Therapeutic potential of systemic brain rejuvenation strategies for neurodegenerative disease

Alana M Horowitz, Saul A Villeda, Alana M Horowitz, Saul A Villeda

Abstract

Neurodegenerative diseases are a devastating group of conditions that cause progressive loss of neuronal integrity, affecting cognitive and motor functioning in an ever-increasing number of older individuals. Attempts to slow neurodegenerative disease advancement have met with little success in the clinic; however, a new therapeutic approach may stem from classic interventions, such as caloric restriction, exercise, and parabiosis. For decades, researchers have reported that these systemic-level manipulations can promote major functional changes that extend organismal lifespan and healthspan. Only recently, however, have the functional effects of these interventions on the brain begun to be appreciated at a molecular and cellular level. The potential to counteract the effects of aging in the brain, in effect rejuvenating the aged brain, could offer broad therapeutic potential to combat dementia-related neurodegenerative disease in the elderly. In particular, results from heterochronic parabiosis and young plasma administration studies indicate that pro-aging and rejuvenating factors exist in the circulation that can independently promote or reverse age-related phenotypes. The recent demonstration that human umbilical cord blood similarly functions to rejuvenate the aged brain further advances this work to clinical translation. In this review, we focus on these blood-based rejuvenation strategies and their capacity to delay age-related molecular and functional decline in the aging brain. We discuss new findings that extend the beneficial effects of young blood to neurodegenerative disease models. Lastly, we explore the translational potential of blood-based interventions, highlighting current clinical trials aimed at addressing therapeutic applications for the treatment of dementia-related neurodegenerative disease in humans.

Keywords: blood plasma administration; brain rejuvenation; caloric restriction; exercise; healthspan; neurodegenerative disease.

Conflict of interest statement

Competing interests: The authors declare that they have no competing interests.No competing interests were disclosed.No competing interests were disclosed.No competing interests were disclosed.

Figures

Figure 1.. Systemic brain rejuvenation strategies.
Figure 1.. Systemic brain rejuvenation strategies.
Hallmarks of brain aging amenable to rejuvenation (middle panel) include decreased regenerative capacity (neurogenesis), impaired synaptic plasticity, increased inflammation, vascular remodeling, increased protein aggregation, and impaired cognitive function. Systemic interventions (top panel), such as caloric restriction, exercise, and blood plasma administration, have been shown to rejuvenate hallmarks of brain aging (left panel) and ameliorate exacerbated pathology in models of neurodegenerative disease (right panel). Cellular or functional rejuvenation elicited by systemic interventions is denoted by a check (✔), lack of rejuvenation is denoted by a dash (), and yet-to-be-determined effects are denoted by a question mark (?).

References

    1. López-Otín C, Blasco MA, Partridge L, et al. : The hallmarks of aging. Cell. 2013;153(6):1194–217. 10.1016/j.cell.2013.05.039
    2. F1000 Recommendation

    1. Mather M, Jacobsen LA, Pollard KM: Aging in the United States.2015;70
    1. Alzheimer's Association: 2016 Alzheimer's disease facts and figures. Alzheimers Dement. 2016;12(4):459–509. 10.1016/j.jalz.2016.03.001
    2. F1000 Recommendation

    1. Wyss-Coray T: Ageing, neurodegeneration and brain rejuvenation. Nature. 2016;539(7628):180–6. 10.1038/nature20411
    2. F1000 Recommendation

    1. Morrison JH, Baxter MG: The ageing cortical synapse: hallmarks and implications for cognitive decline. Nat Rev Neurosci. 2012;13(4):240–50. 10.1038/nrn3200
    1. Farkas E, Luiten PG: Cerebral microvascular pathology in aging and Alzheimer's disease. Prog Neurobiol. 2001;64(6):575–611. 10.1016/S0301-0082(00)00068-X
    1. Conde JR, Streit WJ: Microglia in the aging brain. J Neuropathol Exp Neurol. 2006;65(3):199–203. 10.1097/01.jnen.0000202887.22082.63
    1. Mattson MP, Magnus T: Ageing and neuronal vulnerability. Nat Rev Neurosci. 2006;7(4):278–94. 10.1038/nrn1886
    1. Kuhn HG, Dickinson-Anson H, Gage FH: Neurogenesis in the dentate gyrus of the adult rat: age-related decrease of neuronal progenitor proliferation. J Neurosci. 1996;16(6):2027–33.
    1. Bondolfi L, Ermini F, Long JM, et al. : Impact of age and caloric restriction on neurogenesis in the dentate gyrus of C57BL/6 mice. Neurobiol Aging. 2004;25(3):333–40. 10.1016/S0197-4580(03)00083-6
    1. Kuipers SD, Schroeder JE, Trentani A: Changes in hippocampal neurogenesis throughout early development. Neurobiol Aging. 2015;36(1):365–79. 10.1016/j.neurobiolaging.2014.07.033
    2. F1000 Recommendation

    1. Bond AM, Ming GL, Song H: Adult Mammalian Neural Stem Cells and Neurogenesis: Five Decades Later. Cell Stem Cell. 2015;17(4):385–95. 10.1016/j.stem.2015.09.003
    2. F1000 Recommendation

    1. Alvarez-Buylla A, Garcia-Verdugo JM: Neurogenesis in adult subventricular zone. J Neurosci. 2002;22(3):629–34.
    1. Drapeau E, Mayo W, Aurousseau C, et al. : Spatial memory performances of aged rats in the water maze predict levels of hippocampal neurogenesis. Proc Natl Acad Sci U S A. 2003;100(24):14385–90. 10.1073/pnas.2334169100
    1. Merrill DA, Karim R, Darraq M, et al. : Hippocampal cell genesis does not correlate with spatial learning ability in aged rats. J Comp Neurol. 2003;459(2):201–7. 10.1002/cne.10616
    2. F1000 Recommendation

    1. Riddle DR, Sonntag WE, Lichtenwalner RJ: Microvascular plasticity in aging. Ageing Res Rev. 2003;2(2):149–68. 10.1016/S1568-1637(02)00064-8
    1. Soto I, Graham LC, Richter HJ, et al. : APOE Stabilization by Exercise Prevents Aging Neurovascular Dysfunction and Complement Induction. PLoS Biol. 2015;13(10):e1002279. 10.1371/journal.pbio.1002279
    2. F1000 Recommendation

    1. Hong S, Beja-Glasser VF, Nfonoyim BM, et al. : Complement and microglia mediate early synapse loss in Alzheimer mouse models. Science. 2016;352(6286):712–6. 10.1126/science.aad8373
    2. F1000 Recommendation

    1. Peters A, Sethares C, Luebke JI: Synapses are lost during aging in the primate prefrontal cortex. Neuroscience. 2008;152(4):970–81. 10.1016/j.neuroscience.2007.07.014
    1. Selkoe DJ: Alzheimer's disease is a synaptic failure. Science. 2002;298(5594):789–91. 10.1126/science.1074069
    1. Geiszler PC, Barron MR, Pardon MC: Impaired burrowing is the most prominent behavioral deficit of aging htau mice. Neuroscience. 2016;329:98–111. 10.1016/j.neuroscience.2016.05.004
    1. Murphy CT, McCarroll SA, Bargmann CI, et al. : Genes that act downstream of DAF-16 to influence the lifespan of Caenorhabditis elegans. Nature. 2003;424(6946):277–83. 10.1038/nature01789
    2. F1000 Recommendation

    1. Lin YJ, Seroude L, Benzer S: Extended life-span and stress resistance in the Drosophila mutant methuselah. Science. 1998;282(5390):943–6. 10.1126/science.282.5390.943
    1. Satoh A, Brace CS, Rensing N, et al. : Sirt1 extends life span and delays aging in mice through the regulation of Nk2 homeobox 1 in the DMH and LH. Cell Metab. 2013;18(3):416–30. 10.1016/j.cmet.2013.07.013
    1. Zhang G, Li J, Purkayastha S, et al. : Hypothalamic programming of systemic ageing involving IKK-β, NF-κB and GnRH. Nature. 2013;497(7448):211–6. 10.1038/nature12143
    2. F1000 Recommendation

    1. Ocampo A, Reddy P, Martinez-Redondo P, et al. : In Vivo Amelioration of Age-Associated Hallmarks by Partial Reprogramming. Cell. 2016;167(7):1719–1733.e12. 10.1016/j.cell.2016.11.052
    2. F1000 Recommendation

    1. Rahman MM, Stuchlick O, El-Karim EG, et al. : Intracellular protein glycosylation modulates insulin mediated lifespan in C.elegans. Aging (Albany NY). 2010;2(10):678–90. 10.18632/aging.100208
    1. Harrison DE, Strong R, Sharp ZD, et al. : Rapamycin fed late in life extends lifespan in genetically heterogeneous mice. Nature. 2009;460(7253):392–5. 10.1038/nature08221
    2. F1000 Recommendation

    1. Ryu D, Mouchiroud L, Andreux PA, et al. : Urolithin A induces mitophagy and prolongs lifespan in C. elegans and increases muscle function in rodents. Nat Med. 2016;22(8):879–88. 10.1038/nm.4132
    2. F1000 Recommendation

    1. Zhang H, Ryu D, Wu Y, et al. : NAD + repletion improves mitochondrial and stem cell function and enhances life span in mice. Science. 2016;352(6292):1436–43. 10.1126/science.aaf2693
    2. F1000 Recommendation

    1. Baker DJ, Childs BG, Durik M, et al. : Naturally occurring p16 Ink4a-positive cells shorten healthy lifespan. Nature. 2016;530(7589):184–9. 10.1038/nature16932
    2. F1000 Recommendation

    1. Goodrick CL: The effects of exercise on longevity and behavior of hybrid mice which differ in coat color. J Gerontol. 1974;29(2):129–33. 10.1093/geronj/29.2.129
    1. Ludwig FC, Elashoff RM: Mortality in syngeneic rat parabionts of different chronological age. Trans N Y Acad Sci. 1972;34(7):582–7. 10.1111/j.2164-0947.1972.tb02712.x
    1. Weindruch R, Walford RL: Dietary restriction in mice beginning at 1 year of age: effect on life-span and spontaneous cancer incidence. Science. 1982;215(4538):1415–8. 10.1126/science.7063854
    2. F1000 Recommendation

    1. Loffredo FS, Steinhauser ML, Jay SM, et al. : Growth differentiation factor 11 is a circulating factor that reverses age-related cardiac hypertrophy. Cell. 2013;153(4):828–39. 10.1016/j.cell.2013.04.015
    2. F1000 Recommendation

    1. Conboy IM, Conboy MJ, Wagers AJ, et al. : Rejuvenation of aged progenitor cells by exposure to a young systemic environment. Nature. 2005;433(7027):760–4. 10.1038/nature03260
    2. F1000 Recommendation

    1. Brack AS, Conboy MJ, Roy S, et al. : Increased Wnt signaling during aging alters muscle stem cell fate and increases fibrosis. Science. 2007;317(5839):807–10. 10.1126/science.1144090
    2. F1000 Recommendation

    1. Sinha M, Jang YC, Oh J, et al. : Restoring systemic GDF11 levels reverses age-related dysfunction in mouse skeletal muscle. Science. 2014;344(6184):649–52. 10.1126/science.1251152
    2. F1000 Recommendation

    1. Villeda SA, Plambeck KE, Middeldorp J, et al. : Young blood reverses age-related impairments in cognitive function and synaptic plasticity in mice. Nat Med. 2014;20(6):659–63. 10.1038/nm.3569
    2. F1000 Recommendation

    1. Salpeter SJ, Khalaileh A, Weinberg-Corem N, et al. : Systemic regulation of the age-related decline of pancreatic β-cell replication. Diabetes. 2013;62(8):2843–8. 10.2337/db13-0160
    1. Ruckh JM, Zhao JW, Shadrach JL, et al. : Rejuvenation of regeneration in the aging central nervous system. Cell Stem Cell. 2012;10(1):96–103. 10.1016/j.stem.2011.11.019
    2. F1000 Recommendation

    1. Baht GS, Silkstone D, Vi L, et al. : Exposure to a youthful circulation rejuvenates bone repair through modulation of β-catenin. Nat Commun. 2015;6: 7131. 10.1038/ncomms8131
    2. F1000 Recommendation

    1. Colman RJ, Beasley TM, Kemnitz JW, et al. : Caloric restriction reduces age-related and all-cause mortality in rhesus monkeys. Nat Commun. 2014;5: 3557. 10.1038/ncomms4557
    2. F1000 Recommendation

    1. Weindruch R, Walford RL: The Retardation of aging and disease by dietary restriction. J Nutr. 1988.
    1. Colman RJ, Anderson RM, Johnson SC, et al. : Caloric restriction delays disease onset and mortality in rhesus monkeys. Science. 2009;325(5937):201–4. 10.1126/science.1173635
    2. F1000 Recommendation

    1. Guo J, Bakshi V, Lin AL: Early Shifts of Brain Metabolism by Caloric Restriction Preserve White Matter Integrity and Long-Term Memory in Aging Mice. Front Aging Neurosci. 2015;7:213. 10.3389/fnagi.2015.00213
    2. F1000 Recommendation

    1. Mattson MP: Neuroprotective signaling and the aging brain: take away my food and let me run. Brain Res. 2000;886(1–2):47–53. 10.1016/S0006-8993(00)02790-6
    1. Ferreira-Marques M, Aveleira CA, Carmo-Silva S, et al. : Caloric restriction stimulates autophagy in rat cortical neurons through neuropeptide Y and ghrelin receptors activation. Aging (Albany NY). 2016;8(7):1470–84. 10.18632/aging.100996
    2. F1000 Recommendation

    1. Van Cauwenberghe C, Vandendriessche C, Libert C, et al. : Caloric restriction: beneficial effects on brain aging and Alzheimer's disease. Mamm Genome. 2016;27(7–8):300–19. 10.1007/s00335-016-9647-6
    2. F1000 Recommendation

    1. Lin AL, Zhang W, Gao X, et al. : Caloric restriction increases ketone bodies metabolism and preserves blood flow in aging brain. Neurobiol Aging. 2015;36(7):2296–303. 10.1016/j.neurobiolaging.2015.03.012
    2. F1000 Recommendation

    1. Parikh I, Guo J, Chuang KH, et al. : Caloric restriction preserves memory and reduces anxiety of aging mice with early enhancement of neurovascular functions. Aging (Albany NY). 2016;8(11):2814–26. 10.18632/aging.101094
    2. F1000 Recommendation

    1. Villain N, Picq JL, Aujard F, et al. : Body mass loss correlates with cognitive performance in primates under acute caloric restriction conditions. Behav Brain Res. 2016;305:157–63. 10.1016/j.bbr.2016.02.037
    2. F1000 Recommendation

    1. Dal-Pan A, Pifferi F, Marchal J, et al. : Cognitive performances are selectively enhanced during chronic caloric restriction or resveratrol supplementation in a primate. PLoS One. 2011;6(1):e16581. 10.1371/journal.pone.0016581
    1. Ma L, Zhao Z, Wang R, et al. : Caloric restriction can improve learning ability in C57/BL mice via regulation of the insulin-PI3K/Akt signaling pathway. Neurol Sci. 2014;35(9):1381–6. 10.1007/s10072-014-1717-5
    1. Solon-Biet SM, Cogger VC, Pulpitel T, et al. : Defining the Nutritional and Metabolic Context of FGF21 Using the Geometric Framework. Cell Metab. 2016;24(4):555–65. 10.1016/j.cmet.2016.09.001
    2. F1000 Recommendation

    1. Martin SA, DeMuth TM, Miller KN, et al. : Regional metabolic heterogeneity of the hippocampus is nonuniformly impacted by age and caloric restriction. Aging Cell. 2016;15(1):100–10. 10.1111/acel.12418
    2. F1000 Recommendation

    1. Cardoso A, Marrana F, Andrade JP: Caloric restriction in young rats disturbs hippocampal neurogenesis and spatial learning. Neurobiol Learn Mem. 2016;133:214–24. 10.1016/j.nlm.2016.07.013
    2. F1000 Recommendation

    1. Rühlmann C, Wölk T, Blümel T, et al. : Long-term caloric restriction in ApoE-deficient mice results in neuroprotection via Fgf21-induced AMPK/mTOR pathway. Aging (Albany NY). 2016;8(11):2777–89. 10.18632/aging.101086
    2. F1000 Recommendation

    1. Halagappa VK, Guo Z, Pearson M, et al. : Intermittent fasting and caloric restriction ameliorate age-related behavioral deficits in the triple-transgenic mouse model of Alzheimer's disease. Neurobiol Dis. 2007;26(1):212–20. 10.1016/j.nbd.2006.12.019
    1. Ravussin E, Redman LM, Rochon J, et al. : A 2-Year Randomized Controlled Trial of Human Caloric Restriction: Feasibility and Effects on Predictors of Health Span and Longevity. J Gerontol A Biol Sci Med Sci. 2015;70(9):1097–104. 10.1093/gerona/glv057
    2. F1000 Recommendation

    1. Navarro A, Gomez C, López-Cepero JM, et al. : Beneficial effects of moderate exercise on mice aging: survival, behavior, oxidative stress, and mitochondrial electron transfer. Am J Physiol Regul Integr Comp Physiol. 2004;286(3):R505–11. 10.1152/ajpregu.00208.2003
    1. Samorajski T, Delaney C, Durham L, et al. : Effect of exercise on longevity, body weight, locomotor performance, and passive-avoidance memory of C57BL/6J mice. Neurobiol Aging. 1985;6(1):17–24. 10.1016/0197-4580(85)90066-1
    1. Garcia-Valles R, Gomez-Cabrera MC, Rodriguez-Mañas L, et al. : Life-long spontaneous exercise does not prolong lifespan but improves health span in mice. Longev Healthspan. 2013;2(1):14. 10.1186/2046-2395-2-14
    1. Xue QL: The frailty syndrome: definition and natural history. Clin Geriatr Med. 2011;27(1):1–15. 10.1016/j.cger.2010.08.009
    1. van Praag H, Shubert T, Zhao C, et al. : Exercise enhances learning and hippocampal neurogenesis in aged mice. J Neurosci. 2005;25(38):8680–5. 10.1523/JNEUROSCI.1731-05.2005
    1. Speisman RB, Kumar A, Rani A, et al. : Daily exercise improves memory, stimulates hippocampal neurogenesis and modulates immune and neuroimmune cytokines in aging rats. Brain Behav Immun. 2013;28:25–43. 10.1016/j.bbi.2012.09.013
    1. O'Callaghan RM, Griffin EW, Kelly AM: Long-term treadmill exposure protects against age-related neurodegenerative change in the rat hippocampus. Hippocampus. 2009;19(10):1019–29. 10.1002/hipo.20591
    1. Intlekofer KA, Cotman CW: Exercise counteracts declining hippocampal function in aging and Alzheimer's disease. Neurobiol Dis. 2013;57:47–55. 10.1016/j.nbd.2012.06.011
    1. Stessman J, Hammerman-Rozenberg R, Cohen A, et al. : Physical activity, function, and longevity among the very old. Arch Intern Med. 2009;169(16):1476–83. 10.1001/archinternmed.2009.248
    1. Chakravarty EF, Hubert HB, Lingala VB, et al. : Reduced disability and mortality among aging runners: a 21-year longitudinal study. Arch Intern Med. 2008;168(15):1638–46. 10.1001/archinte.168.15.1638
    1. Colbert LH, Visser M, Simonsick EM, et al. : Physical activity, exercise, and inflammatory markers in older adults: findings from the Health, Aging and Body Composition Study. J Am Geriatr Soc. 2004;52(7):1098–104. 10.1111/j.1532-5415.2004.52307.x
    1. Fiatarone MA, O'Neill EF, Ryan ND, et al. : Exercise training and nutritional supplementation for physical frailty in very elderly people. N Engl J Med. 1994;330(25):1769–75. 10.1056/NEJM199406233302501
    1. Kirk-Sanchez NJ, McGough EL: Physical exercise and cognitive performance in the elderly: current perspectives. Clin Interv Aging. 2014;9:51–62. 10.2147/CIA.S39506
    1. Geda YE, Roberts RO, Knopman DS, et al. : Physical exercise, aging, and mild cognitive impairment: a population-based study. Arch Neurol. 2010;67(1):80–6. 10.1001/archneurol.2009.297
    1. Knecht S, Wersching H, Lohmann H, et al. : High-normal blood pressure is associated with poor cognitive performance. Hypertension. 2008;51(3):663–8. 10.1161/HYPERTENSIONAHA.107.105577
    1. Fujishima M, Ibayashi S, Fujii K, et al. : Cerebral blood flow and brain function in hypertension. Hypertens Res. 1995;18(2):111–7. 10.1291/hypres.18.111
    1. Lautenschlager NT, Cox KL, Flicker L, et al. : Effect of physical activity on cognitive function in older adults at risk for Alzheimer disease: a randomized trial. JAMA. 2008;300(9):1027–37. 10.1001/jama.300.9.1027
    1. Rhodes RE, Martin AD, Taunton JE, et al. : Factors associated with exercise adherence among older adults. An individual perspective. Sports Med. 1999;28(6):397–411. 10.2165/00007256-199928060-00003
    1. Villeda SA, Luo J, Mosher KI, et al. : The ageing systemic milieu negatively regulates neurogenesis and cognitive function. Nature. 2011;477(7362):90–4. 10.1038/nature10357
    2. F1000 Recommendation

    1. Katsimpardi L, Litterman NK, Schein PA, et al. : Vascular and neurogenic rejuvenation of the aging mouse brain by young systemic factors. Science. 2014;344(6184):630–4. 10.1126/science.1251141
    2. F1000 Recommendation

    1. Smith LK, He Y, Park JS, et al. : β2-microglobulin is a systemic pro-aging factor that impairs cognitive function and neurogenesis. Nat Med. 2015;21(8):932–7. 10.1038/nm.3898
    2. F1000 Recommendation

    1. Rebo J, Mehdipour M, Gathwala R, et al. : A single heterochronic blood exchange reveals rapid inhibition of multiple tissues by old blood. Nat Commun. 2016;7: 13363. 10.1038/ncomms13363
    2. F1000 Recommendation

    1. Yousef H, Conboy MJ, Morgenthaler A, et al. : Systemic attenuation of the TGF-β pathway by a single drug simultaneously rejuvenates hippocampal neurogenesis and myogenesis in the same old mammal. Oncotarget. 2015;6(14):11959–78. 10.18632/oncotarget.3851
    2. F1000 Recommendation

    1. Tavazoie M, van der Veken L, Silva-Vargas V, et al. : A specialized vascular niche for adult neural stem cells. Cell Stem Cell. 2008;3(3):279–88. 10.1016/j.stem.2008.07.025
    1. Shen Q, Goderie SK, Jin L, et al. : Endothelial cells stimulate self-renewal and expand neurogenesis of neural stem cells. Science. 2004;304(5675):1338–40. 10.1126/science.1095505
    2. F1000 Recommendation

    1. Castellano JM, Mosher KI, Abbey RJ, et al. : Human umbilical cord plasma proteins revitalize hippocampal function in aged mice. Nature. 2017;544(7651):488–92. 10.1038/nature22067
    2. F1000 Recommendation

    1. Egerman MA, Cadena SM, Gilbert JA, et al. : GDF11 Increases with Age and Inhibits Skeletal Muscle Regeneration. Cell Metab. 2015;22(1):164–74. 10.1016/j.cmet.2015.05.010
    2. F1000 Recommendation

    1. Jucker M: The benefits and limitations of animal models for translational research in neurodegenerative diseases. Nat Med. 2010;16(11):1210–4. 10.1038/nm.2224
    1. Cunningham C, Hennessy E: Co-morbidity and systemic inflammation as drivers of cognitive decline: new experimental models adopting a broader paradigm in dementia research. Alzheimers Res Ther. 2015;7(1):33. 10.1186/s13195-015-0117-2
    2. F1000 Recommendation

    1. Xiang Y, Bu XL, Liu YH, et al. : Physiological amyloid-beta clearance in the periphery and its therapeutic potential for Alzheimer's disease. Acta Neuropathol. 2015;130(4):487–99. 10.1007/s00401-015-1477-1
    2. F1000 Recommendation

    1. Jin WS, Shen LL, Bu XL, et al. : Peritoneal dialysis reduces amyloid-beta plasma levels in humans and attenuates Alzheimer-associated phenotypes in an APP/PS1 mouse model. Acta Neuropathol. 2017;134(2):207–220. 10.1007/s00401-017-1721-y
    2. F1000 Recommendation

    1. Middeldorp J, Lehallier B, Villeda SA, et al. : Preclinical Assessment of Young Blood Plasma for Alzheimer Disease. JAMA Neurol. 2016;73(11):1325–33. 10.1001/jamaneurol.2016.3185
    1. Kazim SF, Blanchard J, Dai CL, et al. : Disease modifying effect of chronic oral treatment with a neurotrophic peptidergic compound in a triple transgenic mouse model of Alzheimer's disease. Neurobiol Dis. 2014;71:110–30. 10.1016/j.nbd.2014.07.001
    1. Baazaoui N, Iqbal K: Prevention of Amyloid-β and Tau Pathologies, Associated Neurodegeneration, and Cognitive Deficit by Early Treatment with a Neurotrophic Compound. J Alzheimers Dis. 2017;58(1):215–30. 10.3233/JAD-170075
    2. F1000 Recommendation

    1. Ferrucci L, National Institute of Aging : Genetic and Epigenetic Signatures of Translational Aging Laboratory Testing (GESTALT). In: . [cited 2017 Apr 10].
    1. Wagner A: Stanford University, Stanford Memory and Aging : . [cited 2017 Apr 10].
    1. Henderson V, Wyss-Cora T: Stanford University, Healthy Brain Aging : [cited 2017 Apr 10].
    1. Stanford University, Alkahest, Inc.,: The PLasma for Alzheimer SymptoM Amelioration (PLASMA) Study. In: . [cited 2017 Apr 10].
    1. Karmazin J, Ambrosia LLC: Young Donor Plasma Transfusion and Age-Related : . [cited 2017 Apr 10].2017.
    1. Kaiser J: Young blood antiaging trial raises questions. Science. 2016. 10.1126/science.aag0716
    1. Bronte-Stewart H: Stanford University, The Stanford Parkinson's Disease Plasma Study (SPDP). In: . [cited 2017 Apr 10].2017.
    1. Tsai R, University of California, San Francisco : Young Plasma Transfusions for Progressive Supranuclear : . [cited 2017 Apr 10].2017.
    1. Xinqiao Hospital of Chongqing: Efficacy and Safety of Young Plasma on Acute . [cited 2017 Apr 10].

Source: PubMed

3
購読する