Skip to main content
F1000Research logoLink to F1000Research
. 2016 Jan 29;5:F1000 Faculty Rev-117. [Version 1] doi: 10.12688/f1000research.7136.1

Dietary restriction with and without caloric restriction for healthy aging

Changhan Lee 1,a, Valter Longo 1,2,b
PMCID: PMC4755412  PMID: 26918181

Abstract

Caloric restriction is the most effective and reproducible dietary intervention known to regulate aging and increase the healthy lifespan in various model organisms, ranging from the unicellular yeast to worms, flies, rodents, and primates. However, caloric restriction, which in most cases entails a 20–40% reduction of food consumption relative to normal intake, is a severe intervention that results in both beneficial and detrimental effects. Specific types of chronic, intermittent, or periodic dietary restrictions without chronic caloric restriction have instead the potential to provide a significant healthspan increase while minimizing adverse effects. Improved periodic or targeted dietary restriction regimens that uncouple the challenge of food deprivation from the beneficial effects will allow a safe intervention feasible for a major portion of the population. Here we focus on healthspan interventions that are not chronic or do not require calorie restriction.

Keywords: Caloric restriction, mechanisms of aging, Dietary restriction, aging, healthspan

Introduction

Aging is the leading risk factor for many among the major diseases and a key factor in the overall decline of physical and mental performance. Interventions that slow down the aging process can delay or prevent multiple chronic diseases and improve productivity and quality of life in older ages. For clarity, here, we use caloric restriction (CR) to refer to a dietary intervention with an overall 20–40% reduction of total caloric intake, and dietary restriction to represent a broader scope of dietary interventions that encompass those with specific macronutrient and feeding pattern restrictions. CR is the most studied and reproducible non-genetic intervention known to extend healthspan and/or lifespan in organisms, ranging from unicellular yeast to monkeys. It started with a simple experiment where a reduction in dietary intake (i.e. caloric restriction) extended the lifespan of rats 1, providing a foundation to experimentally study the relationship between nutrition and the biology of aging. Here we discuss more recent discoveries that have advanced our knowledge of the effects of less invasive and restrictive dietary interventions in aging and healthspan.

Caloric restriction and the conserved mechanisms of aging

In their seminal report in 1935, Crowell and McCay demonstrated that simply reducing caloric intake without causing malnutrition nearly doubled the lifespan of rats 1, providing an experimental model to begin to demonstrate that aging can be slowed down. Nearly half a century later, Walford and Weindruch reported that “adult-initiated” caloric restriction started at 12 months of age not only increased lifespan but also reduced the incidence of spontaneous cancer by more than 50% in rats 2, 3. Several decades later, the effect of caloric restriction on healthspan and lifespan has been confirmed in model organisms ranging from unicellular yeast to worms, flies, rodents, and primates 4, suggesting a highly conserved effect which may involve common genes. Although the molecular mechanisms that mediate the effect of caloric restriction are still being investigated and debated, there is more widespread acceptance of the hypothesis that caloric restriction and lifespan extension involves the down-regulation of insulin and insulin-like signalling (IIS) 5, as well as of the amino signalling target of rapamycin (TOR)-S6 kinase pathway 6, 7, and the glucose signalling Ras-protein kinase A (PKA) pathway 6, 8, 9.

Because an in depth discussion of the anti- and pro-aging pathways conserved in model organisms has been covered elsewhere, here we will only mention the most relevant ones 1012. In yeast, down-regulation of (a) the amino acid-sensing TOR and the ribosomal protein S6 kinase (S6K) ortholog Sch9 pathway 6, and (b) the Ras-AC-PKA pathway 13 are key changes mediating part of the effects of caloric restriction on chronological lifespan, the measurement of cellular survival under non-dividing conditions. In contrast, elevated activity of sirtuin ( SIR2) has been described as a key change in the extension of replicative lifespan, measured by counting the number of buds generated by an individual mother cell 14, 15. In worms, the lifespan extension caused by the inactivation of IIS, or by different forms of caloric restriction, requires Forkhead FoxO transcription factor daf-16 16. In flies, the IIS pathway is involved in the effects of caloric restriction 17 and, although dFoxo is not required for its longevity effect, its activity can affect the response to caloric restriction 18. In rodents, growth hormone (GH) and IGF-1 levels are reduced following caloric restriction 19, but the link between dietary restriction, GH and aging is still being investigated, with focus on the genes and pathways regulating longevity in the simple organisms described above. The long-lived GH receptor knock-out (GHRKO) mice, which are resistant to GH, do not exhibit further lifespan extension or health benefits by caloric restriction 20, 21, but the long-lived GH-deficient Ames mice do 22, suggesting a complex involvement of the GH/IGF-1 axis and periphery pathways in the response to caloric restriction.

Much has been learned about caloric restriction and aging from model organisms, but the ultimate question that lingers is the relevance of these models to human lifespan and healthspan. The rhesus monkeys are the closest model organism to humans in which caloric restriction has been experimentally tested in a controlled environment. Two notable studies performed by independent programs, the National Institute on Aging (NIA) Intramural Research Program and the Wisconsin National Primate Research Center (WNPRC), subjected male and female rhesus monkeys to 30% caloric restriction from levels of baseline caloric intake. The NIA reported no improvement in lifespan but observed a positive trend for the delay of age-related diseases (i.e. healthspan) 23, whereas WNPRC reported significant improvement in both lifespan and healthspan 4, 24. The discrepancies are largely attributed to the different dietary composition and heterogenic genetic background 4, 23, which have been shown to be a significant factor in rodents 25, 26. This underscores the importance of diet composition and genetic background and their compatibility when applying caloric restriction to humans. Nonetheless, several studies provide evidence supporting beneficial health effects of caloric restriction for humans. A notable NIH-sponsored controlled randomized study on non-obese individuals, CALERIE (Comprehensive Assessment of the Long-term Effects of Reducing Intake of Energy), recently reported that a two year 25% caloric restriction is feasible for humans and provides health benefits, such as reduced inflammatory markers and cardiometabolic risk factors 2729. However, caloric restriction was associated with reduced bone mineral density and exercise was recommended to offset such adverse effects 30. Notably, CALERIE was conducted in three independent centers and involved 218 overweight participants, suggesting that caloric restriction can be beneficial even in a very genetically heterogeneous group 31. However, considering the results in monkeys, much longer and larger studies will be needed to know what the effects of CR on human healthspan.

Among the cellular alterations most closely associated with both caloric restriction and longevity mutations is the resistance to multiple stressors, which in most cases includes resistance to oxidative stress. The ability of caloric restriction to prevent the damage caused by exogenous toxins is likely to be associated with the protection, repair and replacement effects that prevent the age-dependent dysfunction caused by endogenous processes and toxic molecules 32. An alternative hypothesis suggests that caloric restriction acts as a mild stressor that promotes hormesis, which refers to the beneficial effects resulting from the cellular responses to mild, repeated stress 33. Stress resistance should also be considered an important criterion for the successful development of caloric restriction-mimetic dietary and pharmacological interventions.

Dietary restriction: macromolecular restriction without caloric restriction

The definition of dietary restriction has been expanded from an alternative description of caloric restriction to also encompass a broader scope of interventions, including short-term starvation, periodic fasting, fasting-mimetic diets, intermittent fasting, normo-caloric diets with planned deficiencies (in particular macronutrients: proteins, carbohydrates, etc.), and time-restricted feeding. Most of these relatively novel interventions are reported to have beneficial effects on overall health and in some cases longevity. Fasting is an extreme dietary intervention describing either a complete lack of food intake or a 60% or higher food restriction. Intermittent fasting refers to practicing this intervention every other day whereas periodic fasting refers to severe restriction for two or more days periodically (every two weeks, month, etc.). Caloric restriction and fasting share similar but often distinct effects on a number of biomarkers (e.g. reduced glucose, and insulin levels) suggesting that partially overlapping mechanisms are involved 19. Both intermittent and periodic fasting can increase lifespan, even when there is little or no overall decrease in calorie intake 34, 50.

Macronutrient restriction

In addition to periodic or intermittent fasting-based strategies as alternatives to caloric restriction, the restriction of specific macronutrients (or macronutrient restriction) without the restriction of calories is among the most promising interventions that have emerged to promote healthy aging in humans. Among the different types of macronutrient restriction, reduced intake of proteins and amino acids is the most effective pro-longevity intervention 35, 36. Simply reducing protein intake can deliver an equally potent impact on lifespan as dietary restriction in multiple model organisms 35. A recent analysis of the National Health and Nutrition Examination Survey (NHANES) showed that low protein intake was associated with reduced overall mortality for those under 65 years of age 37. Also, a high-carbohydrate, low-protein diet resulted in longer lifespan and improved cardiometabolic health, despite increased food intake and body fat 38, 39. Furthermore, the restriction of a single essential amino acid in a normal diet increased lifespan and stress resistance 4044. In flies, adding back essential amino acids to the caloric restriction diet decreased lifespan to that of the normally fed group 36. Laboratory rodents fed a methionine-restricted diet displayed an extended lifespan with decreased age-dependent diseases and increased resistance to oxidative stress, in part due to increased antioxidant capacity 4448. A tryptophan-restricted diet also provided longevity and reduced age-dependent deterioration 42, 43, 49 but has mainly been explored for neurological benefits, due to its role in serotonin synthesis. A fasting-mimicking diet, consisting of very low calorie and protein that leads to similar physiological response to fasting, including reduced levels of glucose and IGF-1 and increased levels of ketone bodies IGFBP-1, enhanced healthspan and rejuvenated the hematopoietic system while promoting adult neurogenesis 50. Further studies on carbohydrate and fat restriction are needed to determine their role in dietary restriction.

Restriction of feeding time

Feeding schedule has also been shown to have a significant impact on health and survival. In flies, time-restricted feeding (limited to 12 daytime hours every day) had profound effects on neural, peripheral, and cardiovascular physiology and improved sleep, body weight maintenance, and delayed signs of cardiac aging, under unchanged caloric intake and activity 53. When mice were given access to food for only 8–9 hours during the active phase of the day, metabolic diseases induced by a high-fat, high-fructose, and high-sucrose diet, were reduced without lowering caloric intake 51. The benefits of time-restricted feeding against such obesogenic diets were proportional to the duration of the fasting each day 52. Ad lib feeding during the weekend did not interfere with the protective effects of time-restricted feeding 52. Notably, the restricted feeding pattern reversed the progression of pre-existing obesity and type II diabetes, suggesting it has the potential to be a clinically relevant and feasible dietary intervention, useful to prevent and treat obesity and metabolic disorders 52. Considering that key metabolic factors, such as 5' AMP-activated protein kinase (AMPK), sirtuins, and protein kinase B (AKT), are regulated by an interplay of circadian rhythm and feeding time 54, 55, dietary schedules should be more carefully studied in the context of dietary restriction.

Conclusion

Dietary interventions that extend healthspan and lifespan have evolved and have become much better characterized since the original caloric restriction experiments performed by McCay in 1935. We now understand that its effects on aging are not simply the result of the reduced amount of calories consumed, but are also determined by diet composition, and can be achieved by periodic interventions which do not require an overall reduction in calorie intake and which can be achieved without a complete lack of food intake during the periodic fasting cycles. Further studies are important to identify even less invasive and more effective dietary interventions that will cause coordinated and beneficial effects on healthspan.

Editorial Note on the Review Process

F1000 Faculty Reviews are commissioned from members of the prestigious F1000 Faculty and are edited as a service to readers. In order to make these reviews as comprehensive and accessible as possible, the referees provide input before publication and only the final, revised version is published. The referees who approved the final version are listed with their names and affiliations but without their reports on earlier versions (any comments will already have been addressed in the published version).

The referees who approved this article are:

  • Rozalyn Anderson, Department of Medicine, University of Wisconsin, Madison, WI, USA

  • Julie Mattison, Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, MD, USA

  • Leanne Redman, Reproductive Endocrinology and Women's Health Lab, Pennington Biomedical Research Center, Baton Rouge, LA, USA

Funding Statement

The author(s) declared that no grants were involved in supporting this work.

[version 1; referees: 3 approved]

References

  • 1. McCay CM, Crowell MF, Maynard LA: The effect of retarded growth upon the length of life span and upon the ultimate body size. 1935. Nutrition. 1989;5(3):155–71; discussion 172. [PubMed] [Google Scholar]; F1000 Recommendation
  • 2. Weindruch R, Walford RL: Dietary restriction in mice beginning at 1 year of age: effect on life-span and spontaneous cancer incidence. Science. 1982;215(4538):1415–8. 10.1126/science.7063854 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 3. Weindruch R, Gottesman SR, Walford RL: Modification of age-related immune decline in mice dietarily restricted from or after midadulthood. Proc Natl Acad Sci U S A. 1982;79(3):898–902. [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 4. Colman RJ, Beasley TM, Kemnitz JW, et al. : Caloric restriction reduces age-related and all-cause mortality in rhesus monkeys. Nat Commun. 2014;5: 3557. 10.1038/ncomms4557 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 5. Kenyon CJ: The genetics of ageing. Nature. 2010;464(7288):504–12. 10.1038/nature08980 [DOI] [PubMed] [Google Scholar]
  • 6. Fabrizio P, Pozza F, Pletcher SD, et al. : Regulation of longevity and stress resistance by Sch9 in yeast. Science. 2001;292(5515):288–90. 10.1126/science.1059497 [DOI] [PubMed] [Google Scholar]
  • 7. Kapahi P, Zid BM, Harper T, et al. : Regulation of lifespan in Drosophila by modulation of genes in the TOR signaling pathway. Curr Biol. 2004;14(10):885–90. 10.1016/j.cub.2004.03.059 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Longo VD: Mutations in signal transduction proteins increase stress resistance and longevity in yeast, nematodes, fruit flies, and mammalian neuronal cells. Neurobiol Aging. 1999;20(5):479–86. 10.1016/S0197-4580(99)00089-5 [DOI] [PubMed] [Google Scholar]
  • 9. Slack C, Alic N, Foley A, et al. : The Ras-Erk-ETS-Signaling Pathway Is a Drug Target for Longevity. Cell. 2015;162(1):72–83. 10.1016/j.cell.2015.06.023 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 10. Fontana L, Partridge L: Promoting health and longevity through diet: from model organisms to humans. Cell. 2015;161(1):106–18. 10.1016/j.cell.2015.02.020 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Longo VD, Finch CE: Evolutionary medicine: from dwarf model systems to healthy centenarians? Science. 2003;299(5611):1342–6. 10.1126/science.1077991 [DOI] [PubMed] [Google Scholar]
  • 12. Fontana L, Partridge L, Longo VD: Extending healthy life span--from yeast to humans. Science. 2010;328(5976):321–6. 10.1126/science.1172539 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Wei M, Fabrizio P, Hu J, et al. : Life span extension by calorie restriction depends on Rim15 and transcription factors downstream of Ras/PKA, Tor, and Sch9. PLoS Genet. 2008;4(1):e13. 10.1371/journal.pgen.0040013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Fabrizio P, Gattazzo C, Battistella L, et al. : Sir2 blocks extreme life-span extension. Cell. 2005;123(4):655–67. 10.1016/j.cell.2005.08.042 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 15. Lin SJ, Defossez PA, Guarente L: Requirement of NAD and SIR2 for life-span extension by calorie restriction in Saccharomyces cerevisiae. Science. 2000;289(5487):2126–8. 10.1126/science.289.5487.2126 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 16. Greer EL, Brunet A: Different dietary restriction regimens extend lifespan by both independent and overlapping genetic pathways in C. elegans. Aging Cell. 2009;8(2):113–27. 10.1111/j.1474-9726.2009.00459.x [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 17. Clancy DJ, Gems D, Hafen E, et al. : Dietary restriction in long-lived dwarf flies. Science. 2002;296(5566):319. 10.1126/science.1069366 [DOI] [PubMed] [Google Scholar]
  • 18. Giannakou ME, Goss M, Partridge L: Role of dFOXO in lifespan extension by dietary restriction in Drosophila melanogaster: not required, but its activity modulates the response. Aging Cell. 2008;7(2):187–98. 10.1111/j.1474-9726.2007.00362.x [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 19. Lee C, Longo VD: Fasting vs dietary restriction in cellular protection and cancer treatment: from model organisms to patients. Oncogene. 2011;30(30):3305–16. 10.1038/onc.2011.91 [DOI] [PubMed] [Google Scholar]
  • 20. Bonkowski MS, Rocha JS, Masternak MM, et al. : Targeted disruption of growth hormone receptor interferes with the beneficial actions of calorie restriction. Proc Natl Acad Sci U S A. 2006;103(20):7901–5. 10.1073/pnas.0600161103 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 21. Westbrook R, Bonkowski MS, Arum O, et al. : Metabolic alterations due to caloric restriction and every other day feeding in normal and growth hormone receptor knockout mice. J Gerontol A Biol Sci Med Sci. 2014;69(1):25–33. 10.1093/gerona/glt080 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 22. Bartke A, Wright JC, Mattison JA, et al. : Extending the lifespan of long-lived mice. Nature. 2001;414(6862):412. 10.1038/35106646 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 23. Mattison JA, Roth GS, Beasley TM, et al. : Impact of caloric restriction on health and survival in rhesus monkeys from the NIA study. Nature. 2012;489(7415):318–21. 10.1038/nature11432 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 24. Colman RJ, Anderson RM, Johnson SC, et al. : Caloric restriction delays disease onset and mortality in rhesus monkeys. Science. 2009;325(5937):201–4. 10.1126/science.1173635 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 25. Swindell WR: Dietary restriction in rats and mice: a meta-analysis and review of the evidence for genotype-dependent effects on lifespan. Ageing Res Rev. 2012;11(2):254–70. 10.1016/j.arr.2011.12.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Liao CY, Rikke BA, Johnson TE, et al. : Genetic variation in the murine lifespan response to dietary restriction: from life extension to life shortening. Aging Cell. 2010;9(1):92–5. 10.1111/j.1474-9726.2009.00533.x [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 27. Ravussin E, Redman LM, Rochon J, et al. : A 2-Year Randomized Controlled Trial of Human Caloric Restriction: Feasibility and Effects on Predictors of Health Span and Longevity. J Gerontol A Biol Sci Med Sci. 2015;70(9):1097–104. 10.1093/gerona/glv057 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 28. Redman LM, Kraus WE, Bhapkar M, et al. : Energy requirements in nonobese men and women: results from CALERIE. Am J Clin Nutr. 2014;99(1):71–8. 10.3945/ajcn.113.065631 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 29. Holloszy JO, Fontana L: Caloric restriction in humans. Exp Gerontol. 2007;42(8):709–12. 10.1016/j.exger.2007.03.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Villareal DT, Fontana L, Weiss EP, et al. : Bone mineral density response to caloric restriction-induced weight loss or exercise-induced weight loss: a randomized controlled trial. Arch Intern Med. 2006;166(22):2502–10. 10.1001/archinte.166.22.2502 [DOI] [PubMed] [Google Scholar]
  • 31. Stewart TM, Bhapkar M, Das S, et al. : Comprehensive Assessment of Long-term Effects of Reducing Intake of Energy Phase 2 (CALERIE Phase 2) screening and recruitment: methods and results. Contemp Clin Trials. 2013;34(1):10–20. 10.1016/j.cct.2012.08.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Shanley DP, Kirkwood TB: Calorie restriction and aging: a life-history analysis. Evolution. 2000;54(3):740–50. 10.1111/j.0014-3820.2000.tb00076.x [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 33. Neafsey PJ: Longevity hormesis. A review. Mech Ageing Dev. 1990;51(1):1–31. 10.1016/0047-6374(90)90158-C [DOI] [PubMed] [Google Scholar]
  • 34. Anson RM, Guo Z, de Cabo R, et al. : Intermittent fasting dissociates beneficial effects of dietary restriction on glucose metabolism and neuronal resistance to injury from calorie intake. Proc Natl Acad Sci U S A. 2003;100(10):6216–20. 10.1073/pnas.1035720100 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 35. Mirzaei H, Suarez JA, Longo VD: Protein and amino acid restriction, aging and disease: from yeast to humans. Trends Endocrinol Metab. 2014;25(11):558–66. 10.1016/j.tem.2014.07.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Grandison RC, Piper MD, Partridge L: Amino-acid imbalance explains extension of lifespan by dietary restriction in Drosophila. Nature. 2009;462(7276):1061–4. 10.1038/nature08619 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 37. Levine ME, Suarez JA, Brandhorst S, et al. : Low protein intake is associated with a major reduction in IGF-1, cancer, and overall mortality in the 65 and younger but not older population. Cell Metab. 2014;19(3):407–17. 10.1016/j.cmet.2014.02.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Solon-Biet SM, McMahon AC, Ballard JW, et al. : The ratio of macronutrients, not caloric intake, dictates cardiometabolic health, aging, and longevity in ad libitum-fed mice. Cell Metab 2014;19(3):418–30. 10.1016/j.cmet.2014.02.009 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 39. Solon-Biet SM, Mitchell SJ, Coogan SC, et al. : Dietary Protein to Carbohydrate Ratio and Caloric Restriction: Comparing Metabolic Outcomes in Mice. Cell Rep. 2015;11(10):1529–34. 10.1016/j.celrep.2015.05.007 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 40. Zimmerman JA, Malloy V, Krajcik R, et al. : Nutritional control of aging. Exp Gerontol. 2003;38(1–2):47–52. 10.1016/S0531-5565(02)00149-3 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 41. Segall P: Long-term tryptophan restriction and aging in the rat. Aktuelle Gerontol. 1977;7(10):535–8. [PubMed] [Google Scholar]
  • 42. De Marte ML, Enesco HE: Influence of low tryptophan diet on survival and organ growth in mice. Mech Ageing Dev. 1986;36(2):161–71. 10.1016/0047-6374(86)90017-5 [DOI] [PubMed] [Google Scholar]
  • 43. Ooka H, Segall PE, Timiras PS: Histology and survival in age-delayed low-tryptophan-fed rats. Mech Ageing Dev. 1988;43(1):79–98. 10.1016/0047-6374(88)90099-1 [DOI] [PubMed] [Google Scholar]
  • 44. Richie JP, Jr, Leutzinger Y, Parthasarathy S, et al. : Methionine restriction increases blood glutathione and longevity in F344 rats. FASEB J. 1994;8(15):1302–7. [DOI] [PubMed] [Google Scholar]
  • 45. Miller RA, Buehner G, Chang Y, et al. : Methionine-deficient diet extends mouse lifespan, slows immune and lens aging, alters glucose, T4, IGF-I and insulin levels, and increases hepatocyte MIF levels and stress resistance. Aging Cell. 2005;4(3):119–25. 10.1111/j.1474-9726.2005.00152.x [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 46. Sun L, Sadighi Akha AA, Miller RA, et al. : Life-span extension in mice by preweaning food restriction and by methionine restriction in middle age. J Gerontol A Biol Sci Med Sci. 2009;64(7):711–22. 10.1093/gerona/glp051 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 47. Malloy VL, Krajcik RA, Bailey SJ, et al. : Methionine restriction decreases visceral fat mass and preserves insulin action in aging male Fischer 344 rats independent of energy restriction. Aging Cell. 2006;5(4):305–14. 10.1111/j.1474-9726.2006.00220.x [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 48. Orentreich N, Matias JR, DeFelice A, et al. : Low methionine ingestion by rats extends life span. J Nutr. 1993;123(2):269–74. [DOI] [PubMed] [Google Scholar]
  • 49. Segall PE, Timiras PS: Patho-physiologic findings after chronic tryptophan deficiency in rats: a model for delayed growth and aging. Mech Ageing Dev. 1976;5(2):109–24. 10.1016/0047-6374(76)90012-9 [DOI] [PubMed] [Google Scholar]
  • 50. Brandhorst S, Choi IY, Wei M, et al. : A Periodic Diet that Mimics Fasting Promotes Multi-System Regeneration, Enhanced Cognitive Performance, and Healthspan. Cell Metab. 2015;22(1):86–99. 10.1016/j.cmet.2015.05.012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Hatori M, Vollmers C, Zarrinpar A, et al. : Time-restricted feeding without reducing caloric intake prevents metabolic diseases in mice fed a high-fat diet. Cell Metab. 2012;15(6):848–60. 10.1016/j.cmet.2012.04.019 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 52. Chaix A, Zarrinpar A, Miu P, et al. : Time-restricted feeding is a preventative and therapeutic intervention against diverse nutritional challenges. Cell Metab. 2014;20(6):991–1005. 10.1016/j.cmet.2014.11.001 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 53. Gill S, Le HD, Melkani GC, et al. : Time-restricted feeding attenuates age-related cardiac decline in Drosophila. Science. 2015;347(6227):1265–9. 10.1126/science.1256682 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 54. Eckel-Mahan K, Sassone-Corsi P: Metabolism and the circadian clock converge. Physiol Rev. 2013;93(1):107–35. 10.1152/physrev.00016.2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Vollmers C, Gill S, DiTacchio L, et al. : Time of feeding and the intrinsic circadian clock drive rhythms in hepatic gene expression. Proc Natl Acad Sci U S A. 2009;106(50):21453–8. 10.1073/pnas.0909591106 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation

Articles from F1000Research are provided here courtesy of F1000 Research Ltd

RESOURCES