mTOR as a central regulator of lifespan and aging
Tóm tắt
Từ khóa
Tài liệu tham khảo
T Niccoli, 2012, Ageing as a risk factor for disease., Curr Biol., 22, R741-52, 10.1016/j.cub.2012.07.024
R Saxton, 2017, mTOR Signaling in Growth, Metabolism, and Disease., Cell., 168, 960-76, 10.1016/j.cell.2017.02.004
C Vézina, 1975, Rapamycin (AY-22,989), a new antifungal antibiotic. I. Taxonomy of the producing streptomycete and isolation of the active principle., J Antibiot (Tokyo)., 28, 721-6, 10.7164/antibiotics.28.721
J Heitman, 1991, Targets for cell cycle arrest by the immunosuppressant rapamycin in yeast., Science., 253, 905-9, 10.1126/science.1715094
E Brown, 1994, A mammalian protein targeted by G1-arresting rapamycin-receptor complex., Nature., 369, 756-8, 10.1038/369756a0
M Bhat, 2015, Targeting the translation machinery in cancer., Nat Rev Drug Discov., 14, 261-78, 10.1038/nrd4505
R Iglesias-Bartolome, 2012, mTOR inhibition prevents epithelial stem cell senescence and protects from radiation-induced mucositis., Cell Stem Cell., 11, 401-14, 10.1016/j.stem.2012.06.007
M Kolesnichenko, 2012, Attenuation of TORC1 signaling delays replicative and oncogenic RAS-induced senescence., Cell Cycle., 11, 2391-401, 10.4161/cc.20683
O Uchenunu, 2019, Oncogenic kinases and perturbations in protein synthesis machinery and energetics in neoplasia., J Mol Endocrinol., 62, R83-R103, 10.1530/JME-18-0058
T Takahara, 2013, Evolutionarily conserved regulation of TOR signalling., J Biochem., 154, 1-10, 10.1093/jb/mvt047
K Hara, 2002, Raptor, a Binding Partner of Target of Rapamycin (TOR), Mediates TOR Action., Cell., 110, 177-89, 10.1016/S0092-8674(02)00833-4
D Kim, 2002, mTOR Interacts with Raptor to Form a Nutrient-Sensitive Complex that Signals to the Cell Growth Machinery., Cell., 110, 163-75, 10.1016/S0092-8674(02)00808-5
D Sarbassov, 2004, Rictor, a novel binding partner of mTOR, defines a rapamycin-insensitive and raptor-independent pathway that regulates the cytoskeleton., Curr Biol., 14, 1296-302, 10.1016/j.cub.2004.06.054
R Loewith, 2002, Two TOR Complexes, Only One of which Is Rapamycin Sensitive, Have Distinct Roles in Cell Growth Control., Mol Cell., 10, 457-68, 10.1016/S1097-2765(02)00636-6
E Caron, 2010, A comprehensive map of the mTOR signaling network., Mol Syst Biol., 6, 453, 10.1038/msb.2010.108
D Kwiatkowski, 2005, Tuberous sclerosis: A GAP at the crossroads of multiple signaling pathways., Hum Mol Genet., 14, R251-8, 10.1093/hmg/ddi260
C Dibble, 2013, Signal integration by mTORC1 coordinates nutrient input with biosynthetic output., Nat Cell Biol., 15, 555-64, 10.1038/ncb2763
K Yamagata, 1994, rheb, a growth factor- and synaptic activity-regulated gene, encodes a novel Ras-related protein., J Biol Chem., 269, 16333-9, 10.1016/S0021-9258(17)34012-7
H Yang, 2017, Mechanisms of mTORC1 activation by RHEB and inhibition by PRAS40., Nature., 552, 368-373, 10.1038/nature25023
P Roux, 2018, Signaling Pathways Involved in the Regulation of mRNA Translation., Mol Cell Biol., 38, 10.1128/MCB.00070-18
S Kang, 2013, mTORC1 phosphorylation sites encode their sensitivity to starvation and rapamycin., Science., 341, 1236566, 10.1126/science.1236566
S Yoon, 2013, Rapamycin Resistance: MTORC1 substrates hold some of the answers., Curr Biol., 23, R880-3, 10.1016/j.cub.2013.08.030
S Li, 2016, Inhibition of mTOR complex 2 induces GSK3/FBXW7-dependent degradation of sterol regulatory element-binding protein 1 (SREBP1) and suppresses lipogenesis in cancer cells., Oncogene., 35, 642-50, 10.1038/onc.2015.123
A Hagiwara, 2012, Hepatic mTORC2 Activates Glycolysis and Lipogenesis through Akt, Glucokinase, and SREBP1c., Cell Metab., 15, 725-38, 10.1016/j.cmet.2012.03.015
K Masui, 2014, mTORC2 in the center of cancer metabolic reprogramming., Trends Endocrinol Metab., 25, 364-73, 10.1016/j.tem.2014.04.002
F Lang, 2016, Regulation of the epithelial Na+ channel by the mTORC2/SGK1 pathway., Nephrol Dial Transplant., 31, 200-5, 10.1093/ndt/gfv270
W Oh, 2011, mTOR complex 2 signaling and functions., Cell Cycle., 10, 2305-16, 10.4161/cc.10.14.16586
N Oshiro, 2004, Dissociation of raptor from mTOR is a mechanism of rapamycin-induced inhibition of mTOR function., Genes Cells., 9, 359-66, 10.1111/j.1356-9597.2004.00727.x
D Sarbassov, 2006, Prolonged Rapamycin Treatment Inhibits mTORC2 Assembly and Akt/PKB., Mol Cell., 22, 159-68, 10.1016/j.molcel.2006.03.029
D Lamming, 2012, Rapamycin-induced insulin resistance is mediated by mTORC2 loss and uncoupled from longevity., Science., 335, 1638-43, 10.1126/science.1215135
K Schreiber, 2015, Rapamycin-mediated mTORC2 inhibition is determined by the relative expression of FK506-binding proteins., Aging Cell., 14, 265-73, 10.1111/acel.12313
A Hinnebusch, 2014, The scanning mechanism of eukaryotic translation initiation., Annu Rev Biochem., 83, 779-812, 10.1146/annurev-biochem-060713-035802
P Fabrizio, 2001, Regulation of longevity and stress resistance by Sch9 in yeast., Science., 292, 288-90, 10.1126/science.1059497
K Jia, 2004, The TOR pathway interacts with the insulin signaling pathway to regulate C. elegans larval development, metabolism and life span., Development., 131, 3897-906, 10.1242/dev.01255
T Vellai, 2003, Genetics: Influence of TOR kinase on lifespan in C. elegans., Nature., 426, 620, 10.1038/426620a
P Kapahi, 2004, Regulation of lifespan in Drosophila by modulation of genes in the TOR signaling pathway., Curr Biol., 14, 885-90, 10.1016/j.cub.2004.03.059
V Anisimov, 2011, Rapamycin increases lifespan and inhibits spontaneous tumorigenesis in inbred female mice., Cell Cycle., 10, 4230-6, 10.4161/cc.10.24.18486
I Bjedov, 2010, Mechanisms of life span extension by rapamycin in the fruit fly Drosophila melanogaster., Cell Metab., 11, 35-46, 10.1016/j.cmet.2009.11.010
D Harrison, 2009, Rapamycin fed late in life extends lifespan in genetically heterogeneous mice., Nature., 460, 392-5, 10.1038/nature08221
O Medvedik, 2007, MSN2 and MSN4 link calorie restriction and TOR to sirtuin-mediated lifespan extension in saccharomyces cerevisiae., PLoS Biol., 5, e261, 10.1371/journal.pbio.0050261
R Miller, 2011, Rapamycin, but not resveratrol or simvastatin, extends life span of genetically heterogeneous mice., J Gerontol A Biol Sci Med Sci., 66, 191-201, 10.1093/gerona/glq178
R Powers, 2006, Extension of chronological life span in yeast by decreased TOR pathway signaling., Genes Dev., 20, 174-84, 10.1101/gad.1381406
S Robida-Stubbs, 2012, TOR signaling and rapamycin influence longevity by regulating SKN-1/Nrf and DAF-16/FoxO., Cell Metab., 15, 713-24, 10.1016/j.cmet.2012.04.007
H Antikainen, 2017, TOR-mediated regulation of metabolism in aging., Aging Cell., 16, 1219-1233, 10.1111/acel.12689
H Pan, 2017, Key proteins and pathways that regulate lifespan., J Biol Chem., 292, 6452-6460, 10.1074/jbc.R116.771915
D Hwangbo, 2004, Drosophila dFOXO controls lifespan and regulates insulin signalling in brain and fat body., Nature., 429, 562-6, 10.1038/nature02549
K Lin, 1997, daf-16: An HNF-3/forkhead family member that can function to double the life-span of Caenorhabditis elegans., Science., 278, 1319-22, 10.1126/science.278.5341.1319
C Selman, 2008, Evidence for lifespan extension and delayed age-related biomarkers in insulin receptor substrate 1 null mice., FASEB J., 22, 807-18, 10.1096/fj.07-9261com
C Selman, 2011, Replication of extended lifespan phenotype in mice with deletion of insulin receptor substrate 1., PLoS One., 6, e16144, 10.1371/journal.pone.0016144
M Tatar, 2001, A mutant Drosophila insulin receptor homolog that extends life-span and impairs neuroendocrine function., Science., 292, 107-10, 10.1126/science.1057987
L Ma, 2005, Phosphorylation and functional inactivation of TSC2 by Erk implications for tuberous sclerosis and cancer pathogenesis., Cell., 121, 179-93, 10.1016/j.cell.2005.02.031
B Manning, 2002, Identification of the tuberous sclerosis complex-2 tumor suppressor gene product tuberin as a target of the phosphoinositide 3-kinase/akt pathway., Mol Cell., 10, 151-62, 10.1016/S1097-2765(02)00568-3
K Inoki, 2002, TSC2 is phosphorylated and inhibited by Akt and suppresses mTOR signalling., Nat Cell Biol., 4, 648-57, 10.1038/ncb839
P Roux, 2004, Tumor-promoting phorbol esters and activated Ras inactivate the tuberous sclerosis tumor suppressor complex via p90 ribosomal S6 kinase., Proc Natl Acad Sci U S A., 101, 13489-94, 10.1073/pnas.0405659101
R Anderson, 2017, Caloric Restriction Research: New Perspectives on the Biology of Aging., J Gerontol A Biol Sci Med Sci., 73, 1-3, 10.1093/gerona/glx212
D Gems, 2013, Genetics of longevity in model organisms: debates and paradigm shifts., Annu Rev Physiol., 75, 621-44, 10.1146/annurev-physiol-030212-183712
J Mattison, 2017, Caloric restriction improves health and survival of rhesus monkeys., Nat Commun., 8, 14063, 10.1038/ncomms14063
C Lee, 2016, Dietary restriction with and without caloric restriction for healthy aging [version 1; peer review: 3 approved]., F1000Res., 5, 10.12688/f1000research.7136.1
M Hansen, 2007, Lifespan extension by conditions that inhibit translation in Caenorhabditis elegans., Aging Cell., 6, 95-110, 10.1111/j.1474-9726.2006.00267.x
M Kaeberlein, 2005, Regulation of yeast replicative life span by TOR and Sch9 in response to nutrients., Science., 310, 1193-6, 10.1126/science.1115535
W Fok, 2014, Short-term rapamycin treatment in mice has few effects on the transcriptome of white adipose tissue compared to dietary restriction., Mech Ageing Dev., 140, 23-9, 10.1016/j.mad.2014.07.004
Z Yu, 2015, Rapamycin and dietary restriction induce metabolically distinctive changes in mouse liver., J Gerontol A Biol Sci Med Sci., 70, 410-20, 10.1093/gerona/glu053
K Choi, 2018, Caloric Restriction and Rapamycin Differentially Alter Energy Metabolism in Yeast., J Gerontol A Biol Sci Med Sci., 73, 29-38, 10.1093/gerona/glx024
C Thoreen, 2009, An ATP-competitive mammalian target of rapamycin inhibitor reveals rapamycin-resistant functions of mTORC1., J Biol Chem., 284, 8023-32, 10.1074/jbc.M900301200
L Hulea, 2018, Translational and HIF-1α-Dependent Metabolic Reprogramming Underpin Metabolic Plasticity and Responses to Kinase Inhibitors and Biguanides., Cell Metab., 28, 817-832.e8, 10.1016/j.cmet.2018.09.001
M Morita, 2013, mTORC1 controls mitochondrial activity and biogenesis through 4E-BP-dependent translational regulation., Cell Metab., 18, 698-711, 10.1016/j.cmet.2013.10.001
M Morita, 2017, mTOR Controls Mitochondrial Dynamics and Cell Survival via MTFP1., Mol Cell., 67, 922-935.e5, 10.1016/j.molcel.2017.08.013
H Mirzaei, 2014, Protein and amino acid restriction, aging and disease: from yeast to humans., Trends Endocrinol Metab., 25, 558-66, 10.1016/j.tem.2014.07.002
S Solon-Biet, 2014, The ratio of macronutrients, not caloric intake, dictates cardiometabolic health, aging, and longevity in ad libitum-fed mice., Cell Metab., 19, 418-30, 10.1016/j.cmet.2014.02.009
M Levine, 2014, 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., 19, 407-17, 10.1016/j.cmet.2014.02.006
L Sun, 2009, Life-span extension in mice by preweaning food restriction and by methionine restriction in middle age., J Gerontol A Biol Sci Med Sci., 64, 711-22, 10.1093/gerona/glp051
P Segall, 1976, Patho-physiologic findings after chronic tryptophan deficiency in rats: a model for delayed growth and aging., Mech Ageing Dev., 5, 109-24, 10.1016/0047-6374(76)90012-9
J Richie, 1994, Methionine restriction increases blood glutathione and longevity in F344 rats., FASEB J., 8, 1302-7, 10.1096/fasebj.8.15.8001743
N Orentreich, 1993, Low methionine ingestion by rats extends life span., J Nutr., 123, 269-74
H Ooka, 1988, Histology and survival in age-delayed low-tryptophan-fed rats., Mech Ageing Dev., 43, 79-98, 10.1016/0047-6374(88)90099-1
R Miller, 2005, 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., 4, 119-25, 10.1111/j.1474-9726.2005.00152.x
V Malloy, 2006, Methionine restriction decreases visceral fat mass and preserves insulin action in aging male Fischer 344 rats independent of energy restriction., Aging Cell., 5, 305-14, 10.1111/j.1474-9726.2006.00220.x
M De Marte, 1986, Influence of low tryptophan diet on survival and organ growth in mice., Mech Ageing Dev., 36, 161-71, 10.1016/0047-6374(86)90017-5
E Lees, 2014, Methionine restriction restores a younger metabolic phenotype in adult mice with alterations in fibroblast growth factor 21., Aging Cell., 13, 817-27, 10.1111/acel.12238
D Yu, 2018, Short-term methionine deprivation improves metabolic health via sexually dimorphic, mTORC1-independent mechanisms., FASEB J., 32, 3471-82, 10.1096/fj.201701211R
N Cummings, 2018, Restoration of metabolic health by decreased consumption of branched-chain amino acids., J Physiol., 596, 623-45, 10.1113/JP275075
L Fontana, 2016, Decreased Consumption of Branched-Chain Amino Acids Improves Metabolic Health., Cell Rep., 16, 520-30, 10.1016/j.celrep.2016.05.092
E Lees, 2017, Direct comparison of methionine restriction with leucine restriction on the metabolic health of C57BL/6J mice., Sci Rep., 7, 9977, 10.1038/s41598-017-10381-3
S Solon-Biet, 2019, Branched-chain amino acids impact health and lifespan indirectly via amino acid balance and appetite control., Nat Metab., 1, 532-45, 10.1038/s42255-019-0059-2
K Khayati, 2017, The amino acid metabolite homocysteine activates mTORC1 to inhibit autophagy and form abnormal proteins in human neurons and mice., FASEB J., 31, 598-609, 10.1096/fj.201600915R
D Lamming, 2015, Restriction of dietary protein decreases mTORC1 in tumors and somatic tissues of a tumor-bearing mouse xenograft model., Oncotarget., 6, 31233-40, 10.18632/oncotarget.5180
D Hardie, 2012, AMPK: a nutrient and energy sensor that maintains energy homeostasis., Nat Rev Mol Cell Biol., 13, 251-62, 10.1038/nrm3311
A Efeyan, 2015, Nutrient-sensing mechanisms and pathways., Nature., 517, 302-10, 10.1038/nature14190
V Albert, 2015, mTOR signaling in cellular and organismal energetics., Curr Opin Cell Biol., 33, 55-66, 10.1016/j.ceb.2014.12.001
E Kim, 2008, Regulation of TORC1 by Rag GTPases in nutrient response., Nat Cell Biol., 10, 935-45, 10.1038/ncb1753
Y Sancak, 2008, The Rag GTPases bind raptor and mediate amino acid signaling to mTORC1., Science., 320, 1496-501, 10.1126/science.1157535
Y Sancak, 2010, Ragulator-Rag complex targets mTORC1 to the lysosomal surface and is necessary for its activation by amino acids., Cell., 141, 290-303, 10.1016/j.cell.2010.02.024
L Bar-Peled, 2012, Ragulator is a GEF for the rag GTPases that signal amino acid levels to mTORC1., Cell., 150, 1196-208, 10.1016/j.cell.2012.07.032
M Fawal, 2015, MCRS1 binds and couples Rheb to amino acid-dependent mTORC1 activation., Dev Cell., 33, 67-81, 10.1016/j.devcel.2015.02.010
L Bar-Peled, 2013, A Tumor suppressor complex with GAP activity for the Rag GTPases that signal amino acid sufficiency to mTORC1., Science., 340, 1100-6, 10.1126/science.1232044
R Wolfson, 2017, KICSTOR recruits GATOR1 to the lysosome and is necessary for nutrients to regulate mTORC1., Nature., 543, 438-42, 10.1038/nature21423
C Petit, 2013, Recruitment of folliculin to lysosomes supports the amino acid-dependent activation of Rag GTPases., J Cell Biol., 202, 1107-22, 10.1083/jcb.201307084
Z Tsun, 2013, The folliculin tumor suppressor is a GAP for the RagC/D GTPases that signal amino acid levels to mTORC1., Mol Cell., 52, 495-505, 10.1016/j.molcel.2013.09.016
M Paquette, 2018, mTOR Pathways in Cancer and Autophagy., Cancers (Basel)., 10, 10.3390/cancers10010018
J Jung, 2015, Amino Acid-Dependent mTORC1 Regulation by the Lysosomal Membrane Protein SLC38A9., Mol Cell Biol., 35, 2479-94, 10.1128/MCB.00125-15
M Rebsamen, 2015, SLC38A9 is a component of the lysosomal amino acid sensing machinery that controls mTORC1., Nature., 519, 477-81, 10.1038/nature14107
S Wang, 2015, Metabolism. Lysosomal amino acid transporter SLC38A9 signals arginine sufficiency to mTORC1., Science., 347, 188-94, 10.1126/science.1257132
G Wyant, 2017, mTORC1 Activator SLC38A9 Is Required to Efflux Essential Amino Acids from Lysosomes and Use Protein as a Nutrient., Cell., 171, 642-654.e12, 10.1016/j.cell.2017.09.046
R Saxton, 2016, Mechanism of arginine sensing by CASTOR1 upstream of mTORC1., Nature., 536, 229-33, 10.1038/nature19079
L Chantranupong, 2016, The CASTOR Proteins Are Arginine Sensors for the mTORC1 Pathway., Cell., 165, 153-64, 10.1016/j.cell.2016.02.035
R Saxton, 2015, Structural basis for leucine sensing by the Sestrin2-mTORC1 pathway., Science., 351, 53-8, 10.1126/science.aad2087
J Kim, 2015, Sestrin2 inhibits mTORC1 through modulation of GATOR complexes., Sci Rep., 5, 9502, 10.1038/srep09502
A Parmigiani, 2014, Sestrins inhibit mTORC1 kinase activation through the GATOR complex., Cell Rep., 9, 1281-91, 10.1016/j.celrep.2014.10.019
R Wolfson, 2016, Sestrin2 is a leucine sensor for the mTORC1 pathway., Science., 351, 43-8, 10.1126/science.aab2674
M Peng, 2014, Sestrins function as guanine nucleotide dissociation inhibitors for Rag GTPases to control mTORC1 signaling., Cell., 159, 122-33, 10.1016/j.cell.2014.08.038
J Han, 2012, Leucyl-tRNA synthetase is an intracellular leucine sensor for the mTORC1-signaling pathway., Cell., 149, 410-24, 10.1016/j.cell.2012.02.044
J Kim, 2017, Control of leucine-dependent mTORC1 pathway through chemical intervention of leucyl-tRNA synthetase and RagD interaction., Nat Commun., 8, 732, 10.1038/s41467-017-00785-0
M Lee, 2018, Coordination of the leucine-sensing Rag GTPase cycle by leucyl-tRNA synthetase in the mTORC1 signaling pathway., Proc Natl Acad Sci U S A., 115, E5279-E5288, 10.1073/pnas.1801287115
M Yoon, 2016, Leucyl-tRNA Synthetase Activates Vps34 in Amino Acid-Sensing mTORC1 Signaling., Cell Rep., 16, 1510-7, 10.1016/j.celrep.2016.07.008
X He, 2018, Sensing and Transmitting Intracellular Amino Acid Signals through Reversible Lysine Aminoacylations., Cell Metab., 27, 151-166.e6, 10.1016/j.cmet.2017.10.015
X Gu, 2017, SAMTOR is an S-adenosylmethionine sensor for the mTORC1 pathway., Science., 358, 813-8, 10.1126/science.aao3265
J Jewell, 2015, Metabolism. Differential regulation of mTORC1 by leucine and glutamine., Science., 347, 194-8, 10.1126/science.1259472
N Quan, 2017, Sestrin2 prevents age-related intolerance to ischemia and reperfusion injury by modulating substrate metabolism., FASEB J., 31, 4153-67, 10.1096/fj.201700063R
N Quan, 2018, Sestrin2 prevents age-related intolerance to post myocardial infarction via AMPK/PGC-1α pathway., J Mol Cell Cardiol., 115, 170-8, 10.1016/j.yjmcc.2018.01.005
C Zhang, 2017, Loss of sestrin 2 potentiates the early onset of age-related sensory cell degeneration in the cochlea., Neuroscience., 361, 179-91, 10.1016/j.neuroscience.2017.08.015
M Lucanic, 2016, Chemical activation of a food deprivation signal extends lifespan., Aging Cell., 15, 832-41, 10.1111/acel.12492
Q Gao, 2008, Neuronal control of energy homeostasis., FEBS Lett., 582, 132-41, 10.1016/j.febslet.2007.11.063
D Cota, 2006, Hypothalamic mTOR signaling regulates food intake., Science., 312, 927-30, 10.1126/science.1124147
C Blouet, 2008, Mediobasal hypothalamic p70 S6 kinase 1 modulates the control of energy homeostasis., Cell Metab., 8, 459-67, 10.1016/j.cmet.2008.10.004
D Cota, 2008, The role of hypothalamic mammalian target of rapamycin complex 1 signaling in diet-induced obesity., J Neurosci., 28, 7202-8, 10.1523/JNEUROSCI.1389-08.2008
M Smith, 2015, Ribosomal S6K1 in POMC and AgRP Neurons Regulates Glucose Homeostasis but Not Feeding Behavior in Mice., Cell Rep., 11, 335-43, 10.1016/j.celrep.2015.03.029
A Caron, 2016, Mediobasal hypothalamic overexpression of DEPTOR protects against high-fat diet-induced obesity., Mol Metab., 5, 102-12, 10.1016/j.molmet.2015.11.005
V Catena, 2017, Deptor: not only a mTOR inhibitor., J Exp Clin Cancer Res., 36, 12, 10.1186/s13046-016-0484-y
L Burke, 2017, mTORC1 in AGRP neurons integrates exteroceptive and interoceptive food-related cues in the modulation of adaptive energy expenditure in mice., eLife., 6, 10.7554/eLife.22848
J Lin, 2005, Metabolic control through the PGC-1 family of transcription coactivators., Cell Metab., 1, 361-70, 10.1016/j.cmet.2005.05.004
F Fisher, 2016, Understanding the Physiology of FGF21., Annu Rev Physiol., 78, 223-41, 10.1146/annurev-physiol-021115-105339
M Cornu, 2014, Hepatic mTORC1 controls locomotor activity, body temperature, and lipid metabolism through FGF21., Proc Natl Acad Sci U S A., 111, 11592-9, 10.1073/pnas.1412047111
P Polak, 2008, Adipose-specific knockout of raptor results in lean mice with enhanced mitochondrial respiration., Cell Metab., 8, 399-410, 10.1016/j.cmet.2008.09.003
H Koga, 2011, Protein homeostasis and aging: The importance of exquisite quality control., Ageing Res Rev., 10, 205-15, 10.1016/j.arr.2010.02.001
E Powers, 2009, Biological and chemical approaches to diseases of proteostasis deficiency., Annu Rev Biochem., 78, 959-91, 10.1146/annurev.biochem.052308.114844
Y Gonskikh, 2017, Alterations of the translation apparatus during aging and stress response., Mech Ageing Dev., 168, 30-6, 10.1016/j.mad.2017.04.003
L Lindqvist, 2018, Cross-talk between protein synthesis, energy metabolism and autophagy in cancer., Curr Opin Genet Dev., 48, 104-11, 10.1016/j.gde.2017.11.003
J Zhao, 2015, mTOR inhibition activates overall protein degradation by the ubiquitin proteasome system as well as by autophagy., Proc Natl Acad Sci U S A., 112, 15790-7, 10.1073/pnas.1521919112
A Rousseau, 2016, An evolutionarily conserved pathway controls proteasome homeostasis., Nature., 536, 184-9, 10.1038/nature18943
Y Zhang, 2014, Coordinated regulation of protein synthesis and degradation by mTORC1., Nature., 513, 440-3, 10.1038/nature13492
C Hetz, 2012, The unfolded protein response: controlling cell fate decisions under ER stress and beyond., Nat Rev Mol Cell Biol., 13, 89-102, 10.1038/nrm3270
M Calfon, 2002, IRE1 couples endoplasmic reticulum load to secretory capacity by processing the XBP-1 mRNA., Nature., 415, 92-6, 10.1038/415092a
K Lee, 2002, IRE1-mediated unconventional mRNA splicing and S2P-mediated ATF6 cleavage merge to regulate XBP1 in signaling the unfolded protein response., Genes Dev., 16, 452-66, 10.1101/gad.964702
H Yoshida, 2001, XBP1 mRNA is induced by ATF6 and spliced by IRE1 in response to ER stress to produce a highly active transcription factor., Cell., 107, 881-91, 10.1016/S0092-8674(01)00611-0
C Hetz, 2015, Proteostasis control by the unfolded protein response., Nat Cell Biol., 17, 829-38, 10.1038/ncb3184
E Bachar, 2009, Glucose amplifies fatty acid-induced endoplasmic reticulum stress in pancreatic beta-cells via activation of mTORC1., PLoS One., 4, e4954, 10.1371/journal.pone.0004954
K Pfaffenbach, 2010, Rapamycin inhibits postprandial-mediated X-box-binding protein-1 splicing in rat liver., J Nutr., 140, 879-84, 10.3945/jn.109.119883
P Walter, 2011, The unfolded protein response: from stress pathway to homeostatic regulation., Science., 334, 1081-6, 10.1126/science.1209038
A Konieczny, 1983, Purification of the eukaryotic initiation factor 2-eukaryotic initiation factor 2B complex and characterization of its guanine nucleotide exchange activity during protein synthesis initiation., J Biol Chem., 258, 3402-8, 10.1016/S0021-9258(18)32875-8
T Krishnamoorthy, 2001, Tight binding of the phosphorylated alpha subunit of initiation factor 2 (eIF2alpha) to the regulatory subunits of guanine nucleotide exchange factor eIF2B is required for inhibition of translation initiation., Mol Cell Biol., 21, 5018-30, 10.1128/MCB.21.15.5018-5030.2001
M Clemens, 1982, Phosphorylation inhibits guanine nucleotide exchange on eukaryotic initiation factor 2., Nature., 296, 93-5, 10.1038/296093a0
T Dever, 1992, Phosphorylation of initiation factor 2 alpha by protein kinase GCN2 mediates gene-specific translational control of GCN4 in yeast., Cell., 68, 585-96, 10.1016/0092-8674(92)90193-G
W B'chir, 2014, Dual role for CHOP in the crosstalk between autophagy and apoptosis to determine cell fate in response to amino acid deprivation., Cell Signal., 26, 1385-91, 10.1016/j.cellsig.2014.03.009
H Harding, 2003, An integrated stress response regulates amino acid metabolism and resistance to oxidative stress., Mol Cell., 11, 619-33, 10.1016/S1097-2765(03)00105-9
J Ye, 2009, ATF4, an ER stress and hypoxia-inducible transcription factor and its potential role in hypoxia tolerance and tumorigenesis., Curr Mol Med., 9, 411-6, 10.2174/156652409788167096
B Guan, 2017, A Unique ISR Program Determines Cellular Responses to Chronic Stress., Mol Cell., 68, 885-900.e6, 10.1016/j.molcel.2017.11.007
D Krokowski, 2013, A self-defeating anabolic program leads to β-cell apoptosis in endoplasmic reticulum stress-induced diabetes via regulation of amino acid flux., J Biol Chem., 288, 17202-13, 10.1074/jbc.M113.466920
B Guan, 2014, Translational control during endoplasmic reticulum stress beyond phosphorylation of the translation initiation factor eIF2α., J Biol Chem., 289, 12593-611, 10.1074/jbc.M113.543215
V Gandin, 2016, mTORC1 and CK2 coordinate ternary and eIF4F complex assembly., Nat Commun., 7, 10.1038/ncomms11127
J Wengrod, 2015, Phosphorylation of eIF2α triggered by mTORC1 inhibition and PP6C activation is required for autophagy and is aberrant in PP6C-mutated melanoma., Sci Signal., 8, ra27, 10.1126/scisignal.aaa0899
J Krishnamoorthy, 2018, Downregulation of PERK activity and eIF2α serine 51 phosphorylation by mTOR complex 1 elicits pro-oxidant and pro-death effects in tuberous sclerosis-deficient cells., Cell Death Dis., 9, 10.1038/s41419-018-0326-2
Y Park, 2017, mTORC1 Balances Cellular Amino Acid Supply with Demand for Protein Synthesis through Post-transcriptional Control of ATF4., Cell Rep., 19, 1083-90, 10.1016/j.celrep.2017.04.042
A Trifunovic, 2004, Premature ageing in mice expressing defective mitochondrial DNA polymerase., Nature., 429, 417-23, 10.1038/nature02517
D Baker, 2017, Biphasic Modeling of Mitochondrial Metabolism Dysregulation during Aging., Trends Biochem Sci., 42, 702-11, 10.1016/j.tibs.2017.06.005
R Dhillon, 2017, Using comparative biology to understand how aging affects mitochondrial metabolism., Mol Cell Endocrinol., 455, 54-61, 10.1016/j.mce.2016.12.020
O Moiseeva, 2009, Mitochondrial dysfunction contributes to oncogene-induced senescence., Mol Cell Biol., 29, 4495-507, 10.1128/MCB.01868-08
V Gandin, 2016, nanoCAGE reveals 5' UTR features that define specific modes of translation of functionally related MTOR-sensitive mRNAs., Genome Res., 26, 636-48, 10.1101/gr.197566.115
J Cunningham, 2007, mTOR controls mitochondrial oxidative function through a YY1-PGC-1alpha transcriptional complex., Nature., 450, 736-40, 10.1038/nature06322
C Betz, 2013, Feature Article: mTOR complex 2-Akt signaling at mitochondria-associated endoplasmic reticulum membranes (MAM) regulates mitochondrial physiology., Proc Natl Acad Sci U S A., 110, 12526-34, 10.1073/pnas.1302455110
I Kim, 2007, Selective degradation of mitochondria by mitophagy., Arch Biochem Biophys., 462, 245-53, 10.1016/j.abb.2007.03.034
D Ryu, 2016, Urolithin A induces mitophagy and prolongs lifespan in C. elegans and increases muscle function in rodents., Nat Med., 22, 879-88, 10.1038/nm.4132
A Schiavi, 2015, Iron-Starvation-Induced Mitophagy Mediates Lifespan Extension upon Mitochondrial Stress in C. elegans., Curr Biol., 25, 1810-22, 10.1016/j.cub.2015.05.059
A Bartolomé, 2017, MTORC1 Regulates both General Autophagy and Mitophagy Induction after Oxidative Phosphorylation Uncoupling., Mol Cell Biol., 37, 10.1128/MCB.00441-17
C Wiley, 2016, From Ancient Pathways to Aging Cells-Connecting Metabolism and Cellular Senescence., Cell Metab., 23, 1013-21, 10.1016/j.cmet.2016.05.010
J Passos, 2010, Feedback between p21 and reactive oxygen production is necessary for cell senescence., Mol Syst Biol., 6, 347, 10.1038/msb.2010.5
J Passos, 2007, Mitochondrial dysfunction accounts for the stochastic heterogeneity in telomere-dependent senescence., PLoS Biol., 5, e110, 10.1371/journal.pbio.0050110
F Rodier, 2009, Persistent DNA damage signalling triggers senescence-associated inflammatory cytokine secretion., Nat Cell Biol., 11, 973-9, 10.1038/ncb1909
P Sapieha, 2018, Cellular Senescence in Postmitotic Cells: Beyond Growth Arrest., Trends Cell Biol., 28, 595-607, 10.1016/j.tcb.2018.03.003
M Serrano, 1997, Oncogenic ras provokes premature cell senescence associated with accumulation of p53 and p16INK4a., Cell., 88, 593-602, 10.1016/S0092-8674(00)81902-9
C Frippiat, 2001, Subcytotoxic H2O2 stress triggers a release of transforming growth factor-beta 1, which induces biomarkers of cellular senescence of human diploid fibroblasts., J Biol Chem., 276, 2531-7, 10.1074/jbc.M006809200
J Karlseder, 2002, Senescence induced by altered telomere state, not telomere loss., Science., 295, 2446-9, 10.1126/science.1069523
F Mallette, 2007, The DNA damage signaling pathway is a critical mediator of oncogene-induced senescence., Genes Dev., 21, 43-8, 10.1101/gad.1487307
F Mallette, 2007, Myc down-regulation as a mechanism to activate the Rb pathway in STAT5A-induced senescence., J Biol Chem., 282, 34938-44, 10.1074/jbc.M707074200
M Demaria, 2014, An essential role for senescent cells in optimal wound healing through secretion of PDGF-AA., Dev Cell., 31, 722-33, 10.1016/j.devcel.2014.11.012
D Baker, 2016, Naturally occurring p16Ink4a-positive cells shorten healthy lifespan., Nature., 530, 184-9, 10.1038/nature16932
D Baker, 2011, Clearance of p16Ink4a-positive senescent cells delays ageing-associated disorders., Nature., 479, 232-6, 10.1038/nature10600
M Milanovic, 2018, Senescence-associated reprogramming promotes cancer stemness., Nature., 553, 96-100, 10.1038/nature25167
M Oubaha, 2016, Senescence-associated secretory phenotype contributes to pathological angiogenesis in retinopathy., Sci Transl Med., 8, 362ra144, 10.1126/scitranslmed.aaf9440
J Campisi, 2013, Aging, Cellular Senescence, and Cancer., Annu Rev Physiol., 75, 685-705, 10.1146/annurev-physiol-030212-183653
J Dörr, 2013, Synthetic lethal metabolic targeting of cellular senescence in cancer therapy., Nature., 501, 421-5, 10.1038/nature12437
M Narita, 2011, Spatial coupling of mTOR and autophagy augments secretory phenotypes., Science., 332, 966-70, 10.1126/science.1205407
R Laberge, 2015, MTOR regulates the pro-tumorigenic senescence-associated secretory phenotype by promoting IL1A translation., Nat Cell Biol., 17, 1049-61, 10.1038/ncb3195
N Herranz, 2015, mTOR regulates MAPKAPK2 translation to control the senescence-associated secretory phenotype., Nat Cell Biol., 17, 1205-17, 10.1038/ncb3225
C Correia-Melo, 2016, Mitochondria are required for pro-ageing features of the senescent phenotype., EMBO J., 35, 724-42, 10.15252/embj.201592862
B Carroll, 2017, Persistent mTORC1 signaling in cell senescence results from defects in amino acid and growth factor sensing., J Cell Biol., 216, 1949-57, 10.1083/jcb.201610113
M Ermolaeva, 2018, Cellular and epigenetic drivers of stem cell ageing., Nat Rev Mol Cell Biol., 19, 594-610, 10.1038/s41580-018-0020-3
P Sampath, 2008, A hierarchical network controls protein translation during murine embryonic stem cell self-renewal and differentiation., Cell Stem Cell., 2, 448-60, 10.1016/j.stem.2008.03.013
M Schultz, 2016, When stem cells grow old: phenotypes and mechanisms of stem cell aging., Development., 143, 3-14, 10.1242/dev.130633
C Chen, 2009, mTOR regulation and therapeutic rejuvenation of aging hematopoietic stem cells., Sci Signal., 2, ra75, 10.1126/scisignal.2000559
O Yilmaz, 2012, mTORC1 in the Paneth cell niche couples intestinal stem-cell function to calorie intake., Nature., 486, 490-5, 10.1038/nature11163
J Oh, 2014, Stem cell aging: Mechanisms, regulators and therapeutic opportunities., Nat Med., 20, 870-80, 10.1038/nm.3651
M Igarashi, 2016, mTORC1 and SIRT1 Cooperate to Foster Expansion of Gut Adult Stem Cells during Calorie Restriction., Cell., 166, 436-50, 10.1016/j.cell.2016.05.044
J Romine, 2015, The proliferation of amplifying neural progenitor cells is impaired in the aging brain and restored by the mTOR pathway activation., Neurobiol Aging., 36, 1716-26, 10.1016/j.neurobiolaging.2015.01.003
K Takayama, 2017, mTOR signaling plays a critical role in the defects observed in muscle-derived stem/progenitor cells isolated from a murine model of accelerated aging., J Orthop Res., 35, 1375-82, 10.1002/jor.23409
C Selman, 2016, Evidence that hematopoietic stem cell function is preserved during aging in long-lived S6K1 mutant mice., Oncotarget., 7, 29937-43, 10.18632/oncotarget.8729
S Sciarretta, 2018, New Insights Into the Role of mTOR Signaling in the Cardiovascular System., Circ Res., 122, 489-505, 10.1161/CIRCRESAHA.117.311147
P Shende, 2011, Cardiac raptor ablation impairs adaptive hypertrophy, alters metabolic gene expression, and causes heart failure in mice., Circulation., 123, 1073-82, 10.1161/CIRCULATIONAHA.110.977066
D Zhang, 2010, MTORC1 regulates cardiac function and myocyte survival through 4E-BP1 inhibition in mice., J Clin Invest., 120, 2805-16, 10.1172/JCI43008
T Tamai, 2013, Rheb (Ras homologue enriched in brain)-dependent mammalian target of rapamycin complex 1 (mTORC1) activation becomes indispensable for cardiac hypertrophic growth after early postnatal period., J Biol Chem., 288, 10176-87, 10.1074/jbc.M112.423640
Y Zhu, 2013, Mechanistic target of rapamycin (Mtor) is essential for murine embryonic heart development and growth., PLoS One., 8, e54221, 10.1371/journal.pone.0054221
L Mazelin, 2016, mTOR inactivation in myocardium from infant mice rapidly leads to dilated cardiomyopathy due to translation defects and p53/JNK-mediated apoptosis., J Mol Cell Cardiol., 97, 213-25, 10.1016/j.yjmcc.2016.04.011
J Flynn, 2013, Late-life rapamycin treatment reverses age-related heart dysfunction., Aging Cell., 12, 851-62, 10.1111/acel.12109
D Dai, 2014, Altered proteome turnover and remodeling by short-term caloric restriction or rapamycin rejuvenate the aging heart., Aging Cell., 13, 529-39, 10.1111/acel.12203
M Laplante, 2012, mTOR signaling in growth control and disease., Cell., 149, 274-93, 10.1016/j.cell.2012.03.017
C Hoeffer, 2010, mTOR signaling: At the crossroads of plasticity, memory and disease., Trends Neurosci., 33, 67-75, 10.1016/j.tins.2009.11.003
C Garza-Lombó, 2016, Mammalian Target of Rapamycin: Its Role in Early Neural Development and in Adult and Aged Brain Function., Front Cell Neurosci., 10, 157, 10.3389/fncel.2016.00157
M Perluigi, 2015, mTOR signaling in aging and neurodegeneration: At the crossroad between metabolism dysfunction and impairment of autophagy., Neurobiol Dis., 84, 39-49, 10.1016/j.nbd.2015.03.014
J Triplett, 2015, Age-related changes in the proteostasis network in the brain of the naked mole-rat: Implications promoting healthy longevity., Biochim Biophys Acta., 1852, 2213-24, 10.1016/j.bbadis.2015.08.002
A Caccamo, 2010, Molecular interplay between mammalian target of rapamycin (mTOR), amyloid-beta, and Tau: Effects on cognitive impairments., J Biol Chem., 285, 13107-20, 10.1074/jbc.M110.100420
P Spilman, 2010, Inhibition of mTOR by rapamycin abolishes cognitive deficits and reduces amyloid-beta levels in a mouse model of Alzheimer's disease., PLoS One., 5, e9979, 10.1371/journal.pone.0009979
A Lin, 2013, Chronic rapamycin restores brain vascular integrity and function through NO synthase activation and improves memory in symptomatic mice modeling Alzheimer's disease., J Cereb Blood Flow Metab., 33, 1412-21, 10.1038/jcbfm.2013.82
S Majumder, 2011, Inducing Autophagy by Rapamycin Before, but Not After, the Formation of Plaques and Tangles Ameliorates Cognitive Deficits., PLoS One., 6, e25416, 10.1371/journal.pone.0025416
A Tramutola, 2018, Intranasal rapamycin ameliorates Alzheimer-like cognitive decline in a mouse model of Down syndrome., Transl Neurodegener., 7, 28, 10.1186/s40035-018-0133-9
U Lang, 2009, Immunosuppression using the mammalian target of rapamycin (mTOR) inhibitor everolimus: pilot study shows significant cognitive and affective improvement., Transplant Proc., 41, 4285-8, 10.1016/j.transproceed.2009.08.050
A Ghaben, 2019, Adipogenesis and metabolic health., Nat Rev Mol Cell Biol., 20, 242-58, 10.1038/s41580-018-0093-z
E Chouchani, 2019, New Advances in Adaptive Thermogenesis: UCP1 and Beyond., Cell Metab., 29, 27-37, 10.1016/j.cmet.2018.11.002
L Kazak, 2017, Genetic Depletion of Adipocyte Creatine Metabolism Inhibits Diet-Induced Thermogenesis and Drives Obesity., Cell Metab., 26, 660-671.e3, 10.1016/j.cmet.2017.08.009
N Rothwell, 1979, A role for brown adipose tissue in diet-induced thermogenesis., Nature., 281, 31-5, 10.1038/281031a0
L Kazak, 2019, Ablation of adipocyte creatine transport impairs thermogenesis and causes diet-induced obesity., Nat Metab., 1, 360-370, 10.1038/s42255-019-0035-x
D Fawcett, 1947, Differences in Physiological Activity in Brown and White Fat as Revealed by Histochemical Reactions., Science., 105, 123, 10.1126/science.105.2718.123
J Wu, 2012, Beige adipocytes are a distinct type of thermogenic fat cell in mouse and human., Cell., 150, 366-76, 10.1016/j.cell.2012.05.016
L Gonçalves, 2017, Ageing is associated with brown adipose tissue remodelling and loss of white fat browning in female C57BL/6 mice., Int J Exp Pathol., 98, 100-108, 10.1111/iep.12228
D Vatner, 2018, Enhanced longevity and metabolism by brown adipose tissue with disruption of the regulator of G protein signaling 14., Aging Cell., 17, e12751, 10.1111/acel.12751
X Xiang, 2015, Tuberous Sclerosis Complex 1–Mechanistic Target of Rapamycin Complex 1 Signaling Determines Brown-to-White Adipocyte Phenotypic Switch., Diabetes., 64, 519-28, 10.2337/db14-0427
X Zhang, 2018, Adipose mTORC1 Suppresses Prostaglandin Signaling and Beige Adipogenesis via the CRTC2-COX-2 Pathway., Cell Rep., 24, 3180-3193, 10.1016/j.celrep.2018.08.055
M Laplante, 2012, DEPTOR cell-autonomously promotes adipogenesis, and its expression is associated with obesity., Cell Metab., 16, 202-12, 10.1016/j.cmet.2012.07.008
J Frey, 2014, A role for Raptor phosphorylation in the mechanical activation of mTOR signaling., Cell Signal., 26, 313-22, 10.1016/j.cellsig.2013.11.009
H Tang, 2019, mTORC1 underlies age-related muscle fiber damage and loss by inducing oxidative stress and catabolism., Aging Cell., 18, e12943, 10.1111/acel.12943
E Baar, 2016, Sex- and tissue-specific changes in mTOR signaling with age in C57BL/6J mice., Aging Cell., 15, 155-66, 10.1111/acel.12425
C Calhoun, 2016, Senescent Cells Contribute to the Physiological Remodeling of Aged Lungs., J Gerontol A Biol Sci Med Sci., 71, 153-60, 10.1093/gerona/glu241
L Harries, 2012, Advancing age is associated with gene expression changes resembling mTOR inhibition: Evidence from two human populations., Mech Ageing Dev., 133, 556-62, 10.1016/j.mad.2012.07.003
O Leontieva, 2014, Fasting levels of hepatic p-S6 are increased in old mice., Cell Cycle., 13, 2656-9, 10.4161/15384101.2014.949150
N Linford, 2007, Transcriptional response to aging and caloric restriction in heart and adipose tissue., Aging Cell., 6, 673-88, 10.1111/j.1474-9726.2007.00319.x
S Sengupta, 2010, mTORC1 controls fasting-induced ketogenesis and its modulation by ageing., Nature., 468, 1100-4, 10.1038/nature09584
K Xie, 2018, Epigenetic alterations in longevity regulators, reduced life span, and exacerbated aging-related pathology in old father offspring mice., Proc Natl Acad Sci U S A., 115, E2348-E2357, 10.1073/pnas.1707337115
S Yang, 2012, Rapamycin ameliorates age-dependent obesity associated with increased mTOR signaling in hypothalamic POMC neurons., Neuron., 75, 425-36, 10.1016/j.neuron.2012.03.043
M Blagosklonny, 2010, Revisiting the antagonistic pleiotropy theory of aging: TOR-driven program and quasi-program., Cell Cycle., 9, 3151-6, 10.4161/cc.9.16.13120
S Murphy, 2013, Deaths: Final data for 2010., Natl Vital Stat Rep., 61, 1-117
2016, Health, United States, 2015: With Special Feature on Racial and Ethnic Health Disparities
J Mannick, 2014, mTOR inhibition improves immune function in the elderly., Sci Transl Med., 6, 268ra179, 10.1126/scitranslmed.3009892
C Lages, 2010, Partial restoration of T-cell function in aged mice by in vitro blockade of the PD-1/PD-L1 pathway., Aging Cell., 9, 785-98, 10.1111/j.1474-9726.2010.00611.x
E Kraig, 2018, A randomized control trial to establish the feasibility and safety of rapamycin treatment in an older human cohort: Immunological, physical performance, and cognitive effects., Exp Gerontol., 105, 53-69, 10.1016/j.exger.2017.12.026
B Zid, 2009, 4E-BP extends lifespan upon dietary restriction by enhancing mitochondrial activity in Drosophila., Cell., 139, 149-60, 10.1016/j.cell.2009.07.034
G Carvalho, 2017, The 4E-BP growth pathway regulates the effect of ambient temperature on Drosophila metabolism and lifespan., Proc Natl Acad Sci U S A., 114, 9737-42, 10.1073/pnas.1618994114
S Tsai, 2016, Increased 4E-BP1 Expression Protects against Diet-Induced Obesity and Insulin Resistance in Male Mice., Cell Rep., 16, 1903-14, 10.1016/j.celrep.2016.07.029
O Le Bacquer, 2007, Elevated sensitivity to diet-induced obesity and insulin resistance in mice lacking 4E-BP1 and 4E-BP2., J Clin Invest., 117, 387-96, 10.1172/JCI29528
Q Fan, 2017, A Kinase Inhibitor Targeted to mTORC1 Drives Regression in Glioblastoma., Cancer Cell., 31, 424-35, 10.1016/j.ccell.2017.01.014
D Lamming, 2014, Depletion of Rictor, an essential protein component of mTORC2, decreases male lifespan., Aging Cell., 13, 911-7, 10.1111/acel.12256
A Soukas, 2009, Rictor/TORC2 regulates fat metabolism, feeding, growth, and life span in Caenorhabditis elegans., Genes Dev., 23, 496-511, 10.1101/gad.1775409
J Mannick, 2018, TORC1 inhibition enhances immune function and reduces infections in the elderly., Sci Transl Med., 10, 10.1126/scitranslmed.aaq1564
A Efeyan, 2014, RagA, but Not RagB, Is Essential for Embryonic Development and Adult Mice., Dev Cell., 29, 321-9, 10.1016/j.devcel.2014.03.017
T Hoshii, 2012, mTORC1 is essential for leukemia propagation but not stem cell self-renewal., J Clin Invest., 122, 2114-29, 10.1172/JCI62279
S Arriola Apelo, 2016, Alternative rapamycin treatment regimens mitigate the impact of rapamycin on glucose homeostasis and the immune system., Aging Cell., 15, 28-38, 10.1111/acel.12405
S Arriola Apelo, 2016, Intermittent Administration of Rapamycin Extends the Life Span of Female C57BL/6J Mice., J Gerontol A Biol Sci Med Sci., 71, 876-81, 10.1093/gerona/glw064
N Barzilai, 2016, Metformin as a Tool to Target Aging., Cell Metab., 23, 1060-5, 10.1016/j.cmet.2016.05.011
Y Kuan, 2017, Effects of metformin exposure on neurodegenerative diseases in elderly patients with type 2 diabetes mellitus., Prog Neuropsychopharmacol Biol Psychiatry., 79, 77-83, 10.1016/j.pnpbp.2017.06.002
M Pollak, 2012, Investigating metformin for cancer prevention and treatment: the end of the beginning., Cancer Discov., 2, 778-90, 10.1158/2159-8290.CD-12-0263
R Dowling, 2011, Understanding the benefit of metformin use in cancer treatment., BMC Med., 9, 33, 10.1186/1741-7015-9-33