genetics MGG. 211 (3): 430–434. doi:10.1007/BF00425696. ISSN 0026-8925.
  42. Tissenbaum, Heidi A.; Guarente, Leonard (March 8, 2001). "Increased dosage of a sir-2 gene extends lifespan in Caenorhabditis elegans". Nature. 410 (6825): 227–230. doi:10.1038/35065638. ISSN 0028-0836. PMID 11242085.
  43. Lin, S. J.; Defossez, P. A.; Guarente, L. (Sep 22, 2000). "Requirement of NAD and SIR2 for life-span extension by calorie restriction in Saccharomyces cerevisiae". Science. 289 (5487): 2126–2128. Bibcode:2000Sci...289.2126L. doi:10.1126/science.289.5487.2126. ISSN 0036-8075. PMID 11000115.
  44. Sinclair, D. A.; Guarente, L. (Dec 26, 1997). "Extrachromosomal rDNA circles--a cause of aging in yeast". Cell. 91 (7): 1033–1042. doi:10.1016/S0092-8674(00)80493-6. ISSN 0092-8674. PMID 9428525.
  45. Guarente L, Picard F (February 2005). "Calorie Restriction— the SIR2 Connection". Cell. 120 (4): 473–482. doi:10.1016/j.cell.2005.01.029. PMID 15734680. Retrieved 2015-05-30.
  46. Lin, Su-Ju; Kaeberlein, Matt; Andalis, Alex A.; Sturtz, Lori A.; Defossez, Pierre-Antoine; Culotta, Valeria C.; Fink, Gerald R.; Guarente, Leonard (July 18, 2002). "Calorie restriction extends Saccharomyces cerevisiae lifespan by increasing respiration". Nature. 418 (6895): 344–348. Bibcode:2002Natur.418..344L. doi:10.1038/nature00829. ISSN 0028-0836. PMID 12124627.
  47. Minor, RK; Allard, JS; Younts, CM; Ward, TM; de Cabo, R (July 2010). "Dietary interventions to extend life span and health span based on calorie restriction". The Journals of Gerontology. Series A, Biological Sciences and Medical Sciences. 65 (7): 695–703. doi:10.1093/gerona/glq042. PMC 2884086. PMID 20371545.
  48. Contestabile, A (2009). "Benefits of caloric restriction on brain aging and related pathological States: understanding mechanisms to devise novel therapies". Current Medicinal Chemistry. 16 (3): 350–61. doi:10.2174/092986709787002637. PMID 19149582.
  49. de Magalhães JP, Wuttke D, Wood SH, Plank M, Vora C (2012). "Genome-environment interactions that modulate aging: powerful targets for drug discovery". Pharmacol Rev. 64 (1): 88–101. doi:10.1124/pr.110.004499. PMC 3250080. PMID 22090473.
  50. Sinclair, David A; Guarente, Leonard (1997). "Extrachromosomal rDNA Circles— A Cause of Aging in Yeast". Cell. 91 (7): 1033–1042. doi:10.1016/S0092-8674(00)80493-6. PMID 9428525.
  51. Cohen, H. Y.; Miller, C; Bitterman, KJ; Wall, NR; Hekking, B; Kessler, B; Howitz, KT; Gorospe, M; et al. (2004). "Calorie Restriction Promotes Mammalian Cell Survival by Inducing the SIRT1 Deacetylase". Science. 305 (5682): 390–2. Bibcode:2004Sci...305..390C. doi:10.1126/science.1099196. PMID 15205477.
  52. Picard, Frédéric; Kurtev, Martin; Chung, Namjin; Topark-Ngarm, Acharawan; Senawong, Thanaset; MacHado De Oliveira, Rita; Leid, Mark; McBurney, Michael W.; Guarente, Leonard (2004). "Sirt1 promotes fat mobilization in white adipocytes by repressing PPAR-γ". Nature. 429 (6993): 771–6. Bibcode:2004Natur.429..771P. doi:10.1038/nature02583. PMC 2820247. PMID 15175761.
  53. Corton, J. C.; Apte, U; Anderson, SP; Limaye, P; Yoon, L; Latendresse, J; Dunn, C; Everitt, JI; et al. (2004). "Mimetics of Caloric Restriction Include Agonists of Lipid-activated Nuclear Receptors". Journal of Biological Chemistry. 279 (44): 46204–12. doi:10.1074/jbc.M406739200. PMID 15302862.
  54. Howitz, Konrad T.; Bitterman, Kevin J.; Cohen, Haim Y.; Lamming, Dudley W.; Lavu, Siva; Wood, Jason G.; Zipkin, Robert E.; Chung, Phuong; et al. (2003). "Small molecule activators of sirtuins extend Saccharomyces cerevisiae lifespan". Nature. 425 (6954): 191–6. Bibcode:2003Natur.425..191H. doi:10.1038/nature01960. PMID 12939617.
  55. Wood, Jason G.; Rogina, Blanka; Lavu, Siva; Howitz, Konrad; Helfand, Stephen L.; Tatar, Marc; Sinclair, David (2004). "Sirtuin activators mimic caloric restriction and delay ageing in metazoans". Nature. 430 (7000): 686–9. Bibcode:2004Natur.430..686W. doi:10.1038/nature02789. PMID 15254550.
  56. Valenzano, Dario R.; Terzibasi, Eva; Genade, Tyrone; Cattaneo, Antonino; Domenici, Luciano; Cellerino, Alessandro (2006). "Resveratrol Prolongs Lifespan and Retards the Onset of Age-Related Markers in a Short-Lived Vertebrate". Current Biology. 16 (3): 296–300. doi:10.1016/j.cub.2005.12.038. PMID 16461283.
  57. Baur, Joseph A.; Pearson, Kevin J.; Price, Nathan L.; Jamieson, Hamish A.; Lerin, Carles; Kalra, Avash; Prabhu, Vinayakumar V.; Allard, Joanne S.; et al. (2006). "Resveratrol improves health and survival of mice on a high-calorie diet". Nature. 444 (7117): 337–42. Bibcode:2006Natur.444..337B. doi:10.1038/nature05354. PMC 4990206. PMID 17086191.
  58. Pearson, Kevin J.; Baur, Joseph A.; Lewis, Kaitlyn N.; Peshkin, Leonid; Price, Nathan L.; Labinskyy, Nazar; Swindell, William R.; Kamara, Davida; et al. (2008). "Resveratrol delays age-related deterioration and mimics transcriptional aspects of dietary restriction without extending lifespan". Cell Metabolism. 8 (2): 157–68. doi:10.1016/j.cmet.2008.06.011. PMC 2538685. PMID 18599363.
  59. Bass, T; Weinkove, D; Houthoofd, K; Gems, D; Partridge, L (2007). "Effects of resveratrol on lifespan in Drosophila melanogaster and Caenorhabditis elegans". Mechanisms of Ageing and Development. 128 (10): 546–52. doi:10.1016/j.mad.2007.07.007. PMID 17875315.
  60. Pacholec, M.; Bleasdale, J. E.; Chrunyk, B.; Cunningham, D.; Flynn, D.; Garofalo, R. S.; Griffith, D.; Griffor, M.; et al. (2010). "SRT1720, SRT2183, SRT1460, and Resveratrol Are Not Direct Activators of SIRT1". Journal of Biological Chemistry. 285 (11): 8340–51. doi:10.1074/jbc.M109.088682. PMC 2832984. PMID 20061378.
  61. Zarse, K.; Schmeisser, S.; Birringer, M.; Falk, E.; Schmoll, D.; Ristow, M. (2010). "Differential Effects of Resveratrol and SRT1720 on Lifespan of AdultCaenorhabditis elegans". Hormone and Metabolic Research. 42 (12): 837–9. doi:10.1055/s-0030-1265225. PMID 20925017.
  62. Kaeberlein, Matt; Kirkland, Kathryn T.; Fields, Stanley; Kennedy, Brian K. (2004). "Sir2-Independent Life Span Extension by Calorie Restriction in Yeast". PLoS Biology. 2 (9): e296. doi:10.1371/journal.pbio.0020296. PMC 514491. PMID 15328540.
  63. Kaeberlein, M; Powersiii, R (2007). "Sir2 and calorie restriction in yeast: A skeptical perspective". Ageing Research Reviews. 6 (2): 128–40. doi:10.1016/j.arr.2007.04.001. PMID 17512264.
  64. Suspended Resveratrol Clinical Trial: More Details Emerge(May 6, 2010)
  65. 1 2 3 Schäfer, Daniel (Mar–Apr 2005). "Aging, Longevity, and Diet: Historical Remarks on Calorie Intake Reduction". Gerontology. 51 (2): 126–30. doi:10.1159/000082198. PMID 15711080.
  66. Cornaro, Luigi (1550). Discourses and Letters on the Sober and Temperate Life. Internet Archive. Retrieved November 13, 2017.
  67. Everitt, Heilbronn & Le Couteur 2010, p. 15.
  68. Benedict, Francis G.; Miles, Walter R.; Roth, Paul; Smith, H. Monmouth (1919). Human Vitality and Efficiency Under Prolonged Restricted Diet. Carnegie Institution of Washington. Retrieved November 13, 2017.
  69. 1 2 Keys et al. 1950, pp. 35–36.
  70. Everitt, Heilbronn & Le Couteur 2010, p. 17.
  71. Vitousek, Kelly M.; Manke1, Frederic P.; Gray, Jennifer A.; Vitousek, Maren N. (2004). "Caloric Restriction for Longevity: II—The Systematic Neglect of Behavioural and Psychological Outcomes in Animal Research". Eur. Eat. Disorders Rev. 12 (6): 338–360. doi:10.1002/erv.604.
  72. 1 2 Everitt, Heilbronn & Le Couteur 2010, p. 18.
  73. Vallejo, EA (January 11, 1957). "Hunger diet on alternate days in the nutrition of the aged". Prensa Med Argent (in Spanish). 44 (2): 119–20. PMID 13453175.
  74. Phelan, J; Rose, M (2005). "Why dietary restriction substantially increases longevity in animal models but won't in humans". Ageing Research Reviews. 4 (3): 339–50. doi:10.1016/j.arr.2005.06.001. PMID 16046282.
  75. Everitt, A. V; Le Couteur, D. G (2007). "Life extension by calorie restriction in humans". Annals of the New York Academy of Sciences. 1114 (1): 428–33. Bibcode:2007NYASA1114..428E. doi:10.1196/annals.1396.005. PMID 17717102.
  76. 1 2 3 4 5 6 Weiss, EP; Fontana, L (Oct 2011). "Caloric restriction: powerful protection for the aging heart and vasculature". Am J Physiol Heart Circ Physiol. 301 (4): H1205–19. doi:10.1152/ajpheart.00685.2011. PMC 3197347. PMID 21841020. Retrieved 27 August 2014.
  77. 1 2 "Calorie restriction lets monkeys live long and prosper". ScienceDirect. 17 January 2017. Retrieved 15 February 2017.
  78. Spindler, S (2005). "Rapid and reversible induction of the longevity, anticancer and genomic effects of caloric restriction". Mechanisms of Ageing and Development. 126 (9): 960–6. doi:10.1016/j.mad.2005.03.016. PMID 15927235.
  79. Kaeberlein, Matt; Burtner, Christopher R.; Kennedy, Brian K. (2007). "Recent Developments in Yeast Aging". PLoS Genetics. 3 (5): e84. doi:10.1371/journal.pgen.0030084. PMC 1877880. PMID 17530929.
  80. Dilova, I.; Easlon, E.; Lin, S. -J. (2007). "Calorie restriction and the nutrient sensing signaling pathways". Cellular and Molecular Life Sciences. 64 (6): 752–67. doi:10.1007/s00018-007-6381-y. PMID 17260088.
  81. Chen, D; Guarente, L (2007). "SIR2: a potential target for calorie restriction mimetics". Trends in Molecular Medicine. 13 (2): 64–71. doi:10.1016/j.molmed.2006.12.004. PMID 17207661.
  82. Piper, Peter W. (2006). "Long-lived yeast as a model for ageing research". Yeast. 23 (3): 215–26. doi:10.1002/yea.1354. PMID 16498698.
  83. Longo, V (2009). "Linking sirtuins, IGF-I signaling, and starvation". Experimental Gerontology. 44 (1–2): 70–4. doi:10.1016/j.exger.2008.06.005. PMID 18638538.
  84. Schleit J.; Wasko B.M.; Kaeberlein M. (2012). "Yeast as a model to understand the interaction between genotype and the response to calorie restriction". FEBS Lett. 586 (18): 2868–2873. doi:10.1016/j.febslet.2012.07.038. PMC 4016815. PMID 22828279.
  85. 1 2 McKiernan, SH; Colman, RJ; Aiken, E; Evans, TD; Beasley, TM; Aiken, JM; Weindruch, R; Anderson, RM (Mar 2012). "Cellular adaptation contributes to calorie restriction-induced preservation of skeletal muscle in aged rhesus monkeys". Exp Gerontol. 47 (3): 229–36. doi:10.1016/j.exger.2011.12.009. PMC 3321729. PMID 22226624.
  86. Colman, RJ; Beasley, TM; Allison, DB; Weindruch, R (2008). "Attenuation of Sarcopenia by Dietary Restriction in Rhesus Monkeys". The Journals of Gerontology. Series A, Biological Sciences and Medical Sciences. 63 (6): 556–9. doi:10.1093/gerona/63.6.556. PMC 2812805. PMID 18559628.
  87. 1 2 Dirks Naylor, AJ; Leeuwenburgh, C (Jan 2008). "Sarcopenia: the role of apoptosis and modulation by caloric restriction". Exerc Sport Sci Rev. 36 (1): 19–24. doi:10.1097/jes.0b013e31815ddd9d. PMID 18156949.
  88. 1 2 Bua, E; McKiernan, SH; Aiken, JM (Mar 2004). "Calorie restriction limits the generation but not the progression of mitochondrial abnormalities in aging skeletal muscle". FASEB J. 18 (3): 582–4. doi:10.1096/fj.03-0668fje. PMID 14734641.
  89. Cerletti, M; Jang, YC; Finley3=LW; Haigis 4=MC; Wagers 5=AJ (May 4, 2012). "Short-term calorie restriction enhances skeletal muscle stem cell function". Cell Stem Cell. 10 (5): 515–9. doi:10.1016/j.stem.2012.04.002. PMC 3561899. PMID 22560075.
  90. Faulks, SC; Turner, N; Else, PL; Hulbert, AJ (Aug 2006). "Calorie restriction in mice: effects on body composition, daily activity, metabolic rate, mitochondrial reactive oxygen species production, and membrane fatty acid composition". J Gerontol a Biol Sci Med Sci. 61 (8): 781–94. doi:10.1093/gerona/61.8.781. PMID 16912094.
  91. 1 2 3 Vitousek K. M.; Manke F. P.; Gray J. A.; Vitousek M. N. (2004). "Caloric Restriction for Longevity: II--The Systematic Neglect of Behavioural and Psychological Outcomes in Animal Research". European Eating Disorders Review. 12 (6): 338–360. doi:10.1002/erv.604.
  92. Weed J. L.; Lane M. A.; Roth G. S.; Speer D. L.; Ingram D. K. (1997). "Activity measures in rhesus monkeys on long-term calorie restriction". Physiology & Behavior. 62: 97–103. doi:10.1016/s0031-9384(97)00147-9.
  93. Sohala Rajindar S.; Forster Michael J. (2014). "Caloric restriction and the aging process: a critique". Free Radical Biology and Medicine. 73: 366–382. doi:10.1016/j.freeradbiomed.2014.05.015. PMC 4111977. PMID 24941891.

Bibliography

  • Everitt, Arthur V.; Heilbronn, Leonie K.; Le Couteur, David G. (2010). "Food Intake, Life Style, Aging and Human Longevity". In Everitt, Arthur V; Rattan, Suresh IS; Le Couteur, David G; de Cabo, Rafael. Calorie Restriction, Aging and Longevity. New York: Springer. ISBN 978-90-481-8555-9.
  • Keys, Ancel; Brozek, Josef; Henschel, Austin; Mickelsen, Olaf; Taylor, Henry Longstreet (1950). The Biology of Human Starvation. I. University of Minnesota Press. ISBN 9780816672349.


This article is issued from Wikipedia. The text is licensed under Creative Commons - Attribution - Sharealike. Additional terms may apply for the media files.