mTOR: A Cellular Regulator Interface in Health and Disease

Cells - Tập 8 Số 1 - Trang 18
Fahd Boutouja1, Christian M. Stiehm1, Harald W. Platta1
1Biochemie Intrazellulärer Transportprozesse, Ruhr-Universität Bochum, 44801 Bochum, Germany;

Tóm tắt

The mechanistic target of Rapamycin (mTOR) is a ubiquitously-conserved serine/threonine kinase, which has a central function in integrating growth signals and orchestrating their physiologic effects on cellular level. mTOR is the core component of differently composed signaling complexes that differ in protein composition and molecular targets. Newly identified classes of mTOR inhibitors are being developed to block autoimmune diseases and transplant rejections but also to treat obesity, diabetes, and different types of cancer. Therefore, the selective and context-dependent inhibition of mTOR activity itself might come into the focus as molecular target to prevent severe diseases and possibly to extend life span. This review provides a general introduction to the molecular composition and physiologic function of mTOR complexes as part of the Special Issue “2018 Select Papers by Cells’ Editorial Board Members”.

Từ khóa


Tài liệu tham khảo

Dann, 2006, The amino acid sensitive TOR pathway from yeast to mammals, FEBS Lett., 580, 2821, 10.1016/j.febslet.2006.04.068

Imseng, 2018, Architecture and activation of phosphatidylinositol 3-kinase related kinases, Curr. Opin. Struct. Biol., 49, 177, 10.1016/j.sbi.2018.03.010

Heitman, 1991, Targets for cell cycle arrest by the immunosuppressant rapamycin in yeast, Science, 253, 905, 10.1126/science.1715094

Sehgal, 1975, Rapamycin (AY-22,989), a new antifungal antibiotic. II. Fermentation, isolation and characterization, J. Antibiot. (Tokyo), 28, 727, 10.7164/antibiotics.28.727

Seto, 2012, Rapamycin and mTOR: A serendipitous discovery and implications for breast cancer, Clin. Transl. Med., 15, 29, 10.1186/2001-1326-1-29

Dechant, 2008, Nutrient signals driving cell growth, Curr. Opin. Cell Biol., 20, 678, 10.1016/j.ceb.2008.09.009

Wu, 2016, F-actin rearrangement is regulated by mTORC2/Akt/Girdin in mouse fertilized eggs, Cell Prolif., 49, 740, 10.1111/cpr.12285

Dennis, 2002, Quick guide: Target of rapamycin, Curr. Biol., 12, R269, 10.1016/S0960-9822(02)00796-0

McMahon, 2002, The rapamycin-binding domain governs substrate selectivity by the mammalian target of rapamycin, Mol. Cell. Biol., 22, 7428, 10.1128/MCB.22.21.7428-7438.2002

Gaubitz, 2015, Molecular basis of the rapamycin insensitivity of target of rapamycin complex 2, Mol. Cell, 58, 977, 10.1016/j.molcel.2015.04.031

Chen, 2018, Cryo-EM structure of human mTOR complex 2, Cell Res., 28, 518, 10.1038/s41422-018-0029-3

Stuttfeld, 2018, Architecture of the human mTORC2 core complex, Elife, 7, e33101, 10.7554/eLife.33101

Lamming, 2012, Rapamycin-induced insulin resistance is mediated by mTORC2 loss and uncoupled from longevity, Science, 335, 1638, 10.1126/science.1215135

Sarbassov, 2006, Prolonged rapamycin treatment inhibits mTORC2 assembly and Akt/PKB, Mol. Cell, 22, 159, 10.1016/j.molcel.2006.03.029

Hara, 2002, Raptor, a binding partner of target of rapamycin (TOR), mediates TOR action, Cell, 110, 177, 10.1016/S0092-8674(02)00833-4

Kim, 2002, mTOR interacts with raptor to form a nutrient-sensitive complex that signals to the cell growth machinery, Cell, 110, 163, 10.1016/S0092-8674(02)00808-5

Kim, 2003, GbetaL, a positive regulator of the rapamycin-sensitive pathway required for the nutrient-sensitive interaction between raptor and mTOR, Mol. Cell, 11, 895, 10.1016/S1097-2765(03)00114-X

Nojima, 2003, The mammalian target of rapamycin (mTOR) partner, raptor, binds the mTOR substrates p70 S6 kinase and 4E-BP1 through their TOR signaling (TOS) motif, J. Biol. Chem., 278, 15461, 10.1074/jbc.C200665200

Schalm, 2003, TOS motif-mediated raptor binding regulates 4E-BP1 multisite phosphorylation and function, Curr. Biol., 13, 797, 10.1016/S0960-9822(03)00329-4

Peterson, 2009, DEPTOR is an mTOR inhibitor frequently overexpressed in multiple myeloma cells and required for their survival, Cell, 137, 873, 10.1016/j.cell.2009.03.046

Sancak, 2007, PRAS40 is an insulin-regulated inhibitor of the mTORC1 protein kinase, Mol. Cell, 25, 903, 10.1016/j.molcel.2007.03.003

Lee, 2007, Insulin signalling to mTOR mediated by the Akt/PKB substrate PRAS40, Nat. Cell Biol., 9, 316, 10.1038/ncb1547

Kakumoto, K., Ikeda, J., Okada, M., Morii, E., and Oneyama, C. (2015). mLST8 promotes mTOR-mediated tumor progression. PLoS One, 10.

Yang, 2013, mTOR kinase structure, mechanism and regulation, Nature, 497, 217, 10.1038/nature12122

Saxton, 2017, mTOR signaling in growth, metabolism, and disease, Cell, 169, 361, 10.1016/j.cell.2017.03.035

Aylett, 2016, Architecture of human mTOR complex 1, Science, 351, 48, 10.1126/science.aaa3870

Berndt, 2016, Tor forms a dimer through an N-terminal helical solenoid with a complex topology, Nat. Commun., 7, 11016, 10.1038/ncomms11016

Yip, 2010, Structure of the human mTOR complex I and its implications for rapamycin inhibition, Mol. Cell, 38, 768, 10.1016/j.molcel.2010.05.017

Williams, 2014, The structural basis for mTOR function, Semin. Cell Dev. Biol., 36, 91, 10.1016/j.semcdb.2014.09.024

Kaizuka, 2010, Tti1 and Tel2 are critical factors in mammalian target of rapamycin complex assembly, J. Biol. Chem., 285, 20109, 10.1074/jbc.M110.121699

Sugimoto, K. (2018). Branching the Tel2 pathway for exact fit on phosphatidylinositol 3-kinase-related kinases. Curr. Genet.

Laplante, 2012, mTOR signaling in growth control and disease, Cell, 149, 274, 10.1016/j.cell.2012.03.017

Jacinto, 2004, Mammalian TOR complex 2 controls the actin cytoskeleton and is rapamycin insensitive, Nat. Cell Biol., 6, 1122, 10.1038/ncb1183

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, 10.1016/j.cub.2004.06.054

Varusai, 2018, Dynamic modelling of the mTOR signalling network reveals complex emergent behaviours conferred by DEPTOR, Sci. Rep., 8, 643, 10.1038/s41598-017-18400-z

Pearce, 2007, Identification of Protor as a novel Rictor-binding component of mTOR complex-2, Biochem. J., 405, 513, 10.1042/BJ20070540

Thedieck, K., Polak, P., Kim, M.L., Molle, K.D., Cohen, A., Jenö, P., Arrieumerlou, C., and Hall, M.N. (2007). PRAS40 and PRR5-like protein are new mTOR interactors that regulate apoptosis. PLoS One, 2.

Woo, 2007, PRR5, a novel component of mTOR complex 2, regulates platelet-derived growth factor receptor beta expression and signaling, J. Biol. Chem., 282, 25604, 10.1074/jbc.M704343200

Frias, 2006, mSin1 is necessary for Akt/PKB phosphorylation, and its isoforms define three distinct mTORC2s, Curr. Biol., 16, 1865, 10.1016/j.cub.2006.08.001

Jacinto, 2006, SIN1/MIP1 maintains rictor-mTOR complex integrity and regulates Akt phosphorylation and substrate specificity, Cell, 127, 125, 10.1016/j.cell.2006.08.033

Yang, 2006, Identification of Sin1 as an essential TORC2 component required for complex formation and kinase activity, Genes Dev., 20, 2820, 10.1101/gad.1461206

Loewith, 2006, The TOR signalling network from yeast to man, Int. J. Biochem. Cell Biol., 38, 1476, 10.1016/j.biocel.2006.02.013

Hall, 2017, Nutrient sensing and TOR signaling in yeast and mammals, EMBO J., 36, 397, 10.15252/embj.201696010

Dibble, 2012, TBC1D7 is a third subunit of the TSC1-TSC2 complex upstream of mTORC1, Mol. Cell, 47, 535, 10.1016/j.molcel.2012.06.009

Demetriades, 2016, Lysosomal recruitment of TSC2 is a universal response to cellular stress, Nat. Commun., 7, 10662, 10.1038/ncomms10662

Garami, 2003, Insulin activation of Rheb, a mediator of mTOR/S6K/4E-BP signaling, is inhibited by TSC1 and 2, Mol. Cell, 11, 1457, 10.1016/S1097-2765(03)00220-X

Inoki, 2003, Rheb GTPase is a direct target of TSC2 GAP activity and regulates mTOR signaling, Genes Dev., 17, 1829, 10.1101/gad.1110003

Long, 2005, Rheb binds and regulates the mTOR kinase, Curr. Biol., 15, 702, 10.1016/j.cub.2005.02.053

Tee, 2003, Tuberous sclerosis complex gene products, Tuberin and Hamartin, control mTOR signaling by acting as a GTPase-activating protein complex toward Rheb, Curr. Biol., 13, 1259, 10.1016/S0960-9822(03)00506-2

Potheraveedu, 2017, The small GTPases Ras and Rheb studied by multidimensional NMR spectroscopy: Structure and function, Biol. Chem., 398, 577, 10.1515/hsz-2016-0276

Inoki, 2002, TSC2 is phosphorylated and inhibited by Akt and suppresses mTOR signalling, Nat. Cell Biol., 4, 648, 10.1038/ncb839

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, 10.1016/S1097-2765(02)00568-3

Menon, 2014, Spatial control of the TSC complex integrates insulin and nutrient regulation of mTORC1 at the lysosome, Cell, 156, 771, 10.1016/j.cell.2013.11.049

Ma, 2005, Phosphorylation and functional inactivation of TSC2 by Erk implications for tuberous sclerosis and cancer pathogenesis, Cell, 121, 179, 10.1016/j.cell.2005.02.031

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. USA, 101, 13489, 10.1073/pnas.0405659101

Mihaylova, 2011, The AMPK signalling pathway coordinates cell growth, autophagy and metabolism, Nat. Cell Biol., 13, 1016, 10.1038/ncb2329

Inoki, 2003, TSC2 mediates cellular energy response to control cell growth and survival, Cell, 115, 577, 10.1016/S0092-8674(03)00929-2

Shaw, 2004, The LKB1 tumor suppressor negatively regulates mTOR signaling, Cancer Cell, 6, 91, 10.1016/j.ccr.2004.06.007

Gwinn, 2008, AMPK phosphorylation of raptor mediates a metabolic checkpoint, Cell, 30, 214

Schneider, 2008, Hypoxia-induced energy stress inhibits the mTOR pathway by activating an AMPK/REDD1 signaling axis in head and neck squamous cell carcinoma, Neoplasia, 10, 1295, 10.1593/neo.08586

Brugarolas, 2004, Regulation of mTOR function in response to hypoxia by REDD1 and the TSC1/TSC2 tumor suppressor complex, Genes Dev., 18, 2893, 10.1101/gad.1256804

Feng, 2007, The regulation of AMPK beta1, TSC2, and PTEN expression by p53: Stress, cell and tissue specificity, and the role of these gene products in modulating the IGF-1-AKT-mTOR pathways, Cancer Res., 67, 3043, 10.1158/0008-5472.CAN-06-4149

Ratnayake, L., Adhvaryu, K.K., Kafes, E., Motavaze, K., and Lakin-Thomas, P. (2018). A component of the TOR (Target of Rapamycin) nutrient-sensing pathway plays a role in circadian rhythmicity in Neurospora crassa. PLoS Genet, 14.

Kim, 2008, Regulation of TORC1 by Rag GTPases in nutrient response, Nat. Cell Biol., 10, 935, 10.1038/ncb1753

Sancak, 2008, The Rag GTPases bind raptor and mediate amino acid signaling to mTORC1, Science, 320, 1496, 10.1126/science.1157535

Kim, 2016, Rag GTPase in amino acid signaling, Amino Acids, 48, 915, 10.1007/s00726-016-2171-x

Su, 2017, Hybrid structure of the RagA/C-ragulator mTORC1 activation complex, Mol. Cell, 68, 835, 10.1016/j.molcel.2017.10.016

Schweitzer, 2012, Ragulator is a GEF for the rag GTPases that signal amino acid levels to mTORC1, Cell, 150, 1196, 10.1016/j.cell.2012.07.032

Sancak, 2010, Ragulator-Rag complex targets mTORC1 to the lysosomal surface and is necessary for its activation by amino acids, Cell, 141, 290, 10.1016/j.cell.2010.02.024

Jung, 2015, Amino acid-dependent mTORC1 regulation by the lysosomal membrane protein SLC38A9, Mol. Cell. Biol., 35, 2479, 10.1128/MCB.00125-15

Rebsamen, 2015, SLC38A9 is a component of the lysosomal amino acid sensing machinery that controls mTORC1, Nature, 519, 477, 10.1038/nature14107

Wang, 2015, Metabolism. lysosomal amino acid transporter SLC38A9 signals arginine sufficiency to mTORC1, Science, 347, 188, 10.1126/science.1257132

Wyant, 2017, mTORC1 activator SLC38A9 is required to efflux essential amino acids from lysosomes and use protein as a nutrient, Cell, 171, 642, 10.1016/j.cell.2017.09.046

Zoncu, 2011, mTORC1 senses lysosomal amino acids through an inside-out mechanism that requires the vacuolar H(+)-ATPase, Science, 334, 678, 10.1126/science.1207056

Chantranupong, 2013, A Tumor suppressor complex with GAP activity for the Rag GTPases that signal amino acid sufficiency to mTORC1, Science, 340, 1100, 10.1126/science.1232044

Peng, 2017, SZT2 dictates GATOR control of mTORC1 signalling, Nature, 543, 433, 10.1038/nature21378

Wolfson, 2017, KICSTOR recruits GATOR1 to the lysosome and is necessary for nutrients to regulate mTORC1, Nature, 543, 438, 10.1038/nature21423

Chantranupong, 2014, The Sestrins interact with GATOR2 to negatively regulate the amino-acid-sensing pathway upstream of mTORC1, Cell Rep., 9, 1, 10.1016/j.celrep.2014.09.014

Parmigiani, 2014, Sestrins inhibit mTORC1 kinase activation through the GATOR complex, Cell Rep., 9, 1281, 10.1016/j.celrep.2014.10.019

Chantranupong, 2016, The CASTOR proteins are arginine sensors for the mTORC1 pathway, Cell, 165, 153, 10.1016/j.cell.2016.02.035

Saxton, 2016, Mechanism of arginine sensing by CASTOR1 upstream of mTORC1, Nature, 536, 229, 10.1038/nature19079

Saxton, 2016, Structural basis for leucine sensing by the Sestrin2-mTORC1 pathway, Science, 351, 53, 10.1126/science.aad2087

Wolfson, 2016, Sestrin2 is a leucine sensor for the mTORC1 pathway, Science, 351, 43, 10.1126/science.aab2674

Ye, 2015, GCN2 sustains mTORC1 suppression upon amino acid deprivation by inducing Sestrin2, Genes Dev., 29, 2331, 10.1101/gad.269324.115

Jewell, 2015, Metabolism. Differential regulation of mTORC1 by leucine and glutamine, Science, 347, 194, 10.1126/science.1259472

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, 10.1016/j.molcel.2013.09.016

Petit, 2013, Recruitment of folliculin to lysosomes supports the amino acid-dependent activation of Rag GTPases, J. Cell Biol., 202, 1107, 10.1083/jcb.201307084

Schmidt, 2018, FLCN: The causative gene for Birt-Hogg-Dubé syndrome, Gene, 640, 28, 10.1016/j.gene.2017.09.044

Liu, 2015, PtdIns(3,4,5)P3-dependent activation of the mTORC2 kinase complex, Cancer Discov., 5, 1194, 10.1158/2159-8290.CD-15-0460

Yang, 2015, A positive feedback loop between Akt and mTORC2 via SIN1 phosphorylation, Cell Rep., 12, 937, 10.1016/j.celrep.2015.07.016

Hsu, 2011, The mTOR-regulated phosphoproteome reveals a mechanism of mTORC1-mediated inhibition of growth factor signaling, Science, 332, 1317, 10.1126/science.1199498

Yu, 2011, Phosphoproteomic analysis identifies Grb10 as an mTORC1 substrate that negatively regulates insulin signaling, Science, 332, 1322, 10.1126/science.1199484

Yoon, M.S. (2017). The role of mammalian target of rapamycin (mTOR) in insulin signaling. Nutrients, 9.

Thedieck, 2017, Differential control of ageing and lifespan by isoforms and splice variants across the mTOR network, Essays Biochem., 61, 349, 10.1042/EBC20160086

Holz, 2005, mTOR and S6K1 mediate assembly of the translation preinitiation complex through dynamic protein interchange and ordered phosphorylation events, Cell, 123, 569, 10.1016/j.cell.2005.10.024

Dorrello, 2006, S6K1- and betaTRCP-mediated degradation of PDCD4 promotes protein translation and cell growth, Science, 314, 467, 10.1126/science.1130276

Brunn, 1997, Phosphorylation of the translational repressor PHAS-I by the mammalian target of rapamycin, Science, 277, 99, 10.1126/science.277.5322.99

Gingras, 1999, Regulation of 4E-BP1 phosphorylation: A novel two-step mechanism, Genes Dev., 13, 1422, 10.1101/gad.13.11.1422

Hsieh, 2012, The translational landscape of mTOR signalling steers cancer initiation and metastasis, Nature, 485, 55, 10.1038/nature10912

Thoreen, 2012, A unifying model for mTORC1-mediated regulation of mRNA translation, Nature, 485, 109, 10.1038/nature11083

Porstmann, 2008, SREBP activity is regulated by mTORC1 and contributes to Akt-dependent cell growth, Cell Metab., 8, 224, 10.1016/j.cmet.2008.07.007

Yecies, 2010, Activation of a metabolic gene regulatory network downstream of mTOR complex 1, Mol. Cell, 39, 171, 10.1016/j.molcel.2010.06.022

Peterson, 2011, mTOR complex 1 regulates lipin 1 localization to control the SREBP pathway, Cell, 146, 408, 10.1016/j.cell.2011.06.034

Lippman, 2009, Protein kinase A and TORC1 activate genes for ribosomal biogenesis by inactivating repressors encoded by Dot6 and its homolog Tod6, Proc. Natl. Acad. Sci. USA, 106, 19928, 10.1073/pnas.0907027106

Kos-Braun, I.C., Jung, I., and Koš, M. (2017). Tor1 and CK2 kinases control a switch between alternative ribosome biogenesis pathways in a growth-dependent manner. PLoS Biol., 15.

Hoxhaj, 2016, mTORC1 induces purine synthesis through control of the mitochondrial tetrahydrofolate cycle, Science, 351, 728, 10.1126/science.aad0489

Howell, 2013, Stimulation of de novo pyrimidine synthesis by growth signaling through mTOR and S6K1, Science, 339, 1323, 10.1126/science.1228792

Robitaille, 2013, Quantitative phosphoproteomics reveal mTORC1 activates de novo pyrimidine synthesis, Science, 339, 1320, 10.1126/science.1228771

Tsouko, 2014, Regulation of the pentose phosphate pathway by an androgen receptor-mTOR-mediated mechanism and its role in prostate cancer cell growth, Oncogenesis, 26, e103, 10.1038/oncsis.2014.18

Boutouja, F., Brinkmeier, R., Mastalski, T., El Magraoui, F., and Platta, H.W. (2017). Regulation of the tumor-suppressor BECLIN 1 by distinct ubiquitination cascades. Int. J. Mol. Sci., 18.

Kim, 2011, AMPK and mTOR regulate autophagy through direct phosphorylation of Ulk1, Nat. Cell Biol., 13, 132, 10.1038/ncb2152

Alers, 2014, ATG13: Just a companion, or an executor of the autophagic program?, Autophagy, 10, 944, 10.4161/auto.28987

Martina, 2012, MTORC1 functions as a transcriptional regulator of autophagy by preventing nuclear transport of TFEB, Autophagy, 8, 903, 10.4161/auto.19653

Petit, 2012, The transcription factor TFEB links mTORC1 signaling to transcriptional control of lysosome homeostasis, Sci. Signal., 5, ra42

Settembre, 2012, A lysosome-to-nucleus signalling mechanism senses and regulates the lysosome via mTOR and TFEB, EMBO J., 31, 1095, 10.1038/emboj.2012.32

Rousseau, 2016, An evolutionarily conserved pathway controls proteasome homeostasis, Nature, 536, 184, 10.1038/nature18943

Zhao, 2015, mTOR inhibition activates overall protein degradation by the ubiquitin proteasome system as well as by autophagy, Proc. Natl. Acad. Sci. USA, 112, 15790, 10.1073/pnas.1521919112

Zhang, 2016, Zhang & Manning reply, Nature, 529, E2, 10.1038/nature16473

Zhang, 2014, Coordinated regulation of protein synthesis and degradation by mTORC1, Nature, 513, 440, 10.1038/nature13492

Zhao, 2016, Control of proteasomal proteolysis by mTOR, Nature, 529, E1, 10.1038/nature16472

Gan, 2012, PRR5L degradation promotes mTORC2-mediated PKC-δ phosphorylation and cell migration downstream of Gα12, Nat. Cell Biol., 14, 686, 10.1038/ncb2507

Li, 2014, mTORC2 phosphorylates protein kinase Cζ to regulate its stability and activity, EMBO Rep., 15, 191, 10.1002/embr.201338119

Thomanetz, 2013, Ablation of the mTORC2 component rictor in brain or Purkinje cells affects size and neuron morphology, J. Cell Biol., 201, 293, 10.1083/jcb.201205030

Masri, 2007, mTORC2 activity is elevated in gliomas and promotes growth and cell motility via overexpression of rictor, Cancer Res., 67, 11712, 10.1158/0008-5472.CAN-07-2223

Alessi, 2008, mTOR complex 2 (mTORC2) controls hydrophobic motif phosphorylation and activation of serum- and glucocorticoid-induced protein kinase 1 (SGK1), Biochem. J., 416, 375, 10.1042/BJ20081668

Lyo, 2010, Phospholipase D stabilizes HDM2 through an mTORC2/SGK1 pathway, Biochem. Biophys. Res. Commun., 396, 562, 10.1016/j.bbrc.2010.04.148

Sarbassov, 2005, Phosphorylation and regulation of Akt/PKB by the rictor-mTOR complex, Science, 307, 1098, 10.1126/science.1106148

Guertin, 2006, Ablation in mice of the mTORC components raptor, rictor, or mLST8 reveals that mTORC2 is required for signaling to Akt-FOXO and PKCalpha, but not S6K1, Dev. Cell, 11, 859, 10.1016/j.devcel.2006.10.007

Kwiatkowski, 2003, Tuberous sclerosis: From tubers to mTOR, Ann. Hum. Genet., 67, 87, 10.1046/j.1469-1809.2003.00012.x

Roberts, 2003, A 34 bp deletion within TSC2 is a rare polymorphism, not a pathogenic mutation, Ann. Hum. Genet., 67, 495, 10.1046/j.1529-8817.2003.00059.x

Li, 2014, Estradiol and mTORC2 cooperate to enhance prostaglandin biosynthesis and tumorigenesis in TSC2-deficient LAM cells, J. Exp. Med., 211, 15, 10.1084/jem.20131080

Feng, 2005, The coordinate regulation of the p53 and mTOR pathways in cells, Proc. Natl. Acad. Sci. USA, 102, 8204, 10.1073/pnas.0502857102

Sato, 2014, Improving type 2 diabetes through a distinct adrenergic signaling pathway involving mTORC2 that mediates glucose uptake in skeletal muscle, Diabetes, 63, 4115, 10.2337/db13-1860

Vogelstein, 2000, Surfing the p53 network, Nature, 408, 307, 10.1038/35042675

Sun, 2011, Mammalian target of rapamycin up-regulation of pyruvate kinase isoenzyme type M2 is critical for aerobic glycolysis and tumor growth, Proc. Natl. Acad. Sci. USA, 108, 4129, 10.1073/pnas.1014769108

Warburg, 1956, On the origin of cancer cells, Science, 123, 309, 10.1126/science.123.3191.309

Burns, J.S., and Manda, G. (2017). Metabolic pathways of the warburg effect in health and disease: Perspectives of choice, chain or chance. Int. J. Mol. Sci., 18.

Liu, 2014, SIRT1 limits the function and fate of myeloid-derived suppressor cells in tumors by orchestrating HIF-1α-dependent glycolysis, Cancer Res., 74, 727, 10.1158/0008-5472.CAN-13-2584

Semenza, 2013, HIF-1 mediates metabolic responses to intratumoral hypoxia and oncogenic mutations, J. Clin. Invest., 123, 3664, 10.1172/JCI67230

Mathupala, 2006, Hexokinase II: cancer’s double-edged sword acting as both facilitator and gatekeeper of malignancy when bound to mitochondria, Oncogenesis, 25, 4777, 10.1038/sj.onc.1209603

Roberts, 2015, Hexokinase II integrates energy metabolism and cellular protection: Akting on mitochondria and TORCing to autophagy, Cell Death Differ., 22, 248, 10.1038/cdd.2014.173

Shintani, 2004, Autophagy in health and disease: A double-edged sword, Science, 306, 990, 10.1126/science.1099993

Thorburn, A. (2014). Autophagy and its effects: Making sense of double-edged swords. PLoS Biol., 12.

Nazio, 2013, mTOR inhibits autophagy by controlling ULK1 ubiquitylation, self-association and function through AMBRA1 and TRAF6, Nat. Cell Biol., 15, 406, 10.1038/ncb2708

Abrahamsen, 2012, Ubiquitination and phosphorylation of Beclin 1 and its binding partners: Tuning class III phosphatidylinositol 3-kinase activity and tumor suppression, FEBS Lett., 586, 1584, 10.1016/j.febslet.2012.04.046

Stenmark, H. (2010, January 29). The Sir Hans Krebs Lecture. How a lipid mediates tumour suppression. Proceedings of the 35th FEBS Congress in Gothenburg, Gothenburg, Sweden.

Janji, 2016, The multifaceted role of autophagy in tumor evasion from immune surveillance, Oncotarget, 7, 17591, 10.18632/oncotarget.7540

Perl, 2015, mTOR activation is a biomarker and a central pathway to autoimmune disorders, cancer, obesity, and aging, Ann. NY. Acad. Sci., 1346, 33, 10.1111/nyas.12756

Qi, W.X., Huang, Y.J., Yao, Y., Shen, Z., and Min, D.L. (2013). Incidence and risk of treatment-related mortality with mTOR inhibitors everolimus and temsirolimus in cancer patients: A meta-analysis. PLoS ONE, 8.

Guertin, 2009, mTOR complex 2 is required for the development of prostate cancer induced by Pten loss in mice, Cancer Cell, 15, 148, 10.1016/j.ccr.2008.12.017

Guri, 2017, mTORC2 promotes tumorigenesis via lipid synthesis, Cancer Cell, 32, 807, 10.1016/j.ccell.2017.11.011

Chen, 2016, mTOR complex-2 stimulates acetyl-CoA and de novo lipogenesis through ATP citrate lyase in HER2/PIK3CA-hyperactive breast cancer, Oncotarget, 7, 25224, 10.18632/oncotarget.8279

Khamzina, 2005, Increased activation of the mammalian target of rapamycin pathway in liver and skeletal muscle of obese rats: Possible involvement in obesity-linked insulin resistance, Endocrinology, 146, 1473, 10.1210/en.2004-0921

Krebs, 2007, The Mammalian target of rapamycin pathway regulates nutrient-sensitive glucose uptake in man, Diabetes, 56, 1600, 10.2337/db06-1016

Dibble, 2009, Characterization of Rictor phosphorylation sites reveals direct regulation of mTOR complex 2 by S6K1, Mol. Cell. Biol., 29, 5657, 10.1128/MCB.00735-09

Bell, 2000, Rapamycin inhibits human adipocyte differentiation in primary culture, Obes. Res., 8, 249, 10.1038/oby.2000.29

Yeh, 1995, Rapamycin inhibits clonal expansion and adipogenic differentiation of 3T3-L1 cells, Proc. Natl. Acad. Sci. USA, 92, 11086, 10.1073/pnas.92.24.11086

Houde, 2010, Chronic rapamycin treatment causes glucose intolerance and hyperlipidemia by upregulating hepatic gluconeogenesis and impairing lipid deposition in adipose tissue, Diabetes, 59, 1338, 10.2337/db09-1324

Chang, 2009, Rapamycin protects against high fat diet-induced obesity in C57BL/6J mice, J. Pharmacol. Sci., 109, 496, 10.1254/jphs.08215FP

Fang, 2013, Duration of rapamycin treatment has differential effects on metabolism in mice, Cell Metab., 17, 456, 10.1016/j.cmet.2013.02.008

Rajan, 2018, Inhibition of FOXO1 transcription factor in primary human adipocytes mimics the insulin resistant state of type 2 diabetes, Biochem. J., 475, 1807, 10.1042/BCJ20180144

Kim, 2012, mTOR complex 2 regulates proper turnover of insulin receptor substrate-1 via the ubiquitin ligase subunit Fbw8, Mol. Cell, 48, 875, 10.1016/j.molcel.2012.09.029

Vellai, 2003, Genetics: Influence of TOR kinase on lifespan in C. elegans, Nature, 426, 620, 10.1038/426620a

Kapahi, 2004, Regulation of lifespan in Drosophila by modulation of genes in the TOR signaling pathway, Curr. Biol., 14, 885, 10.1016/j.cub.2004.03.059

Bai, H., Post, S., Kang, P., and Tatar, M. (2015). drosophila longevity assurance conferred by reduced insulin receptor substrate chico partially requires d4eBP. PLoS ONE, 10.

Kaeberlein, 2005, Regulation of yeast replicative life span by TOR and Sch9 in response to nutrients, Science, 310, 1193, 10.1126/science.1115535

Deprez, 2018, The TORC1-Sch9 pathway as a crucial mediator of chronological lifespan in the yeast Saccharomyces cerevisiae, FEMS Yeast Res., 18, 18, 10.1093/femsyr/foy048

Wu, 2013, Increased mammalian lifespan and a segmental and tissue-specific slowing of aging after genetic reduction of mTOR expression, Cell Rep., 4, 913, 10.1016/j.celrep.2013.07.030

Selman, 2009, Ribosomal protein S6 kinase 1 signaling regulates mammalian life span, Science, 326, 140, 10.1126/science.1177221

Bjedov, 2010, Mechanisms of life span extension by rapamycin in the fruit fly Drosophila melanogaster, Cell Metab., 11, 35, 10.1016/j.cmet.2009.11.010

Harrison, 2009, Rapamycin fed late in life extends lifespan in genetically heterogeneous mice, Nature, 460, 392, 10.1038/nature08221

Powers, 2006, Extension of chronological life span in yeast by decreased TOR pathway signaling, Genes Dev., 20, 174, 10.1101/gad.1381406

Lamming, 2012, TOR signaling and rapamycin influence longevity by regulating SKN-1/Nrf and DAF-16/FoxO, Cell Metab., 15, 713, 10.1016/j.cmet.2012.04.007

Hansen, 2007, Lifespan extension by conditions that inhibit translation in Caenorhabditis elegans, Aging Cell, 6, 95, 10.1111/j.1474-9726.2006.00267.x

Filer, 2017, DNA polymerase III limits longevity downstream of TORC1, Nature, 552, 263, 10.1038/nature25007

Nakamura, 2018, Autophagy and Longevity, Mol. Cells, 41, 65

Reidick, 2015, Autophagy-related deubiquitinating enzymes involved in health and disease, Cells, 4, 596, 10.3390/cells4040596

Chen, 2009, mTOR regulation and therapeutic rejuvenation of aging hematopoietic stem cells, Sci. Signal., 2, ra75, 10.1126/scisignal.2000559

Yilmaz, 2012, mTORC1 in the Paneth cell niche couples intestinal stem-cell function to calorie intake, Nature, 486, 490, 10.1038/nature11163

Artoni, 2017, Loss of foxo rescues stem cell aging in Drosophila germ line, Elife, 6, e27842, 10.7554/eLife.27842

Neuman, 2016, Alternative rapamycin treatment regimens mitigate the impact of rapamycin on glucose homeostasis and the immune system, Aging Cell, 15, 28, 10.1111/acel.12405

Benjamin, 2011, Rapamycin passes the torch: A new generation of mTOR inhibitors, Nat. Rev. Drug Discov., 10, 868, 10.1038/nrd3531

Pierzynowska, 2018, Autophagy stimulation as a promising approach in treatment of neurodegenerative diseases, Metab. Brain Dis., 33, 989, 10.1007/s11011-018-0214-6

Frake, 2015, Autophagy and neurodegeneration, J. Clin. Invest., 125, 65, 10.1172/JCI73944

Caccamo, 2010, Molecular interplay between mammalian target of rapamycin (mTOR), amyloid-beta, and Tau: Effects on cognitive impairments, J. Biol. Chem., 285, 13107, 10.1074/jbc.M110.100420

Majumder, S., Richardson, A., Strong, R., and Oddo, S. (2011). Inducing autophagy by rapamycin before, but not after, the formation of plaques and tangles ameliorates cognitive deficits. PLoS ONE, 6.

Rahman, 2017, Therapeutic implication of autophagy in neurodegenerative diseases, BMB Rep., 50, 345, 10.5483/BMBRep.2017.50.7.069

Jiang, 2013, Curcumin ameliorates the neurodegenerative pathology in A53T α-synuclein cell model of Parkinson’s disease through the downregulation of mTOR/p70S6K signaling and the recovery of macroautophagy, J. Neuroimmune. Pharmacol., 8, 356, 10.1007/s11481-012-9431-7

Lin, 2017, “mTOR Signaling Pathway”: A potential target of curcumin in the treatment of spinal cord injury, Biomed. Res. Int., 2017, 1634801, 10.1155/2017/1634801

Xu, X.Y., Meng, X., Li, S., Gan, R.Y., Li, Y., and Li, H.B. (2018). Bioactivity, health benefits, and related molecular mechanisms of curcumin: Current progress, challenges, and perspectives. Nutrients.

Sciarretta, 2018, New insights into the role of mtor signaling in the cardiovascular system, Circ. Res., 122, 489, 10.1161/CIRCRESAHA.117.311147

McMullen, 2004, Inhibition of mTOR signaling with rapamycin regresses established cardiac hypertrophy induced by pressure overload, Circulation, 109, 3050, 10.1161/01.CIR.0000130641.08705.45

Shioi, 2003, Rapamycin attenuates load-induced cardiac hypertrophy in mice, Circulation, 107, 1664, 10.1161/01.CIR.0000057979.36322.88

Ikeda, 2015, The Akt-mTOR axis is a pivotal regulator of eccentric hypertrophy during volume overload, Sci. Rep., 5, 15881, 10.1038/srep15881

Kurdi, 2016, Potential therapeutic effects of mTOR inhibition in atherosclerosis, Br. J. Clin. Pharmacol., 82, 1267, 10.1111/bcp.12820

Kurdi, 2018, mTOR inhibition and cardiovascular diseases: Dyslipidemia and atherosclerosis, Transplantation, 102, S44, 10.1097/TP.0000000000001693

Zha, X.Q., Zhang, W.N., Peng, F.H., Xue, L., Liu, J., and Luo, J.P. (2017). Alleviating VLDL overproduction is an important mechanism for Laminaria japonica polysaccharide to inhibit atherosclerosis in LDLr-/- mice with diet-induced insulin resistance. Mol. Nutr. Food Res., 61.

Faivre, 2006, Current development of mTOR inhibitors as anticancer agents, Nat. Rev. Drug Discov., 5, 671, 10.1038/nrd2062

Fine, 2016, Recent advances in mammalian target of rapamycin inhibitor use in heart and lung transplantation, Transplantation, 100, 2558, 10.1097/TP.0000000000001432

Giordano, 2011, Inhibition of human in-stent restenosis: A molecular view, Curr. Opin. Pharmacol., 11, 372, 10.1016/j.coph.2011.03.006

Shirooie, S., Nabavi, S.F., Dehpour, A.R., Belwal, T., Habtemariam, S., Argüelles, S., Sureda, A., Daglia, M., Tomczyk, M., and Sobarzo-Sanchez, E. (2018). Targeting mTORs by omega-3 fatty acids: A possible novel therapeutic strategy for neurodegeneration?. Pharmacol. Res.

Zheng, 2015, mTOR inhibitors at a glance, Mol. Cell. Pharmacol., 7, 15

Neil, 2016, ATP-site binding inhibitor effectively targets mTORC1 and mTORC2 complexes in glioblastoma, Int. J. Oncol., 48, 1045, 10.3892/ijo.2015.3311

Ballou, 2008, Rapamycin and mTOR kinase inhibitors, J. Chem. Biol., 1, 27, 10.1007/s12154-008-0003-5

Zaytseva, 2012, mTOR inhibitors in cancer therapy, Cancer Lett., 319, 1, 10.1016/j.canlet.2012.01.005

Vilar, 2011, Pushing the envelope in the mTOR pathway: The second generation of inhibitors, J. Mol. Cancer Ther., 10, 395, 10.1158/1535-7163.MCT-10-0905

Conciatori, F., Ciuffreda, L., Bazzichetto, C., Falcone, I., Pilotto, S., Bria, E., Cognetti, F., and Milella, M. (2018). mTOR cross-talk in cancer and potential for combination therapy. Cancers (Basel), 10.

Mahoney, 2018, A small molecule inhibitor of Rheb selectively targets mTORC1 signaling, Nat. Commun., 9, 548, 10.1038/s41467-018-03035-z

Okaniwa, 2016, Overcoming mTOR resistance mutations with a new-generation mTOR inhibitor, Nature, 534, 272, 10.1038/nature17963

Fan, 2017, A kinase inhibitor targeted to mtorc1 drives regression in glioblastoma, Cancer Cell, 31, 424, 10.1016/j.ccell.2017.01.014

Fan, 2018, Inhibiting 4EBP1 in Glioblastoma, Clin. Cancer Res., 24, 14, 10.1158/1078-0432.CCR-17-0042

Renna, 2016, Commentary: Overcoming mTOR resistance mutations with a new-generation mTOR inhibitor, Front. Pharmacol., 7, 431, 10.3389/fphar.2016.00431