Skip to main navigation menu Skip to main content Skip to site footer

Review article: Biomedical intelligence

Vol. 144 No. 0708 (2014)

Selective manipulation of aging: a novel strategy for the treatment of neurodegenerative disorders

  • Ehud Cohen
  • Lorna Moll
  • Tayir El-Ami
DOI
https://doi.org/10.4414/smw.2014.13917
Cite this as:
Swiss Med Wkly. 2014;144:w13917
Published
09.02.2014

Summary

Aging is the major risk factor for the development of human neurodegenerative maladies such as Alzheimer’s, Huntington’s and Parkinson’s diseases, and prion disorders, all of which stem from toxic protein aggregation. Although sporadic cases typically onset during the patient’s seventh decade of life or later, mutation-linked, familial disorders manifest during the fifth or sixth decade of life. This common temporal emergence pattern suggests that slowing aging can postpone the onset of these maladies and alleviate their symptoms once emerged. Studies in worms and flies that express disease-linked aggregative proteins revealed that reducing the activity of the insulin / insulin-like growth factor (IGF) signalling (IIS), a prominent aging regulatory pathway, protects these animals from toxic protein aggregation. The therapeutic potential of this approach has been tested and confirmed in mammals as reducing the activity of the IGF1 signalling cascade partially protects Alzheimer’s-model mice from premature death, and behavioural and pathological impairments associated with the disorder. Here we review the recent advances in the field, describe the known mechanistic links between toxic protein aggregation, neurodegenerative disorders and the aging process and delineate recent studies that point at IGF1 signalling inhibitors as promising therapies for the treatment of various late-onset neurodegenerative disorders.

References

  1. Hartl FU, Bracher A, Hayer-Hartl M. Molecular chaperones in protein folding and proteostasis. Nature. 2011;475:324–32.
  2. Ciechanover A. The ubiquitin proteolytic system: from a vague idea, through basic mechanisms, and onto human diseases and drug targeting. Neurology. 2006;66:S7–19.
  3. Wong E, Cuervo AM. Autophagy gone awry in neurodegenerative diseases. Nat Neurosci. 2010;13:805–11.
  4. Balch WE, Morimoto RI, Dillin A, Kelly JW. Adapting proteostasis for disease intervention. Science. 2008;319:916–9.
  5. Kaganovich D, Kopito R, Frydman J. Misfolded proteins partition between two distinct quality control compartments. Nature. 2008;454:1088–95.
  6. Arrasate M, Mitra S, Schweitzer ES, Segal MR, Finkbeiner S. Inclusion body formation reduces levels of mutant huntingtin and the risk of neuronal death. Nature. 2004;31:805-–10.
  7. Ben-Gedalya T, Cohen E. Quality control compartments coming of age. Traffic. 2012;13:635–42.
  8. Kopito RR, Ron D. Conformational disease. Nat Cell Biol. 2000;2:E207–209.
  9. Soto C. Unfolding the role of protein misfolding in neurodegenerative diseases. Nat Rev Neurosci. 2003;4:49–60.
  10. Selkoe DJ. Alzheimer’s disease. Cold Spring Harb Perspect Biol. 2011;3(7).
  11. Shankar GM, Li S, Mehta TH, Garcia-Munoz A, Shepardson NE, et al. (2008) Amyloid-beta protein dimers isolated directly from Alzheimer’s brains impair synaptic plasticity and memory. Nat Med. 2008;14(8):837‒42.
  12. O’Nuallain B, Freir DB, Nicoll AJ, Risse E, Ferguson N, et al. Amyloid beta-protein dimers rapidly form stable synaptotoxic protofibrils. J Neurosci. 2010;30:14411–9.
  13. Campion D, Flaman JM, Brice A, Hannequin D, Dubois B, Martin C, et al. Mutations of the presenilin/gene in families with early-onset Alzheimer’s disease. Hum Mol Genet. 1995;4:2373–7.
  14. Jonsson T, Atwal JK, Steinberg S, Snaedal J, Jonsson PV, et al. A mutation in APP protects against Alzheimer’s disease and age-related cognitive decline. Nature. 2012;488(7409):96‒9.
  15. Chavez-Gutierrez L, Bammens L, Benilova I, Vandersteen A, Benurwar M, et al. The mechanism of gamma-Secretase dysfunction in familial Alzheimer disease. Embo J. 2012;31:2261–74.
  16. Shen J, Kelleher RJ, 3rd. The presenilin hypothesis of Alzheimer’s disease: evidence for a loss-of-function pathogenic mechanism. Proc Natl Acad Sci U S A 2007;104:403–9.
  17. Coen K, Flannagan RS, Baron S, Carraro-Lacroix LR, Wang D, et al. Lysosomal calcium homeostasis defects, not proton pump defects, cause endo-lysosomal dysfunction in PSEN-deficient cells. J Cell Biol. 2012;198:23–35.
  18. Lee JH, Yu WH, Kumar A, Lee S, Mohan PS, et al. Lysosomal proteolysis and autophagy require presenilin 1 and are disrupted by Alzheimer-related PS1 mutations. Cell. 2010;141:1146–58.
  19. Area-Gomez E, Del Carmen Lara Castillo M, Tambini MD, Guardia-Laguarta C, de Groof AJ, et al. Upregulated function of mitochondria-associated ER membranes in Alzheimer disease. Embo J. 2012;31:4106–23.
  20. Bates G. Huntingtin aggregation and toxicity in Huntington’s disease. Lancet. 2003;361:1642–4.
  21. Durcan TM, Fon EA. Ataxin-3 and its e3 partners: implications for machado-joseph disease. Front Neurol. 2013;4:46.
  22. Robberecht W, Philips T. The changing scene of amyotrophic lateral sclerosis. Nat Rev Neurosci. 2013;14:248–64.
  23. Neumann M, Sampathu DM, Kwong LK, Truax AC, Micsenyi MC, et al. Ubiquitinated TDP-43 in frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Science. 2006;314:130–3.
  24. Rosen DR, Siddique T, Patterson D, Figlewicz DA, Sapp P, et al. Mutations in Cu/Zn superoxide dismutase gene are associated with familial amyotrophic lateral sclerosis. Nature. 1993;362:59–62.
  25. Amaducci L, Tesco G. Aging as a major risk for degenerative diseases of the central nervous system. Curr Opin Neurol. 1994;7:283–6.
  26. Gems D, Partridge L. Genetics of longevity in model organisms: debates and paradigm shifts. Annu Rev Physiol. 2013;75:621‒44.
  27. McCay CM, Crowell MF, Maynard LA. The effect of retarded growth upon the length of life span and upon the ultimate body size. 1935. Nutrition 1989;5:155–71; discussion 172.
  28. Mair W, Dillin A. Aging and Survival: The Genetics of Life Span Extension by Dietary Restriction. Annu Rev Biochem. 2008;77:727‒54.
  29. Bishop NA, Guarente L. Two neurons mediate diet-restriction-induced longevity in C. elegans. Nature. 2007;447:545–9.
  30. Panowski SH, Wolff S, Aguilaniu H, Durieux J, Dillin A. PHA-4/Foxa mediates diet-restriction-induced longevity of C. elegans. Nature. 2007;447:550–5.
  31. Dillin A, Hsu AL, Arantes-Oliveira N, Lehrer-Graiwer J, Hsin H, et al. Rates of behavior and aging specified by mitochondrial function during development. Science. 2002;298:2398–401.
  32. Feng J, Bussiere F, Hekimi S. Mitochondrial electron transport is a key determinant of life span in Caenorhabditis elegans. Dev Cell. 2001;1:633–44.
  33. Durieux J, Wolff S, Dillin A. The cell-non-autonomous nature of electron transport chain-mediated longevity. Cell. 2011;144:79–91.
  34. Giannakou ME, Goss M, Jacobson J, Vinti G, Leevers SJ, et al. Dynamics of the action of dFOXO on adult mortality in Drosophila. Aging Cell. 2007;6(4):429‒38.
  35. Kenyon C, Chang J, Gensch E, Rudner A, Tabtiang R. A C. elegans mutant that lives twice as long as wild type. Nature. 1993;366:461–4.
  36. Holzenberger M, Dupont J, Ducos B, Leneuve P, Geloen A, et al. IGF-1 receptor regulates lifespan and resistance to oxidative stress in mice. Nature. 2003;421:182–7.
  37. Lee RY, Hench J, Ruvkun G. Regulation of C. elegans DAF-16 and its human ortholog FKHRL1 by the daf-2 insulin-like signaling pathway. Curr Biol. 2001;11:1950–7.
  38. Tullet JM, Hertweck M, An JH, Baker J, Hwang JY, et al. Direct inhibition of the longevity-promoting factor SKN-1 by insulin-like signaling in C. elegans. Cell. 2008;132:1025–38.
  39. Chiang WC, Ching TT, Lee HC, Mousigian C, Hsu AL. HSF-1 Regulators DDL-1/2 Link Insulin-like Signaling to Heat-Shock Responses and Modulation of Longevity. Cell. 2012;148:322–34.
  40. Tepper RG, Ashraf J, Kaletsky R, Kleemann G, Murphy CT, et al. PQM-1 Complements DAF-16 as a Key Transcriptional Regulator of DAF-2-Mediated Development and Longevity. Cell. 2013;154:676–90.
  41. Kenyon C. The plasticity of aging: insights from long-lived mutants. Cell. 2005;120:449–60.
  42. Kimura KD, Tissenbaum HA, Liu Y, Ruvkun G. daf-2, an insulin receptor-like gene that regulates longevity and diapause in Caenorhabditis elegans. Science. 1997;277:942–6.
  43. Partridge L, Bruning JC. Forkhead transcription factors and ageing. Oncogene. 2008;27:2351–63.
  44. Selman C, Lingard S, Choudhury AI, Batterham RL, Claret M, et al. Evidence for lifespan extension and delayed age-related biomarkers in insulin receptor substrate 1 null mice. Faseb J. 2008;22:807–18.
  45. Flachsbart F, Caliebe A, Kleindorp R, Blanche H, von Eller-Eberstein H, et al. Association of FOXO3A variation with human longevity confirmed in German centenarians. Proc Natl Acad Sci U S A 2009;106:2700–5.
  46. Suh Y, Atzmon G, Cho MO, Hwang D, Liu B, et al. Functionally significant insulin-like growth factor I receptor mutations in centenarians. Proc Natl Acad Sci U S A. 2008;105:3438–42.
  47. Willcox BJ, Donlon TA, He Q, Chen R, Grove JS, et al. FOXO3A genotype is strongly associated with human longevity. Proc Natl Acad Sci U S A. 2008;105:13987–92.
  48. McElwee J, Bubb K, Thomas JH. Transcriptional outputs of the Caenorhabditis elegans forkhead protein DAF-16. Aging Cell. 2003;2:111–21.
  49. Murphy CT, McCarroll SA, Bargmann CI, Fraser A, Kamath RS, et al. Genes that act downstream of DAF-16 to influence the lifespan of Caenorhabditis elegans. Nature. 2003;424:277–83.
  50. Halaschek-Wiener J, Khattra JS, McKay S, Pouzyrev A, Stott JM, et al. Analysis of long-lived C. elegans daf-2 mutants using serial analysis of gene expression. Genome Res. 2005;15:603–15.
  51. Oh SW, Mukhopadhyay A, Dixit BL, Raha T, Green MR, et al. Identification of direct DAF-16 targets controlling longevity, metabolism and diapause by chromatin immunoprecipitation. Nat Genet. 2006;38:251–7.
  52. Dong MQ, Venable JD, Au N, Xu T, Park SK, et al. Quantitative mass spectrometry identifies insulin signaling targets in C. elegans. Science. 2007;317:660–3.
  53. Morley JF, Brignull HR, Weyers JJ, Morimoto RI. The threshold for polyglutamine-expansion protein aggregation and cellular toxicity is dynamic and influenced by aging in Caenorhabditis elegans. Proc Natl Acad Sci U S A. 2002;99:10417–22.
  54. Link C. Expression of human beta-amyloid peptide in transgenic Caenorhabditis elegans. Proc Natl Acad Sci U S A. 1995;92:9368–72.
  55. Cohen E, Bieschke J, Perciavalle RM, Kelly JW, Dillin A. Opposing activities protect against age-onset proteotoxicity. Science. 2006;313:1604–10.
  56. Cohen E, Du D, Joyce D, Kapernick EA, Volovik Y, et al. Temporal requirements of insulin/IGF-1 signaling for proteotoxicity protection. Aging Cell. 2010;9:126–34.
  57. Steinkraus KA, Smith ED, Davis C, Carr D, Pendergrass WR, et al. Dietary restriction suppresses proteotoxicity and enhances longevity by an hsf-1-dependent mechanism in Caenorhabditis elegans. Aging Cell. 2008;7:394–404.
  58. Mouton PR, Chachich ME, Quigley C, Spangler E, Ingram DK. Caloric restriction attenuates amyloid deposition in middle-aged dtg APP/PS1 mice. Neurosci Lett. 2009;464:184–7.
  59. Arai T, Hasegawa M, Akiyama H, Ikeda K, Nonaka T, et al. TDP-43 is a component of ubiquitin-positive tau-negative inclusions in frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Biochem Biophys Res Commun. 2006;351:602–11.
  60. Zhang T, Mullane PC, Periz G, Wang J. (2011) TDP-43 neurotoxicity and protein aggregation modulated by heat shock factor and insulin/IGF-1 signaling. Hum Mol Genet.
  61. Boccitto M, Lamitina T, Kalb RG. Daf-2 signaling modifies mutant SOD1 toxicity in C. elegans. PLoS One. 2012;7:e33494.
  62. Teixeira-Castro A, Ailion M, Jalles A, Brignull HR, Vilaca JL, et al. (2011) Neuron-specific proteotoxicity of mutant ataxin-3 in C. elegans: rescue by the DAF-16 and HSF-1 pathways. Hum Mol Genet. 2011;20(15):2996-3009.
  63. Killick R, Scales G, Leroy K, Causevic M, Hooper C, et al. Deletion of Irs2 reduces amyloid deposition and rescues behavioural deficits in APP transgenic mice. Biochem Biophys Res Commun. 2009;386:257–62.
  64. Hsiao K, Chapman P, Nilsen S, Eckman C, Harigaya Y, et al. Correlative memory deficits, Abeta elevation, and amyloid plaques in transgenic mice. Science. 1996;274:99–102.
  65. Freude S, Hettich MM, Schumann C, Stohr O, Koch L, et al. Neuronal IGF-1 resistance reduces Abeta accumulation and protects against premature death in a model of Alzheimer’s disease. Faseb J. 2009;23:3315–24.
  66. Jankowsky JL, Slunt HH, Ratovitski T, Jenkins NA, Copeland NG, et al. Co-expression of multiple transgenes in mouse CNS: a comparison of strategies. Biomol Eng. 2001;17:157–65.
  67. Reiserer RS, Harrison FE, Syverud DC, McDonald MP. Impaired spatial learning in the APPSwe + PSEN1DeltaE9 bigenic mouse model of Alzheimer’s disease. Genes Brain Behav. 2007;6:54–65.
  68. Cohen E, Paulsson JF, Blinder P, Burstyn-Cohen T, Du D, et al. Reduced IGF-1 signaling delays age-associated proteotoxicity in mice. Cell. 2009;139:1157–69.
  69. Haass C, Selkoe DJ. Soluble protein oligomers in neurodegeneration: lessons from the Alzheimer’s amyloid beta-peptide. Nat Rev Mol Cell Biol. 2007;8:101–12.
  70. Carro E, Trejo JL, Gomez-Isla T, LeRoith D, Torres-Aleman I. Serum insulin-like growth factor I regulates brain amyloid-beta levels. Nat Med. 2002;8:1390–7.
  71. Poirier R, Fernandez AM, Torres-Aleman I, Metzger F. Early brain amyloidosis in APP/PS1 mice with serum insulin-like growth factor-I deficiency. Neurosci Lett. 2012;509:101–4.
  72. Fernandez AM, Torres-Aleman I. The many faces of insulin-like peptide signalling in the brain. Nat Rev Neurosci. 2012;13:225–39.
  73. Carro E, Trejo JL, Gerber A, Loetscher H, Torrado J, et al. Therapeutic actions of insulin-like growth factor I on APP/PS2 mice with severe brain amyloidosis. Neurobiol Aging. 2006;27:1250–7.
  74. Calamini B, Silva MC, Madoux F, Hutt DM, Khanna S, et al. Small-molecule proteostasis regulators for protein conformational diseases. Nat Chem Biol. 2012;8:185–96.
  75. Honda Y, Tanaka M, Honda S. Trehalose extends longevity in the nematode Caenorhabditis elegans. Aging Cell. 2010;9:558–69.
  76. Alavez S, Vantipalli MC, Zucker DJ, Klang IM, Lithgow GJ. Amyloid-binding compounds maintain protein homeostasis during ageing and extend lifespan. Nature. 2011;472:226–9.
  77. Mojsilovic-Petrovic J, Nedelsky N, Boccitto M, Mano I, Georgiades SN, et al. FOXO3a is broadly neuroprotective in vitro and in vivo against insults implicated in motor neuron diseases. J Neurosci. 2009;29:8236–47.
  78. El-Ami T, Moll L, Marques CF, Volovik Y, Reuveni H, et al. (2013) A Novel Inhibitor of the Insulin/IGF Signaling Pathway Protects from Age-Onset, Neurodegeneration-linked Proteotoxicity. Aging Cell. 2014;13(1):165–74.