Is Rapamycin a Dietary Restriction Mimetic?

Abstract Since the initial suggestion that rapamycin, an inhibitor of target of rapamycin (TOR) nutrient signaling, increased lifespan comparable to dietary restriction, investigators have viewed rapamycin as a potential dietary restriction mimetic. Both dietary restriction and rapamycin increase lifespan across a wide range of evolutionarily diverse species (including yeast, Caenorhabditis elegans, Drosophila, and mice) as well as reducing pathology and improving physiological functions that decline with age in mice. The purpose of this article is to review the research comparing the effect of dietary restriction and rapamycin in mice. The current data show that dietary restriction and rapamycin have different effects on many pathways and molecular processes. In addition, these interventions affect the lifespan of many genetically manipulated mouse models differently. In other words, while dietary restriction and rapamycin may have similar effects on some pathways and processes; overall, they affect many pathways/processes quite differently. Therefore, rapamycin is likely not a true dietary restriction mimetic. Rather dietary restriction and rapamycin appear to be increasing lifespan and retarding aging largely through different mechanisms/pathways, suggesting that a combination of dietary restriction and rapamycin will have a greater effect on lifespan than either manipulation alone.

[1]  R. Weiss,et al.  Induction of fat apoptosis by a non‐thermal device: Mechanism of action of non‐invasive high‐intensity electromagnetic technology in a porcine model , 2018, Lasers in surgery and medicine.

[2]  S. Subramanian,et al.  Chronic oral rapamycin decreases adiposity, hepatic triglycerides and insulin resistance in male mice fed a diet high in sucrose and saturated fat , 2018, Experimental physiology.

[3]  Jie Chen,et al.  Effects of rapamycin on growth hormone receptor knockout mice , 2018, Proceedings of the National Academy of Sciences.

[4]  Anindita Das,et al.  Chronic treatment with novel nanoformulated micelles of rapamycin, Rapatar, protects diabetic heart against ischaemia/reperfusion injury , 2017, British journal of pharmacology.

[5]  John J. Cole,et al.  Diverse interventions that extend mouse lifespan suppress shared age-associated epigenetic changes at critical gene regulatory regions , 2017, Genome Biology.

[6]  Peter D. Adams,et al.  Epigenetic aging signatures in mice livers are slowed by dwarfism, calorie restriction and rapamycin treatment , 2017, Genome Biology.

[7]  D. Seals,et al.  Dietary rapamycin supplementation reverses age‐related vascular dysfunction and oxidative stress, while modulating nutrient‐sensing, cell cycle, and senescence pathways , 2016, Aging cell.

[8]  K. Kim,et al.  Caloric Restriction and Rapamycin Differentially Alter Energy Metabolism in Yeast , 2017, The journals of gerontology. Series A, Biological sciences and medical sciences.

[9]  D. Harrison,et al.  Rapamycin treatment benefits glucose metabolism in mouse models of type 2 diabetes , 2016, Aging.

[10]  A. Young,et al.  Prolonged Calorie Restriction Downregulates Skeletal Muscle mTORC1 Signaling Independent of Dietary Protein Intake and Associated microRNA Expression , 2016, Front. Physiol..

[11]  Kathleen F. Kerr,et al.  Transient rapamycin treatment can increase lifespan and healthspan in middle-aged mice , 2016, eLife.

[12]  S. A. Arriola Apelo,et al.  Rapamycin: An InhibiTOR of Aging Emerges From the Soil of Easter Island. , 2016, The journals of gerontology. Series A, Biological sciences and medical sciences.

[13]  D. Féliers,et al.  Rapamycin Increases Mortality in db/db Mice, a Mouse Model of Type 2 Diabetes. , 2016, The journals of gerontology. Series A, Biological sciences and medical sciences.

[14]  L. Ferrucci,et al.  Effects of Sex, Strain, and Energy Intake on Hallmarks of Aging in Mice. , 2016, Cell metabolism.

[15]  Dudley Lamming,et al.  Longer lifespan in male mice treated with a weakly estrogenic agonist, an antioxidant, an α‐glucosidase inhibitor or a Nrf2‐inducer , 2016, Aging cell.

[16]  Dudley Lamming Inhibition of the Mechanistic Target of Rapamycin (mTOR)-Rapamycin and Beyond. , 2016, Cold Spring Harbor perspectives in medicine.

[17]  R. Kondratov,et al.  Circadian clocks govern calorie restriction—mediated life span extension through BMAL1‐ and IGF‐1‐dependent mechanisms , 2016, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[18]  E. Ravussin,et al.  Effects of 2‐year calorie restriction on circulating levels of IGF‐1, IGF‐binding proteins and cortisol in nonobese men and women: a randomized clinical trial , 2015, Aging cell.

[19]  A. Richardson,et al.  How longevity research can lead to therapies for Alzheimer's disease: The rapamycin story , 2015, Experimental Gerontology.

[20]  P. Rabinovitch,et al.  Dose-dependent effects of mTOR inhibition on weight and mitochondrial disease in mice , 2015, Front. Genet..

[21]  Yun Li,et al.  Effect of caloric restriction on the SIRT1/mTOR signaling pathways in senile mice , 2015, Brain Research Bulletin.

[22]  C. Livi,et al.  p53 and rapamycin are additive , 2015, Oncotarget.

[23]  W. Fok,et al.  Rapamycin and dietary restriction induce metabolically distinctive changes in mouse liver. , 2015, The journals of gerontology. Series A, Biological sciences and medical sciences.

[24]  Nolan G. Ericson,et al.  Subacute calorie restriction and rapamycin discordantly alter mouse liver proteome homeostasis and reverse aging effects , 2015, Aging cell.

[25]  B. Phillips-Farfán,et al.  Caloric restriction protects against electrical kindling of the amygdala by inhibiting the mTOR signaling pathway , 2015, Front. Cell. Neurosci..

[26]  Dudley Lamming,et al.  Rapamycin-induced metabolic defects are reversible in both lean and obese mice , 2014, Aging.

[27]  C. Livi,et al.  Short-term rapamycin treatment in mice has few effects on the transcriptome of white adipose tissue compared to dietary restriction , 2014, Mechanisms of Ageing and Development.

[28]  M. Antoch,et al.  Metabolic clock generates nutrient anticipation rhythms in mTOR signaling , 2014, Aging.

[29]  B. Vollmar,et al.  Lifelong caloric restriction reprograms hepatic fat metabolism in mice. , 2014, The journals of gerontology. Series A, Biological sciences and medical sciences.

[30]  M. Mirisola,et al.  Calorie Restriction in Mammals and Simple Model Organisms , 2014, BioMed research international.

[31]  Z. D. Sharp,et al.  Rapamycin-mediated lifespan increase in mice is dose and sex dependent and metabolically distinct from dietary restriction , 2014, Aging cell.

[32]  S. Austad,et al.  Rapamycin extends life and health in C57BL/6 mice. , 2014, The journals of gerontology. Series A, Biological sciences and medical sciences.

[33]  W. Wood,et al.  Mice Fed Rapamycin Have an Increase in Lifespan Associated with Major Changes in the Liver Transcriptome , 2014, PloS one.

[34]  W. Wood,et al.  Combined treatment of rapamycin and dietary restriction has a larger effect on the transcriptome and metabolome of liver , 2013, Aging cell.

[35]  Yimin Fang,et al.  Growth hormone-releasing hormone disruption extends lifespan and regulates response to caloric restriction in mice , 2013, eLife.

[36]  A. Richardson Rapamycin, anti-aging, and avoiding the fate of Tithonus. , 2013, The Journal of clinical investigation.

[37]  Dudley Lamming,et al.  Young and old genetically heterogeneous HET3 mice on a rapamycin diet are glucose intolerant but insulin sensitive , 2013, Aging cell.

[38]  G. Enikolopov,et al.  Calorie restriction alleviates the age‐related decrease in neural progenitor cell division in the aging brain , 2013, The European journal of neuroscience.

[39]  Nektarios Tavernarakis,et al.  Metabolic Control by Target of Rapamycin and Autophagy during Ageing - A Mini-Review , 2013, Gerontology.

[40]  Jie Chen,et al.  Duration of rapamycin treatment has differential effects on metabolism in mice. , 2013, Cell metabolism.

[41]  M. Kaeberlein Faculty Opinions recommendation of The effect of retarded growth upon the length of life span and upon the ultimate body size. 1935. , 2013 .

[42]  C. Livi,et al.  Rapamycin extends life span of Rb1+/− mice by inhibiting neuroendocrine tumors , 2013, Aging.

[43]  P. Rabinovitch,et al.  mTOR is a key modulator of ageing and age-related disease , 2013, Nature.

[44]  Dean P. Jones,et al.  Short-Term Treatment With Rapamycin and Dietary Restriction Have Overlapping and Distinctive Effects in Young Mice , 2012, The journals of gerontology. Series A, Biological sciences and medical sciences.

[45]  S. Kliewer,et al.  The starvation hormone, fibroblast growth factor-21, extends lifespan in mice , 2012, eLife.

[46]  M. Antoch,et al.  Rapamycin extends lifespan and delays tumorigenesis in heterozygous p53+/− mice , 2012, Aging.

[47]  M. Antoch,et al.  New nanoformulation of rapamycin Rapatar extends lifespan in homozygous p53−/− mice by delaying carcinogenesis , 2012, Aging.

[48]  T. von Zglinicki,et al.  Male mice retain a metabolic memory of improved glucose tolerance induced during adult onset, short-term dietary restriction , 2012, Longevity & healthspan.

[49]  C. Selman,et al.  Evidence of a metabolic memory to early-life dietary restriction in male C57BL/6 mice , 2012, Longevity & healthspan.

[50]  Dudley Lamming,et al.  Rapamycin has a biphasic effect on insulin sensitivity in C2C12 myotubes due to sequential disruption of mTORC1 and mTORC2 , 2012, Front. Gene..

[51]  F. Muller,et al.  Dietary restriction but not rapamycin extends disease onset and survival of the H46R/H48Q mouse model of ALS , 2012, Neurobiology of Aging.

[52]  Dudley Lamming,et al.  mTORC1 in the Paneth cell niche couples intestinal stem cell function to calorie intake , 2012, Nature.

[53]  Dudley Lamming,et al.  Rapamycin-Induced Insulin Resistance Is Mediated by mTORC2 Loss and Uncoupled from Longevity , 2012, Science.

[54]  S. Kliewer,et al.  Endocrine fibroblast growth factors 15/19 and 21: from feast to famine. , 2012, Genes & development.

[55]  Y. Jan,et al.  Rapamycin induces glucose intolerance in mice by reducing islet mass, insulin content, and insulin sensitivity , 2011, Journal of Molecular Medicine.

[56]  J. Grande,et al.  Effects of Intermittent and Chronic Calorie Restriction on Mammalian Target of Rapamycin (mTOR) and IGF-I Signaling Pathways in Mammary Fat Pad Tissues and Mammary Tumors , 2011, Nutrition and Cancer.

[57]  R. de Cabo,et al.  Rapamycin, but not resveratrol or simvastatin, extends life span of genetically heterogeneous mice. , 2011, The journals of gerontology. Series A, Biological sciences and medical sciences.

[58]  K. Flurkey,et al.  Life extension by diet restriction and N-acetyl-L-cysteine in genetically heterogeneous mice. , 2010, The journals of gerontology. Series A, Biological sciences and medical sciences.

[59]  L. Partridge,et al.  Mechanisms of Life Span Extension by Rapamycin in the Fruit Fly Drosophila melanogaster , 2010, Cell metabolism.

[60]  H. Hsu,et al.  Long-term administration of rapamycin reduces adiposity, but impairs glucose tolerance in high-fat diet-fed KK/HlJ mice. , 2009, Basic & clinical pharmacology & toxicology.

[61]  B. Kennedy,et al.  Ageing: A midlife longevity drug? , 2009, Nature.

[62]  Marco Pahor,et al.  Rapamycin fed late in life extends lifespan in genetically heterogeneous mice , 2009, Nature.

[63]  V. Monnier,et al.  Plasma glucose and the action of calorie restriction on aging. , 2007, The journals of gerontology. Series A, Biological sciences and medical sciences.

[64]  C. Álvarez,et al.  Effect of age and moderate food restriction on insulin sensitivity in Wistar rats: role of adiposity. , 2007, The Journal of endocrinology.

[65]  Seung-Jae V. Lee,et al.  Lifespan extension by conditions that inhibit translation in Caenorhabditis elegans , 2007, Aging cell.

[66]  M. Antoch,et al.  Early aging and age-related pathologies in mice deficient in BMAL1, the core componentof the circadian clock. , 2006, Genes & development.

[67]  K. Coschigano Aging-related characteristics of growth hormone receptor/binding protein gene-disrupted mice , 2006, AGE.

[68]  E. Ravussin,et al.  Effect of calorie restriction with or without exercise on insulin sensitivity, beta-cell function, fat cell size, and ectopic lipid in overweight subjects. , 2006, Diabetes care.

[69]  Andrzej Bartke,et al.  Targeted disruption of growth hormone receptor interferes with the beneficial actions of calorie restriction , 2006, Proceedings of the National Academy of Sciences of the United States of America.

[70]  Matt Kaeberlein,et al.  Regulation of Yeast Replicative Life Span by TOR and Sch9 in Response to Nutrients , 2005, Science.

[71]  Z. D. Sharp,et al.  Evidence for down-regulation of phosphoinositide 3-kinase/Akt/mammalian target of rapamycin (PI3K/Akt/mTOR)-dependent translation regulatory signaling pathways in Ames dwarf mice. , 2005, The journals of gerontology. Series A, Biological sciences and medical sciences.

[72]  S. Benzer,et al.  Regulation of Lifespan in Drosophila by Modulation of Genes in the TOR Signaling Pathway , 2004, Current Biology.

[73]  Tibor Vellai,et al.  Genetics: Influence of TOR kinase on lifespan in C. elegans , 2003, Nature.

[74]  S. Del Prato,et al.  Reducing insulin resistance with metformin: the evidence today. , 2003, Diabetes & metabolism.

[75]  Wen-Hwa Lee,et al.  Minimal effects of dietary restriction on neuroendocrine carcinogenesis in Rb+/- mice. , 2003, Carcinogenesis.

[76]  Andrzej Bartke,et al.  Longevity: Extending the lifespan of long-lived mice , 2001, Nature.

[77]  D. Allison,et al.  Dietary restriction and glucose regulation in aging rhesus monkeys: a follow-up report at 8.5 yr. , 2001, American journal of physiology. Endocrinology and metabolism.

[78]  F. Dominici,et al.  Compensatory alterations of insulin signal transduction in liver of growth hormone receptor knockout mice. , 2000, The Journal of endocrinology.

[79]  S. Bennett,et al.  Pleiotropic effects of growth hormone and insulin-like growth factor (IGF)-1 on biological aging: inferences from moderate caloric-restricted animals. , 1999, The journals of gerontology. Series A, Biological sciences and medical sciences.

[80]  B. Hass,et al.  Growth curves and survival characteristics of the animals used in the Biomarkers of Aging Program. , 1999, The journals of gerontology. Series A, Biological sciences and medical sciences.

[81]  A. Gingras,et al.  Rapamycin blocks the phosphorylation of 4E‐BP1 and inhibits cap‐dependent initiation of translation. , 1996, The EMBO journal.

[82]  I. Stansfield,et al.  An MBoC Favorite: TOR controls translation initiation and early G1 progression in yeast , 2012, Molecular biology of the cell.

[83]  S. Perkins,et al.  Calorie restriction delays spontaneous tumorigenesis in p53-knockout transgenic mice. , 1994, Proceedings of the National Academy of Sciences of the United States of America.

[84]  C. McMahan,et al.  Dietary restriction alters characteristics of glucose fuel use. , 1992, Journal of gerontology.

[85]  L. Groop,et al.  Weight loss by very-low-calorie diets: effects on substrate oxidation, energy expenditure, and insulin sensitivity in obese subjects. , 1992, The American journal of clinical nutrition.

[86]  C M McCay,et al.  The effect of retarded growth upon the length of life span and upon the ultimate body size. 1935. , 1935, Nutrition.

[87]  D. Harrison,et al.  Effects of food restriction on aging: separation of food intake and adiposity. , 1984, Proceedings of the National Academy of Sciences of the United States of America.