全部 标题 作者
关键词 摘要

OALib Journal期刊
ISSN: 2333-9721
费用:99美元

查看量下载量

相关文章

更多...
Scientifica  2013 

mTOR Inhibition: From Aging to Autism and Beyond

DOI: 10.1155/2013/849186

Full-Text   Cite this paper   Add to My Lib

Abstract:

The mechanistic target of rapamycin (mTOR) is a highly conserved protein that regulates growth and proliferation in response to environmental and hormonal cues. Broadly speaking, organisms are constantly faced with the challenge of interpreting their environment and making a decision between “grow or do not grow.” mTOR is a major component of the network that makes this decision at the cellular level and, to some extent, the tissue and organismal level as well. Although overly simplistic, this framework can be useful when considering the myriad functions ascribed to mTOR and the pleiotropic phenotypes associated with genetic or pharmacological modulation of mTOR signaling. In this review, I will consider mTOR function in this context and attempt to summarize and interpret the growing body of literature demonstrating interesting and varied effects of mTOR inhibitors. These include robust effects on a multitude of age-related parameters and pathologies, as well as several other processes not obviously linked to aging or age-related disease. 1. Introduction mTOR regulates a diverse array of cellular processes through its catalytic function as a serine/threonine protein kinase of the phosphoinositide-3-kinase-related family [1]. It acts within at least two distinct molecular complexes: mTOR complex 1 (mTORC1) and mTOR complex 2 (mTORC2) [2]. The composition of each complex is highly studied, and many of the distinct components of each complex have been characterized [3, 4]. mTORC1 consists of mTOR, the regulatory-associated protein of mTOR (raptor), the mammalian lethal with Sec13 protein 8 (mLST8), the DEP domain containing mTOR-interacting protein (deptor), and the proline-rich Akt substrate of 40?kDa (PRAS40). mTORC2 also contains mTOR and mLST8, but the remaining mTORC2 components are distinct from mTORC1. These include the rapamycin-insensitive companion of mTOR (rictor), protein observed with rictor (protor), mammalian stress-activated protein kinase-interacting protein 1 (mSin1), and proline-rich protein 5 (PRR5). Both mTOR complexes are essential, as loss of either raptor or rictor results in loss of viability [5, 6]. mTOR was first identified from studies in the budding yeast Saccharomyces cerevisiae of mutations that conferred altered sensitivity to the macrolide antibiotic rapamycin (also known as sirolimus) [7, 8]. Analysis of rapamycin resistant mutants led to the identification of two yeast genes, TOR1 and TOR2, that both encode mTOR kinases. Yeast Tor1 is found exclusively in mTORC1, while yeast Tor2 functions in both mTOR complexes. Thus,

References

[1]  C. T. Keith and S. L. Schreiber, “PIK-related kinases: DNA repair, recombination, and cell cycle checkpoints,” Science, vol. 270, no. 5233, pp. 50–51, 1995.
[2]  R. Loewith, E. Jacinto, S. Wullschleger et al., “Two TOR complexes, only one of which is rapamycin sensitive, have distinct roles in cell growth control,” Molecular Cell, vol. 10, no. 3, pp. 457–468, 2002.
[3]  H. Zhou and S. Huang, “The complexes of mammalian target of rapamycin,” Current Protein and Peptide Science, vol. 11, no. 6, pp. 409–424, 2010.
[4]  I. Bracho-Valdés, P. Moreno-Alvarez, I. Valencia-Martínez, E. Robles-Molina, L. Chávez-Vargas, and J. Vázquez-Prado, “MTORC1- and mTORC2-interacting proteins keep their multifunctional partners focused,” IUBMB Life, vol. 63, no. 10, pp. 880–898, 2011.
[5]  D. A. Guertin, D. M. Stevens, C. C. Thoreen et al., “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,” Developmental Cell, vol. 11, no. 6, pp. 859–871, 2006.
[6]  S. B. Helliwell, I. Howald, N. Barbet, and M. N. Hall, “TOR2 is part of two related signaling pathways coordinating cell growth in Saccharomyces cerevisiae,” Genetics, vol. 148, no. 1, pp. 99–112, 1998.
[7]  J. Heitman, N. R. Movva, and M. N. Hall, “Targets for cell cycle arrest by the immunosuppressant rapamycin in yeast,” Science, vol. 253, no. 5022, pp. 905–909, 1991.
[8]  R. Cafferkey, P. R. Young, M. M. McLaughlin et al., “Dominant missense mutations in a novel yeast protein related to mammalian phosphatidylinositol 3-kinase and VPS34 abrogate rapamycin cytotoxicity,” Molecular and Cellular Biology, vol. 13, no. 10, pp. 6012–6023, 1993.
[9]  C. J. Sabers, M. M. Martin, G. J. Brunn et al., “Isolation of a protein target of the FKBP12-rapamycin complex in mammalian cells,” Journal of Biological Chemistry, vol. 270, no. 2, pp. 815–822, 1995.
[10]  E. J. Brown, M. W. Albers, T. B. S. Tae Bum Shin et al., “A mammalian protein targeted by G1-arresting rapamycin-receptor complex,” Nature, vol. 369, no. 6483, pp. 756–758, 1994.
[11]  M. I. Chiu, H. Katz, and V. Berlin, “RAPT1, a mammalian homolog of yeast Tor, interacts with the FKBP12/rapamycin complex,” Proceedings of the National Academy of Sciences of the United States of America, vol. 91, no. 26, pp. 12574–12578, 1994.
[12]  D. M. Sabatini, H. Erdjument-Bromage, M. Lui, P. Tempst, and S. H. Snyder, “RAFT1: a mammalian protein that binds to FKBP12 in a rapamycin-dependent fashion and is homologous to yeast TORs,” Cell, vol. 78, no. 1, pp. 35–43, 1994.
[13]  Y. Chen, H. Chen, A. E. Rhoad et al., “A putative sirolimus (rapamycin) effector protein,” Biochemical and Biophysical Research Communications, vol. 203, no. 1, pp. 1–7, 1994.
[14]  M. Laplante and D. M. Sabatini, “Regulation of mTORC1 and its impact on gene expression at a glance,” Journal of Cell Science, vol. 126, pp. 1713–1719, 2013.
[15]  M. Laplante and D. M. Sabatini, “mTOR signaling at a glance,” Journal of Cell Science, vol. 122, no. 20, pp. 3589–3594, 2009.
[16]  X. M. Ma and J. Blenis, “Molecular mechanisms of mTOR-mediated translational control,” Nature Reviews Molecular Cell Biology, vol. 10, no. 5, pp. 307–318, 2009.
[17]  K. Inoki, Y. Li, T. Xu, and K.-L. Guan, “Rheb GTpase is a direct target of TSC2 GAP activity and regulates mTOR signaling,” Genes and Development, vol. 17, no. 15, pp. 1829–1834, 2003.
[18]  K. Inoki, Y. Li, T. Zhu, J. Wu, and K.-L. Guan, “TSC2 is phosphorylated and inhibited by Akt and suppresses mTOR signalling,” Nature Cell Biology, vol. 4, no. 9, pp. 648–657, 2002.
[19]  B. D. Manning, A. R. Tee, M. N. Logsdon, J. Blenis, and L. C. Cantley, “Identification of the tuberous sclerosis complex-2 tumor suppressor gene product tuberin as a target of the phosphoinositide 3-kinase/Akt pathway,” Molecular Cell, vol. 10, no. 1, pp. 151–162, 2002.
[20]  X. Long, F. Müller, and J. Avruch, “TOR action in mammalian cells and in Caenorhabditis elegans,” Current Topics in Microbiology and Immunology, vol. 279, pp. 115–138, 2003.
[21]  X. Long, C. Spycher, Z. S. Han, A. M. Rose, F. Müller, and J. Avruch, “TOR deficiency in C. elegans causes developmental arrest and intestinal atrophy by inhibition of mRNA translation,” Current Biology, vol. 12, no. 17, pp. 1448–1461, 2002.
[22]  C. J. Potter, L. G. Pedraza, and T. Xu, “Akt regulates growth by directly phosphorylating Tsc2,” Nature Cell Biology, vol. 4, no. 9, pp. 658–665, 2002.
[23]  L. Ma, Z. Chen, H. Erdjument-Bromage, P. Tempst, and P. P. Pandolfi, “Phosphorylation and functional inactivation of TSC2 by Erk: implications for tuberous sclerosis and cancer pathogenesis,” Cell, vol. 121, no. 2, pp. 179–193, 2005.
[24]  K. Inoki, T. Zhu, and K.-L. Guan, “TSC2 mediates cellular energy response to control cell growth and survival,” Cell, vol. 115, no. 5, pp. 577–590, 2003.
[25]  D. M. Gwinn, D. B. Shackelford, D. F. Egan et al., “AMPK phosphorylation of raptor mediates a metabolic checkpoint,” Molecular Cell, vol. 30, no. 2, pp. 214–226, 2008.
[26]  Y. Sancak, T. R. Peterson, Y. D. Shaul et al., “The rag GTPases bind raptor and mediate amino acid signaling to mTORC1,” Science, vol. 320, no. 5882, pp. 1496–1501, 2008.
[27]  Y. Sancak, L. Bar-Peled, R. Zoncu, A. L. Markhard, S. Nada, and D. M. Sabatini, “Ragulator-rag complex targets mTORC1 to the lysosomal surface and is necessary for its activation by amino acids,” Cell, vol. 141, no. 2, pp. 290–303, 2010.
[28]  R. Zoncu, L. Bar-Peled, A. Efeyan, S. Wang, Y. Sancak, and D. M. Sabatini, “mTORC1 senses lysosomal amino acids through an inside-out mechanism that requires the vacuolar H+-ATPase,” Science, vol. 334, no. 6056, pp. 678–683, 2011.
[29]  E. Kim, P. Goraksha-Hicks, L. Li, T. P. Neufeld, and K.-L. Guan, “Regulation of TORC1 by Rag GTPases in nutrient response,” Nature Cell Biology, vol. 10, no. 8, pp. 935–945, 2008.
[30]  M. Laplante and D. M. Sabatini, “mTOR signaling,” Cold Spring Harbor Perspectives in Biology, vol. 4, 2012.
[31]  M. Laplante and D. M. Sabatini, “mTOR signaling in growth control and disease,” Cell, vol. 149, no. 2, pp. 274–293, 2012.
[32]  S. G. Kim, G. R. Buel, and J. Blenis, “Nutrient regulation of the mTOR complex 1 signaling pathway,” Molecules and Cells, vol. 35, pp. 463–473, 2013.
[33]  D. E. Martin and M. N. Hall, “The expanding TOR signaling network,” Current Opinion in Cell Biology, vol. 17, no. 2, pp. 158–166, 2005.
[34]  S. N. Sehgal, H. Baker, and C. Vezina, “Rapamycin (AY 22,989), a new antifungal antibiotic—II. Fermentation, isolation and characterization,” Journal of Antibiotics, vol. 28, no. 10, pp. 727–732, 1975.
[35]  C. Vezina, A. Kudelski, and S. N. Sehgal, “Rapamycin (AY 22,989), a new antifungal antibiotic—I. Taxonomy of the producing streptomycete and isolation of the active principle,” Journal of Antibiotics, vol. 28, no. 10, pp. 721–726, 1975.
[36]  J. Douros and M. Suffness, “New antitumor substances of natural origin,” Cancer Treatment Reviews, vol. 8, no. 1, pp. 63–87, 1981.
[37]  C. P. Eng, S. N. Sehgal, and C. Vezina, “Activity of rapamycin (AY-22,989) against transplanted tumors,” Journal of Antibiotics, vol. 37, no. 10, pp. 1231–1237, 1984.
[38]  J. Chen, X.-F. Zheng, E. J. Brown, and S. L. Schreiber, “Identification of an 11-kDa FKBP12-rapamycin-binding domain within the 289-kDa FKBP12-rapamycin-associated protein and characterization of a critical serine residue,” Proceedings of the National Academy of Sciences of the United States of America, vol. 92, no. 11, pp. 4947–4951, 1995.
[39]  J. Choi, J. Chen, S. L. Schreiber, and J. Clardy, “Structure of the FKBP12-rapamycin complex interacting with the binding domain of human FRAP,” Science, vol. 273, no. 5272, pp. 239–242, 1996.
[40]  S. C. Johnson, P. S. Rabinovitch, and M. Kaeberlein, “mTOR is a key modulator of ageing and age-related disease,” Nature, vol. 493, pp. 338–345, 2013.
[41]  M. Kaeberlein, “Longevity and aging,” F1000prime Reports, vol. 5, p. 5, 2013.
[42]  G. M. Martin, “The biology of aging: 1985–2010 and beyond,” FASEB Journal, vol. 25, no. 11, pp. 3756–3762, 2011.
[43]  M. Kaeberlein, “Lessons on longevity from budding yeast,” Nature, vol. 464, no. 7288, pp. 513–519, 2010.
[44]  M. E. Yanos, C. F. Bennett, and M. Kaeberlein, “Genome-wide RNAi longevity screens in Caenorhabditis elegans,” Current Genomics, vol. 13, pp. 508–518, 2012.
[45]  T. E. Johnson, “25 years after age-1: genes, interventions and the revolution in aging research,” Experimental Gerontology, vol. 48, p. 640, 2013.
[46]  A. Bartke, “Single-gene mutations and healthy ageing in mammals,” Philosophical Transactions of the Royal Society B, vol. 366, p. 28, 2011.
[47]  L. Partridge, “Some highlights of research on aging with invertebrates, 2010,” Aging Cell, vol. 10, no. 1, pp. 5–9, 2011.
[48]  L. Fontana, L. Partridge, and V. D. Longo, “Extending healthy life span-from yeast to humans,” Science, vol. 328, no. 5976, pp. 321–326, 2010.
[49]  C. J. Kenyon, “The genetics of ageing,” Nature, vol. 464, no. 7288, pp. 504–512, 2010.
[50]  B. K. Kennedy, K. K. Steffen, and M. Kaeberlein, “Ruminations on dietary restriction and aging,” Cellular and Molecular Life Sciences, vol. 64, no. 11, pp. 1323–1328, 2007.
[51]  V. D. Longo, G. S. Shadel, M. Kaeberlein, and B. Kennedy, “Replicative and chronological aging in Saccharomyces cerevisiae,” Cell Metabolism, vol. 16, p. 18, 2012.
[52]  K. A. Steinkraus, M. Kaeberlein, and B. K. Kennedy, “Replicative aging in yeast: the means to the end,” Annual Review of Cell and Developmental Biology, vol. 24, pp. 29–54, 2008.
[53]  P. Fabrizio, F. Pozza, S. D. Pletcher, C. M. Gendron, and V. D. Longo, “Regulation of longevity and stress resistance by Sch9 in yeast,” Science, vol. 292, no. 5515, pp. 288–290, 2001.
[54]  J. Urban, A. Soulard, A. Huber et al., “Sch9 is a major target of TORC1 in Saccharomyces cerevisiae,” Molecular Cell, vol. 26, no. 5, pp. 663–674, 2007.
[55]  P. Fabrizio, L.-L. Liou, V. N. Moy et al., “SOD2 functions downstream of Sch9 to extend longevity in yeast,” Genetics, vol. 163, no. 1, pp. 35–46, 2003.
[56]  P. Fabrizio, S. D. Pletcher, N. Minois, J. W. Vaupel, and V. D. Longo, “Chronological aging-independent replicative life span regulation by Msn2/Msn4 and Sod2 in Saccharomyces cerevisiae,” FEBS Letters, vol. 557, no. 1-3, pp. 136–142, 2004.
[57]  M. Kaeberlein, R. W. Powers III, K. K. Steffen et al., “Regulation of yeast replicative life span by TOR and Sch9 response to nutrients,” Science, vol. 310, no. 5751, pp. 1193–1196, 2005.
[58]  R. W. Powers III, M. Kaeberlein, S. D. Caldwell, B. K. Kennedy, and S. Fields, “Extension of chronological life span in yeast by decreased TOR pathway signaling,” Genes and Development, vol. 20, no. 2, pp. 174–184, 2006.
[59]  T. Vellai, K. Takacs-Vellai, Y. Zhang, A. L. Kovacs, L. Orosz, and F. Müller, “Influence of TOR kinase on lifespan in C. elegans,” Nature, vol. 426, no. 6967, p. 620, 2003.
[60]  K. Jia, D. Chen, and D. L. Riddle, “The TOR pathway interacts with the insulin signaling pathway to regulate C. elegans larval development, metabolism and life span,” Development, vol. 131, no. 16, pp. 3897–3906, 2004.
[61]  P. Kapahi, B. M. Zid, T. Harper, D. Koslover, V. Sapin, and S. Benzer, “Regulation of lifespan in Drosophila by modulation of genes in the TOR signaling pathway,” Current Biology, vol. 14, p. 885, 2004.
[62]  S. Huang and P. J. Houghton, “Inhibitors of mammalian target of rapamycin as novel antitumor agents: from bench to clinic,” Current Opinion in Investigational Drugs, vol. 3, no. 2, pp. 295–304, 2002.
[63]  C. M. Hartford and M. J. Ratain, “Rapamycin: something old, something new, sometimes borrowed and now renewed,” Clinical Pharmacology and Therapeutics, vol. 82, no. 4, pp. 381–388, 2007.
[64]  M. Mita, K. Sankhala, I. Abdel-Karim, A. Mita, and F. Giles, “Deforolimus (AP23573) a novel mTOR inhibitor in clinical development,” Expert Opinion on Investigational Drugs, vol. 17, no. 12, pp. 1947–1954, 2008.
[65]  L. Buellesfeld and E. Grube, “ABT-578-eluting stents: the promising successor of sirolimus- and paclitaxel-eluting stent concepts?” Herz, vol. 29, no. 2, pp. 167–170, 2004.
[66]  S. E. Burke, R. E. Kuntz, and L. B. Schwartz, “Zotarolimus (ABT-578) eluting stents,” Advanced Drug Delivery Reviews, vol. 58, no. 3, pp. 437–446, 2006.
[67]  K. G. Pike, K. Malagu, M. G. Hummersone, et al., “Optimization of potent and selective dual mTORC1 and mTORC2 inhibitors: the discovery of AZD8055 and AZD2014,” Bioorganic & Medicinal Chemistry Letters, vol. 23, pp. 1212–1216, 2013.
[68]  A. Naing, C. Aghajanian, E. Raymond, et al., “Safety, tolerability, pharmacokinetics and pharmacodynamics of AZD8055 in advanced solid tumours and lymphoma,” British Journal of Cancer, vol. 107, pp. 1093–1099, 2012.
[69]  H. Asahina, H. Nokihara, N. Yamamoto, et al., “Safety and tolerability of AZD8055 in Japanese patients with advanced solid tumors; a dose-finding phase I study,” Investigational New Drugs, vol. 31, pp. 677–684, 2013.
[70]  C. M. Chresta, B. R. Davies, and I. Hickson, “AZD8055 is a potent, selective, and orally bioavailable ATP-competitive mammalian target of rapamycin kinase inhibitor with in vitro and in vivo antitumor activity,” Cancer Research, vol. 70, p. 288, 2010.
[71]  N. Carayol, E. Vakana, A. Sassano et al., “Critical roles for mTORC2- and rapamycin-insensitive mTORC1-complexes in growth and survival of BCR-ABL-expressing leukemic cells,” Proceedings of the National Academy of Sciences of the United States of America, vol. 107, no. 28, pp. 12469–12474, 2010.
[72]  S. Schenone, C. Brullo, F. Musumeci, M. Radi, and M. Botta, “ATP-competitive inhibitors of mTOR: an update,” Current Medicinal Chemistry, vol. 18, no. 20, pp. 2995–3014, 2011.
[73]  K. Yu, C. Shi, L. Toral-Barza et al., “Beyond rapalog therapy: preclinical pharmacology and antitumor activity of WYE-125132, an ATP-competitive and specific inhibitor of mTORC1 and mTORC2,” Cancer Research, vol. 70, no. 2, pp. 621–631, 2010.
[74]  C. C. Thoreen, S. A. Kang, J. W. Chang et al., “An ATP-competitive mammalian target of rapamycin inhibitor reveals rapamycin-resistant functions of mTORC1,” Journal of Biological Chemistry, vol. 284, no. 12, pp. 8023–8032, 2009.
[75]  Q.-W. Fan, Z. A. Knight, D. D. Goldenberg et al., “A dual PI3 kinase/mTOR inhibitor reveals emergent efficacy in glioma,” Cancer Cell, vol. 9, no. 5, pp. 341–349, 2006.
[76]  V. R. Agarwal, A. Joshi, and M. Venkataraman, “Abstract 3759: P7170, a novel inhibitor of phosphoinositide 3-kinase (PI3K)-mammalian target of Rapamycin (mTOR) and activin receptor-like kinase 1 (ALK1) as a new therapeutic option for Kras mutated non small cell lung cancer (NSCLC),” Cancer Research, vol. 72, Abstract 3759, 2012.
[77]  J. Yuan, P. P. Mehta, M.-J. Yin et al., “PF-04691502, a potent and selective oral inhibitor of PI3K and mTOR kinases with antitumor activity,” Molecular Cancer Therapeutics, vol. 10, no. 11, pp. 2189–2199, 2011.
[78]  S. J. Shuttleworth, F. A. Silva, A. R. L. Cecil et al., “Progress in the preclinical discovery and clinical development of class I and dual class I/IV phosphoinositide 3-kinase (PI3K) inhibitors,” Current Medicinal Chemistry, vol. 18, no. 18, pp. 2686–2714, 2011.
[79]  A. M. Venkatesan, C. M. Dehnhardt, E. D. Delos Santos et al., “Bis(morpholino-l,3,5-triazine) derivatives: potent adenosine 5′-triphosphate competitive phosphatidylinositol-3-kinase/mammalian target of rapamycin inhibitors: discovery of compound 26 (PKI-587), a highly efficacious dual inhibitor,” Journal of Medicinal Chemistry, vol. 53, no. 6, pp. 2636–2645, 2010.
[80]  A. M. Venkatesan, Z. Chen, O. D. Santos et al., “PKI-179: an orally efficacious dual phosphatidylinositol-3-kinase (PI3K)/mammalian target of rapamycin (mTOR) inhibitor,” Bioorganic and Medicinal Chemistry Letters, vol. 20, no. 19, pp. 5869–5873, 2010.
[81]  G. Prasad, T. Sottero, X. Yang et al., “Inhibition of PI3K/mTOR pathways in glioblastoma and implications for combination therapy with temozolomide,” Neuro-Oncology, vol. 13, no. 4, pp. 384–392, 2011.
[82]  S.-M. Maira, F. Stauffer, J. Brueggen et al., “Identification and characterization of NVP-BEZ235, a new orally available dual phosphatidylinositol 3-kinase/mammalian target of rapamycin inhibitor with potent in vivo antitumor activity,” Molecular Cancer Therapeutics, vol. 7, no. 7, pp. 1851–1863, 2008.
[83]  E. Leung, J. E. Kim, G. W. Rewcastle, G. J. Finlay, and B. C. Baguley, “Comparison of the effects of the PI3K/mTOR inhibitors NVP-BEZ235 and GSK2126458 on tamoxifen-resistant breast cancer cells,” Cancer Biology and Therapy, vol. 11, no. 11, pp. 938–946, 2011.
[84]  C. G. Sanchez, C. X. Ma, R. J. Crowder et al., “Preclinical modeling of combined phosphatidylinositol-3-kinase inhibition with endocrine therapy for estrogen receptor-positive breast cancer,” Breast Cancer Research, vol. 13, no. 2, article R21, 2011.
[85]  B. Markman, J. Tabernero, I. Krop, et al., “Phase I safety, pharmacokinetic, and pharmacodynamic study of the oral phosphatidylinositol-3-kinase and mTOR inhibitor BGT226 in patients with advanced solid tumors,” Annals of Oncology, vol. 23, pp. 2399–2408, 2012.
[86]  D. Mahadevan, E. G. Chiorean, D. D. Von Hoff, et al., “Phase I pharmacokinetic and pharmacodynamic study of the pan-PI3K/mTORC vascular targeted pro-drug SF1126 in patients with advanced solid tumours and B-cell malignancies,” European Journal of Cancer, vol. 48, pp. 3319–3327, 2012.
[87]  M. Kaeberlein and B. K. Kennedy, “Hot topics in aging research: protein translation and TOR signaling, 2010,” Aging Cell, vol. 10, no. 2, pp. 185–190, 2011.
[88]  S. D. Katewa and P. Kapahi, “Dietary restriction and aging, 2009,” Aging Cell, vol. 9, no. 2, pp. 105–112, 2010.
[89]  P. Kapahi, D. Chen, A. N. Rogers et al., “With TOR, less is more: a key role for the conserved nutrient-sensing TOR pathway in aging,” Cell Metabolism, vol. 11, no. 6, pp. 453–465, 2010.
[90]  J. Gallinetti, E. Harputlugil, and J. R. Mitchell, “Amino acid sensing in dietary-restriction-mediated longevity: roles of signal-transducing kinases GCN2 and TOR,” The Biochemical Journal, vol. 449, p. 1, 2013.
[91]  U. Kruegel, B. Robison, T. Dange et al., “Elevated proteasome capacity extends replicative lifespan in Saccharomyces cerevisiae,” PLoS Genetics, vol. 7, no. 9, Article ID e1002253, 2011.
[92]  N. D. Bonawitz, M. Chatenay-Lapointe, Y. Pan, and G. S. Shadel, “Reduced TOR signaling extends chronological life span via increased respiration and upregulation of mitochondrial gene expression,” Cell Metabolism, vol. 5, no. 4, pp. 265–277, 2007.
[93]  Y. Pan and G. S. Shadel, “Extension of chronological life span by reduced TOR signaling requires down-regulation of Sch9p and involves increased mitochondrial OXPHOS complex density,” Aging, vol. 1, no. 1, pp. 131–145, 2009.
[94]  M. Hansen, A. Chandra, L. L. Mitic, B. Onken, M. Driscoll, and C. Kenyon, “A role for autophagy in the extension of lifespan by dietary restriction in C. elegans,” PLoS Genetics, vol. 4, no. 2, article e24, 2008.
[95]  M. Hansen, S. Taubert, D. Crawford, N. Libina, S.-J. Lee, and C. Kenyon, “Lifespan extension by conditions that inhibit translation in Caenorhabditis elegans,” Aging Cell, vol. 6, no. 1, pp. 95–110, 2007.
[96]  T.-T. Ching, A. B. Paal, A. Mehta, L. Zhong, and A.-L. Hsu, “drr-2 encodes an eIF4H that acts downstream of TOR in diet-restriction-induced longevity of C. elegans,” Aging Cell, vol. 9, no. 4, pp. 545–557, 2010.
[97]  D. W. Lamming, L. Ye, P. Katajisto et al., “Rapamycin-induced insulin resistance is mediated by mTORC2 loss and uncoupled from longevity,” Science, vol. 335, no. 6076, pp. 1638–1643, 2012.
[98]  J. J. Wu, J. Liu, E. B. Chen, et al., “Increased mammalian lifespan and a segmental and tissue-specific slowing of aging after genetic reduction of mTOR expression,” Cell Reports, vol. 4, no. 5, pp. 913–920, 2013.
[99]  O. Medvedik, D. W. Lamming, K. D. Kim, and D. A. Sinclair, “MSN2 and MSN4 link calorie restriction and TOR to sirtuin-mediated lifespan extension in Saccharomyces cerevisiae,” PLoS Biology, vol. 5, no. 10, article e261, 2007.
[100]  C. Rallis, S. Codlin, and J. Bahler, “TORC1 signaling inhibition by rapamycin and caffeine affect lifespan, global gene expression, and cell proliferation of fission yeast,” Aging Cell, vol. 12, p. 563, 2013.
[101]  S. Robida-Stubbs, K. Glover-Cutter, D. W. Lamming et al., “TOR signaling and rapamycin influence longevity by regulating SKN-1/Nrf and DAF-16/FoxO,” Cell Metabolism, vol. 15, no. 5, pp. 713–724, 2012.
[102]  K. Seo, E. Choi, D. Lee, D. E. Jeong, S. K. Jang, and S. J. Lee, “Heat shock factor 1 mediates the longevity conferred by inhibition of TOR and insulin/IGF-1 signaling pathways in C. elegans,” Aging Cell, 2013.
[103]  I. Bjedov, J. M. Toivonen, F. Kerr et al., “Mechanisms of life span extension by rapamycin in the fruit fly Drosophila melanogaster,” Cell Metabolism, vol. 11, no. 1, pp. 35–46, 2010.
[104]  A. A. Moskalev and M. V. Shaposhnikov, “Pharmacological inhibition of phosphoinositide 3 and TOR kinases improves survival of drosophila melanogaster,” Rejuvenation Research, vol. 13, no. 2-3, pp. 246–247, 2010.
[105]  D. E. Harrison, R. Strong, Z. D. Sharp et al., “Rapamycin fed late in life extends lifespan in genetically heterogeneous mice,” Nature, vol. 460, no. 7253, pp. 392–395, 2009.
[106]  R. A. Miller, D. E. Harrison, C. M. Astle, et al., “Rapamycin, but not resveratrol or simvastatin, extends life span of genetically heterogeneous mice,” Journals of Gerontology, vol. 66, pp. 191–201, 2011.
[107]  J. E. Wilkinson, L. Burmeister, and S. V. Brooks, “Rapamycin slows aging in mice,” Aging Cell, vol. 11, pp. 675–682, 2012.
[108]  V. N. Anisimov, M. A. Zabezhinski, I. G. Popovich et al., “Rapamycin increases lifespan and inhibits spontaneous tumorigenesis in inbred female mice,” Cell Cycle, vol. 10, no. 24, pp. 4230–4236, 2011.
[109]  F. Neff, et al., “Rapamycin extends murine lifespan but has limited effects on aging,” The Journal of Clinical Investigation, vol. 123, pp. 3272–3291, 2013.
[110]  Y. Zhang, A. Bokov, J. Gelfond, et al., “Rapamycin extends life and health in C57BL/6 mice,” Journal of Gerontology, 2013.
[111]  C. Chen, Y. Liu, Y. Liu, and P. Zheng, “MTOR regulation and therapeutic rejuvenation of aging hematopoietic stem cells,” Science Signaling, vol. 2, no. 98, p. ra75, 2009.
[112]  A. E. Roux, A. Quissac, P. Chartrand, G. Ferbeyre, and L. A. Rokeach, “Regulation of chronological aging in Schizosaccharomyces pombe by the protein kinases Pka1 and Sck2,” Aging Cell, vol. 5, no. 4, pp. 345–357, 2006.
[113]  K. Z. Pan, J. E. Palter, A. N. Rogers et al., “Inhibition of mRNA translation extends lifespan in Caenorhabditis elegans,” Aging Cell, vol. 6, no. 1, pp. 111–119, 2007.
[114]  D. Chen, E. L. Thomas, and P. Kapahi, “HIF-1 modulates dietary restriction-mediated lifespan extension via IRE-1 in Caenorhabditis elegans,” PLoS Genetics, vol. 5, no. 5, Article ID e1000486, 2009.
[115]  B. M. Zid, A. N. Rogers, S. D. Katewa et al., “4E-BP extends lifespan upon dietary restriction by enhancing mitochondrial activity in Drosophila,” Cell, vol. 139, no. 1, pp. 149–160, 2009.
[116]  C. Selman, J. M. Tullet, D. Wieser, et al., “Ribosomal protein S6 kinase 1 signaling regulates mammalian life span,” Science, vol. 326, pp. 140–144, 2009.
[117]  R. Mehta, D. Chandler-Brown, F. J. Ramos, L. S. Shamieh, and M. Kaeberlein, “Regulation of mRNA translation as a conserved mechanism of longevity control,” Advances in Experimental Medicine and Biology, vol. 694, pp. 14–29, 2010.
[118]  B. K. Kennedy and M. Kaeberlein, “Hot topics in aging research: protein translation, 2009,” Aging Cell, vol. 8, no. 6, pp. 617–623, 2009.
[119]  P. Syntichaki, K. Troulinaki, and N. Tavernarakis, “eIF4E function in somatic cells modulates ageing in Caenorhabditis elegans,” Nature, vol. 445, no. 7130, pp. 922–926, 2007.
[120]  S. P. Curran and G. Ruvkun, “Lifespan regulation by evolutionarily conserved genes essential for viability,” PLoS Genetics, vol. 3, no. 4, e56, 2007.
[121]  J. R. Managbanag, T. M. Witten, D. Bonchev et al., “Shortest-path network analysis is a useful approach toward indentifying genetic determinants of longevity,” PLoS ONE, vol. 3, no. 11, Article ID e3802, 2008.
[122]  E. D. Smith, M. Tsuchiya, L. A. Fox et al., “Quantitative evidence for conserved longevity pathways between divergent eukaryotic species,” Genome Research, vol. 18, no. 4, pp. 564–570, 2008.
[123]  K. K. Steffen, V. L. MacKay, E. O. Kerr et al., “Yeast life span extension by depletion of 60S ribosomal subunits is mediated by Gcn4,” Cell, vol. 133, no. 2, pp. 292–302, 2008.
[124]  K. K. Steffen, M. A. McCormick, K. M. Pham et al., “Ribosome deficiency protects against ER stress in Saccharomyces cerevisiae,” Genetics, vol. 191, no. 1, pp. 107–118, 2012.
[125]  A. Chiocchetti, J. Zhou, H. Zhu et al., “Ribosomal proteins Rpl10 and Rps6 are potent regulators of yeast replicative life span,” Experimental Gerontology, vol. 42, no. 4, pp. 275–286, 2007.
[126]  S. Gelino and M. Hansen, “Autophagy—an emerging anti-aging mechanism,” Journal of Clinical & Experimental Pathology, 2012.
[127]  E. Lionaki, M. Markaki, and N. Tavernarakis, “Autophagy and ageing: insights from invertebrate model organisms,” Ageing Research Reviews, vol. 12, p. 413, 2013.
[128]  A. M. Cuervo, “Autophagy and aging: keeping that old broom working,” Trends in Genetics, vol. 24, no. 12, pp. 604–612, 2008.
[129]  J.-O. Pyo, S.-M. Yoo, and H. H. Ahn, “Overexpression of Atg5 in mice activates autophagy and extends lifespan,” Nature Communications, vol. 4, p. 2300, 2013.
[130]  R. A. Miller, D. E. Harrison, C. M. Astle et al., “An aging interventions testing program: study design and interim report,” Aging Cell, vol. 6, no. 4, pp. 565–575, 2007.
[131]  N. L. Nadon, R. Strong, R. A. Miller et al., “Design of aging intervention studies: the NIA interventions testing program,” Age, vol. 30, no. 4, pp. 187–199, 2008.
[132]  M. Kaeberlein and B. K. Kennedy, “Ageing: a midlife longevity drug?” Nature, 2009.
[133]  M. Kaeberlein, “Resveratrol and rapamycin: are they anti-aging drugs?” BioEssays, vol. 32, no. 2, pp. 96–99, 2010.
[134]  M. Kaeberlein and P. Kapahi, “Aging is RSKY business,” Science, vol. 326, no. 5949, pp. 55–56, 2009.
[135]  J. L. Kirkland and C. Peterson, “Healthspan, translation, and new outcomes for animal studies of aging,” Journals of Gerontology A, vol. 64, no. 2, pp. 209–212, 2009.
[136]  M. Tatar, “Can we develop genetically tractable models to assess healthspan (rather than life span) in animal models?” Journals of Gerontology A, vol. 64, no. 2, pp. 161–163, 2009.
[137]  M. V. Blagosklonny, “Prospective treatment of age-related diseases by slowing down aging,” The American Journal of Pathology, vol. 181, p. 1142, 2012.
[138]  D. S. Evans, P. Kapahi, W.-C. Hsueh, and L. Kockel, “TOR signaling never gets old: aging, longevity and TORC1 activity,” Ageing Research Reviews, vol. 10, no. 2, pp. 225–237, 2011.
[139]  M. Kaeberlein and B. K. Kennedy, “Protein translation, 2008,” Aging Cell, vol. 7, no. 6, pp. 777–782, 2008.
[140]  M. Cornu, V. Albert, and M. N. Hall, “mTOR in aging, metabolism, and cancer,” Current Opinion in Genetics & Development, vol. 23, p. 53, 2013.
[141]  P. Hasty, Z. D. Sharp, T. J. Curiel, and J. Campisi, “mTORC1 and p53: clash of the gods?” Cell Cycle, vol. 12, p. 20, 2013.
[142]  F. J. Ramos and M. Kaeberlein, “Ageing: a healthy diet for stem cells,” Nature, vol. 486, p. 477, 2012.
[143]  C. Lerner, A. Bitto, D. Pulliam, et al., “Reduced mammalian target of rapamycin activity facilitates mitochondrial retrograde signaling and increases life span in normal human fibroblasts,” Aging Cell, 2013.
[144]  T. V. Pospelova, “Suppression of replicative senescence by rapamycin in rodent embryonic cells,” Cell Cycle, vol. 11, pp. 2402–2407, 2012.
[145]  V. Dulic, “Senescence regulation by mTOR,” Methods in Molecular Biology, vol. 965, p. 15, 2013.
[146]  E. Dazert and M. N. Hall, “MTOR signaling in disease,” Current Opinion in Cell Biology, vol. 23, no. 6, pp. 744–755, 2011.
[147]  Z. Yang and X. F. Ming, “mTOR signalling: the molecular interface connecting metabolic stress, aging and cardiovascular diseases,” Obesity Reviews, vol. 13, supplement 2, p. 58, 2012.
[148]  J. Bové, M. Martínez-Vicente, and M. Vila, “Fighting neurodegeneration with rapamycin: mechanistic insights,” Nature Reviews Neuroscience, vol. 12, no. 8, pp. 437–452, 2011.
[149]  E. Aso and I. Ferrer, “It may be possible to delay the onset of neurodegenerative diseases with an immunosuppressive drug (rapamycin),” Expert Opinion on Biological Therapy, vol. 13, p. 1215, 2013.
[150]  E. Wong and A. M. Cuervo, “Autophagy gone awry in neurodegenerative diseases,” Nature Neuroscience, vol. 13, no. 7, pp. 805–811, 2010.
[151]  D. C. Rubinsztein, “The roles of intracellular protein-degradation pathways in neurodegeneration,” Nature, vol. 443, no. 7113, pp. 780–786, 2006.
[152]  C. Malagelada, Z. H. Jin, V. Jackson-Lewis, S. Przedborski, and L. A. Greene, “Rapamycin protects against neuron death in in vitro and in vivo models of Parkinson's disease,” Journal of Neuroscience, vol. 30, no. 3, pp. 1166–1175, 2010.
[153]  L. S. Tain, H. Mortiboys, R. N. Tao, E. Ziviani, O. Bandmann, and A. J. Whitworth, “Rapamycin activation of 4E-BP prevents parkinsonian dopaminergic neuron loss,” Nature Neuroscience, vol. 12, no. 9, pp. 1129–1135, 2009.
[154]  K. Liu, N. Shi, Y. Sun, T. Zhang, and X. Sun, “Therapeutic effects of rapamycin on MPTP-induced Parkinsonism in mice,” Neurochemical Research, vol. 38, p. 201, 2013.
[155]  B. Ravikumar, C. Vacher, Z. Berger et al., “Inhibition of mTOR induces autophagy and reduces toxicity of polyglutamine expansions in fly and mouse models of Huntington disease,” Nature Genetics, vol. 36, no. 6, pp. 585–595, 2004.
[156]  A. Roscic, B. Baldo, C. Crochemore, D. Marcellin, and P. Paganetti, “Induction of autophagy with catalytic mTOR inhibitors reduces huntingtin aggregates in a neuronal cell model,” Journal of Neurochemistry, vol. 119, no. 2, pp. 398–407, 2011.
[157]  S. Oddo, “The role of mTOR signaling in Alzheimer disease,” Frontiers in Bioscience, vol. 4, p. 941, 2012.
[158]  Y. X. Sun, X. Ji, X. Mao, et al., “Differential activation of mTOR complex 1 signaling in human brain with mild to severe Alzheimer's disease,” Journal of Alzheimer's Disease, 2013.
[159]  S. Majumder, A. Richardson, R. Strong, and S. Oddo, “Inducing autophagy by rapamycin before, but not after, the formation of plaques and tangles ameliorates cognitive deficits,” PLoS ONE, vol. 6, no. 9, Article ID e25416, 2011.
[160]  P. Spilman, N. Podlutskaya, M. J. Hart et al., “Inhibition of mTOR by rapamycin abolishes cognitive deficits and reduces amyloid-β levels in a mouse model of alzheimer's disease,” PLoS ONE, vol. 5, no. 4, Article ID e9979, 2010.
[161]  A. L. Lin, W. Zheng, J. J. Halloran, et al., “Chronic rapamycin restores brain vascular integrity and function through NO synthase activation and improves memory in symptomatic mice modeling Alzheimer's disease,” Journal of Cerebral Blood Flow and Metabolism, vol. 33, p. 1412, 2013.
[162]  C. A. Hoeffer and E. Klann, “mTOR signaling: at the crossroads of plasticity, memory and disease,” Trends in Neurosciences, vol. 33, no. 2, pp. 67–75, 2010.
[163]  J. Halloran, S. A. Hussong, R. Burbank, et al., “Chronic inhibition of mammalian target of rapamycin by rapamycin modulates cognitive and non-cognitive components of behavior throughout lifespan in mice,” Neuroscience, vol. 223, pp. 102–113, 2012.
[164]  S. Majumder, A. Caccamo, D. X. Medina et al., “Lifelong rapamycin administration ameliorates age-dependent cognitive deficits by reducing IL-1β and enhancing NMDA signaling,” Aging Cell, vol. 11, no. 2, pp. 326–335, 2012.
[165]  J. Xie and T. P. Herbert, “The role of mammalian target of rapamycin (mTOR) in the regulation of pancreatic β-cell mass: implications in the development of type-2 diabetes,” Cellular and Molecular Life Sciences, vol. 69, pp. 1289–1304, 2012.
[166]  K. Garber, “Targeting mTOR: something old, something new,” Journal of the National Cancer Institute, vol. 101, no. 5, pp. 288–290, 2009.
[167]  M. V. Blagosklonny, “Rapalogs in cancer prevention: anti-aging or anticancer?” Cancer Biology & Therapy, vol. 13, p. 1349, 2012.
[168]  Z. D. Sharp and A. Richardson, “Aging and cancer: can mTOR inhibitors kill two birds with one drug?” Targeted Oncology, vol. 6, no. 1, pp. 41–51, 2011.
[169]  E. A. Komarova, M. P. Antoch, L. R. Novototskaya, et al., “Rapamycin extends lifespan and delays tumorigenesis in heterozygous p53+/- mice,” Aging, vol. 4, pp. 709–714, 2012.
[170]  C. B. Livi, R. L. Hardman, B. A. Christy, et al., “Rapamycin extends life span of Rb1+/- mice by inhibiting neuroendocrine tumors,” Aging, vol. 5, pp. 100–110, 2013.
[171]  A. Arcella, F. Biagioni, M. Antonietta Oliva, et al., “Rapamycin inhibits the growth of glioblastoma,” Brain Research, vol. 1495, pp. 37–51, 2013.
[172]  O. Ekshyyan, T. N. Moore-Medlin, M. C. Raley, et al., “Anti-lymphangiogenic properties of mTOR inhibitors in head and neck squamous cell carcinoma experimental models,” BMC Cancer, vol. 13, article 320, 2013.
[173]  J. E. Hartwich, W. S. Orr, and C.Y. Ng, “Rapamycin increases neuroblastoma xenograft and host stromal derived osteoprotegerin inhibiting osteolytic bone disease in a bone metastasis model,” Journal of Pediatric Surgery, vol. 48, pp. 47–55, 2013.
[174]  S. Z. Kaylani, J. Xu, R. K. Srivastava, et al., “Rapamycin targeting mTOR and hedgehog signaling pathways blocks human rhabdomyosarcoma growth in xenograft murine model,” Biochemical and Biophysical Research Communications, vol. 435, pp. 557–561, 2013.
[175]  T. Nishikawa, M. Takaoka, T. Ohara, et al., “Antiproliferative effect of a novel mTOR inhibitor temsirolimus contributes to the prolonged survival of orthotopic esophageal cancer-bearing mice,” Cancer Biology & Therapy, vol. 14, pp. 230–236, 2013.
[176]  D. Pachow, N. Andrae, N. Kliese, et al., “mTORC1 inhibitors suppress meningioma growth in mouse models,” Clinical Cancer Research, vol. 19, p. 1180, 2013.
[177]  R. J. O. Dowling, I. Topisirovic, B. D. Fonseca, and N. Sonenberg, “Dissecting the role of mTOR: lessons from mTOR inhibitors,” Biochimica et Biophysica Acta, vol. 1804, no. 3, pp. 433–439, 2010.
[178]  A. Fasolo and C. Sessa, “Targeting mTOR pathways in human malignancies,” Current Pharmaceutical Design, vol. 18, p. 2766, 2012.
[179]  J. M. Flynn, “Late-life rapamycin treatment reverses age-related heart dysfunction,” Aging Cell, vol. 12, no. 5, pp. 851–862, 2013.
[180]  J. Sadoshima and S. Izumo, “Rapamycin selectively inhibits angiotensin II-induced increase in protein synthesis in cardiac myocytes in vitro: potential role of 70-kD S6 kinase in angiotensin II-induced cardiac hypertrophy,” Circulation Research, vol. 77, no. 6, pp. 1040–1052, 1995.
[181]  T. Shioi, J. R. McMullen, O. Tarnavski et al., “Rapamycin attenuates load-induced cardiac hypertrophy in mice,” Circulation, vol. 107, no. 12, pp. 1664–1670, 2003.
[182]  J. R. McMullen, M. C. Sherwood, O. Tarnavski et al., “Inhibition of mTOR signaling with rapamycin regresses established cardiac hypertrophy induced by pressure overload,” Circulation, vol. 109, no. 24, pp. 3050–3055, 2004.
[183]  J. A. Kuzman, T. D. O'Connell, and A. M. Gerdes, “Rapamycin prevents thyroid hormone-induced cardiac hypertrophy,” Endocrinology, vol. 148, no. 7, pp. 3477–3484, 2007.
[184]  S. A. Khan, F. Salloum, A. Das, L. Xi, G. W. Vetrovec, and R. C. Kukreja, “Rapamycin confers preconditioning-like protection against ischemia-reperfusion injury in isolated mouse heart and cardiomyocytes,” Journal of Molecular and Cellular Cardiology, vol. 41, no. 2, pp. 256–264, 2006.
[185]  A. Das, F. N. Salloum, D. Durrant, R. Ockaili, and R. C. Kukreja, “Rapamycin protects against myocardial ischemia-reperfusion injury through JAK2-STAT3 signaling pathway,” Journal of Molecular and Cellular Cardiology, vol. 53, p. 858, 2012.
[186]  T. M. Marin, K. Keith, B. Davies et al., “Rapamycin reverses hypertrophic cardiomyopathy in a mouse model of LEOPARD syndrome-associated PTPN11 mutation,” Journal of Clinical Investigation, vol. 121, no. 3, pp. 1026–1043, 2011.
[187]  K. Xie, B. Jin, Y. Li et al., “Modulating autophagy improves cardiac function in a rat model of early-stage dilated cardiomyopathy,” Cardiology, vol. 125, no. 1, pp. 60–68, 2013.
[188]  F. J. Ramos, S. C. Chen, M. G. Garelick, et al., “Rapamycin reverses elevated mTORC1 signaling in lamin A/C-deficient mice, rescues cardiac and skeletal muscle function, and extends survival,” Science Translational Medicine, vol. 4, p. 144ra103, 2012.
[189]  J. C. Choi and H. J. Worman, “Reactivation of autophagy ameliorates LMNA cardiomyopathy,” Autophagy, vol. 9, p. 110, 2013.
[190]  J. C. Choi, A. Muchir, W. Wu, et al., “Temsirolimus activates autophagy and ameliorates cardiomyopathy caused by lamin A/C gene mutation,” Science Translational Medicine, vol. 4, p. 144ra102, 2012.
[191]  Y. Ding, X. Sun, W. Huang et al., “Haploinsufficiency of target of rapamycin attenuates cardiomyopathies in adult zebrafish,” Circulation Research, vol. 109, no. 6, pp. 658–669, 2011.
[192]  J. M. Carrascosa, M. Ros, A. Andrés, T. Fernández-Agulló, and C. Arribas, “Changes in the neuroendocrine control of energy homeostasis by adiposity signals during aging,” Experimental Gerontology, vol. 44, no. 1-2, pp. 20–25, 2009.
[193]  A. M. Chang and J. B. Halter, “Effects of aging on glucose homeostasis,” in DiabetesMellitus: A Fundamental and Clinical Text, D. Le Roith, S. I. Taylor, and J. M. Olefsky, Eds., pp. 869–877, Lippincott Williams & Wilkins, 2004.
[194]  J. Chen, “Multiple signal pathways in obesity-associated cancer,” Obesity Reviews, vol. 12, no. 12, pp. 1063–1070, 2011.
[195]  I. Bakan and M. Laplante, “Connecting mTORC1 signaling to SREBP-1 activation,” Current Opinion in Lipidology, vol. 2, p. 226, 2012.
[196]  M. Pende, S. C. Kozma, M. Jaquet et al., “Hypoinsulinaemia, glucose intolerance and diminished β-cell size in S6K1-deficient mice,” Nature, vol. 408, no. 6815, pp. 994–997, 2000.
[197]  P. Polak, N. Cybulski, J. N. Feige, J. Auwerx, M. A. Rüegg, and M. N. Hall, “Adipose-specific knockout of raptor results in lean mice with enhanced mitochondrial respiration,” Cell Metabolism, vol. 8, no. 5, pp. 399–410, 2008.
[198]  S. H. Um, F. Frigerio, M. Watanabe, et al., “Absence of S6K1 protects against age- and diet-induced obesity while enhancing insulin sensitivity,” Nature, vol. 431, pp. 200–205, 2004.
[199]  A. D. Barlow, M. L. Nicholson, and T. P. Herbert, “Evidence for rapamycin toxicity in pancreatic beta-cells and a review of the underlying molecular mechanisms,” Diabetes, vol. 62, p. 2674, 2013.
[200]  S. S. Deepa, M. E. Walsh, R. T. Hamilton, et al., “Rapamycin modulates markers of mitochondrial biogenesis and fatty acid oxidation in the adipose tissue of db/db mice,” Journal of Biochemical and Pharmacological Research, vol. 1, pp. 114–123, 2013.
[201]  M. V. Blagosklonny, “Once again on rapamycin-induced insulin resistance and longevity: despite of or owing to,” Aging, vol. 4, p. 350, 2012.
[202]  C. Jagannath and P. Bakhru, “Rapamycin-induced enhancement of vaccine efficacy in mice,” Methods in Molecular Biology, vol. 821, pp. 295–303, 2012.
[203]  E. Amiel, B. Everts, and T. C. Freitas, “Inhibition of mechanistic target of rapamycin promotes dendritic cell activation and enhances therapeutic autologous vaccination in mice,” Journal of Immunology, vol. 189, no. 5, pp. 2151–2158, 2012.
[204]  K. Araki, A. P. Turner, V. O. Shaffer et al., “mTOR regulates memory CD8 T-cell differentiation,” Nature, vol. 460, no. 7251, pp. 108–112, 2009.
[205]  S. Anand, K. L. Johansen, and M. Kurella Tamura, “Aging and chronic kidney disease: the impact on physical function and cognition,” Journal of Gerontology, 2013.
[206]  P. Stenvinkel and T. E. Larsson, “Chronic kidney disease: a clinical model of premature aging,” American Journal of Kidney Diseases, vol. 62, p. 339, 2013.
[207]  W. Lieberthal and J. S. Levine, “Mammalian target of rapamycin and the kidney—II. Pathophysiology and therapeutic implications,” American Journal of Physiology: Renal Physiology, vol. 303, p. F180, 2012.
[208]  E. Leung and G. Landa, “Update on current and future novel therapies for dry age-related macular degeneration,” Expert Review of Clinical Pharmacology, 2013.
[209]  A. Stahl, L. Paschek, G. Martin et al., “Rapamycin reduces VEGF expression in retinal pigment epithelium (RPE) and inhibits RPE-induced sprouting angiogenesis in vitro,” FEBS Letters, vol. 582, no. 20, pp. 3097–3102, 2008.
[210]  N. G. Kolosova, N. A. Muraleva, A. A. Zhdankina, N. A. Stefanova, A. Z. Fursova, and M. V. Blagosklonny, “Prevention of age-related macular degeneration-like retinopathy by rapamycin in rats,” American Journal of Pathology, vol. 181, pp. 472–477, 2012.
[211]  R. B. Nussenblatt, G. Byrnes, H. N. Sen et al., “A randomized pilot study of systemic immunosuppression in the treatment of age-related macular degeneration with choroidal neovascularization,” Retina, vol. 30, no. 10, pp. 1579–1587, 2010.
[212]  C. R. Burtner and B. K. Kennedy, “Progeria syndromes and ageing: what is the connection?” Nature Reviews Molecular Cell Biology, vol. 11, no. 8, pp. 567–578, 2010.
[213]  K. Cao, J. J. Graziotto, C. D. Blair et al., “Rapamycin reverses cellular phenotypes and enhances mutant protein clearance in Hutchinson-Gilford progeria syndrome cells,” Science Translational Medicine, vol. 3, no. 89, p. 89ra58, 2011.
[214]  J. J. Graziotto, K. Cao, F. S. Collins, and D. Krainc, “Rapamycin activates autophagy in Hutchinson-Gilford progeria syndrome: implications for normal aging and age-dependent neurodegenerative disorders,” Autophagy, vol. 8, no. 1, pp. 147–151, 2012.
[215]  P. Curatolo, R. Bombardieri, and S. Jozwiak, “Tuberous sclerosis,” The Lancet, vol. 372, no. 9639, pp. 657–668, 2008.
[216]  J. P. Osborne, A. Fryer, and D. Webb, “Epidemiology of tuberous sclerosis,” Annals of the New York Academy of Sciences, vol. 615, pp. 125–127, 1991.
[217]  P. B. Crino, K. L. Nathanson, and E. P. Henske, “The tuberous sclerosis complex,” The New England Journal of Medicine, vol. 355, no. 13, pp. 1345–1356, 2006.
[218]  K. Ridler, J. Suckling, N. J. Higgins et al., “Neuroanatomical correlates of memory deficits in tuberous sclerosis complex,” Cerebral Cortex, vol. 17, no. 2, pp. 261–271, 2007.
[219]  S. L. Smalley, “Autism and tuberous sclerosis,” Journal of Autism and Developmental Disorders, vol. 28, no. 5, pp. 407–414, 1998.
[220]  P. Prather and P. J. de Vries, “Behavioral and cognitive aspects of tuberous sclerosis complex,” Journal of Child Neurology, vol. 19, no. 9, pp. 666–674, 2004.
[221]  P. Curatolo and R. Moavero, “mTOR inhibitors as a new therapeutic option for epilepsy,” Expert Review of Neurotherapeutics, vol. 13, p. 627, 2013.
[222]  J. R. Sampson, “Therapeutic targeting of mTOR in tuberous sclerosis,” Biochemical Society Transactions, vol. 37, no. 1, pp. 259–264, 2009.
[223]  L. Meikle, K. Pollizzi, A. Egnor et al., “Response of a neuronal model of tuberous sclerosis to mammalian target of rapamycin (mTOR) inhibitors: effects on mTORC1 and Akt signaling lead to improved survival and function,” Journal of Neuroscience, vol. 28, no. 21, pp. 5422–5432, 2008.
[224]  N. Lee, C. L. Woodrum, A. M. Nobil, A. E. Rauktys, M. P. Messina, and S. L. Dabora, “Rapamycin weekly maintenance dosing and the potential efficacy of combination sorafenib plus rapamycin but not atorvastatin or doxycycline in tuberous sclerosis preclinical models,” BMC Pharmacology, vol. 9, p. 8, 2009.
[225]  M. Wong, “A critical review of mTOR inhibitors and epilepsy: from basic science to clinical trials,” Expert Review of Neurotherapeutics, vol. 13, p. 657, 2013.
[226]  D. A. Krueger, M. M. Care, K. Holland et al., “Everolimus for subependymal giant-cell astrocytomas in tuberous sclerosis,” New England Journal of Medicine, vol. 363, no. 19, pp. 1801–1811, 2010.
[227]  R. Moavero, M. Pinci, R. Bombardieri, and P. Curatolo, “The management of subependymal giant cell tumors in tuberous sclerosis: a clinician's perspective,” Child's Nervous System, vol. 27, no. 8, pp. 1203–1210, 2011.
[228]  J. J. Bissler, F. X. McCormack, L. R. Young et al., “Sirolimus for angiomyolipoma in tuberous sclerosis complex or lymphangioleiomyomatosis,” New England Journal of Medicine, vol. 358, no. 2, pp. 140–151, 2008.
[229]  D. M. Davies, P. J. De Vries, S. R. Johnson et al., “Sirolimus therapy for angiomyolipoma in tuberous sclerosis and sporadic lymphangioleiomyomatosis: a phase 2 trial,” Clinical Cancer Research, vol. 17, no. 12, pp. 4071–4081, 2011.
[230]  D. M. Davies, S. R. Johnson, A. E. Tattersfield et al., “Sirolimus therapy in tuberous sclerosis or sporadic lymphangioleiomyomatosis,” New England Journal of Medicine, vol. 358, no. 2, pp. 200–203, 2008.
[231]  T. T. Gipson and M. V. Johnston, “Plasticity and mTOR: towards restoration of impaired synaptic plasticity in mTOR-related neurogenetic disorders,” Neural Plasticity, vol. 2012, Article ID 486402, 10 pages, 2012.
[232]  X. F. Meng, J. T. Yu, J. H. Song, S. Chi, and L. Tan, “Role of the mTOR signaling pathway in epilepsy,” Journal of the Neurological Sciences, vol. 332, p. 4, 2013.
[233]  M. Wong and P. B. Crino, Jasper's Basic Mechanisms of the Epilepsies, edited by J. L. Noebels, M. Avoli, M. A. Rogawski, R. W. Olsen, A. V. Delgado-Escueta, Bethesda, Md, USA, 2012.
[234]  C.-H. Kwon, X. Zhu, J. Zhang, and S. J. Baker, “mTor is required for hypertrophy of Pten-deficient neuronal soma in vivo,” Proceedings of the National Academy of Sciences of the United States of America, vol. 100, no. 22, pp. 12923–12928, 2003.
[235]  A. S. Galanopoulou, J. A. Gorter, and C. Cepeda, “Finding a better drug for epilepsy: the mTOR pathway as an antiepileptogenic target,” Epilepsia, vol. 53, p. 1119, 2012.
[236]  K. Heng, M. M. Haney, and P. S. Buckmaster, “High-dose rapamycin blocks mossy fiber sprouting but not seizures in a mouse model of temporal lobe epilepsy,” Epilepsia, vol. 54, p. 1535, 2013.
[237]  P. F. Bolton, R. J. Park, J. N. P. Higgins, P. D. Griffiths, and A. Pickles, “Neuro-epileptic determinants of autism spectrum disorders in tuberous sclerosis complex,” Brain, vol. 125, no. 6, pp. 1247–1255, 2002.
[238]  E. Fombonne, “Epidemiological surveys of autism and other pervasive developmental disorders: an update,” Journal of Autism and Developmental Disorders, vol. 33, no. 4, pp. 365–382, 2003.
[239]  M. G. Butler, M. J. Dazouki, X.-P. Zhou et al., “Subset of individuals with autism spectrum disorders and extreme macrocephaly associated with germline PTEN tumour suppressor gene mutations,” Journal of Medical Genetics, vol. 42, no. 4, pp. 318–321, 2005.
[240]  M. Neves-Pereira, B. Müller, D. Massie et al., “Deregulation of EIF4E: a novel mechanism for autism,” Journal of Medical Genetics, vol. 46, no. 11, pp. 759–765, 2009.
[241]  D. Ehninger and A. J. Silva, “Rapamycin for treating Tuberous sclerosis and Autism spectrum disorders,” Trends in Molecular Medicine, vol. 17, no. 2, pp. 78–87, 2011.
[242]  J. Hughes, C. J. Ward, B. Peral et al., “The polycystic kidney disease 1 (PKD1) gene encodes a novel protein with multiple cell recognition domains,” Nature Genetics, vol. 10, no. 2, pp. 151–160, 1995.
[243]  T. Weimbs, “Regulation of mTOR by polycystin-1: is polycystic kidney disease a case of futile repair?” Cell Cycle, vol. 5, no. 21, pp. 2425–2429, 2006.
[244]  J. M. Shillingford, N. S. Murcia, C. H. Larson et al., “The mTOR pathway is regulated by polycystin-1, and its inhibition reverses renal cystogenesis in polycystic kidney disease,” Proceedings of the National Academy of Sciences of the United States of America, vol. 103, no. 14, pp. 5466–5471, 2006.
[245]  S. M. Flechner, D. Goldfarb, C. Modlin et al., “Kidney transplantation without calcineurin inhibitor drugs: a prospective, randomized trial of sirolimus versus cyclosporine,” Transplantation, vol. 74, no. 8, pp. 1070–1076, 2002.
[246]  G. Stallone, B. Infante, G. Grandaliano, et al., “Rapamycin for treatment of type I autosomal dominant polycystic kidney disease (RAPYD-study): a randomized, controlled study,” Nephrology, Dialysis, Transplantation, vol. 27, pp. 3560–3567, 2012.
[247]  N. Perico and G. Remuzzi, “Do mTOR inhibitors still have a future in ADPKD?” Nature Reviews, vol. 6, no. 12, pp. 696–698, 2010.
[248]  N. Perico, L. Antiga, A. Caroli et al., “Sirolimus therapy to halt the progression of ADPKD,” Journal of the American Society of Nephrology, vol. 21, no. 6, pp. 1031–1040, 2010.
[249]  I. Obara, S. M. Géranton, and S. P. Hunt, “Axonal protein synthesis: a potential target for pain relief?” Current Opinion in Pharmacology, vol. 12, no. 1, pp. 42–48, 2012.
[250]  S. M. Géranton, L. Jiménez-Díaz, C. Torsney et al., “A rapamycin-sensitive signaling pathway is essential for the full expression of persistent pain states,” Journal of Neuroscience, vol. 29, no. 47, pp. 15017–15027, 2009.
[251]  E. Norsted Gregory, S. Codeluppi, J. A. Gregory, J. Steinauer, and C. I. Svensson, “Mammalian target of rapamycin in spinal cord neurons mediates hypersensitivity induced by peripheral inflammation,” Neuroscience, vol. 169, no. 3, pp. 1392–1402, 2010.
[252]  I. Obara, K. K. Tochiki, S. M. Géranton et al., “Systemic inhibition of the mammalian target of rapamycin (mTOR) pathway reduces neuropathic pain in mice,” Pain, vol. 152, no. 11, pp. 2582–2595, 2011.
[253]  W. Zhang, X. F. Sun, and J. H. Bo, “Activation of mTOR in the spinal cord is required for pain hypersensitivity induced by chronic constriction injury in mice,” Pharmacology, Biochemistry, and Behavior, vol. 111, pp. 64–70, 2013.
[254]  L. Jiménez-Díaz, S. M. Géranton, G. M. Passmore et al., “Local translation in primary afferent fibers regulates nociception,” PLoS ONE, vol. 3, no. 4, Article ID e1961, 2008.
[255]  D. Guo, L. Zeng, D. L. Brody, and M. Wong, “Rapamycin attenuates the development of posttraumatic epilepsy in a mouse model of traumatic brain injury,” PLoS ONE, vol. 8, Article ID e64078, 2013.
[256]  A. S. Don, C. K. Tsang, et al., “Targeting mTOR as a novel therapeutic strategy for traumatic CNS injuries,” Drug Discovery Today, vol. 17, pp. 861–868, 2012.
[257]  J. Park, J. Zhang, J. Qiu et al., “Combination therapy targeting Akt and mammalian target of rapamycin improves functional outcome after controlled cortical impact in mice,” Journal of Cerebral Blood Flow and Metabolism, vol. 32, no. 2, pp. 330–340, 2012.
[258]  S. Erlich, A. Alexandrovich, E. Shohami, and R. Pinkas-Kramarski, “Rapamycin is a neuroprotective treatment for traumatic brain injury,” Neurobiology of Disease, vol. 26, no. 1, pp. 86–93, 2007.
[259]  J. Wang, K. Lu, F. Liang, et al., “Decreased autophagy contributes to myocardial dysfunction in rats subjected to nonlethal mechanical trauma,” PLoS ONE, vol. 8, Article ID e71400, 2013.
[260]  P. Tang, H. Hou, L. Zhang, et al., “Autophagy reduces neuronal damage and promotes locomotor recovery via inhibition of apoptosis after spinal cord injury in rats,” Molecular Neurobiology, 2013.

Full-Text

comments powered by Disqus

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133