Neural mechanisms of ageing and cognitive decline

Nature - Tập 464 Số 7288 - Trang 529-535 - 2010
Nicholas A. Bishop1, Tao Lu2, Bruce A. Yankner2
1Department of Pathology, Harvard Medical School, Boston, Massachusetts 02115 USA
2Dept. of Pathology, Harvard Medical School, Boston, USA

Tóm tắt

Từ khóa


Tài liệu tham khảo

Hebert, L. E., Scherr, P. A., Bienias, J. L., Bennett, D. A. & Evans, D. A. Alzheimer disease in the US population: prevalence estimates using the 2000 census. Arch. Neurol. 60, 1119–1122 (2003).

Andrews-Hanna, J. R. et al. Disruption of large-scale brain systems in advanced aging. Neuron 56, 924–935 (2007). This paper reports that coordination of brain activity between different brain regions becomes less robust in the ageing brain, suggesting a systems-level breakdown of integrated function that correlates with poor cognitive performance.

Cabeza, R. Hemispheric asymmetry reduction in older adults: the HAROLD model. Psychol. Aging 17, 85–100 (2002). This paper shows that spreading activation of the human prefrontal cortex from one hemisphere to both hemispheres may be a compensatory mechanism that preserves function against age-related degenerative changes.

Park, D. C. & Reuter- Lorenz, P. The adaptive brain: aging and neurocognitive scaffolding. Annu. Rev. Psychol. 60, 173–196 (2009).

Cabeza, R., Anderson, N. D., Locantore, J. K. & McIntosh, A. R. Aging gracefully: compensatory brain activity in high-performing older adults. Neuroimage 17, 1394–1402 (2002).

Yankner, B. A., Lu, T. & Loerch, P. The aging brain. Annu. Rev. Pathol. 3, 41–66 (2008).

Lu, T. et al. Gene regulation and DNA damage in the ageing human brain. Nature 429, 883–891 (2004). This paper shows that the ageing of the human cortex is characterized by a distinct transcriptional signature that includes reduced expression of genes that mediate synaptic plasticity and that correlates with age-dependent DNA damage to the promoters of these genes.

Lee, C. K., Weindruch, R. & Prolla, T. A. Gene-expression profile of the ageing brain in mice. Nature Genet. 25, 294–297 (2000).

Jiang, C. H., Tsien, J. Z., Schultz, P. G. & Hu, Y. The effects of aging on gene expression in the hypothalamus and cortex of mice. Proc. Natl Acad. Sci. USA 98, 1930–1934 (2001).

Blalock, E. M. et al. Gene microarrays in hippocampal aging: statistical profiling identifies novel processes correlated with cognitive impairment. J. Neurosci. 23, 3807–3819 (2003).

Fraser, H. B., Khaitovich, P., Plotkin, J. B., Paabo, S. & Eisen, M. B. Aging and gene expression in the primate brain. PLoS Biol. 3, e274 (2005).

Erraji-Benchekroun, L. et al. Molecular aging in human prefrontal cortex is selective and continuous throughout adult life. Biol. Psychiatry 57, 549–558 (2005).

Loerch, P. M. et al. Evolution of the aging brain transcriptome and synaptic regulation. PLoS ONE 3, e3329 (2008).

Small, S. A., Tsai, W. Y., DeLaPaz, R., Mayeux, R. & Stern, Y. Imaging hippocampal function across the human life span: is memory decline normal or not? Ann. Neurol. 51, 290–295 (2002).

West, M. J., Coleman, P. D., Flood, D. G. & Troncoso, J. C. Differences in the pattern of hippocampal neuronal loss in normal ageing and Alzheimer's disease. Lancet 344, 769–772 (1994).

Price, J. L. et al. Neuron number in the entorhinal cortex and CA1 in preclinical Alzheimer disease. Arch. Neurol. 58, 1395–1402 (2001).

Gomez- Isla, T. et al. Profound loss of layer II entorhinal cortex neurons occurs in very mild Alzheimer's disease. J. Neurosci. 16, 4491–4500 (1996).

Rodrigue, K. M. & Raz, N. Shrinkage of the entorhinal cortex over five years predicts memory performance in healthy adults. J. Neurosci. 24, 956–963 (2004).

Zahn, J. M. et al. AGEMAP: a gene expression database for aging in mice. PLoS Genet. 3, e201 (2007).

Blalock, E. M. et al. Incipient Alzheimer's disease: microarray correlation analyses reveal major transcriptional and tumor suppressor responses. Proc. Natl Acad. Sci. USA 101, 2173–2178 (2004).

Liang, W. S. et al. Alzheimer's disease is associated with reduced expression of energy metabolism genes in posterior cingulate neurons. Proc. Natl Acad. Sci. USA 105, 4441–4446 (2008).

Miller, J. A., Oldham, M. C. & Geschwind, D. H. A systems level analysis of transcriptional changes in Alzheimer's disease and normal aging. J. Neurosci. 28, 1410–1420 (2008).

Walker, D. W., Muffat, J., Rundel, C. & Benzer, S. Overexpression of a Drosophila homolog of apolipoprotein D leads to increased stress resistance and extended lifespan. Curr. Biol. 16, 674–679 (2006).

Sanchez, D. et al. Loss of glial lazarillo, a homolog of apolipoprotein D, reduces lifespan and stress resistance in Drosophila . Curr. Biol. 16, 680–686 (2006).

Kalman, J., McConathy, W., Araoz, C., Kasa, P. & Lacko, A. G. Apolipoprotein D in the aging brain and in Alzheimer's dementia. Neurol. Res. 22, 330–336 (2000).

Sedensky, M. M. & Morgan, P. G. Mitochondrial respiration and reactive oxygen species in mitochondrial aging mutants. Exp. Gerontol. 41, 237–245 (2006).

Rea, S. L., Ventura, N. & Johnson, T. E. Relationship between mitochondrial electron transport chain dysfunction, development, and life extension in Caenorhabditis elegans . PLoS Biol. 5, e259 (2007).

Trifunovic, A. et al. Premature ageing in mice expressing defective mitochondrial DNA polymerase. Nature 429, 417–423 (2004).

Kujoth, G. C. et al. Mitochondrial DNA mutations, oxidative stress, and apoptosis in mammalian aging. Science 309, 481–484 (2005).

Schriner, S. E. et al. Extension of murine life span by overexpression of catalase targeted to mitochondria. Science 308, 1909–1911 (2005).

Lin, S. J. et al. Calorie restriction extends Saccharomyces cerevisiae lifespan by increasing respiration. Nature 418, 344–348 (2002).

Wallace, D. C. et al. Familial mitochondrial encephalomyopathy (MERRF): genetic, pathophysiological, and biochemical characterization of a mitochondrial DNA disease. Cell 55, 601–610 (1988).

Holt, I. J., Harding, A. E. & Morgan-Hughes, J. A. Deletions of muscle mitochondrial DNA in patients with mitochondrial myopathies. Nature 331, 717–719 (1988).

Sanchez-Blanco, A., Fridell, Y. W. & Helfand, S. L. Involvement of Drosophila uncoupling protein 5 in metabolism and aging. Genetics 172, 1699–1710 (2006).

Branicky, R., Bénard, C. & Hekimi, S. clk-1, mitochondria, and physiological rates. BioEssays 22, 48–56 (2000).

Lee, S. S. et al. A systematic RNAi screen identifies a critical role for mitochondria in C. elegans longevity. Nature Genet. 33, 40–48 (2003).

Dillin, A. et al. Rates of behavior and aging specified by mitochondrial function during development. Science 298, 2398–2401 (2002). This paper provided the first systematic demonstration of a role for the electron transport chain in metazoan lifespan control, through RNAi-mediated knockdown of several of the chain's components.

Cristina, D., Cary, M., Lunceford, A., Clarke, C. & Kenyon, C. A. Regulated response to impaired respiration slows behavioral rates and increases lifespan in Caenorhabditis elegans. PLoS Genet. 5, e1000450 (2009).

Copeland, J. M. et al. Extension of Drosophila life span by RNAi of the mitochondrial respiratory chain. Curr. Biol. 19, 1591–1598 (2009).

Liu, X. et al. Evolutionary conservation of the clk-1-dependent mechanism of longevity: loss of mclk1 increases cellular fitness and lifespan in mice. Genes Dev. 19, 2424–2434 (2005).

Dell'agnello, C. et al. Increased longevity and refractoriness to Ca2+-dependent neurodegeneration in Surf1 knockout mice. Hum. Mol. Genet. 16, 431–444 (2007).

Muller, F. L., Lustgarten, M. S., Jang, Y., Richardson, A. & Van Remmen, H. Trends in oxidative aging theories. Free Radic. Biol. Med. 43, 477–503 (2007).

Park, S.-K. et al. Gene expression profiling of aging in multiple mouse strains: identification of aging biomarkers and impact of dietary antioxidants. Aging Cell 8, 484–495 (2009).

Liu, J. et al. Memory loss in old rats is associated with brain mitochondrial decay and RNA/DNA oxidation: partial reversal by feeding acetyl-L-carnitine and/or R-α-lipoic acid. Proc. Natl Acad. Sci. USA 99, 2356–2361 (2002). This paper reports that memory loss in aged rats can be reversed by restoring mitochondrial function with dietary mitochondrial substrates and antioxidants.

Oberdoerffer, P. et al. SIRT1 redistribution on chromatin promotes genomic stability but alters gene expression during aging. Cell 135, 907–918 (2008).

O' Hagan, H. M., Mohammad, H. P. & Baylin, S. B. Double strand breaks can initiate gene silencing and SIRT1-dependent onset of DNA methylation in an exogenous promoter CpG island. PLoS Genet. 4, e1000155 (2008).

Guarente, L. Sir2 links chromatin silencing, metabolism, and aging. Genes Dev. 14, 1021–1026 (2000).

Curran, S. P. & Ruvkun, G. Lifespan regulation by evolutionarily conserved genes essential for viability. PLoS Genet. 3, e56 (2007).

Fischer, A., Sananbenesi, F., Wang, X., Dobbin, M. & Tsai, L. H. Recovery of learning and memory is associated with chromatin remodelling. Nature 447, 178–182 (2007). This paper shows that memory loss may be related to altered chromatin structure in a mouse model of neurodegeneration and can be reversed in part with histone deacetylase inhibitors. This raises the possibility of epigenetic approaches to the treatment of human neurodegenerative disorders.

Melendez, A. et al. Autophagy genes are essential for dauer development and life-span extension in C. elegans . Science 301, 1387–1391 (2003).

Hansen, M. et al. A role for autophagy in the extension of lifespan by dietary restriction in C. elegans . PLoS Genet. 4, e24 (2008).

Simonsen, A. et al. Promoting basal levels of autophagy in the nervous system enhances longevity and oxidant resistance in adult Drosophila . Autophagy 4, 176–184 (2008).

Juhasz, G., Erdi, B., Sass, M. & Neufeld, T. P. Atg7-dependent autophagy promotes neuronal health, stress tolerance, and longevity but is dispensable for metamorphosis in Drosophila . Genes Dev. 21, 3061–3066 (2007).

Hara, T. et al. Suppression of basal autophagy in neural cells causes neurodegenerative disease in mice. Nature 441, 885–889 (2006).

Komatsu, M. et al. Loss of autophagy in the central nervous system causes neurodegeneration in mice. Nature 441, 880–884 (2006). References 54 and 55 describe mouse models deficient in key autophagy regulatory genes that demonstrate the essential role of autophagy in protection against age-related neurodegeneration and protein aggregation.

Shibata, M. et al. Regulation of intracellular accumulation of mutant Huntingtin by Beclin 1. J. Biol. Chem. 281, 14474–14485 (2006).

Schieke, S. M. & Finkel, T. Mitochondrial signaling, TOR, and life span. Biol. Chem. 387, 1357–1361 (2006).

Harrison, D. E. et al. Rapamycin fed late in life extends lifespan in genetically heterogeneous mice. Nature 460, 392–395 (2009).

Ravikumar, B. et al. Inhibition of mTOR induces autophagy and reduces toxicity of polyglutamine expansions in fly and mouse models of Huntington disease. Nature Genet. 36, 585–595 (2004).

Fischer, D. F. et al. Long-term proteasome dysfunction in the mouse brain by expression of aberrant ubiquitin. Neurobiol. Aging 30, 847–863 (2009).

Broughton, S. & Partridge, L. Insulin/IGF-like signalling, the central nervous system and aging. Biochem. J. 418, 1–12 (2009).

Flachsbart, F. et al. Association of FOXO3A variation with human longevity confirmed in German centenarians. Proc. Natl Acad. Sci. USA 106, 2700–2705 (2009).

Suh, Y. et al. Functionally significant insulin-like growth factor I receptor mutations in centenarians. Proc. Natl Acad. Sci. USA 105, 3438–3442 (2008).

Willcox, B. J. et al. FOXO3A genotype is strongly associated with human longevity. Proc. Natl Acad. Sci. USA 105, 13987–13992 (2008).

Libina, N., Berman, J. R. & Kenyon, C. Tissue-specific activities of C. elegans DAF-16 in the regulation of lifespan. Cell 115, 489–502 (2003).

Iser, W. B., Gami, M. S. & Wolkow, C. A. Insulin signaling in Caenorhabditis elegans regulates both endocrine-like and cell-autonomous outputs. Dev. Biol. 303, 434–447 (2007).

Broughton, S. J. et al. Longer lifespan, altered metabolism, and stress resistance in Drosophila from ablation of cells making insulin-like ligands. Proc. Natl Acad. Sci. USA 102, 3105–3110 (2005).

van der Heide, L. P., Ramakers, G. M. J. & Smidt, M. P. Insulin signaling in the central nervous system: learning to survive. Prog. Neurobiol. 79, 205–221 (2006).

Taguchi, A., Wartschow, L. M. & White, M. F. Brain IRS2 signaling coordinates life span and nutrient homeostasis. Science 317, 369–372 (2007).

Cohen, E., Bieschke, J., Perciavalle, R. M., Kelly, J. W. & Dillin, A. Opposing activities protect against age-onset proteotoxicity. Science 313, 1604–1610 (2006).

Freude, S. et al. Neuronal IGF-1 resistance reduces Aβ accumulation and protects against premature death in a model of Alzheimer's disease. FASEB J. 23, 3315–3324 (2009).

Cohen, E. et al. Reduced IGF-1 signaling delays age-associated proteotoxicity in mice. Cell 139, 1157–1169 (2009).

Moloney, A. M. et al. Defects in IGF-1 receptor, insulin receptor and IRS-1/2 in Alzheimer's disease indicate possible resistance to IGF-1 and insulin signalling. Neurobiol. Aging 31, 224–243 (2008).

Haigis, M. C. & Guarente, L. P. Mammalian sirtuins — emerging roles in physiology, aging, and calorie restriction. Genes Dev. 20, 2913–2921 (2006).

Colman, R. J. et al. Caloric restriction delays disease onset and mortality in rhesus monkeys. Science 325, 201–204 (2009). This paper reports that caloric restriction increases longevity in primates, delays the onset of age-related diseases and reduces age-related brain atrophy.

Ingram, D. K., Weindruch, R., Spangler, E. L., Freeman, J. R. & Walford, R. L. Dietary restriction benefits learning and motor performance of aged mice. J. Gerontol. 42, 78–81 (1987).

Stewart, J., Mitchell, J. & Kalant, N. The effects of life-long food restriction on spatial memory in young and aged Fischer 344 rats measured in the eight-arm radial and the Morris water mazes. Neurobiol. Aging 10, 669–675 (1989).

Witte, A. V., Fobker, M., Gellner, R., Knecht, S. & Flöel, A. Caloric restriction improves memory in elderly humans. Proc. Natl Acad. Sci. USA 106, 1255–1260 (2009).

Halagappa, V. K. et al. Intermittent fasting and caloric restriction ameliorate age-related behavioral deficits in the triple-transgenic mouse model of Alzheimer's disease. Neurobiol. Dis. 26, 212–220 (2007).

Mair, W. & Dillin, A. Aging and survival: the genetics of life span extension by dietary restriction. Annu. Rev. Biochem. 77, 727–754 (2008).

Lin, S. J., Defossez, P. A. & Guarente, L. Requirement of NAD and SIR2 for life-span extension by calorie restriction in Saccharomyces cerevisiae . Science 289, 2126–2128 (2000).

Rogina, B. & Helfand, S. L. Sir2 mediates longevity in the fly through a pathway related to calorie restriction. Proc. Natl Acad. Sci. USA 101, 15998–16003 (2004).

Boily, G. et al. SirT1 regulates energy metabolism and response to caloric restriction in mice. PLoS ONE 3, e1759 (2008).

Chen, D., Steele, A. D., Lindquist, S. & Guarente, L. Increase in activity during calorie restriction requires Sirt1. Science 310, 1641 (2005).

Kaeberlein, M., Kirkland, K. T., Fields, S. & Kennedy, B. K. Sir2-independent life span extension by calorie restriction in yeast. PLoS. Biol. 2, e296 (2004).

Chen, D. et al. Tissue-specific regulation of SIRT1 by calorie restriction. Genes Dev. 22, 1753–1757 (2008).

Araki, T., Sasaki, Y. & Milbrandt, J. Increased nuclear NAD biosynthesis and SIRT1 activation prevent axonal degeneration. Science 305, 1010–1013 (2004).

Kim, D. et al. SIRT1 deacetylase protects against neurodegeneration in models for Alzheimer's disease and amyotrophic lateral sclerosis. EMBO J. 26, 3169–3179 (2007).

Li, Y., Xu, W., McBurney, M. W. & Longo, V. D. SirT1 inhibition reduces IGF-I/IRS-2/Ras/ERK1/2 signaling and protects neurons. Cell Metab. 8, 38–48 (2008).

Apfeld, J. & Kenyon, C. Regulation of lifespan by sensory perception in Caenorhabditis elegans . Nature 402, 804–809 (1999).

Alcedo, J. & Kenyon, C. Regulation of C. elegans longevity by specific gustatory and olfactory neurons. Neuron 41, 45–55 (2004).

Bishop, N. A. & Guarente, L. Two neurons mediate diet-restriction-induced longevity in C. elegans . Nature 447, 545–549 (2007). This paper demonstrates the essential role of two C. elegans neurons in coordinating the organismal response to caloric restriction and argues for a central role of the nervous system in this form of increased longevity.

Flurkey, K., Papaconstantinou, J., Miller, R. A. & Harrison, D. E. Lifespan extension and delayed immune and collagen aging in mutant mice with defects in growth hormone production. Proc. Natl Acad. Sci. USA 98, 6736–6741 (2001).

Conti, B. et al. Transgenic mice with a reduced core body temperature have an increased life span. Science 314, 825–828 (2006).

Jankord, R. & Herman, J. P. Limbic regulation of hypothalamo-pituitary-adrenocortical function during acute and chronic stress. Ann. NY Acad. Sci. 1148, 64–73 (2008).

Conrad, C. D. Chronic stress-induced hippocampal vulnerability: the glucocorticoid vulnerability hypothesis. Rev. Neurosci. 19, 395–411 (2008).

Bao, A. M., Meynen, G. & Swaab, D. F. The stress system in depression and neurodegeneration: focus on the human hypothalamus. Brain Res. Rev. 57, 531–553 (2008).

Zahn, J. M. et al. Transcriptional profiling of aging in human muscle reveals a common aging signature. PLoS Genet. 2, e115 (2006).

Rodwell, G. E. et al. A transcriptional profile of aging in the human kidney. PLoS Biol. 2, e427 (2004).

Maalouf, M., Rho, J. M. & Mattson, M. P. The neuroprotective properties of calorie restriction, the ketogenic diet, and ketone bodies. Brain Res. Rev. 59, 293–315 (2009).