Calorie restriction

from Wikipedia, the free encyclopedia

Calorie restriction , also called caloric restriction or calorie restriction , is the designation for a diet with a 10 to 50 percent reduction in energy from food (see: physiological calorific value ) via diet compared to diet ad libitum ("at will") . The aim is to achieve a longer life expectancy or at least delayed aging or a health-promoting effect without causing malnutrition . In the English-language specialist literature, the terms caloric restriction , calorie restriction or dietary restriction are used for calorie restriction.

In this way, a health-promoting and life-prolonging effect could be demonstrated in a number of model organisms . For some species or cultivars , however, no increase in life expectancy was found. The increase in life expectancy of some rodent strains by up to 50% depends , among other things, on genome and gender.

Reliable data on the life-prolonging effect of calorie restriction in humans are not available. While the effect is largely recognized in the animal model, the transferability to humans is controversial. The mechanisms of action are hypothetical and unexplained.

history

An engraving supposed to show the calorie restricted Luigi Cornaro.

In his autobiographical treatise Discorsi della vita sobria ( On the moderate life ), which he wrote at the age of 83, the Venetian Luigi Cornaro (1467–1565) attributed his old age and health to the fact that he was on a strict diet (calorie restriction ) followed. Cornaro ate just as much as was necessary to survive. He chose the food not according to his taste, but according to health aspects.

Francis Peyton Rous found in animal experiments in 1914 that reducing the food supply in rats could reduce the incidence of tumors and prolong the life of the animals. In the 1930s, after similar animal experiments , Clive McCay and colleagues hypothesized that the prolongation of life through calorie restriction in rats was due to a reduced growth rate. They had no molecular biological explanation for this.

The life-prolonging effect of calorie restriction was described in 1934 by the US biochemist Clive Maine McCay (1898-1967) and Mary F. Crowell (both Cornell University ). McCay and Crowell found in their experiments with rats that the permanent reduction in the amount of food - while ensuring adequate nutrition - significantly increases the life expectancy of the animals. Some of the test animals received 33 percent less food than the comparison group, which was fed ad libitum (" ad libitum "), which increased the life expectancy of the first-named animals by almost 50 percent.

Morris Ross carried out further experiments with rats in the 1960s and 1970s and was able to support McCay's hypothesis.

In his test series, Ross was able to determine that the composition of the food - compared to the calorie restriction - has a significantly lower effect on extending life. Richard Weindruch and Roy Walford showed in experiments with mice in the 1980s that calorie restriction, even if it is only initiated in adult animals , can prolong life. They concluded that delayed ripening is not the main cause of life extension, but that calorie restriction apparently positively mediates the process of aging itself. In the second half of the 20th century, the Soviet doctor Galina Schatalowa experimented with calorie restriction in combination with raw food .

During the 1980s and early 1990s, researchers began a serious discussion of human participation studies of calorie restriction studies . As a result, studies were carried out with rhesus monkeys ( Macaca mulatta ), among others , which showed that the positive results from other animal species can also be transferred to primates.

Intensive research into the effects of calorie restriction on the nematode Caenorhabditis elegans began in the 1990s . In 1998, Bernard Lakowski and Siegfried Hekimi published that mutations that alter the worm's throat and restrict food intake increase life expectancy by 50%.

In the Biosphere 2 experiment, the study participants were supposed to feed themselves completely from the inner cycle of materials in a closed building complex. Roy Walford participated in the experiment from 1991 to 1993. When the study participants discovered that they had overestimated their ability to produce food, Walford suggested a calorie restriction. During seven eighths of their two-year stay, the eight test subjects consumed a low-calorie (7300 to 8900 kJ per day) diet consisting primarily of vegetables , fruit , nuts , grains and legumes . Walford reported that the healthy, lean test participants reacted with physiological, hematological, hormonal and biochemical changes comparable to those of the test animals in calorie restriction studies.

Effect on various living things

Effects of calorie restriction on the survival rate of laboratory mice (KR = calorie restriction).
Calorie restriction can significantly increase the life expectancy of the black-bellied fruit fly ( Drosophila melanogaster ).

Experiments similar to those with mice have been carried out since 1934 with a wide variety of species and varied experimental conditions. In the case of many species, not only the mean lifespan of the test animals but also their maximum lifespan is increased. The frequency of age-related diseases decreases accordingly. The effect of the increase in maximum life expectancy occurs in rodents both when starting the diet in the early phase of life (1st to 3rd month) and in the middle phase of life (12th month). If, on the other hand, the calorie restriction is started at a later stage in the life of the test animals, for example in the 17th or 24th month of mice, the effect is reversed and the life span of the test animals is shortened.

The calorie restriction was investigated on baker's yeast ( Saccharomyces cerevisiae ), the nematode Caenorhabditis elegans , the fruit fly Drosophila melanogaster , mice , rats , dogs and non-human primates .

Both in a study with rhesus monkeys by the US National Institute on Aging , as well as in a study on Drosophila , it was assumed that the prolongation of life depends not only on the calorie restriction, but also on the composition of the diet.

General criticism

Various findings cast doubts on the idea that calorie restriction slows down the aging process, delays the age-related decline in physiological fitness, or extends the lifespan of organisms from different phylogenetic groups. The positive effects of calorie restriction are not universal:

In fruit flies, the positive effects of calorie restriction are not reproduced if the nutrient fractions are carefully controlled.

The increase in life expectancy caused by calorie restriction is in some cases not even reproducible among the different strains of the same species.

The calorie restriction does not extend the life span of all mice. In the graphic above, a significant effect can be observed in C57BL / 6 mice (“laboratory mice”), while this is not seen in the DBA / 2 mice (“wild type”) below (AL = ad libitum , KR = calorie restriction).

Thus, calorie restriction does not lead to a life extension in all mouse strains. In 19 to 27% of the examined mouse breeds, a calorie restriction of 40% was found to even reduce life expectancy.

The C57BL / 6 mouse, which is often used in experiments, has a tendency to become overweight if there is an unlimited supply of food ( ad libitum ). In these animals the effect of the calorie restriction is significant. DBA / 2 mice, on the other hand, stay slim even when fed ad libitum. In mice from this strain, the calorie restriction does not lead to an extension of life. DBA / 2 mice consume more oxygen than C57BL / 6 mice with the same energy supply, which means that their metabolic rate is increased - they are poorer "feed converters". In previous experiments it was found that the calorie restriction is most successful in mice that have put on weight in early adulthood. The results of these studies are interpreted to mean that the life span is more influenced by the balance of energy intake and energy consumption. The calorie restriction can only bring about a life extension in experimental animals with a tendency to overweight or obesity.

The above NIA study in rhesus monkeys found no life extension. In a long-term study carried out at the Wisconsin National Primate Research Center over a period of 20 years on rhesus monkeys, on the other hand, a significantly better state of health and a significantly increased life expectancy were found in the group of animals that received only a reduced food supply during this period . In this group, 80% of the animals were still alive, while it was only 50% in the normally fed comparison group. In addition, a significantly delayed occurrence ( late-onset ) of age-associated diseases such as diabetes, cancer and brain atrophy , as well as cardiovascular incidents, could be observed in the animals with calorie restriction . The authors of the study come to the conclusion that calorie restriction in this primate species delays the aging process.

Effect on humans

There is currently no scientific evidence that permanent calorie restriction - with adequate human nutrition - leads to an increase in life expectancy compared to a slim adult. It is undisputed that being very overweight , i.e. obese , leads to a shortening of the mean and maximum life expectancy.

The hormonal and metabolic effects of calorie restriction observed in the test animals , such as lower body temperature , reduced metabolic rate and lower oxidative stress , could also be demonstrated in humans. Furthermore, lower values ​​for basal insulin ("fasting insulin "), profibrotic proteins , various growth factors - such as PDGF and TGF-α - as well as cytokines such as tumor necrosis factor -α were detected in the serum . It is also confirmed that long-term calorie restriction is an effective prevention against type II diabetes mellitus , high blood pressure and arteriosclerosis , which together are the main causes of morbidity , disability and mortality in humans.

Evolutionary counter-arguments

There are evolutionary theoretical arguments for the fact that calorie restriction in primates, the order of which also includes great apes including humans, has little or no positive effect on lifespan:

According to the disposable soma theory on the evolution of aging , a drastic reduction in the supply of energy would have a negative effect on reproduction or the lifespan of an organism, since the energy consumed is either used for metabolism, reproduction or for repair and maintenance of the body (physical integrity). must be spent. According to the disposable soma theory, an organism that channels its energy in a state of deficiency to repair and maintenance while suppressing other characteristics such as reproductive effort can live longer at the expense of reproductive capacity.

This fact can also be explained evolutionarily, since a famine has a negative effect on the survival of offspring and it makes less sense to invest energy in offspring who would probably not survive to sexual maturity anyway than in repairing and maintaining your own body .

If rodents' caloric intake is restricted, it actually affects the reproductive system: the increased longevity that rodents result from caloric restriction is always accompanied by a dramatic decline in reproductive capacity. Usually rodents reach sexual maturity around one month of age, and females can produce their body weight in offspring about every month and a half. Rodents are exposed to strong natural selection with regard to early, intensive reproductive investments, accompanied by a short life expectancy .

Primates, on the other hand, experience slow individual development and a low rate of reproduction, combined with a long life span compared to rodents or even similarly sized non-primates. It is therefore assumed that the calorie restriction in humans does not work in the same way as invertebrates and small mammals, because in humans the reproductive investment is low, but the life span is high.

It could be that the absolute life extension through calorie restriction is about one year in all species and not roughly proportional to the life span. The reason for this could be that the variability in food availability depends particularly on the annual decline in plant biomass in winter and is therefore independent of the size of an organism, its phylogenesis or the absolute lifespan. [This thesis is occasionally referenced as weather hypothesis after the publication by Aubrey de Gray from 2005. ] The theory should therefore only lead to an expectation of a life extension of 5 to 14 months, which also agrees with studies on various model organisms.

Risks of calorie restriction in humans

In the USA in particular, the results of animal experiments have led to the fact that calorie restriction has found many practicing followers , especially in California . Some of the followers have joined forces to form the Calorie Restriction Society . Excessive calorie reduction always carries the risk of malnutrition, which can have negative effects on physical and mental health. There are repeated warnings about possible eating disorders associated with calorie restriction. On the other hand, one study showed that calorie restriction does not lead to anorexia or bulimia accumulation . The psychological effects of calorie restriction were rated as positive in this study.

Long-term malnutrition can not only have positive effects but also lead to various deficiency diseases . Developmental disorders can occur in minors. The sensitivity to cold may increase. The ovulation may be suspended in women with very low BMI, resulting in temporary infertility. In the Minnesota Starvation Experiment , anemia , lower extremity edema , muscle wasting, weakness, neurological impairment, dizziness, irritability, lethargy, and depression were observed when a 90% carbohydrate calorie restriction was performed in adult males for six months . Short-term calorie restriction can lead to muscle wasting and decreased bone density . Restricting calories can be harmful in people with low body fat.

mechanism

The causes of the prolongation of the life of model organisms through the caloric restriction have not yet been clarified. The mechanism underlying this effect is unknown. It is possible that the extension of the lifespan results from the improvement of the state of health through the absence of obesity and the later onset of age-related diseases of the metabolic syndrome such as cardiovascular diseases and type II diabetes mellitus .

Studies done in mice suggest that the increase in life that comes with calorie restriction is not simply a result of slimness brought about by the calorie restriction. The maximum lifespan of male rats that kept a low body fat mass through exercise did not increase, but it did that of mice that kept a low body weight simply by restricting calories in a sedentary lifestyle.

The calorie restriction in rats creates soluble factors in the blood serum that prolong life in human cell cultures . Different mechanisms are discussed:

Reduction of oxidative stress

There is evidence that oxidative stress is reduced by reduced food intake, thereby delaying primary aging. Primary aging is the process of cells and organs that - in the absence of disease - defines the maximum life span (inevitable aging). Secondary aging is determined by external factors such as illnesses, environmental factors, lifestyle and physical activity (preventable aging). The oxidative stress mainly takes place in the mitochondria , the power plants of the cells. By resveratrol , the effect of calorie restriction can be induced in certain strains of mice partially. In yeast, the protein Rim15 , a glucose- inhibited protein kinase , serves as a sensor for nutrient concentrations as well as the initiation of meiosis and is necessary for a lifetime extension in yeast. However, in a meta-analysis it was also reported that a calorie restriction - contrary to earlier results - does not lead to an extension of the lifespan in yeast, but that the results in yeast are partly based on methodological artifacts .

Hormesis

A contrary hypothesis is oxidative stress should Reactive oxygen species (Engl. Reactive oxygen species , ROS) cell metabolism positively stimulate ( hormesis ), which as well as the health benefit of calorie restriction by fasting , oxidative plant substances in cruciferous vegetables , and body workout explain.

In contrast to the theory of free radicals , it is assumed that an increased formation of reactive oxygen species in the mitochondria associated with calorie restriction causes an adaptive reaction that increases stress resistance.

Activation of sirtuin-1 and decreased expression of the mTOR receptor

The signal-regulating enzymes Sirtuin-1 (Sirt1) in mammals and Sirtuin Sir2 in yeasts could play a role. The cells of the calorically restrictive fed test animals produce Sirt1 in larger quantities. An increased production of Sirt1 in turn reduces the expression of the mTOR receptor ( mammalian target of rapamycin ), which is also related to the aging process. The life expectancy of mice can be significantly increased by administering rapamycin , which docks at the mTOR receptor. Melatonin is also being studied for sirtuin activation.

"Reprogramming" metabolism and gene expression

According to another theory, the long-term reduced food intake "reprogrammes" the metabolism. A change in gene expression was found in mice under caloric restriction . On the one hand, the genes that are involved in the energy metabolism are overexpressed, while on the other hand, over 50 pro-inflammatory genes are downregulated. The regeneration of some stem cells may be increased. By metformin , a similar effect can be induced in certain strains of mice.

Increased formation of ketone bodies

Both calorie restriction and the ketogenic diet have therapeutic potential in various animal models of neurological diseases. With calorie restriction, there is a transition from glucose metabolism to the use of ketone bodies . Ketone bodies can be used as an alternative source of energy for brain cells when glucose availability is inadequate.

Ketone bodies protect neurons against different types of neural injury. This is an explanatory approach for the beneficial effect of calorie restriction in the animal model of neurological diseases.

Increased autophagocytosis

Autophagocytosis or autophagy , also known as "cellular self-digestion", is a cellular signaling pathway that is involved in the breakdown of proteins and organelles and plays a role in various diseases. Autophagy malfunction has been linked to neurodegeneration , microbial infection, and aging .

Several pieces of evidence suggest that autophagy is important for the effects of calorie restriction: The efficiency of autophagy decreases with increasing age; the decrease in autophagy is associated with changes in the biomarkers of aging; the age-dependent change in autophagy is prevented in the experiment by calorie restriction; if a decrease in the efficiency of autophagy is prevented, the effects are similar to those of calorie restriction; a long-lasting inhibition of autophagy accelerates the aging process; Long-term stimulation of autophagy, on the other hand, delays the aging process in rats ; stimulation of autophagy can save older cells from the accumulation of altered mitochondrial DNA ; stimulation of autophagy alleviates age-related hypercholesterolemia in rodents.

A comparable effect was observed in plants whose exposure was reduced.

Decreased thyroid hormones

The plasma levels of the thyroid hormones triiodothyronine (T 3 ), thyroxine (T 4 ) and thyroid-stimulating hormone (TSH) were measured in rhesus monkeys ( Macaca mulatta ) that were subjected to a 30% CR diet. The T 3 value in the plasma decreased compared to the control group. Given the effects of the thyroid axis on metabolism, this could be one mechanism by which a CR diet communicates its health benefits.

Calorie Restriction Mimetics

Even if the results of human studies should prove a positive effect of calorie restriction on the life expectancy of people, the necessary reduction in the supply of food energy over the corresponding duration and with the corresponding degree is not practicable or desired for the majority of people. Therefore, so-called calorie restriction mimetics ( CR mimetics ) are also being researched. The aim of this strategy is to discover compounds that mimic the effects of calorie restriction in the human body, for example by acting on the same metabolic pathways without the need for actual calorie restriction.

However, more studies are needed to determine whether calorie restriction mimetics actually have an impact on people's life expectancy.

Selection of possible calorie restriction mimetics

According to Ingram, various substances can be used as substances to mimic the calorie restriction in the human body:

  • 2-Deoxy-D-glucose is a substance that can initiate ketogenesis , causes rats to gain slightly less body weight than control animals and leads to a significant reduction in body temperature and the fasting serum insulin level, which simulates certain effects of calorie restriction.
  • Metformin , an oral antidiabetic drug , reduces the incidence of cancer in rats and slows the progression of the disease. It also reduces the incidence of cardiovascular disease and extends life span.
  • Like metformin, glipizide is an oral anti-diabetic drug that helps control blood sugar levels. It works by partially blocking the potassium channels of the beta cells of the islets of Langerhans .
  • Rosiglitazone prevents fatty acid-induced insulin resistance by reducing the glucose infusion rate and improves the insulin-mediated suppression of hepatic glucose production. In addition, it improves the systemic elimination of non-esterified fatty acids.
  • Like rosiglitazone, pioglitazone belongs to the class of thiazolidinediones / glitazones.
  • Soy isoflavones appear to have cardioprotective effects similar to those of calorie restriction, such as a reduction in LDL cholesterol, an inhibition of proinflammatory cytokines , stimulation of nitric oxide production, potential reduction of LDL particles, inhibition of platelet aggregation and an improvement vascular reactivity.
  • Resveratrol increases the survival rate of obese mice compared to a control group of lean, untreated animals. However, adding resveratrol to the diet of lean mice does not result in any further increase in lifespan.
  • Rimonabant is one of the endocannabinoids , cannabis-like substances that can regulate appetite and the energy balance. Rimonabant is a cannabinoid 1 receptor blocker. By overstimulating the endocannabinoid receptor in the hypothalamus , it stimulates fatty acid synthesis (lipogenesis), presumably by increasing adiponectin levels. This lipogenesis reduces intra-abdominal fat. Rimonabant also improves lipid profile and glucose tolerance.
  • Adiponectin , a hormone secreted by fat cells, reduces insulin resistance in obese mice by reducing the levels of triglycerides in the muscles and liver.
  • Sirolimus / rapamycin inhibited the mTOR signaling pathway when fed to mice and resulted in a significantly increased lifespan compared to control mice.
  • Acipimox inhibits the release of fatty acids from adipose tissue and reduces the blood concentration of LDL particles, accompanied by a reduction in triglyceride and cholesterol levels.

Adverse drug effects of calorie restriction mimetics

Depending on their class of active ingredients, the above-mentioned CR mimetics have different side effects / adverse drug effects:

  • Sirolimus inhibits IL-2 and other cytokine receptor-dependent signal cascades and thus develops a strong immunosuppressive effect.
  • Metformin can rarely cause lactic acidosis.
  • Rimonabant can cause diarrhea, dizziness and nausea as well as promote respiratory infections. Other possible undesirable effects are suicidal ideation, memory disorders and probably also seizures.
  • Rosiglitazone - especially in combination with insulin - can cause weight gain and hypercholesterolemia, anemia and edema.
  • Pioglitazone can increase both body weight and the amount of water in the body, and it also increases the risk of fractures. It could also potentially increase your risk of bladder cancer.
  • In women with elevated plasma levels, adiponectin is associated with an increased risk of dementia.

See also

literature

Trade journals (reviews)

  • L. Fontana: Extending Healthy Life Span From Yeast to Humans. In: Science 328, 2010, pp. 321-326. PMID 20395504 .
  • JE Morley et al. a .: Antiaging, longevity and calorie restriction. In: Current Opinion in Clinical Nutrition and Metabolic Care 13, 2010, pp. 40-45. doi: 10.1097 / MCO.0b013e3283331384 PMID 19851100 .
  • J. Skrha: Effect of caloric restriction on oxidative markers. In: Adv Clin Chem 47, 2009, pp. 223-247. PMID 19634782 .
  • JV Smith et al. a .: Energy restriction and aging. In: Curr Opin Clin Nutr Metab Care 7, 2004, pp. 615-622. PMID 15534428 .

Reference books

  • EJ Masoro: Caloric Restriction: A Key to Understanding and Modulating Aging. Elsevier Health Sciences Verlag, 2002, ISBN 0-444-51162-8 .

Popular science articles

  • R. Weindruch: Living longer with poor food? In: Spectrum of Science Dossier 4, 2008, pp. 18–26.
  • J. Blech: healing power of hunger . In: Der Spiegel . No. 50 , 2006, p. 154 ( online ).
  • P. Bethge, L. Höflinger, J. Koch: The biology of hunger . In: Der Spiegel . No. 13 , 2011, p. 154 ( online ).

Web links

Individual evidence

  1. ^ H. Pijl: Longevity. The allostatic load of dietary restriction. In: Physiology & behavior. Volume 106, Number 1, April 2012, ISSN  1873-507X , pp. 51-57, doi: 10.1016 / j.physbeh.2011.05.030 , PMID 21663754 .
  2. K. Dorshkind et al. a .: The aging immune system: is it ever too old to become young again? In: Nat Rev Immunol 9, 2009, pp. 57-62. PMID 19104499 .
  3. GD Kolovou, V. Kolovou, S. Mavrogeni: We are aging. In: BioMed Research International. Volume 2014, 2014, ISSN  2314-6141 , p. 808307, doi: 10.1155 / 2014/808307 , PMID 25045704 , PMC 4090574 (free full text).
  4. ^ JR Speakman, SE Mitchell: Caloric restriction. In: Molecular Aspects of Medicine. Volume 32, Number 3, June 2011, ISSN  1872-9452 , pp. 159-221, doi: 10.1016 / j.mam.2011.07.001 , PMID 21840335 .
  5. a b L. Mulvey, A. Sinclair, C. Selman: Lifespan modulation in mice and the confounding effects of genetic background. In: Journal of genetics and genomics = Yi chuan xue bao. Volume 41, number 9, September 2014, ISSN  1673-8527 , pp. 497-503, doi: 10.1016 / j.jgg.2014.06.002 , PMID 25269675 , PMC 4257991 (free full text).
  6. ^ Louis Cornaro: The Art of Living Long. Translated into English by WF Butler, Verlag Springer, 1903, ISBN 978-0-8261-2695-5 p. XXIII ( limited preview in Google book search).
  7. ^ AJ Carlson, F. Hoelzel: Apparent Prolongation of the Life Span of Rats by Intermittent Fasting. In: Journal of Nutrition , Volume 31, 1946, pp. 363-375, PMID 21021020 .
  8. ^ A b C. M. McCay and MF Crowell: Prolonging the Life Span . In: The Scientific Monthly 39, 1934, pp. 405-414; JSTOR 15813 .
  9. CM McCay et al. a .: The effect of retarded growth upon the length of the life span and upon the ultimate body size. In: J Nutr 79, 1935, pp. 63-79.
  10. CM McCay et al. a .: The effect of retarded growth upon the length of life span and upon the ultimate body size. 1935. In: Nutrition 5, 1989, pp. 155-171. PMID 2520283 .
  11. a b L. Fontana: The scientific basis of caloric restriction leading to longer life. In: Curr Opin Gastroenterol 25, 2009, pp. 144-150. PMID 19262201 (Review).
  12. ^ MH Ross: Length of life and nutrition in the rat. In: The Journal of Nutrition. Volume 75, October 1961, ISSN  0022-3166 , pp. 197-210, PMID 14494200 .
  13. ^ MH Ross: Length of life and caloric intake. In: The American Journal of Clinical Nutrition. Volume 25, Number 8, August 1972, ISSN  0002-9165 , pp. 834-838, PMID 5046728 .
  14. a b R. B. McDonald, JJ Ramsey: Honoring Clive McCay and 75 years of calorie restriction research. In: The Journal of Nutrition. Volume 140, number 7, July 2010, ISSN  1541-6100 , pp. 1205-1210, doi: 10.3945 / jn.110.122804 , PMID 20484554 , PMC 2884327 (free full text).
  15. R. Weindruch, RL Walford: Dietary restriction in mice beginning at 1 year of age: effect on life-span and spontaneous cancer incidence. In: Science. Volume 215, Number 4538, March 1982, ISSN  0036-8075 , pp. 1415-1418, PMID 7063854 .
  16. ^ RJ Colman, TM Beasley et al. a .: Caloric restriction reduces age-related and all-cause mortality in rhesus monkeys. In: Nature Communications. Volume 5, 2014, ISSN  2041-1723 , p. 3557, doi: 10.1038 / ncomms4557 , PMID 24691430 , PMC 3988801 (free full text).
  17. a b B. Lakowski, S. Hekimi: The genetics of caloric restriction in Caenorhabditis elegans. In: PNAS. Volume 95, Number 22, October 1998, ISSN  0027-8424 , pp. 13091-13096, PMID 9789046 , PMC 23719 (free full text).
  18. ^ RL Walford, D. Mock et al. a .: Calorie restriction in biosphere 2: alterations in physiologic, hematologic, hormonal, and biochemical parameters in humans restricted for a 2-year period. In: The Journals of Gerontology. Series A, Biological sciences and medical sciences. Volume 57, Number 6, June 2002, ISSN  1079-5006 , pp. B211-B224, PMID 12023257 .
  19. a b R. Weindruch u. a .: The retardation of aging in mice by dietary restriction: longevity, cancer, immunity and lifetime energy intake. In: Journal of Nutrition Volume 116, 1986, pp. 641-654. PMID 3958810 .
  20. a b T. Hofer u. a .: One year of caloric restriction and exercise in humans and the effects on DNA and RNA oxidation levels in white blood cells and urine. In: Rejuvenation Res 11, 2008, pp. 793-799. PMID 18729811 .
  21. a b M. A. Lane et al. a .: Caloric restriction and aging in primates: Relevance to humans and possible CR mimetics. In: Microsc Res Tech 59, 2002, pp. 335-338. PMID 12424798 (Review).
  22. a b J. Wanagat u. a .: Caloric intake and aging: mechanisms in rodents and a study in nonhuman primates. In: Toxicol Sci 52, 1999, pp 35-40. PMID 10630588 (Review).
  23. a b c M. J. Forster u. a .: Genotype and age influence the effect of caloric intake on mortality in mice. In: FASEB J 17, 2003, pp. 690-692. PMID 12586746 .
  24. SJ Lin et al. a .: Requirement of NAD and SIR2 for life-span extension by calorie restriction in Saccharomyces cerevisiae. In: Science 289, 2000, pp. 2126-2128. PMID 11000115 .
  25. SJ Lin et al. a .: Calorie restriction extends Saccharomyces cerevisiae lifespan by increasing respiration. In: Nature 418, 2002, pp. 344-348. PMID 12124627 .
  26. W. Mair et al. a .: Calories do not explain extension of life span by dietary restriction in Drosophila. In: PLoS Biol 3, 2005, e223. PMID 16000018 .
  27. DF Lawler et al. a .: Diet restriction and aging in the dog: major observations over two decades. In: Br J Nutr 99, 2007, pp. 1-13. PMID 1806283 .
  28. R. Weindruch: The retardation of aging by caloric restriction: studies in rodents and primates. In: Toxicol Pathol 24, 1996, pp. 742-745. PMID 8994305 .
  29. a b J. A. Mattison, GS Roth u. a .: Impact of caloric restriction on health and survival in rhesus monkeys from the NIA study. In: Nature. Volume 489, Number 7415, September 2012, pp. 318-321. doi: 10.1038 / nature11432 . PMID 22932268 .
  30. ^ W. Mair, MD Piper, L. Partridge: Calories do not explain extension of life span by dietary restriction in Drosophila. In: PLoS Biology. Volume 3, number 7, July 2005, ISSN  1545-7885 , p. E223, doi: 10.1371 / journal.pbio.0030223 , PMID 16000018 , PMC 1140680 (free full text).
  31. a b R. S. Sohal, MJ Forster: Caloric restriction and the aging process: a critique. In: Free radical biology & medicine. Volume 73, August 2014, ISSN  1873-4596 , pp. 366-382, doi: 10.1016 / j.freeradbiomed.2014.05.015 , PMID 24941891 , PMC 4111977 (free full text) (review).
  32. KP Lee, SJ Simpson et al. a .: Lifespan and reproduction in Drosophila: New insights from nutritional geometry. In: Proceedings of the National Academy of Sciences . Volume 105, number 7, February 2008, ISSN  1091-6490 , pp. 2498-2503, doi: 10.1073 / pnas.0710787105 , PMID 18268352 , PMC 2268165 (free full text).
  33. ^ CY Liao, BA Rikke, TE Johnson, V. Diaz, JF Nelson: Genetic variation in the murine lifespan response to dietary restriction: from life extension to life shortening. In: Aging Cell. Volume 9, number 1, February 2010, ISSN  1474-9726 , pp. 92-95, doi: 10.1111 / j.1474-9726.2009.00533.x , PMID 19878144 , PMC 3476836 (free full text).
  34. BA Rikke, CY Liao, MB McQueen, JF Nelson, TE Johnson: Genetic dissection of dietary restriction in mice supports the metabolic efficiency model of life extension. In: Experimental Gerontology. Volume 45, number 9, September 2010, ISSN  1873-6815 , pp. 691-701, doi: 10.1016 / j.exger.2010.04.008 , PMID 20452416 , PMC 2926251 (free full text).
  35. RS Sohal: Life span extension in mice by food restriction depends on an energy imbalance. In: J Nutr 139, 2009, pp. 533-539. doi: 10.3945 / jn.108.100313 PMID 19141702 .
  36. MH Ross et al. a .: Dietary practices and growth responses as predictors of longevity. In: Nature 262, 1976, pp. 548-553. PMID 958413 .
  37. a b Life extension: the myth of calorie restriction refuted. (No longer available online.) In: aerzteblatt.de . January 26, 2009, archived from the original on May 18, 2015 ; accessed on May 1, 2015 . Info: The archive link was inserted automatically and has not yet been checked. Please check the original and archive link according to the instructions and then remove this notice. @1@ 2Template: Webachiv / IABot / www.aerzteblatt.de
  38. RJ Colman, RM Anderson et al. a .: Caloric restriction delays disease onset and mortality in rhesus monkeys. In: Science. Volume 325, Number 5937, July 2009, pp. 201-204, ISSN  1095-9203 . doi: 10.1126 / science.1173635 . PMID 19590001 . PMC 2812811 (free full text).
  39. rme: Anti-Aging: Calorie restriction lets monkeys live longer. ( Memento of the original from October 15, 2013 in the Internet Archive ) Info: The archive link was inserted automatically and has not yet been checked. Please check the original and archive link according to the instructions and then remove this notice. In: aerzteblatt.de of July 10, 2009. @1@ 2Template: Webachiv / IABot / www.aerzteblatt.de
  40. Stephan Schleim: In search of the fountain of youth: the example of calorie reduction. In: heise.de . August 31, 2012, accessed May 11, 2015 .
  41. ^ LK Heilbronn u. a .: Effect of 6-month calorie restriction on biomarkers of longevity, metabolic adaptation, and oxidative stress in overweight individuals: a randomized controlled trial. In: JAMA 295, 2006, pp. 1539-1548. PMID 16595757
  42. L. Fontana, TE Meyer et al. a .: Long-term calorie restriction is highly effective in reducing the risk for atherosclerosis in humans. In: Proceedings of the National Academy of Sciences . Volume 101, number 17, April 2004, ISSN  0027-8424 , pp. 6659-6663, doi: 10.1073 / pnas.0308291101 , PMID 15096581 , PMC 404101 (free full text).
  43. L. Fontana et al. a .: Calorie restriction or exercise: effects on coronary heart disease risk factors. A randomized, controlled trial. In: Am J Physiol Endocrinol Metab 293, 2007, pp. E197-202. PMID 17389710
  44. EP Weiss u. a .: Improvements in glucose tolerance and insulin action induced by increasing energy expenditure or decreasing energy intake: a randomized controlled trial. In: Am J Clin Nutr , Volume 84, 2006, pp. 1033-1042. PMID 17093155
  45. ^ DE Larson-Meyer u. a .: Effect of calorie restriction with or without exercise on insulin sensitivity, beta-cell function, fat cell size, and ectopic lipid in overweight subjects. In: Diabetes Care 29, 2006, pp. 1337-1344. PMID 16732018
  46. TE Meyer u. a .: Long-term caloric restriction ameliorates the decline in diastolic function in humans. In: J Am College Cardiol 47, 2006, pp. 398-402. PMID 16412867 .
  47. a b D. P. Shanley, TB Kirkwood: Calorie restriction and aging: a life-history analysis. In: evolution; international journal of organic evolution. Volume 54, Number 3, June 2000, ISSN  0014-3820 , pp. 740-750, PMID 10937249 .
  48. EJ Masoro, SN Austad: The evolution of the anti-aging action of dietary restriction: a hypothesis. In: The journals of gerontology. Series A, Biological sciences and medical sciences. Volume 51, Number 6, November 1996, ISSN  1079-5006 , pp. B387-B391, PMID 8914486 .
  49. a b c J. P. Phelan, MR Rose: Why dietary restriction substantially increases longevity in animal models but won't in humans. In: Aging research reviews. Volume 4, Number 3, August 2005, ISSN  1568-1637 , pp. 339-350, doi: 10.1016 / j.arr.2005.06.001 , PMID 16046282 (review).
  50. ^ A b J. R. Speakman, SE Mitchell: Caloric restriction. In: Molecular aspects of medicine. Volume 32, number 3, June 2011, ISSN  1872-9452 , pp. 159-221, doi: 10.1016 / j.mam.2011.07.001 , PMID 21840335 (review).
  51. a b A. D. de Gray: The unfortunate influence of the weather on the rate of aging: why human caloric restriction or its emulation may only extend life expectancy by 2-3 years. In: Gerontology. Volume 51, Number 2, 2005 Mar-Apr, ISSN  0304-324X , pp. 73-82, doi: 10.1159 / 000082192 , PMID 15711074 (review).
  52. DA Williamson et al. a .: Is Caloric Restriction Associated With Development of Eating-Disorder Symptoms? Results From the CALERIE Trial. In: Health Psychology , Volume 27, 2008, pp. S32-S42. PMID 18248104 .
  53. a b E. Marzetti, SE Wohlgemuth, SD Anton, R Bernabei, CS Carter, C. Leeuwenburgh: Cellular mechanisms of cardiovascular protection by calorie restriction: state of the science and future perspectives . In: Ncbi.nlm.nih.gov (Ed.): Clin. Geriatrician Med. . 25, No. 4, October 19, 2012, pp. 715-32, ix. doi : 10.1016 / j.cger.2009.07.002 . PMID 19944269 . PMC 2786899 (free full text).
  54. ^ A. Keys, J. Brozek, A. Henschels, O. Mickelsen, H. Taylor: The Biology of Human Starvation , 1950, Volume 2, University of Minnesota Press, Minneapolis. P. 1133.
  55. John E Morley, Elie Chahla, Saad Alkaade: Antiaging, longevity and calorie restriction . In: Current Opinion in Clinical Nutrition and Metabolic Care . 13, No. 1, 2010, pp. 40-5. doi : 10.1097 / MCO.0b013e3283331384 . PMID 19851100 .
  56. L. Fontana, S. Klein: Aging, Adiposity, and Calorie Restriction . In: JAMA . 297, No. 9, 2007, pp. 986-94. doi : 10.1001 / jama.297.9.986 . PMID 17341713 .
  57. a b L. Fontana, S. Klein: Aging, adiposity, and calorie restriction. In: JAMA. Volume 297, Number 9, March 2007, ISSN  1538-3598 , pp. 986-994, doi: 10.1001 / jama.297.9.986 , PMID 17341713 (review).
  58. ^ R. de Cabo, L. Liu, A. Ali, N. Price, J. Zhang, M. Wang, E. Lakatta, PM Irusta: Serum from calorie-restricted animals delays senescence and extends the lifespan of normal human fibroblasts in vitro. In: Aging. Volume 7, Number 3, March 2015, ISSN  1945-4589 , pp. 152-166, PMID 25855056 .
  59. M. Tostlebe: Disproportionality of the activities of the mitochondrial respiratory chain complexes in the myocardium and in the skeletal muscles in old age. Dissertation, Martin Luther University Halle-Wittenberg, 2005.
  60. A. Csiszar et al. a .: Anti-oxidative and anti-inflammatory vasoprotective effects of caloric restriction in aging: role of circulating factors and SIRT1. In: Mech Aging Dev 130, 2009, pp. 518-527. PMID 19549533 .
  61. J. Skrha: Effect of caloric restriction on oxidative markers. In: Adv Clin Chem 47, 2009, pp. 223-247. PMID 19634782 .
  62. a b M. R. Ramis, S. Esteban u. a .: Caloric restriction, resveratrol and melatonin: Role of SIRT1 and implications for aging and related diseases. In: Mechanisms of aging and development. [Electronic publication before printing] March 2015, ISSN  1872-6216 , doi: 10.1016 / j.mad.2015.03.008 , PMID 25824609 .
  63. S. Nagarajan, Kruckeberg AL, KH Schmidt, E. Kroll, M. Hamilton, K. McInnerney, R. Summers, T. Taylor, F. Rosenzweig: Uncoupling reproduction from metabolism extends chronological life span in yeast. In: PNAS. Volume 111, number 15, April 2014, ISSN  1091-6490 , pp. E1538 – E1547, doi: 10.1073 / pnas.1323918111 , PMID 24706810 , PMC 3992631 (free full text).
  64. DH Huberts, J. González, SS Lee, A. Litsios, G. Hubmann, EC Wit, M. Heinemann: Calorie restriction does not elicit a robust extension of replicative lifespan in Saccharomyces cerevisiae. In: PNAS. Volume 111, number 32, August 2014, ISSN  1091-6490 , pp. 11727-11731, doi: 10.1073 / pnas.1410024111 , PMID 25071164 , PMC 4136557 (free full text).
  65. PC Tapia: Sublethal mitochondrial stress with an attendant stoichiometric augmentation of reactive oxygen species may precipitate many of the beneficial alterations in cellular physiology produced by caloric restriction, intermittent fasting, exercise and dietary phytonutrients: “Mitohormesis” for health and vitality. In: Medical Hypotheses. Volume 66, Number 4, October 19, 2005, pp. 832-843. PMID 16242247
  66. M. Ristow, K. Zarse: How increased oxidative stress promotes longevity and metabolic health: The concept of mitochondrial hormesis (mitohormesis). In: Experimental Gerontology . Volume 45, Number 6, June 2010, ISSN  1873-6815 , pp. 410-418, doi: 10.1016 / j.exger.2010.03.014 , PMID 20350594 (review).
  67. ^ Y. Wang: Molecular Links between Caloric Restriction and Sir2 / SIRT1 Activation. In: Diabetes & Metabolism Journal. Volume 38, number 5, October 2014, ISSN  2233-6079 , pp. 321-329, doi: 10.4093 / dmj.2014.38.5.321 , PMID 25349818 , PMC 4209345 (free full text).
  68. C. Cantó and J. Auwerx: Caloric restriction, SIRT1 and longevity. In: Trends Endocrinol Metab 20, 2009, pp. 325–331. PMID 19713122 .
  69. HS Ghosh et al. a .: SIRT1 negatively regulates the mammalian target of rapamycin . In: PLoS One 5, 2010, e9199, PMID 20169165 .
  70. ^ S. Austad: Recent advances in vertebrate aging research 2009. In: Aging Cell 9, 2010, pp. 297-303 PMID 20331443 .
  71. ^ DE Harrison et al. a .: Rapamycin fed late in life extends lifespan in genetically heterogeneous mice. In: Nature 460, 2009, pp. 392-395. PMID 19587680 , PMC 2786175 (free full text).
  72. RM Anderson, R. Weindruch: Metabolic reprogramming in dietary restriction. In: Interdiscip Top Gerontol 35, 2007, pp. 18-38. PMID 17063031 (Review).
  73. Y. Higami et al. a .: Adipose tissue energy metabolism: altered gene expression profile of mice subjected to long-term caloric restriction. In: FASEB J 18, 2004, pp. 415-417. PMID 14688200 .
  74. Y. Higami et al. a .: Energy restriction lowers the expression of genes linked to inflammation, the cytoskeleton, the extracellular matrix, and angiogenesis in mouse adipose tissue. In: J Nutr 136, 2006, pp. 343-352. PMID 16424110 .
  75. RM Anderson et al. a .: Caloric restriction and aging: studies in mice and monkeys. In: Toxicol Pathol 37, 2009, pp. 47-51. PMID 19075044 (Review).
  76. G. Mazzoccoli, MF Tevy, M. Borghesan, MR Delle Vergini, M. Vinciguerra: Caloric restriction and aging stem cells: the stick and the carrot? In: Experimental Gerontology. Volume 50, February 2014, ISSN  1873-6815 , pp. 137-148, doi: 10.1016 / j.exger.2013.10.014 , PMID 24211426 .
  77. M. Maalouf, JM Rho, MP Mattson: The neuroprotective properties of calorie restriction, the ketogenic diet, and ketone bodies. In: Brain research reviews. Volume 59, number 2, March 2009, ISSN  0165-0173 , pp. 293-315, doi: 10.1016 / j.brainresrev.2008.09.002 , PMID 18845187 , PMC 2649682 (free full text) (review).
  78. a b A. L. Lin, W. Zhang u. a .: Caloric restriction increases ketone bodies metabolism and preserves blood flow in aging brain. In: Neurobiology of aging. [Electronic publication before printing] March 2015, ISSN  1558-1497 , doi: 10.1016 / j.neurobiolaging.2015.03.012 , PMID 25896951 .
  79. ^ EA Minina, V. Sanchez-Vera et al. a .: Autophagy mediates caloric restriction-induced lifespan extension in Arabidopsis. In: Aging cell. Volume 12, Number 2, April 2013, ISSN  1474-9726 , pp. 327-329, doi: 10.1111 / acel.12048 , PMID 23331488 .
  80. G. Cavallini, A. Donati et al. a .: Towards an understanding of the anti-aging mechanism of caloric restriction. In: Current aging science. Volume 1, Number 1, March 2008, ISSN  1874-6128 , pp. 4-9, PMID 20021367 .
  81. GS Roth, AM Handy u. a .: Effects of dietary caloric restriction and aging on thyroid hormones of rhesus monkeys. In: Hormone and metabolic research. Volume 34, Number 7, July 2002, ISSN  0018-5043 , pp. 378-382, doi: 10.1055 / s-2002-33469 , PMID 12189585 .
  82. Hans-Jörg Ehni: Ethics of Biogerontology. Springer-Verlag, 2014, ISBN 978-3-658-03378-1 , p. 56 ( limited preview in Google book search).
  83. a b DK Ingram, M. Zhu u. a .: Calorie restriction mimetics: an emerging research field. In: Aging cell. Volume 5, Number 2, April 2006, ISSN  1474-9718 , pp. 97-108, doi: 10.1111 / j.1474-9726.2006.00202.x , PMID 16626389 (review).
  84. G. Testa, F. Biasi, G. Poli, E. Chiarpotto: Calorie restriction and dietary restriction mimetics: a strategy for improving healthy aging and longevity. In: Current Pharmaceutical Design. Volume 20, Number 18, 2014, ISSN  1873-4286 , pp. 2950-2977, PMID 24079773 .
  85. J. Yao, S. Chen et al. a .: 2-Deoxy-D-glucose treatment induces ketogenesis, sustains mitochondrial function, and reduces pathology in female mouse model of Alzheimer's disease. In: PloS one. Volume 6, number 7, 2011, ISSN  1932-6203 , p. E21788, doi: 10.1371 / journal.pone.0021788 , PMID 21747957 , PMC 3128612 (free full text).
  86. MA Lane, DK Ingram et al. a .: 2-Deoxy-D-Glucose Feeding in Rats Mimics Physiologic Effects of Calorie Restriction. In: Journal of Anti-Aging Medicine. 1, 1998, pp. 327-337, doi: 10.1089 / rej.1.1998.1.327 .
  87. DL Smith, CF Elam et al. a .: Metformin supplementation and life span in Fischer-344 rats. In: The journals of gerontology. Series A, Biological sciences and medical sciences. Volume 65, number 5, May 2010, ISSN  1758-535X , pp. 468-474, doi: 10.1093 / gerona / glq033 , PMID 20304770 , PMC 2854888 (free full text).
  88. HE Lebovitz, MN fine Glos: Mechanism of action of the second-generation sulfonylurea glipizide. In: The American journal of medicine. Volume 75, Number 5B, November 1983, ISSN  0002-9343 , pp. 46-54, PMID 6369967 .
  89. JM Ye, N. Dzamko et al. a .: Direct demonstration of lipid sequestration as a mechanism by which rosiglitazone prevents fatty-acid-induced insulin resistance in the rat: comparison with metformin. In: Diabetologia. Volume 47, Number 7, July 2004, ISSN  0012-186X , pp. 1306-1313, doi: 10.1007 / s00125-004-1436-1 , PMID 15232684 .
  90. G. Rimbach, C. Boesch-Saadatmandi u. a .: Dietary isoflavones in the prevention of cardiovascular disease - a molecular perspective. In: Food and chemical toxicology: an international journal published for the British Industrial Biological Research Association. Volume 46, Number 4, April 2008, ISSN  0278-6915 , pp. 1308-1319, doi: 10.1016 / j.fct.2007.06.029 , PMID 17689850 (review).
  91. B. Agarwal, JA Baur: Resveratrol and life extension. In: Annals of the New York Academy of Sciences. Volume 1215, January 2011, ISSN  1749-6632 , pp. 138-143, doi: 10.1111 / j.1749-6632.2010.05850.x , PMID 21261652 (review).
  92. a b M. Kyriazis: Calorie restriction mimetics: Examples and mode of action. In: Open Longevity Sci. Volume 3, 2009, pp. 17-21. doi: 10.2174 / 1876326X00903010017
  93. T. Yamauchi, J. Kamon et al. a .: The fat-derived hormone adiponectin reverses insulin resistance associated with both lipoatrophy and obesity. In: Nature medicine. Volume 7, Number 8, August 2001, ISSN  1078-8956 , pp. 941-946, doi: 10.1038 / 90984 , PMID 11479627 .
  94. T. Chiba, T. Tsuchiya et al. a .: Development of calorie restriction mimetics as therapeutics for obesity, diabetes, inflammatory and neurodegenerative diseases. In: Current genomics. Volume 11, number 8, December 2010, ISSN  1875-5488 , pp. 562-567, doi: 10.2174 / 138920210793360934 , PMID 21629433 , PMC 3078680 (free full text).
  95. ^ J. Rice: First Drug Shown to Extend Life Span in Mammals . In: Massachusetts Institute of Technology (Ed.): Technology Review . July 8, 2009, pp. 1-2. Retrieved July 9, 2009.
  96. Madiraju et al. Metformin suppresses gluconeogenesis by inhibiting mitochondrial glycerophosphate dehydrogenase. Nature 2014; 510: 542-546. doi: 10.1038 / nature13270
  97. Dt. Ärzteblatt online: Weight loss drug Rimonabant can trigger suicidal thoughts ( Memento of the original from February 26, 2009 in the Internet Archive ) Info: The archive link was inserted automatically and has not yet been checked. Please check the original and archive link according to the instructions and then remove this notice. , June 12, 2007. @1@ 2Template: Webachiv / IABot / www.aerzteblatt.de
  98. Avandia: Product information texts (PDF; 849 kB), as of July 2010.
  99. Meier, Juris J .; Nauck, Michael A .; Schmidt, Wolfgang E .; Gallwitz, Baptist: Diagnosis of limited glucose tolerance and diabetes prevention: Can the diabetes epidemic be stopped? Dtsch Arztebl 2002; 99 (47): A-3182 / B-2685 / C-2500.
  100. Fiche Médicament sous surveillance renforcée: Actos, Competact - Traitement du diabète de type 2 of June 9, 2011.
  101. Thomas M. van Himbergen, Alexa S. Beiser, Masumi Ai, Sudha Seshadri, Seiko Otokozawa, Rhoda Au, Nuntakorn Thongtang, Philip A. Wolf, Ernst J. Schaefer: Biomarkers for Insulin Resistance and Inflammation and the Risk for All-Cause Dementia and Alzheimer's Disease. In: Arch Neurol . 69, 2012, p. 594, doi: 10.1001 / archneurol.2011.670 .