Hostname: page-component-8448b6f56d-qsmjn Total loading time: 0 Render date: 2024-04-17T23:37:22.203Z Has data issue: false hasContentIssue false

Effects of lifespan-extending interventions on cognitive healthspan

Published online by Cambridge University Press:  15 November 2022

Luka Culig
Affiliation:
Section on DNA Repair, National Institute on Aging, NIH, Baltimore, MD 21224, USA
Burcin Duan Sahbaz
Affiliation:
Section on DNA Repair, National Institute on Aging, NIH, Baltimore, MD 21224, USA
Vilhelm A. Bohr*
Affiliation:
Section on DNA Repair, National Institute on Aging, NIH, Baltimore, MD 21224, USA
*
Author for correspondence: Vilhelm A. Bohr, E-mail: vbohr@nih.gov

Abstract

Ageing is known to be the primary risk factor for most neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease and Huntington's disease. They are currently incurable and worsen over time, which has broad implications in the context of lifespan and healthspan extension. Adding years to life and even to physical health is suboptimal or even insufficient, if cognitive ageing is not adequately improved. In this review, we will examine how interventions that have the potential to extend lifespan in animals affect the brain, and if they would be able to thwart or delay the development of cognitive dysfunction and/or neurodegeneration. These interventions range from lifestyle (caloric restriction, physical exercise and environmental enrichment) through pharmacological (nicotinamide adenine dinucleotide precursors, resveratrol, rapamycin, metformin, spermidine and senolytics) to epigenetic reprogramming. We argue that while many of these interventions have clear potential to improve cognitive health and resilience, large-scale and long-term randomised controlled trials are needed, along with studies utilising washout periods to determine the effects of supplementation cessation, particularly in aged individuals.

Type
Review
Creative Commons
This is a work of the US Government and is not subject to copyright protection within the United States. Published by Cambridge University Press
Copyright
Copyright © NIH, (2022)

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

López-Otín, C et al. (2013) The hallmarks of aging. Cell 153, 1194.CrossRefGoogle ScholarPubMed
Lemoine, M (2021) The evolution of the hallmarks of aging. Frontiers in Genetics 12, 124. https://doi.org/10.3389/fgene.2021.693071.CrossRefGoogle ScholarPubMed
Longo, VD et al. (2015) Interventions to slow aging in humans: are we ready? Aging Cell 14, 497510.CrossRefGoogle ScholarPubMed
Zhavoronkov, A et al. (2019) Artificial intelligence for aging and longevity research: recent advances and perspectives. Ageing Research Reviews 49, 4966.CrossRefGoogle ScholarPubMed
Fabris, F et al. (2019) Using deep learning to associate human genes with age-related diseases. Bioinformatics 36, 22022208. https://doi.org/10.1093/bioinformatics/btz887.CrossRefGoogle Scholar
Zhavoronkov, A, Bischof, E and Lee, K-F (2021) Artificial intelligence in longevity medicine. Nature Aging 1, 57.CrossRefGoogle Scholar
Kennedy, BK et al. (2014) Geroscience: linking aging to chronic disease. Cell 159, 709713.CrossRefGoogle ScholarPubMed
Tinetti, ME, Fried, TR and Boyd, CM (2012) Designing health care for the most common chronic condition – multimorbidity. JAMA 307, 24932494.CrossRefGoogle ScholarPubMed
Marengoni, A et al. (2011) Aging with multimorbidity: a systematic review of the literature. Ageing Research Reviews 10, 430439.CrossRefGoogle ScholarPubMed
Zealley, B and De Grey, ADNJ (2013) Strategies for engineered negligible senescence. Gerontology 59, 183189.CrossRefGoogle ScholarPubMed
Fabbri, E et al. (2015) Aging and multimorbidity: new tasks, priorities, and frontiers for integrated gerontological and clinical research. Journal of the American Medical Directors Association 16, 640647.CrossRefGoogle ScholarPubMed
Yun, M (2015) Changes in regenerative capacity through lifespan. International Journal of Molecular Sciences 16, 2539225432.CrossRefGoogle ScholarPubMed
Oberman, L and Pascual-Leone, A (2013) Changes in plasticity across the lifespan: cause of disease and target for intervention. Progress in Brain Research 207, 91120.CrossRefGoogle ScholarPubMed
Ginhoux, F and Prinz, M (2015) Origin of microglia: current concepts and past controversies. Cold Spring Harbor Perspectives in Biology 7, a020537.CrossRefGoogle ScholarPubMed
Mehl, LC et al. (2022) Microglia in brain development and regeneration. Development 149, dev200425.CrossRefGoogle ScholarPubMed
Zhang, G et al. (2021) Microglia in Alzheimer's disease: a target for therapeutic intervention. Frontiers in Cellular Neuroscience 15, 749587.CrossRefGoogle ScholarPubMed
Feigin, VL et al. (2017) Global, regional, and national burden of neurological disorders during 1990–2015: a systematic analysis for the global burden of disease study 2015. The Lancet. Neurology 16, 877897.CrossRefGoogle Scholar
Alzheimer's Association (2021) Alzheimer's disease facts and figures special report race, ethnicity and Alzheimer's in America. Alzheimer's & Dementia: The Journal of the Alzheimer's Association 17, 327406.Google Scholar
Salthouse, TA (2009) When does age-related cognitive decline begin? Neurobiology of Aging 30, 507514.CrossRefGoogle ScholarPubMed
Hayano, M et al. (2019) DNA break-induced epigenetic drift as a cause of mammalian aging. SSRN Electronic Journal. https://doi.org/10.2139/ssrn.3466338.Google Scholar
Partridge, L, Deelen, J and Slagboom, PE (2018) Facing up to the global challenges of ageing. Nature 561, 4556.CrossRefGoogle Scholar
Pearce, M and Raftery, AE (2021) Probabilistic forecasting of maximum human lifespan by 2100 using Bayesian population projections. Demographic Research 44, 12711294.CrossRefGoogle Scholar
Berkel, C and Cacan, E (2021) A collective analysis of lifespan-extending compounds in diverse model organisms, and of species whose lifespan can be extended the most by the application of compounds. Biogerontology 22, 639653.CrossRefGoogle ScholarPubMed
Sharma, G et al. (2021) CRISPR-Cas9: a preclinical and clinical perspective for the treatment of human diseases. Molecular Therapy 29, 571586.CrossRefGoogle ScholarPubMed
Rohn, TT et al. (2018) The potential of CRISPR/Cas9 gene editing as a treatment strategy for Alzheimer's disease. Journal of Alzheimer's Disease & Parkinsonism 8, 207221. https://doi.org/10.4172/2161-0460.1000439.Google ScholarPubMed
Newberry, RC (1995) Environmental enrichment: increasing the biological relevance of captive environments. Applied Animal Behaviour Science 44, 229243.CrossRefGoogle Scholar
Sztainberg, Y and Chen, A (2010) An environmental enrichment model for mice. Nature Protocols 5, 15351539.CrossRefGoogle ScholarPubMed
Yamashita, Y et al. (2018) Induction of prolonged natural lifespans in mice exposed to acoustic environmental enrichment. Scientific Reports 8, 18.CrossRefGoogle ScholarPubMed
Arranz, L et al. (2010) Environmental enrichment improves age-related immune system impairment: long-term exposure since adulthood increases life span in mice. Rejuvenation Research 13, 415428.CrossRefGoogle ScholarPubMed
Ziegler-Waldkirch, S et al. (2018) Environmental enrichment reverses Aβ pathology during pregnancy in a mouse model of Alzheimer's disease. Acta Neuropathologica Communications 6, 44.CrossRefGoogle Scholar
Griñán-Ferré, C et al. (2018) Environmental enrichment improves cognitive deficits, AD hallmarks and epigenetic alterations presented in 5xFAD mouse model. Frontiers in Cellular Neuroscience 12, 1114. https://doi.org/10.3389/fncel.2018.00224.CrossRefGoogle ScholarPubMed
Rodriguez, JJ et al. (2011) Voluntary running and environmental enrichment restores impaired hippocampal neurogenesis in a triple transgenic mouse model of Alzheimer's disease. Current Alzheimer Research 8, 707717.CrossRefGoogle Scholar
van Dellen, A et al. (2000) Delaying the onset of Huntington's in mice. Nature 404, 721722.CrossRefGoogle ScholarPubMed
Couly, S et al. (2021) Exposure of R6/2 mice in an enriched environment augments P42 therapy efficacy on Huntington's disease progression. Neuropharmacology 186, 108467.CrossRefGoogle Scholar
Hockly, E et al. (2002) Environmental enrichment slows disease progression in R6/2 Huntington's disease mice. Annals of Neurology 51, 235242.CrossRefGoogle ScholarPubMed
Zajac, MS et al. (2018) Short-term environmental stimulation spatiotemporally modulates specific serotonin receptor gene expression and behavioral pharmacology in a sexually dimorphic manner in Huntington's disease transgenic mice. Frontiers in Molecular Neuroscience 11, 114. https://doi.org/10.3389/fnmol.2018.00433.CrossRefGoogle Scholar
Bezard, E et al. (2003) Enriched environment confers resistance to 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine and cocaine: involvement of dopamine transporter and trophic factors. The Journal of Neuroscience 23, 1099911007.CrossRefGoogle ScholarPubMed
Jungling, A et al. (2017) Effects of postnatal enriched environment in a model of Parkinson's disease in adult rats. International Journal of Molecular Sciences 18, 406.CrossRefGoogle Scholar
Silva, BA et al. (2020) Environmental enrichment improves cognitive symptoms and pathological features in a focal model of cortical damage of multiple sclerosis. Brain Research 1727, 146520.CrossRefGoogle Scholar
Suemaru, K et al. (2018) Environmental enrichment alleviates cognitive and behavioral impairments in EL mice. Epilepsy & Behavior 85, 227233.CrossRefGoogle ScholarPubMed
Fratiglioni, L, Marseglia, A and Dekhtyar, S (2020) Ageing without dementia: can stimulating psychosocial and lifestyle experiences make a difference? The Lancet. Neurology 19, 533543.CrossRefGoogle Scholar
Wahl, D et al. (2019) Aging, lifestyle and dementia. Neurobiology of Disease 130, 104481.CrossRefGoogle ScholarPubMed
Clemenson, GD, Gage, FH and Stark, CEL (2018) Environmental enrichment and neuronal plasticity. In Chao, MV (ed.), The Oxford Handbook of Developmental Neural Plasticity. New York, NY: Oxford University Press, pp. 142, https://doi.org/10.1093/oxfordhb/9780190635374.013.13.Google Scholar
Figuracion, KCF and Lewis, FM (2021) Environmental enrichment: a concept analysis. Nursing Forum 56, 703709.CrossRefGoogle ScholarPubMed
Caspersen, CJ, Powell, KE and Christenson, GM (1985) Physical activity, exercise, and physical fitness: definitions and distinctions for health-related research. Public Health Reports 100, 126131.Google ScholarPubMed
Manzanares, G, Brito-da-Silva, G and Gandra, PG (2019) Voluntary wheel running: patterns and physiological effects in mice. Brazilian Journal of Medical and Biological Research 52, 19. https://doi.org/10.1590/1414-431x20187830.CrossRefGoogle Scholar
Holloszy, JO (1997) Mortality rate and longevity of food-restricted exercising male rats: a reevaluation. Journal of Applied Physiology 82, 399403.CrossRefGoogle ScholarPubMed
Laranjeiro, R et al. (2019) Swim exercise in Caenorhabditis elegans extends neuromuscular and gut healthspan, enhances learning ability, and protects against neurodegeneration. Proceedings of the National Academy of Sciences of the USA 116, 2382923839.CrossRefGoogle ScholarPubMed
Garcia-Valles, R et al. (2013) Life-long spontaneous exercise does not prolong lifespan but improves health span in mice. Longevity & Healthspan 2, 14.CrossRefGoogle Scholar
Karvinen, S et al. (2015) Physical activity in adulthood: genes and mortality. Scientific Reports 5, 19. https://doi.org/10.1038/srep18259.CrossRefGoogle ScholarPubMed
Lee, D-C et al. (2014) Leisure-time running reduces all-cause and cardiovascular mortality risk. Journal of the American College of Cardiology 64, 472481.CrossRefGoogle ScholarPubMed
Pasanen, T et al. (2017) Exercise therapy for functional capacity in chronic diseases: an overview of meta-analyses of randomised controlled trials. British Journal of Sports Medicine 51, 14591465.CrossRefGoogle ScholarPubMed
O'Keefe, JH et al. (2012) Potential adverse cardiovascular effects from excessive endurance exercise. In Mayo Clin Proc. Elsevier Ltd, pp. 587595. https://doi.org/10.1016/j.mayocp.2012.04.005.Google Scholar
Huxley, RR (2015) Physical activity: can there be too much of a good thing? Circulation 131, 692694.CrossRefGoogle ScholarPubMed
Schnohr, P et al. (2015) Dose of jogging and long-term mortality: the Copenhagen City heart study. Journal of the American College of Cardiology 65, 411419.CrossRefGoogle Scholar
O'Keefe, JH, O'Keefe, EL and Lavie, CJ (2018) The Goldilocks zone for exercise: not too little, not too much. Missouri Medicine 115, 98105.Google Scholar
García-Mesa, Y et al. (2011) Physical exercise protects against Alzheimer's disease in 3xTg-AD mice. Journal of Alzheimer's Disease 24, 421454.CrossRefGoogle ScholarPubMed
Cho, J et al. (2015) Treadmill running reverses cognitive declines due to Alzheimer disease. Medicine & Science in Sports & Exercise 47, 18141824.CrossRefGoogle ScholarPubMed
Souza, LC et al. (2013) Neuroprotective effect of physical exercise in a mouse model of Alzheimer's disease induced by β-amyloid1–40 peptide. Neurotoxicity Research 24, 148163.CrossRefGoogle Scholar
Liu, Y et al. (2020) Short-term resistance exercise inhibits neuroinflammation and attenuates neuropathological changes in 3xTg Alzheimer's disease mice. Journal of Neuroinflammation 17, 4.CrossRefGoogle ScholarPubMed
Richter, H et al. (2008) Wheel-running in a transgenic mouse model of Alzheimer's disease: protection or symptom? Behavioural Brain Research 190, 7484.CrossRefGoogle ScholarPubMed
Sim, Y-J (2014) Treadmill exercise alleviates impairment of spatial learning ability through enhancing cell proliferation in the streptozotocin-induced Alzheimer's disease rats. Journal of Exercise Rehabilitation 10, 8188.CrossRefGoogle ScholarPubMed
de Sousa, RAL et al. (2021) Physical exercise protocols in animal models of Alzheimer's disease: a systematic review. Metabolic Brain Disease 36, 8595.CrossRefGoogle ScholarPubMed
Lai, J-H et al. (2019) Voluntary exercise delays progressive deterioration of markers of metabolism and behavior in a mouse model of Parkinson's disease. Brain Research 1720, 146301.CrossRefGoogle Scholar
Zhou, W, Barkow, JC and Freed, CR (2017) Running wheel exercise reduces α-synuclein aggregation and improves motor and cognitive function in a transgenic mouse model of Parkinson's disease. PLoS ONE 12, e0190160.CrossRefGoogle Scholar
Lau, Y-S et al. (2011) Neuroprotective effects and mechanisms of exercise in a chronic mouse model of Parkinson's disease with moderate neurodegeneration. European Journal of Neuroscience 33, 12641274.CrossRefGoogle Scholar
Jang, YH, Chae, HS and Kim, YJ (2017) Female-specific myoinhibitory peptide neurons regulate mating receptivity in Drosophila melanogaster. Nature Communications 8, 112. https://doi.org/10.1038/s41467-017-01794-9.CrossRefGoogle ScholarPubMed
Ji, E-S et al. (2015) Treadmill exercise enhances spatial learning ability through suppressing hippocampal apoptosis in Huntington's disease rats. Journal of Exercise Rehabilitation 11, 133139.CrossRefGoogle ScholarPubMed
Pothakos, K, Kurz, MJ and Lau, Y-S (2009) Restorative effect of endurance exercise on behavioral deficits in the chronic mouse model of Parkinson's disease with severe neurodegeneration. BMC Neuroscience 10, 6.CrossRefGoogle ScholarPubMed
Palasz, E et al. (2019) Exercise-induced neuroprotection and recovery of motor function in animal models of Parkinson's disease. Frontiers in Neurology 10, 115. https://doi.org/10.3389/fneur.2019.01143.CrossRefGoogle ScholarPubMed
Pang, TYC et al. (2006) Differential effects of voluntary physical exercise on behavioral and brain-derived neurotrophic factor expression deficits in Huntington's disease transgenic mice. Neuroscience 141, 569584.CrossRefGoogle ScholarPubMed
van Dellen, A et al. (2008) Wheel running from a juvenile age delays onset of specific motor deficits but does not alter protein aggregate density in a mouse model of Huntington's disease. BMC Neuroscience 9, 34.CrossRefGoogle ScholarPubMed
Caldwell, CC et al. (2020) Treadmill exercise rescues mitochondrial function and motor behavior in the CAG140 knock-in mouse model of Huntington's disease. Chemico–Biological Interactions 315, 108907.CrossRefGoogle ScholarPubMed
Stefanko, DP et al. (2017) Treadmill exercise delays the onset of non-motor behaviors and striatal pathology in the CAG140 knock-in mouse model of Huntington's disease. Neurobiology of Disease 105, 1532.CrossRefGoogle ScholarPubMed
Potter, MC et al. (2010) Exercise is not beneficial and may accelerate symptom onset in a mouse model of Huntington's disease. PLoS Currents 2, RRN1201.CrossRefGoogle ScholarPubMed
Corrochano, S et al. (2018) A genetic modifier suggests that endurance exercise exacerbates Huntington's disease. Human Molecular Genetics 27, 17231731.CrossRefGoogle ScholarPubMed
de la Rosa, A et al. (2020) Physical exercise in the prevention and treatment of Alzheimer's disease. Journal of Sport and Health Science 9, 394404.CrossRefGoogle ScholarPubMed
Paillard, T, Rolland, Y and de Souto Barreto, P (2015) Protective effects of physical exercise in Alzheimer's disease and Parkinson's disease: a narrative review. Journal of Clinical Neurology 11, 212.CrossRefGoogle ScholarPubMed
Ahlskog, JE (2011) Does vigorous exercise have a neuroprotective effect in Parkinson disease? Neurology 77, 288294.CrossRefGoogle ScholarPubMed
Frese, S et al. (2017) Exercise effects in Huntington disease. Journal of Neurology 264, 3239.CrossRefGoogle ScholarPubMed
Mueller, SM, Petersen, JA and Jung, HH (2019) Exercise in Huntington's disease: current state and clinical significance. Tremor and Other Hyperkinetic Movements 9, 601.CrossRefGoogle ScholarPubMed
Madeo, F et al. (2019) Caloric restriction mimetics against age-associated disease: targets, mechanisms, and therapeutic potential. Cell Metabolism 29, 592610.CrossRefGoogle ScholarPubMed
López-Lluch, G and Navas, P (2016) Calorie restriction as an intervention in ageing. Journal of Physiology 594, 20432060.CrossRefGoogle ScholarPubMed
Solon-Biet, SM et al. (2014) The ratio of macronutrients, not caloric intake, dictates cardiometabolic health, aging, and longevity in ad libitum-fed mice. Cell Metabolism 19, 418430.CrossRefGoogle Scholar
Brown-Borg, HM and Buffenstein, R (2017) Cutting back on the essentials: can manipulating intake of specific amino acids modulate health and lifespan? Ageing Research Reviews 39, 8795.CrossRefGoogle ScholarPubMed
Juricic, P, Grönke, S and Partridge, L (2020) Branched-chain amino acids have equivalent effects to other essential amino acids on lifespan and aging-related traits in Drosophila. The Journals of Gerontology, Series A: Biological Sciences and Medical Sciences 75, 2431.CrossRefGoogle ScholarPubMed
Solon-Biet, SM et al. (2019) Branched-chain amino acids impact health and lifespan indirectly via amino acid balance and appetite control. Nature Metabolism 1, 532545.CrossRefGoogle ScholarPubMed
Mitchell, SJ et al. (2019) Daily fasting improves health and survival in male mice independent of diet composition and calories. Cell Metabolism 29, 221228.e3.CrossRefGoogle ScholarPubMed
Simpson, SJ et al. (2017) Dietary protein, aging and nutritional geometry. Ageing Research Reviews 39, 7886.CrossRefGoogle ScholarPubMed
Green, CL, Lamming, DW and Fontana, L (2021) Molecular mechanisms of dietary restriction promoting health and longevity. Nature Reviews Molecular Cell Biology 23, 5673.CrossRefGoogle ScholarPubMed
Duregon, E et al. (2021) Intermittent fasting: from calories to time restriction. Geroscience 43, 10831092.CrossRefGoogle ScholarPubMed
Most, J et al. (2017) Calorie restriction in humans: an update. Ageing Research Reviews 39, 3645.CrossRefGoogle ScholarPubMed
Dorling, JL et al. (2021) Effects of caloric restriction on human physiological, psychological, and behavioral outcomes: highlights from CALERIE phase 2. Nutrition Reviews 79, 98113.CrossRefGoogle ScholarPubMed
Di Francesco, A et al. (2018) A time to fast. Science (1979) 362, 770775.Google Scholar
Yang, Y and Zhang, L (2020) The effects of caloric restriction and its mimetics in Alzheimer's disease through autophagy pathways. Food & Function 11, 12111224.CrossRefGoogle ScholarPubMed
Halagappa, VKM et al. (2007) Intermittent fasting and caloric restriction ameliorate age-related behavioral deficits in the triple-transgenic mouse model of Alzheimer's disease. Neurobiology of Disease 26, 212220.CrossRefGoogle ScholarPubMed
Wu, P et al. (2008) Calorie restriction ameliorates neurodegenerative phenotypes in forebrain-specific presenilin-1 and presenilin-2 double knockout mice. Neurobiology of Aging 29, 15021511.CrossRefGoogle ScholarPubMed
Brownlow, ML et al. (2014) Partial rescue of memory deficits induced by calorie restriction in a mouse model of tau deposition. Behavioural Brain Research 271, 7988.CrossRefGoogle Scholar
Pedersen, WA and Mattson, MP (1999) No benefit of dietary restriction on disease onset or progression in amyotrophic lateral sclerosis Cu/Zn-superoxide dismutase mutant mice. Brain Research 833, 117120.CrossRefGoogle ScholarPubMed
van Cauwenberghe, C et al. (2016) Caloric restriction: beneficial effects on brain aging and Alzheimer's disease. Mammalian Genome 27, 300319.CrossRefGoogle ScholarPubMed
Fontana, L et al. (2021) Effects of dietary restriction on neuroinflammation in neurodegenerative diseases. Journal of Experimental Medicine 218, 114. https://doi.org/10.1084/jem.20190086.CrossRefGoogle ScholarPubMed
Bayliss, JA et al. (2016) Ghrelin-AMPK signaling mediates the neuroprotective effects of calorie restriction in Parkinson's disease. Journal of Neuroscience 36, 30493063.CrossRefGoogle ScholarPubMed
Duan, W and Mattson, MP (1999) Dietary restriction and 2-deoxyglucose administration improve behavioral outcome and reduce degeneration of dopaminergic neurons in models of Parkinson's disease. Journal of Neuroscience Research 57, 195206.3.0.CO;2-P>CrossRefGoogle ScholarPubMed
Maswood, N et al. (2004) Caloric restriction increases neurotrophic factor levels and attenuates neurochemical and behavioral deficits in a primate model of Parkinson's disease. Proceedings of the National Academy of Sciences 101, 1817118176.CrossRefGoogle Scholar
Bruce-Keller, AJ et al. (1999) Food restriction reduces brain damage and improves behavioral outcome following excitotoxic and metabolic insults. Annals of Neurology 45, 815.3.0.CO;2-V>CrossRefGoogle ScholarPubMed
Duan, W et al. (2003) Dietary restriction normalizes glucose metabolism and BDNF levels, slows disease progression, and increases survival in huntingtin mutant mice. Proceedings of the National Academy of Sciences 100, 29112916.CrossRefGoogle ScholarPubMed
Duan, W et al. (2001) Dietary restriction stimulates BDNF production in the brain and thereby protects neurons against excitotoxic injury. Journal of Molecular Neuroscience 16, 112.CrossRefGoogle ScholarPubMed
Yang, F et al. (2014) mTOR and autophagy in normal brain aging and caloric restriction ameliorating age-related cognition deficits. Behavioural Brain Research 264, 8290.CrossRefGoogle ScholarPubMed
Ajoolabady, A et al. (2021) Targeting autophagy in neurodegenerative diseases: from molecular mechanisms to clinical therapeutics. Clinical and Experimental Pharmacology and Physiology 48, 943953.CrossRefGoogle ScholarPubMed
Luchsinger, JA et al. (2002) Caloric intake and the risk of Alzheimer disease. Archives of Neurology 59. 1258.CrossRefGoogle ScholarPubMed
Horie, NC et al. (2016) Cognitive effects of intentional weight loss in elderly obese individuals with mild cognitive impairment. Journal of Clinical Endocrinology and Metabolism 101, 11041112.CrossRefGoogle ScholarPubMed
Agim, ZS and Cannon, JR (2015) Dietary factors in the etiology of Parkinson's disease. Biomed Research International 2015, 116.CrossRefGoogle ScholarPubMed
Logroscino, G et al. (1996) Dietary lipids and antioxidants in Parkinson's disease: a population-based, case-control study. Annals of Neurology 39, 8994.CrossRefGoogle ScholarPubMed
Trejo, A et al. (2004) Assessment of the nutrition status of patients with Huntington's disease. Nutrition 20, 192196.CrossRefGoogle ScholarPubMed
Pratley, RE et al. (2000) Higher sedentary energy expenditure in patients with Huntington's disease. Annals of Neurology 47, 6470.3.0.CO;2-S>CrossRefGoogle ScholarPubMed
Chu, C-Q et al. (2021) Dietary patterns affect Parkinson's disease via the microbiota–gut–brain axis. Trends in Food Science & Technology 116, 90101.CrossRefGoogle Scholar
Covarrubias, AJ et al. (2021) NAD+ metabolism and its roles in cellular processes during ageing. Nature Reviews Molecular Cell Biology 22, 119.CrossRefGoogle ScholarPubMed
Rajman, L, Chwalek, K and Sinclair, DA (2018) Therapeutic potential of NAD-boosting molecules: the in vivo evidence. Cell Metabolism 27, 529547.CrossRefGoogle ScholarPubMed
Fang, EF et al. (2019) NAD+ augmentation restores mitophagy and limits accelerated aging in Werner syndrome. https://doi.org/10.1038/s41467-019-13172-8.CrossRefGoogle Scholar
Zhang, H et al. (2016) NAD+ repletion improves mitochondrial and stem cell function and enhances life span in mice. Science (1979) 352, 14361443.Google ScholarPubMed
Fang, EF et al. (2016) NAD+ replenishment improves lifespan and healthspan in ataxia telangiectasia models via mitophagy and DNA repair. Cell Metabolism 24, 566581.CrossRefGoogle ScholarPubMed
Mills, KF et al. (2016) Long-term administration of nicotinamide mononucleotide mitigates age-associated physiological decline in mice. Cell Metabolism 24, 795806.CrossRefGoogle ScholarPubMed
Mitchell, SJ et al. (2018) Nicotinamide improves aspects of healthspan, but not lifespan, in mice. Cell Metabolism 27, 667676.e4.CrossRefGoogle Scholar
Harrison, DE et al. (2021) 17-a-estradiol late in life extends lifespan in aging UM-HET3 male mice; nicotinamide riboside and three other drugs do not affect lifespan in either sex. Aging Cell 20, 110. https://doi.org/10.1111/acel.13328.CrossRefGoogle Scholar
Hou, Y et al. (2018) NAD+ supplementation normalizes key Alzheimer's features and DNA damage responses in a new AD mouse model with introduced DNA repair deficiency. Proceedings of the National Academy of Sciences 115, 16. https://doi.org/10.1073/pnas.1718819115.CrossRefGoogle Scholar
Gong, B et al. (2013) Nicotinamide riboside restores cognition through an upregulation of proliferator-activated receptor-γ coactivator 1α regulated β-secretase 1 degradation and mitochondrial gene expression in Alzheimer's mouse models. Neurobiology of Aging 34, 15811588.CrossRefGoogle ScholarPubMed
Wang, X et al. (2016) Nicotinamide mononucleotide protects against β-amyloid oligomer-induced cognitive impairment and neuronal death. Brain Research 1643, 19.CrossRefGoogle ScholarPubMed
Yao, Z et al. (2017) Nicotinamide mononucleotide inhibits JNK activation to reverse Alzheimer disease. Neuroscience Letters 647, 133140.CrossRefGoogle ScholarPubMed
Lautrup, S et al. (2019) NAD+ in brain aging and neurodegenerative disorders. Cell Metabolism 30, 630655.CrossRefGoogle ScholarPubMed
Lu, L et al. (2014) Nicotinamide mononucleotide improves energy activity and survival rate in an in vitro model of Parkinson's disease. Experimental and Therapeutic Medicine 8, 943950.CrossRefGoogle Scholar
Schöndorf, DC et al. (2018) The NAD+ precursor nicotinamide riboside rescues mitochondrial defects and neuronal loss in iPSC and fly models of Parkinson's disease. Cell Reports 23, 29762988.CrossRefGoogle ScholarPubMed
Braidy, N and Liu, Y (2020) Can nicotinamide riboside protect against cognitive impairment? Current Opinion in Clinical Nutrition and Metabolic Care 23, 413420.CrossRefGoogle ScholarPubMed
Martens, CR et al. (2018) Chronic nicotinamide riboside supplementation is well-tolerated and elevates NAD+ in healthy middle-aged and older adults. Nature Communications 9, 1286.CrossRefGoogle ScholarPubMed
Dollerup, OL et al. (2018) A randomized placebo-controlled clinical trial of nicotinamide riboside in obese men: safety, insulin-sensitivity, and lipid-mobilizing effects. American Journal of Clinical Nutrition 108, 343353.CrossRefGoogle ScholarPubMed
Brakedal, B et al. (2022) The NADPARK study: a randomized phase I trial of nicotinamide riboside supplementation in Parkinson's disease. Cell Metabolism 34, 396407.e6.CrossRefGoogle ScholarPubMed
Pezzuto, JM (2019) Resveratrol: twenty years of growth, development and controversy. Biomolecules and Therapeutics 27, 114.CrossRefGoogle ScholarPubMed
Fischer-Posovszky, P et al. (2010) Resveratrol regulates human adipocyte number and function in a Sirt1-dependent manner. American Journal of Clinical Nutrition 92, 515.CrossRefGoogle Scholar
Howitz, KT et al. (2003) Small molecule activators of sirtuins extend Saccharomyces cerevisiae lifespan. Nature 425, 191196.CrossRefGoogle ScholarPubMed
Pacholec, M et al. (2010) SRT1720, SRT2183, SRT1460, and resveratrol are not direct activators of SIRT1. Journal of Biological Chemistry 285, 83408351.CrossRefGoogle Scholar
Imai, S-I and Guarente, L (2014) NAD+ and sirtuins in aging and disease. Trends in Cell Biology 24, 464471.CrossRefGoogle ScholarPubMed
Godoy, JA et al. (2014) Role of Sirt1 during the ageing process: relevance to protection of synapses in the brain. Molecular Neurobiology 50, 744756.CrossRefGoogle ScholarPubMed
Walle, T (2011) Bioavailability of resveratrol. Annals of the New York Academy of Sciences 1215, 915.CrossRefGoogle ScholarPubMed
Smoliga, JM, Baur, JA and Hausenblas, HA (2011) Resveratrol and health – a comprehensive review of human clinical trials. Molecular Nutrition & Food Research 55, 11291141.CrossRefGoogle Scholar
Li, Y-R, Li, S and Lin, C-C (2018) Effect of resveratrol and pterostilbene on aging and longevity. BioFactors 44, 6982.CrossRefGoogle ScholarPubMed
Hector, KL, Lagisz, M and Nakagawa, S (2012) The effect of resveratrol on longevity across species: a meta-analysis. Biology Letters 8, 790793.CrossRefGoogle ScholarPubMed
Pallauf, K et al. (2016) Resveratrol and lifespan in model organisms. Current Medicinal Chemistry 23, 46394680.CrossRefGoogle ScholarPubMed
Semba, RD et al. (2014) Resveratrol levels and all-cause mortality in older community-dwelling adults. JAMA Internal Medicine 174, 1077.CrossRefGoogle ScholarPubMed
Fernández, AF and Fraga, MF (2011) The effects of the dietary polyphenol resveratrol on human healthy aging and lifespan. Epigenetics 6, 870874.CrossRefGoogle ScholarPubMed
Kim, D et al. (2007) SIRT1 deacetylase protects against neurodegeneration in models for Alzheimer's disease and amyotrophic lateral sclerosis. EMBO Journal 26, 31693179.CrossRefGoogle ScholarPubMed
Zhao, H et al. (2012) Long-term resveratrol consumption protects ovariectomized rats chronically treated with d-galactose from developing memory decline without effects on the uterus, Brain Research 1467, 6780.CrossRefGoogle ScholarPubMed
Zhao, HF et al. (2015) Resveratrol decreases the insoluble Aβ1–42 level in hippocampus and protects the integrity of the blood–brain barrier in AD rats. Neuroscience 310, 641649.CrossRefGoogle ScholarPubMed
Lofrumento, DD et al. (2014) Neuroprotective effects of resveratrol in an MPTP mouse model of Parkinson's-like disease: possible role of SOCS-1 in reducing pro-inflammatory responses. Innate Immunity 20, 249260.CrossRefGoogle Scholar
Blanchet, J et al. (2008) Resveratrol, a red wine polyphenol, protects dopaminergic neurons in MPTP-treated mice. Progress in Neuro-Psychopharmacology & Biological Psychiatry 32, 12431250.CrossRefGoogle ScholarPubMed
Jin, F et al. (2008) Neuroprotective effect of resveratrol on 6-OHDA-induced Parkinson's disease in rats. European Journal of Pharmacology 600, 7882.CrossRefGoogle ScholarPubMed
Andrade, S et al. (2018) Resveratrol brain delivery for neurological disorders prevention and treatment. Frontiers in Pharmacology 9, 119. https://doi.org/10.3389/fphar.2018.01261.CrossRefGoogle ScholarPubMed
Naia, L et al. (2017) Comparative mitochondrial-based protective effects of resveratrol and nicotinamide in Huntington's disease models. Molecular Neurobiology 54, 53855399.CrossRefGoogle ScholarPubMed
Ho, DJ et al. (2010) Resveratrol protects against peripheral deficits in a mouse model of Huntington's disease. Experimental Neurology 225, 7484.CrossRefGoogle Scholar
Tallaksen-Greene, SJ and Albin, RL (2011) Treating mouse models of Huntington disease. Neuropsychopharmacology 36, 23732374.CrossRefGoogle ScholarPubMed
Kumar, P et al. (2006) Effect of resveratrol on 3-nitropropionic acid-induced biochemical and behavioural changes: possible neuroprotective mechanisms. Behavioural Pharmacology 17, 485492.CrossRefGoogle ScholarPubMed
Bernier, M et al. (2016) Resveratrol supplementation confers neuroprotection in cortical brain tissue of nonhuman primates fed a high-fat/sucrose diet. Aging 8, 899916.CrossRefGoogle ScholarPubMed
Köbe, T et al. (2017) Impact of resveratrol on glucose control, hippocampal structure and connectivity, and memory performance in patients with mild cognitive impairment. Frontiers in Neuroscience 11, 111.CrossRefGoogle ScholarPubMed
Turner, RS et al. (2015) A randomized, double-blind, placebo-controlled trial of resveratrol for Alzheimer disease. Neurology 85, 13831391.CrossRefGoogle ScholarPubMed
Moussa, C et al. (2017) Resveratrol regulates neuro-inflammation and induces adaptive immunity in Alzheimer's disease. Journal of Neuroinflammation 14, 110.CrossRefGoogle ScholarPubMed
Zhu, CW et al. (2018) A randomized, double-blind, placebo-controlled trial of resveratrol with glucose and malate (RGM) to slow the progression of Alzheimer's disease: a pilot study. Alzheimer's & Dementia: Translational Research & Clinical Interventions 4, 609616.CrossRefGoogle ScholarPubMed
Hambright, WS, Philippon, MJ and Huard, J (2020) Rapamycin for aging stem cells. Aging 12, 1518415185.CrossRefGoogle ScholarPubMed
Martel, J et al. (2021) Recent advances in the field of caloric restriction mimetics and anti-aging molecules. Ageing Research Reviews 66, 101240.CrossRefGoogle ScholarPubMed
Unnikrishnan, A et al. (2020) Is rapamycin a dietary restriction mimetic? The Journals of Gerontology, Series A: Biological Sciences and Medical Sciences 75, 413.CrossRefGoogle ScholarPubMed
Weichhart, T (2018) mTOR as regulator of lifespan, aging, and cellular senescence: a mini-review. Gerontology 64, 127134.CrossRefGoogle ScholarPubMed
Harrison, DE et al. (2009) Rapamycin fed late in life extends lifespan in genetically heterogeneous mice. Nature 460, 392395.CrossRefGoogle ScholarPubMed
Miller, RA et al. (2014) Rapamycin-mediated lifespan increase in mice is dose and sex dependent and metabolically distinct from dietary restriction. Aging Cell 13, 468477.CrossRefGoogle ScholarPubMed
Bitto, A et al. (2016) Transient rapamycin treatment can increase lifespan and healthspan in middle-aged mice. eLife 5, 117.CrossRefGoogle ScholarPubMed
Zhang, Y et al. (2014) Rapamycin extends life and health in C57BL/6 mice. The Journals of Gerontology, Series A: Biological Sciences and Medical Sciences 69, 119130.CrossRefGoogle ScholarPubMed
Blagosklonny, MV (2019) Rapamycin for longevity: opinion article. Aging 11, 80488067.CrossRefGoogle ScholarPubMed
A Quick Update on PEARL, ARX's Upcoming Clinical Trial (2021) Available at https://www.agelessrx.com/post/a-quick-update-on-pearl-arx-s-upcoming-clinical-trial (Accessed 28 August 2021).Google Scholar
Johnson, SC and Kaeberlein, M (2016) Rapamycin in aging and disease: maximizing efficacy while minimizing side effects. Oncotarget 7, 4487644878.CrossRefGoogle ScholarPubMed
Spilman, P et al. (2010) Inhibition of mTOR by rapamycin abolishes cognitive deficits and reduces amyloid-β levels in a mouse model of Alzheimer's disease. PLoS ONE 5, 18.CrossRefGoogle Scholar
Lin, AL et al. (2013) 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 33, 14121421.CrossRefGoogle ScholarPubMed
Caccamo, A et al. (2010) Molecular interplay between mammalian target of rapamycin (mTOR), amyloid-β, and tau: effects on cognitive impairments. Journal of Biological Chemistry 285, 1310713120.CrossRefGoogle ScholarPubMed
Malagelada, C et al. (2010) Rapamycin protects against neuron death in in vitro and in vivo models of Parkinson's disease. Journal of Neuroscience 30, 11661175.CrossRefGoogle ScholarPubMed
Zhang, Y et al. (2017) Rapamycin upregulates glutamate transporter and IL-6 expression in astrocytes in a mouse model of Parkinson's disease. Cell Death & Disease 8, 113.Google Scholar
Jiang, J et al. (2013) Rapamycin protects the mitochondria against oxidative stress and apoptosis in a rat model of Parkinson's disease. International Journal of Molecular Medicine 31, 825832.CrossRefGoogle Scholar
Ravikumar, B et al. (2004) Inhibition of mTOR induces autophagy and reduces toxicity of polyglutamine expansions in fly and mouse models of Huntington disease. Nature Genetics 36, 585595.CrossRefGoogle ScholarPubMed
Fox, JH et al. (2010) The mTOR kinase inhibitor everolimus decreases S6 kinase phosphorylation but fails to reduce mutant huntingtin levels in brain and is not neuroprotective in the R6/2 mouse model of Huntington's disease. Molecular Neurodegeneration 5, 112.CrossRefGoogle Scholar
Sarkar, S et al. (2008) A rational mechanism for combination treatment of Huntington's disease using lithium and rapamycin. Human Molecular Genetics 17, 170178.CrossRefGoogle ScholarPubMed
Kraig, E et al. (2018) A randomized control trial to establish the feasibility and safety of rapamycin treatment in an older human cohort: immunological, physical performance, and cognitive effects. Experimental Gerontology 105, 5369.CrossRefGoogle Scholar
resTORbio announces interim results for phase 1b/2a trial of RTB101 in patients with Parkinson's disease and provides corporate update, resTORbio, Inc. (2020) Available at https://ir.restorbio.com/news-releases/news-release-details/restorbio-announces-interim-results-phase-1b2a-trial-rtb101.Google Scholar
Kaeberlein, M and Galvan, V (2019) Rapamycin and Alzheimer's disease: time for a clinical trial? Science Translational Medicine 11, eaar4289.CrossRefGoogle ScholarPubMed
Carosi, JM and Sargeant, TJ (2019) Rapamycin and Alzheimer disease: a double-edged sword? Autophagy 15, 14601462.CrossRefGoogle ScholarPubMed
Flory, J and Lipska, K (2019) Metformin in 2019. JAMA – Journal of the American Medical Association 321, 19261927.CrossRefGoogle ScholarPubMed
Anisimov, VN et al. (2010) Gender differences in metformin effect on aging, life span and spontaneous tumorigenesis in 129/Sv mice. Aging 2, 945958.CrossRefGoogle ScholarPubMed
Smith, DL et al. (2010) Metformin supplementation and life span in Fischer-344 rats. The Journals of Gerontology, Series A: Biological Sciences and Medical Sciences 65, 468474.CrossRefGoogle ScholarPubMed
Onken, B et al. (2021) Metformin treatment of diverse Caenorhabditis species reveals the importance of genetic background in longevity and healthspan extension outcomes. Aging Cell 21, 111. https://doi.org/10.1111/ACEL.13488.Google Scholar
Ma, TC et al. (2007) Metformin therapy in a transgenic mouse model of Huntington's disease. Neuroscience Letters 411, 98103.CrossRefGoogle Scholar
Anisimov, VN et al. (2008) Metformin slows down aging and extends life span of female SHR mice. Cell Cycle 7, 27692773.CrossRefGoogle ScholarPubMed
Anisimov, VN et al. (2011) If started early in life, metformin treatment increases life span and postpones tumors in female SHR mice. Aging 3, 148157.CrossRefGoogle ScholarPubMed
Martin-Montalvo, A et al. (2013) Metformin improves healthspan and lifespan in mice. Nature Communications 4, 19. https://doi.org/10.1038/ncomms3192.CrossRefGoogle ScholarPubMed
Slack, C, Foley, A and Partridge, L (2012) Activation of AMPK by the putative dietary restriction mimetic metformin is insufficient to extend lifespan in Drosophila. PLoS ONE 7, 17.CrossRefGoogle ScholarPubMed
Smith, DL et al. (2010) Metformin supplementation and life span in Fischer-344 rats. The Journals of Gerontology, Series A: Biological Sciences and Medical Sciences 65, 468474.CrossRefGoogle ScholarPubMed
Strong, R et al. (2016) Longer lifespan in male mice treated with a weakly estrogenic agonist, an antioxidant, an α-glucosidase inhibitor or a Nrf2-inducer. Aging Cell 15, 872884.CrossRefGoogle ScholarPubMed
Mohammed, I et al. (2021) A critical review of the evidence that metformin is a putative anti-aging drug that enhances healthspan and extends lifespan. Frontiers in Endocrinology 12, 933.CrossRefGoogle ScholarPubMed
Bannister, CA et al. (2014) Can people with type 2 diabetes live longer than those without? A comparison of mortality in people initiated with metformin or sulphonylurea monotherapy and matched, non-diabetic controls. Diabetes, Obesity & Metabolism 16, 11651173.CrossRefGoogle ScholarPubMed
Campbell, JM et al. (2017) Metformin reduces all-cause mortality and diseases of ageing independent of its effect on diabetes control: a systematic review and meta-analysis. Ageing Research Reviews 40, 3144.CrossRefGoogle ScholarPubMed
Barzilai, N et al. (2016) Metformin as a tool to target aging. Cell Metabolism 23, 10601065.CrossRefGoogle ScholarPubMed
Lee, CG et al. (2021) Diabetes prevention program research group, diabetes prevention program research group: effect of metformin and lifestyle interventions on mortality in the diabetes prevention program and diabetes prevention program outcomes study. Diabetes Care 44, 27752782.CrossRefGoogle Scholar
Adak, T et al. (2018) A reappraisal on metformin. Regulatory Toxicology and Pharmacology 92, 324332.CrossRefGoogle ScholarPubMed
Chaudhari, K, Reynolds, CD and Yang, SH (2020) Metformin and cognition from the perspectives of sex, age, and disease. Geroscience 42, 97116.CrossRefGoogle ScholarPubMed
Ou, Z et al. (2018) Metformin treatment prevents amyloid plaque deposition and memory impairment in APP/PS1 mice. Brain Behavior and Immunity 69, 351363.CrossRefGoogle ScholarPubMed
DiTacchio, KA, Heinemann, SF and Dziewczapolski, G (2015) Metformin treatment alters memory function in a mouse model of Alzheimer's disease. Journal of Alzheimer's Disease 44, 4348.CrossRefGoogle Scholar
Asadbegi, M et al. (2016) Neuroprotective effects of metformin against Aβ-mediated inhibition of long-term potentiation in rats fed a high-fat diet. Brain Research Bulletin 121, 178185.CrossRefGoogle ScholarPubMed
Patil, SP et al. (2014) Neuroprotective effect of metformin in MPTP-induced Parkinson's disease in mice. Neuroscience 277, 747754.CrossRefGoogle ScholarPubMed
Lu, M et al. (2016) Metformin prevents dopaminergic neuron death in MPTP/P-induced mouse model of Parkinson's disease via autophagy and mitochondrial ROS clearance. International Journal of Neuropsychopharmacology 19, 137.CrossRefGoogle ScholarPubMed
Tayara, K et al. (2018) Divergent effects of metformin on an inflammatory model of Parkinson's disease. Frontiers in Cellular Neuroscience 12, 116.CrossRefGoogle Scholar
Ismaiel, AAK et al. (2016) Metformin, besides exhibiting strong in vivo anti-inflammatory properties, increases MPTP-induced damage to the nigrostriatal dopaminergic system. Toxicology and Applied Pharmacology 298, 1930.CrossRefGoogle Scholar
Sanchis, A et al. (2019) Metformin treatment reduces motor and neuropsychiatric phenotypes in the zQ175 mouse model of Huntington disease. Experimental and Molecular Medicine 51, 116. https://doi.org/10.1038/s12276-019-0264-9.CrossRefGoogle ScholarPubMed
Imfeld, P et al. (2012) Metformin, other antidiabetic drugs, and risk of Alzheimer's disease: a population-based case-control study. Journal of the American Geriatrics Society 60, 916921.CrossRefGoogle ScholarPubMed
Kuan, Y-C et al. (2017) Effects of metformin exposure on neurodegenerative diseases in elderly patients with type 2 diabetes mellitus. Progress in Neuro-Psychopharmacology & Biological Psychiatry 79, 7783.CrossRefGoogle ScholarPubMed
Moore, EM et al. (2013) Increased risk of cognitive impairment in patients with diabetes is associated with metformin. Diabetes Care 36, 29812987.CrossRefGoogle ScholarPubMed
Huang, CC et al. (2014) Diabetes mellitus and the risk of Alzheimer's disease: a nationwide population-based study. PLoS ONE 9, 17. https://doi.org/10.1371/journal.pone.0087095.Google ScholarPubMed
Koenig, AM et al. (2017) Effects of the insulin sensitizer metformin in Alzheimer disease: pilot data from a randomized placebo-controlled crossover study. Alzheimer Disease and Associated Disorders 31, 107113.CrossRefGoogle ScholarPubMed
Ng, TP et al. (2014) Long-term metformin usage and cognitive function among older adults with diabetes. Journal of Alzheimer's Disease 41, 6168.CrossRefGoogle ScholarPubMed
Campbell, JM et al. (2018) Metformin use associated with reduced risk of dementia in patients with diabetes: a systematic review and meta-analysis. Journal of Alzheimer's Disease 65, 12251236.CrossRefGoogle ScholarPubMed
Wahl, D et al. (2019) Cognitive impairment, and dementia. The Journals of Gerontology, Series A: Biological Sciences and Medical Sciences XX, 110.Google Scholar
Wahlqvist, ML et al. (2012) Metformin-inclusive sulfonylurea therapy reduces the risk of Parkinson's disease occurring with type 2 diabetes in a Taiwanese population cohort. Parkinsonism & Related Disorders 18, 753758.CrossRefGoogle Scholar
Hervás, D et al. (2017) Metformin intake associates with better cognitive function in patients with Huntington's disease. PLoS ONE 12, 111.CrossRefGoogle ScholarPubMed
Tang, BL (2019) Could metformin be therapeutically useful in Huntington's disease? Reviews in the Neurosciences 204, 113. https://doi.org/10.1515/revneuro-2019-0072.Google Scholar
Pucciarelli, S et al. (2012) Spermidine and spermine are enriched in whole blood of nona/centenarians. Rejuvenation Research 15, 590595.CrossRefGoogle ScholarPubMed
Gupta, VK et al. (2013) Restoring polyamines protects from age-induced memory impairment in an autophagy-dependent manner. Nature Neuroscience 16, 14531460.CrossRefGoogle Scholar
Tain, LS et al. (2020) Longevity in response to lowered insulin signaling requires glycine N-methyltransferase-dependent spermidine production. Aging Cell 19, 111. https://doi.org/10.1111/acel.13043.CrossRefGoogle ScholarPubMed
Eisenberg, T et al. (2009) Induction of autophagy by spermidine promotes longevity. Nature Cell Biology 11, 13051314.CrossRefGoogle ScholarPubMed
Eisenberg, T et al. (2016) Cardioprotection and lifespan extension by the natural polyamine spermidine. Nature Medicine 22, 14281438.CrossRefGoogle ScholarPubMed
Morselli, E et al. (2011) Spermidine and resveratrol induce autophagy by distinct pathways converging on the acetylproteome. Journal of Cell Biology 192, 615629.CrossRefGoogle ScholarPubMed
Yue, F et al. (2017) Spermidine prolongs lifespan and prevents liver fibrosis and hepatocellular carcinoma by activating MAP1S-mediated autophagy. Cancer Research 77, 29382951.CrossRefGoogle ScholarPubMed
Kiechl, S et al. (2018) Higher spermidine intake is linked to lower mortality: a prospective population-based study. American Journal of Clinical Nutrition 108, 371380.CrossRefGoogle ScholarPubMed
Freitag, K et al. (2020) The autophagy activator spermidine ameliorates Alzheimer's disease pathology and neuroinflammation in mice. BioRxiv. https://doi.org/10.1101/2020.12.27.424477.Google Scholar
Sandusky-Beltran, LA et al. (2019) Spermidine/spermine-N1-acetyltransferase ablation impacts tauopathy-induced polyamine stress response. Alzheimer's Research & Therapy 11, 58.CrossRefGoogle Scholar
Yang, X et al. (2020) Spermidine inhibits neurodegeneration and delays aging via the PINK1-PDR1-dependent mitophagy pathway in C. elegans. Aging 12, 1685216866.CrossRefGoogle ScholarPubMed
Büttner, S et al. (2014) Spermidine protects against α-synuclein neurotoxicity. Cell Cycle 13, 39033908.CrossRefGoogle ScholarPubMed
Sharma, S, Kumar, P and Deshmukh, R (2018) Neuroprotective potential of spermidine against rotenone induced Parkinson's disease in rats. Neurochemistry International 116, 104111.CrossRefGoogle ScholarPubMed
Velloso, NA et al. (2009) Spermine improves recognition memory deficit in a rodent model of Huntington's disease. Neurobiology of Learning and Memory 92, 574580.CrossRefGoogle Scholar
Xu, T-T et al. (2020) Spermidine and spermine phosphorylate AMPK and induce autophagy in SAMP8. Aging 12, 64016414.CrossRefGoogle ScholarPubMed
Inoue, K et al. (2013) Metabolic profiling of Alzheimer's disease brains. Scientific Reports 3, 19. https://doi.org/10.1038/srep02364.CrossRefGoogle ScholarPubMed
Joaquim, HPG et al. (2019) Decreased plasmatic spermidine and increased spermine in mild cognitive impairment and Alzheimer's disease patients. Revista de Psiquiatria Clinica 46, 120124.CrossRefGoogle Scholar
Mahajan, UV et al. (2020) Dysregulation of multiple metabolic networks related to brain transmethylation and polyamine pathways in Alzheimer disease: a targeted metabolomic and transcriptomic study. PLoS Medicine 17, 131. https://doi.org/10.1371/JOURNAL.PMED.1003012.Google ScholarPubMed
Polis, B, Karasik, D and Samson, AO (2021) Alzheimer's disease as a chronic maladaptive polyamine stress response. Aging 13, 1077010795.CrossRefGoogle ScholarPubMed
Schwarz, C et al. (2018) Safety and tolerability of spermidine supplementation in mice and older adults with subjective cognitive decline. Aging (Albany NY) 10(1), 1933. https://doi.org/10.18632/aging.101354.CrossRefGoogle ScholarPubMed
Wirth, M et al. (2019) Effects of spermidine supplementation on cognition and biomarkers in older adults with subjective cognitive decline (SmartAge) – study protocol for a randomized controlled trial. Alzheimer's Research & Therapy 11, 117. https://doi.org/10.1186/s13195-019-0484-1.CrossRefGoogle ScholarPubMed
Schwarz, C et al. (2020) Spermidine intake is associated with cortical thickness and hippocampal volume in older adults. NeuroImage 221, 18. https://doi.org/10.1016/j.neuroimage.2020.117132.CrossRefGoogle ScholarPubMed
Acosta, JC et al. (2013) A complex secretory program orchestrated by the inflammasome controls paracrine senescence. Nature Cell Biology 15, 978990.CrossRefGoogle ScholarPubMed
Baker, DJ et al. (2016) Naturally occurring p16 Ink4a-positive cells shorten healthy lifespan. Nature 530, 184189.CrossRefGoogle Scholar
Zhang, P et al. (2019) Senolytic therapy alleviates Aβ-associated oligodendrocyte progenitor cell senescence and cognitive deficits in an Alzheimer's disease model. Nature Neuroscience 22, 719728.CrossRefGoogle Scholar
Xu, M et al. (2018) Senolytics improve physical function and increase lifespan in old age. Nature Medicine 24, 12461256.CrossRefGoogle ScholarPubMed
Yousefzadeh, MJ et al. (2018) Fisetin is a senotherapeutic that extends health and lifespan. EBioMedicine 36, 1828.CrossRefGoogle ScholarPubMed
Suda, M et al. (2021) Senolytic vaccination improves normal and pathological age-related phenotypes and increases lifespan in progeroid mice. Nature Aging 1, 11171126.CrossRefGoogle Scholar
Xu, Q et al. (2021) The flavonoid procyanidin C1 has senotherapeutic activity and increases lifespan in mice. Nature Metabolism 3, 17061726.CrossRefGoogle ScholarPubMed
Ahmad, A et al. (2017) Neuroprotective effect of fisetin against amyloid-beta-induced cognitive/synaptic dysfunction, neuroinflammation, and neurodegeneration in adult mice. Molecular Neurobiology 54, 22692285.CrossRefGoogle ScholarPubMed
Ahmad, A et al. (2019) Phytomedicine-based potent antioxidant, fisetin protects CNS-insult LPS-induced oxidative stress-mediated neurodegeneration and memory impairment. Journal of Clinical Medicine 8, 123. https://doi.org/10.3390/jcm8060850.CrossRefGoogle ScholarPubMed
Justice, JN et al. (2019) Senolytics in idiopathic pulmonary fibrosis: results from a first-in-human, open-label, pilot study. EBioMedicine 40, 554563.CrossRefGoogle ScholarPubMed
Jurk, D et al. (2012) Postmitotic neurons develop a p21-dependent senescence-like phenotype driven by a DNA damage response. Aging Cell 11, 9961004.CrossRefGoogle ScholarPubMed
Bhat, R et al. (2012) Astrocyte senescence as a component of Alzheimer's disease. PLoS ONE 7, 110. https://doi.org/10.1371/journal.pone.0045069.CrossRefGoogle ScholarPubMed
Chinta, SJ et al. (2018) Cellular senescence is induced by the environmental neurotoxin paraquat and contributes to neuropathology linked to Parkinson's disease. Cell Reports 22, 930940.CrossRefGoogle ScholarPubMed
di Micco, R et al. (2021) Cellular senescence in ageing: from mechanisms to therapeutic opportunities. Nature Reviews Molecular Cell Biology 22, 7595.CrossRefGoogle ScholarPubMed
Palmer, AK, Tchkonia, T and Kirkland, JL (2021) Senolytics: potential for alleviating diabetes and its complications. Endocrinology (United States) 162, 17. https://doi.org/10.1210/endocr/bqab058.Google ScholarPubMed
Wissler Gerdes, EO et al. (2020) Cellular senescence in aging and age-related diseases: Implications for neurodegenerative diseases. Int Rev Neurobiol 155, 203234. doi: 10.1016/bs.irn.2020.03.019.CrossRefGoogle ScholarPubMed
Lee, S et al. (2021) A guide to senolytic intervention in neurodegenerative disease. Mechanisms of Ageing and Development 200, 114. https://doi.org/10.1016/j.mad.2021.111585.CrossRefGoogle ScholarPubMed
Bocklandt, S et al. (2011) Epigenetic predictor of age. PLoS ONE 6, 16. https://doi.org/10.1371/journal.pone.0014821.CrossRefGoogle ScholarPubMed
Hannum, G et al. (2013) Genome-wide methylation profiles reveal quantitative views of human aging rates. Molecular Cell 49, 359367.CrossRefGoogle ScholarPubMed
Horvath, H and Horvath, S (2013) DNA methylation age of human tissues and cell types. http://genomebiology.com//14/10/R115.CrossRefGoogle Scholar
Trapp, A, Kerepesi, C and Gladyshev, VN (2021) Profiling epigenetic age in single cells. Nature Aging 1, 11891201.CrossRefGoogle ScholarPubMed
Wang, W et al. (2021) A genome-wide CRISPR-based screen identifies KAT7 as a driver of cellular senescence. Science Translational Medicine 13, 113. doi: 10.1126/scitranslmed.abd2655.CrossRefGoogle ScholarPubMed
Takahashi, K and Yamanaka, S (2006) Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 126, 663676.CrossRefGoogle ScholarPubMed
Hébert, JM and Vijg, J (2018) Cell replacement to reverse brain aging: challenges, pitfalls, and opportunities. Trends in Neurosciences 41, 267279.CrossRefGoogle ScholarPubMed
Schweitzer, JS et al. (2020) Personalized iPSC-derived dopamine progenitor cells for Parkinson's disease. New England Journal of Medicine 382, 19261932.CrossRefGoogle ScholarPubMed
Csobonyeiova, M, Polak, S and Danisovic, L (2020) Recent overview of the use of iPSCs Huntington's disease modeling and therapy. International Journal of Molecular Sciences 21, 115. https://doi.org/10.3390/ijms21062239.CrossRefGoogle ScholarPubMed
Spitzhorn, LS et al. (2019) Human iPSC-derived MSCs (iMSCs) from aged individuals acquire a rejuvenation signature. Stem Cell Research & Therapy 10, 118. https://doi.org/10.1186/s13287-019-1209-x.CrossRefGoogle ScholarPubMed
Marion, RM et al. (2009) Telomeres acquire embryonic stem cell characteristics in induced pluripotent stem cells. Cell Stem Cell 4, 141154.CrossRefGoogle ScholarPubMed
Lapasset, L et al. (2011) Rejuvenating senescent and centenarian human cells by reprogramming through the pluripotent state. Genes & Development 25, 22482253.CrossRefGoogle ScholarPubMed
Ocampo, A, Reddy, P and Belmonte, JCI (2016) Anti-aging strategies based on cellular reprogramming. Trends in Molecular Medicine 22, 725738.CrossRefGoogle ScholarPubMed
Bailly, A, Milhavet, O and Lemaitre, JM (2022) RNA-based strategies for cell reprogramming toward pluripotency. Pharmaceutics 14, 125. https://doi.org/10.3390/pharmaceutics14020317.CrossRefGoogle ScholarPubMed
Simpson, DJ, Olova, NN and Chandra, T (2021) Cellular reprogramming and epigenetic rejuvenation. Clinical Epigenetics 13, 110. https://doi.org/10.1186/s13148-021-01158-7.CrossRefGoogle ScholarPubMed
Guan, J et al. (2022) Chemical reprogramming of human somatic cells to pluripotent stem cells. Nature, 325331. https://doi.org/10.1038/s41586-022-04593-5.CrossRefGoogle ScholarPubMed
Ocampo, A et al. (2016) In vivo amelioration of age-associated hallmarks by partial reprogramming. Cell 167, 17191733.e12.CrossRefGoogle ScholarPubMed
Alle, Q et al. (2022) A single short reprogramming early in life initiates and propagates an epigenetically related mechanism improving fitness and promoting an increased healthy lifespan. Aging Cell, 112. https://doi.org/10.1111/acel.13714.Google ScholarPubMed
Browder, KC et al. (2022) In vivo partial reprogramming alters age-associated molecular changes during physiological aging in mice. Nature Aging 2, 243253.CrossRefGoogle Scholar
Sarkar, TJ et al. (2020) Transient non-integrative expression of nuclear reprogramming factors promotes multifaceted amelioration of aging in human cells. Nature Communications 11, 112. https://doi.org/10.1038/s41467-020-15174-3.CrossRefGoogle ScholarPubMed
Fischer, A et al. (2007) Recovery of learning and memory is associated with chromatin remodelling. Nature 447, 178182.CrossRefGoogle ScholarPubMed
de Jager, PL et al. (2014) Alzheimer's disease: early alterations in brain DNA methylation at ANK1, BIN1, RHBDF2 and other loci. Nature Neuroscience 17, 11561163.CrossRefGoogle ScholarPubMed
W.T.C.C.C. (WTCCC2) International Parkinson's Disease Genomics Consortium (IPDGC) (2011) A two-stage meta-analysis identifies several new loci for Parkinson's disease. PLoS Genetics 7, 19. https://doi.org/10.1371/journal.pgen.1002142.Google Scholar
Shireby, GL et al. (2020) Recalibrating the epigenetic clock: implications for assessing biological age in the human cortex. Brain 143, 37633775.CrossRefGoogle ScholarPubMed
Grodstein, F et al. (2021) Characteristics of epigenetic clocks across blood and brain tissue in older women and men. Frontiers in Neuroscience 14, 112. https://doi.org/10.3389/fnins.2020.555307.CrossRefGoogle ScholarPubMed
Fujiwara, N et al. (2013) Restoration of spatial memory dysfunction of human APP transgenic mice by transplantation of neuronal precursors derived from human iPS cells. Neuroscience Letters 557, 129134.CrossRefGoogle ScholarPubMed
Han, B et al. (2015) Quantitative neuropeptidome analysis reveals neuropeptides are correlated with social behavior regulation of the honeybee workers. Journal of Proteome Research 14, 43824393.CrossRefGoogle ScholarPubMed
Doi, D et al. (2020) Pre-clinical study of induced pluripotent stem cell-derived dopaminergic progenitor cells for Parkinson's disease. Nature Communications 11, 114. https://doi.org/10.1038/s41467-020-17165-w.CrossRefGoogle ScholarPubMed
Yoon, Y et al. (2020) Neural transplants from human induced pluripotent stem cells rescue the pathology and behavioral defects in a rodent model of Huntington's disease. Frontiers in Neuroscience 14, 114. https://doi.org/10.3389/fnins.2020.558204.CrossRefGoogle Scholar
Kondo, T et al. (2014) Focal transplantation of human iPSC-derived glial-rich neural progenitors improves lifespan of ALS mice. Stem Cell Reports 3, 242249.CrossRefGoogle ScholarPubMed
Rodríguez-Matellán, A et al. (2020) In vivo reprogramming ameliorates aging features in dentate gyrus cells and improves memory in mice. Stem Cell Reports 15, 10561066.CrossRefGoogle ScholarPubMed
Rebok, GW et al. (2014) Ten-year effects of the advanced cognitive training for independent and vital elderly cognitive training trial on cognition and everyday functioning in older adults. Journal of the American Geriatrics Society 62, 1624.CrossRefGoogle ScholarPubMed
Lee, MB et al. (2021) Antiaging diets: separating fact from fiction. Science 374, 18. https://doi.org/10.1126/science.abe7365.CrossRefGoogle ScholarPubMed
Kempermann, G (2019) Environmental enrichment, new neurons and the neurobiology of individuality. Nature Reviews Neuroscience 20, 235245.CrossRefGoogle ScholarPubMed
von Schwartzenberg, RJ et al. (2021) Caloric restriction disrupts the microbiota and colonization resistance. Nature 595, 272277.CrossRefGoogle ScholarPubMed
Greathouse, KL and Johnson, AJ (2021) Does caloric restriction prime the microbiome for pathogenic bacteria? Cell Host & Microbe 29, 12091211.CrossRefGoogle ScholarPubMed
Shaito, A et al. (2020) Potential adverse effects of resveratrol: a literature review. International Journal of Molecular Sciences 21, 2084.CrossRefGoogle ScholarPubMed
Conze, D, Brenner, C and Kruger, CL (2019) Safety and metabolism of long-term administration of NIAGEN (nicotinamide riboside chloride) in a randomized, double-blind, placebo-controlled clinical trial of healthy overweight adults. Scientific Reports 9, 9772.CrossRefGoogle Scholar
Zong, L et al. (2021) NAD+ augmentation with nicotinamide riboside improves lymphoid potential of Atm−/− and old mice HSCs. NPJ Aging and Mechanisms of Disease 7, 25.CrossRefGoogle ScholarPubMed
Pérez, MJ, Baden, P and Deleidi, M (2021) Progresses in both basic research and clinical trials of NAD+ in Parkinson's disease. Mechanisms of Ageing and Development 197, 111499.CrossRefGoogle ScholarPubMed
Dollerup, OL et al. (2018) A randomized placebo-controlled clinical trial of nicotinamide riboside in obese men: safety, insulin-sensitivity, and lipid-mobilizing effects. American Journal of Clinical Nutrition 108, 343353.CrossRefGoogle ScholarPubMed
Shi, W et al. (2019) High dose of dietary nicotinamide riboside induces glucose intolerance and white adipose tissue dysfunction in mice fed a mildly obesogenic diet. Nutrients 11, 2439.CrossRefGoogle ScholarPubMed
Wang, W et al. (2020) Resveratrol: multi-targets mechanism on neurodegenerative diseases based on network pharmacology. Frontiers in Pharmacology 11, 112. https://doi.org/10.3389/fphar.2020.00694.Google ScholarPubMed
Le, HH et al. (2021) Molecular modelling of the FOXO4-TP53 interaction to design senolytic peptides for the elimination of senescent cancer cells. EBioMedicine 73, 103646.CrossRefGoogle ScholarPubMed
d'Errico, P et al. (2022) Microglia contribute to the propagation of Aβ into unaffected brain tissue. Nature Neuroscience 25, 2025.CrossRefGoogle Scholar
Horvath, S (2013) DNA methylation age of human tissues and cell types. Genome Biology 14, R115.CrossRefGoogle ScholarPubMed
de Magalhães, JP and Ocampo, A (2022) Cellular reprogramming and the rise of rejuvenation biotech. Trends in Biotechnology 40, 639642. https://doi.org/10.1016/j.tibtech.2022.01.011.CrossRefGoogle ScholarPubMed
Gonzalez-Freire, M et al. (2020) The road ahead for health and lifespan interventions. Ageing Research Reviews 59, 101037.CrossRefGoogle ScholarPubMed
Culig, L, Chu, X and Bohr, VA (2022) Neurogenesis in aging and age-related neurodegenerative diseases. Ageing Research Reviews, 101636.CrossRefGoogle ScholarPubMed
Kujala, UM (2018) Is physical activity a cause of longevity? It is not as straightforward as some would believe. A critical analysis. British Journal of Sports Medicine 52, 914918.CrossRefGoogle Scholar
Garmany, A, Yamada, S and Terzic, A (2021) Longevity leap: mind the healthspan gap. NPJ Regenerative Medicine 6, 57. http://dx.doi.org/10.1038/s41536-021-00169-5.CrossRefGoogle ScholarPubMed