Dietary Restriction against Parkinson’s Disease: What We Know So Far
Abstract
:1. Introduction
2. Dietary Restriction
2.1. Methods of DR
2.1.1. Calorie Restriction
2.1.2. Intermittent Fasting
- (a)
- Alternate day fasting (ADF)
- (b)
- Time-restricted feeding
- (c)
- Periodic fasting
2.1.3. Fasting Mimicking Diet (FMD)
2.2. The Physiological Benefits of DR
2.3. Life-Extending Effect of DR and the Influencing Factors
2.4. Delay Aging and Age-Related Diseases
3. General Neuroprotective Effects of DR
3.1. Improve Cognitive and Motor Function
3.2. Promote Neurotrophic Factor Levels
3.3. Improve Neuronal Plasticity
4. Effects of DR on PD and the Underlying Mechanisms
4.1. DR and PD Animal Models
4.2. DR and PD-Related Risk Factors
4.3. Possible Mechanisms of DR on PD
4.3.1. Ameliorate Neuroinflammation
4.3.2. Reduce Oxidative Stress
4.3.3. Preserve Mitochondrial Function and Reduce Mitochondrial Damage
4.3.4. Maintain Autophagy Homeostasis
4.3.5. Regulate Gut Microbiota Composition and Richness
5. Other Dietary Interventions on PD
5.1. Low Fat Diet
5.2. Protein-Restricted Diet and Amino-Acid-Restricted Diet
6. Perspectives
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
Abbreviations
dietary restriction | DR |
Parkinson’s disease | PD |
central nervous system | CNS |
Alzheimer’s disease | AD |
multiple sclerosis | MS |
calorie restriction | CR |
intermittent fasting | IF |
alternate day fasting | ADF |
time-restricted feeding | TRF |
every-other-day fasting | EODF |
intermittent calorie restriction | ICR |
fasting mimicking diet | FMD |
glucagon-like peptide-1 | GLP-1 |
peptide tyrosine-tyrosine | PYY |
cholecystokinin | CCK |
platelet-activating factor acetyl hydrolase | PLA2G7 |
AMP-activated protein kinase | AMPK |
nuclear respiratory factor | NRF |
peroxisome proliferator-activated receptor | PPAR |
ad libitum | AL |
intermittent TRF | iTRF |
glial-cell-line-derived neurotrophic factor | GDNF |
brain-derived neurotrophic factor | BDNF |
indole-3-propionic acid | IPA |
1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine | MPTP |
substantia nigra | SN |
substantia nigra pars compacta | SNpc |
6-hydroxydopamine | 6-OHDA |
α-synuclein preformed fibers | PFF |
dorsal motor nucleus of the vagus | DMV |
high-fat diet | HFD |
type 2 diabetes | T2DM |
intermittent energy restriction | IER |
continuous energy restriction | CER |
multiple sclerosis | MS |
reactive oxygen species | ROS |
sirtuins | SIRTs |
nicotinamide adenine dinucleotide | NAD |
calcium/calmodulin-dependent protein kinase II | CaMKII |
electron transport chain | ETC |
old ad libitum | OAL |
ubiquitin-proteasome system | UPS |
short-chain fatty acids | SCFAs |
blood–brain barrier | BBB |
high-fat diet | HFD |
low-fat diet | LFD |
large amounts of neutral amino acids | LNAA |
branched-chain amino acids | BCAAs |
References
- Tysnes, O.B.; Storstein, A. Epidemiology of Parkinson’s disease. J. Neural. Transm. 2017, 124, 901–905. [Google Scholar] [CrossRef]
- Kalia, L.V.; Lang, A.E. Parkinson’s disease. Lancet 2015, 386, 896–912. [Google Scholar] [CrossRef]
- Lees, A.J.; Hardy, J.; Revesz, T. Parkinson’s disease. Lancet 2009, 373, 2055–2066. [Google Scholar] [CrossRef]
- Connolly, B.S.; Lang, A.E. Pharmacological treatment of Parkinson disease: A review. JAMA 2014, 311, 1670–1683. [Google Scholar] [CrossRef] [PubMed]
- Pajares, M.; Rojo, A.I.; Manda, G.; Bosca, L.; Cuadrado, A. Inflammation in Parkinson’s Disease: Mechanisms and Therapeutic Implications. Cells 2020, 9, 687. [Google Scholar] [CrossRef]
- Macdonald, R.; Barnes, K.; Hastings, C.; Mortiboys, H. Mitochondrial abnormalities in Parkinson’s disease and Alzheimer’s disease: Can mitochondria be targeted therapeutically? Biochem. Soc. Trans. 2018, 46, 891–909. [Google Scholar] [CrossRef]
- Sampson, T.R.; Debelius, J.W.; Thron, T.; Janssen, S.; Shastri, G.G.; Ilhan, Z.E.; Challis, C.; Schretter, C.E.; Rocha, S.; Gradinaru, V.; et al. Gut Microbiota Regulate Motor Deficits and Neuroinflammation in a Model of Parkinson’s Disease. Cell 2016, 167, 1469–1480.e12. [Google Scholar] [CrossRef] [Green Version]
- Chen, Z.; Li, G.; Liu, J. Autonomic dysfunction in Parkinson’s disease: Implications for pathophysiology, diagnosis, and treatment. Neurobiol. Dis. 2020, 134, 104700. [Google Scholar] [CrossRef]
- Kim, S.; Kwon, S.H.; Kam, T.I.; Panicker, N.; Karuppagounder, S.S.; Lee, S.; Lee, J.H.; Kim, W.R.; Kook, M.; Foss, C.A.; et al. Transneuronal Propagation of Pathologic alpha-Synuclein from the Gut to the Brain Models Parkinson’s Disease. Neuron 2019, 103, 627–641.e7. [Google Scholar] [CrossRef]
- Armstrong, M.J.; Okun, M.S. Diagnosis and Treatment of Parkinson Disease: A Review. JAMA 2020, 323, 548–560. [Google Scholar] [CrossRef]
- Singleton, A.B.; Farrer, M.J.; Bonifati, V. The genetics of Parkinson’s disease: Progress and therapeutic implications. Mov. Disord. 2013, 28, 14–23. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Reich, S.G.; Savitt, J.M. Parkinson’s Disease. Med. Clin. N. Am. 2019, 103, 337–350. [Google Scholar] [CrossRef] [PubMed]
- Tambasco, N.; Romoli, M.; Calabresi, P. Levodopa in Parkinson’s Disease: Current Status and Future Developments. Curr. Neuropharmacol. 2018, 16, 1239–1252. [Google Scholar] [CrossRef]
- Salat, D.; Tolosa, E. Levodopa in the treatment of Parkinson’s disease: Current status and new developments. J. Parkinsons Dis. 2013, 3, 255–269. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Alonso Canovas, A.; Luquin Piudo, R.; Garcia Ruiz-Espiga, P.; Burguera, J.A.; Campos Arillo, V.; Castro, A.; Linazasoro, G.; Lopez Del Val, J.; Vela, L.; Martinez Castrillo, J.C. Dopaminergic agonists in Parkinson’s disease. Neurologia 2014, 29, 230–241. [Google Scholar] [CrossRef]
- Perez-Lloret, S.; Rascol, O. Dopamine receptor agonists for the treatment of early or advanced Parkinson’s disease. CNS Drugs 2010, 24, 941–968. [Google Scholar] [CrossRef]
- Stocchi, F.; Torti, M.; Fossati, C. Advances in dopamine receptor agonists for the treatment of Parkinson’s disease. Expert Opin. Pharm. 2016, 17, 1889–1902. [Google Scholar] [CrossRef]
- Braga, C.A.; Follmer, C.; Palhano, F.L.; Khattar, E.; Freitas, M.S.; Romao, L.; Di Giovanni, S.; Lashuel, H.A.; Silva, J.L.; Foguel, D. The anti-Parkinsonian drug selegiline delays the nucleation phase of alpha-synuclein aggregation leading to the formation of nontoxic species. J. Mol. Biol. 2011, 405, 254–273. [Google Scholar] [CrossRef] [Green Version]
- Tan, Y.Y.; Jenner, P.; Chen, S.D. Monoamine Oxidase-B Inhibitors for the Treatment of Parkinson’s Disease: Past, Present, and Future. J. Parkinsons Dis. 2022, 12, 477–493. [Google Scholar] [CrossRef]
- Kakish, J.; Tavassoly, O.; Lee, J.S. Rasagiline, a suicide inhibitor of monoamine oxidases, binds reversibly to alpha-synuclein. ACS Chem. Neurosci. 2015, 6, 347–355. [Google Scholar] [CrossRef]
- Brocchi, A.; Rebelos, E.; Dardano, A.; Mantuano, M.; Daniele, G. Effects of Intermittent Fasting on Brain Metabolism. Nutrients 2022, 14, 1275. [Google Scholar] [CrossRef] [PubMed]
- Fontana, L.; Ghezzi, L.; Cross, A.H.; Piccio, L. Effects of dietary restriction on neuroinflammation in neurodegenerative diseases. J. Exp. Med. 2021, 218, e20190086. [Google Scholar] [CrossRef] [PubMed]
- Mattson, M.P.; Allison, D.B.; Fontana, L.; Harvie, M.; Longo, V.D.; Malaisse, W.J.; Mosley, M.; Notterpek, L.; Ravussin, E.; Scheer, F.A.; et al. Meal frequency and timing in health and disease. Proc. Natl. Acad. Sci. USA 2014, 111, 16647–16653. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Patterson, R.E.; Laughlin, G.A.; LaCroix, A.Z.; Hartman, S.J.; Natarajan, L.; Senger, C.M.; Martinez, M.E.; Villasenor, A.; Sears, D.D.; Marinac, C.R.; et al. Intermittent Fasting and Human Metabolic Health. J. Acad. Nutr. Diet. 2015, 115, 1203–1212. [Google Scholar] [CrossRef] [Green Version]
- Barazzoni, R.; Gortan Cappellari, G.; Zanetti, M.; Klaus, K.A.; Semolic, A.; Johnson, M.L.; Nair, K.S. Higher unacylated ghrelin and insulin sensitivity following dietary restriction and weight loss in obese humans. Clin. Nutr. 2021, 40, 638–644. [Google Scholar] [CrossRef]
- Fontana, L.; Klein, S.; Holloszy, J.O. Effects of long-term calorie restriction and endurance exercise on glucose tolerance, insulin action, and adipokine production. Age 2010, 32, 97–108. [Google Scholar] [CrossRef] [Green Version]
- Spadaro, O.; Youm, Y.; Shchukina, I.; Ryu, S.; Sidorov, S.; Ravussin, A.; Nguyen, K.; Aladyeva, E.; Predeus, A.N.; Smith, S.R.; et al. Caloric restriction in humans reveals immunometabolic regulators of health span. Science 2022, 375, 671–677. [Google Scholar] [CrossRef]
- Shen, H.; Guan, Q.; Zhang, X.; Yuan, C.; Tan, Z.; Zhai, L.; Hao, Y.; Gu, Y.; Han, C. New mechanism of neuroinflammation in Alzheimer’s disease: The activation of NLRP3 inflammasome mediated by gut microbiota. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2020, 100, 109884. [Google Scholar] [CrossRef]
- Ghezzi, L.; Cantoni, C.; Pinget, G.V.; Zhou, Y.; Piccio, L. Targeting the gut to treat multiple sclerosis. J. Clin. Investig. 2021, 131, e143774. [Google Scholar] [CrossRef]
- Green, C.L.; Lamming, D.W.; Fontana, L. Molecular mechanisms of dietary restriction promoting health and longevity. Nat. Rev. Mol. Cell Biol. 2022, 23, 56–73. [Google Scholar] [CrossRef]
- Dorling, J.L.; Martin, C.K.; Redman, L.M. Calorie restriction for enhanced longevity: The role of novel dietary strategies in the present obesogenic environment. Ageing Res. Rev. 2020, 64, 101038. [Google Scholar] [CrossRef] [PubMed]
- Neth, B.J.; Bauer, B.A.; Benarroch, E.E.; Savica, R. The Role of Intermittent Fasting in Parkinson’s Disease. Front. Neurol. 2021, 12, 682184. [Google Scholar] [CrossRef] [PubMed]
- Most, J.; Tosti, V.; Redman, L.M.; Fontana, L. Calorie restriction in humans: An update. Ageing Res. Rev. 2017, 39, 36–45. [Google Scholar] [CrossRef] [PubMed]
- Hofer, S.J.; Carmona-Gutierrez, D.; Mueller, M.I.; Madeo, F. The ups and downs of caloric restriction and fasting: From molecular effects to clinical application. EMBO Mol. Med. 2022, 14, e14418. [Google Scholar] [CrossRef] [PubMed]
- Speakman, J.R.; Mitchell, S.E. Caloric restriction. Mol. Asp. Med. 2011, 32, 159–221. [Google Scholar] [CrossRef] [PubMed]
- Martin, C.K.; Bhapkar, M.; Pittas, A.G.; Pieper, C.F.; Das, S.K.; Williamson, D.A.; Scott, T.; Redman, L.M.; Stein, R.; Gilhooly, C.H.; et al. Effect of Calorie Restriction on Mood, Quality of Life, Sleep, and Sexual Function in Healthy Nonobese Adults: The CALERIE 2 Randomized Clinical Trial. JAMA Intern. Med. 2016, 176, 743–752. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hashimoto, T.; Horikawa, M.; Nomura, T.; Sakamoto, K. Nicotinamide adenine dinucleotide extends the lifespan of Caenorhabditis elegans mediated by sir-2.1 and daf-16. Biogerontology 2010, 11, 31–43. [Google Scholar] [CrossRef] [Green Version]
- Wang, X.; Wang, X.; Li, L.; Wang, D. Lifespan extension in Caenorhabditis elegans by DMSO is dependent on sir-2.1 and daf-16. Biochem. Biophys. Res. Commun. 2010, 400, 613–618. [Google Scholar] [CrossRef]
- Bamps, S.; Wirtz, J.; Savory, F.R.; Lake, D.; Hope, I.A. The Caenorhabditis elegans sirtuin gene, sir-2.1, is widely expressed and induced upon caloric restriction. Mech. Ageing Dev. 2009, 130, 762–770. [Google Scholar] [CrossRef]
- Mattson, M.P.; Longo, V.D.; Harvie, M. Impact of intermittent fasting on health and disease processes. Ageing Res. Rev. 2017, 39, 46–58. [Google Scholar] [CrossRef]
- Varady, K.A.; Hellerstein, M.K. Alternate-day fasting and chronic disease prevention: A review of human and animal trials. Am. J. Clin. Nutr. 2007, 86, 7–13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Stekovic, S.; Hofer, S.J.; Tripolt, N.; Aon, M.A.; Royer, P.; Pein, L.; Stadler, J.T.; Pendl, T.; Prietl, B.; Url, J.; et al. Alternate Day Fasting Improves Physiological and Molecular Markers of Aging in Healthy, Non-obese Humans. Cell Metab. 2019, 30, 462–476.e6. [Google Scholar] [CrossRef] [PubMed]
- Gill, S.; Le, H.D.; Melkani, G.C.; Panda, S. Time-restricted feeding attenuates age-related cardiac decline in Drosophila. Science 2015, 347, 1265–1269. [Google Scholar] [CrossRef] [Green Version]
- Long, H.; Panda, S. Time-restricted feeding and circadian autophagy for long life. Nat. Rev. Endocrinol. 2022, 18, 5–6. [Google Scholar] [CrossRef] [PubMed]
- Schubel, R.; Nattenmuller, J.; Sookthai, D.; Nonnenmacher, T.; Graf, M.E.; Riedl, L.; Schlett, C.L.; von Stackelberg, O.; Johnson, T.; Nabers, D.; et al. Effects of intermittent and continuous calorie restriction on body weight and metabolism over 50 wk: A randomized controlled trial. Am. J. Clin. Nutr. 2018, 108, 933–945. [Google Scholar] [CrossRef]
- Longo, V.D.; Mattson, M.P. Fasting: Molecular mechanisms and clinical applications. Cell Metab. 2014, 19, 181–192. [Google Scholar] [CrossRef] [Green Version]
- Longo, V.D.; Panda, S. Fasting, Circadian Rhythms, and Time-Restricted Feeding in Healthy Lifespan. Cell Metab. 2016, 23, 1048–1059. [Google Scholar] [CrossRef] [Green Version]
- Mishra, A.; Mirzaei, H.; Guidi, N.; Vinciguerra, M.; Mouton, A.; Linardic, M.; Rappa, F.; Barone, R.; Navarrete, G.; Wei, M.; et al. Fasting-mimicking diet prevents high-fat diet effect on cardiometabolic risk and lifespan. Nat. Metab. 2021, 3, 1342–1356. [Google Scholar] [CrossRef]
- Choi, I.Y.; Piccio, L.; Childress, P.; Bollman, B.; Ghosh, A.; Brandhorst, S.; Suarez, J.; Michalsen, A.; Cross, A.H.; Morgan, T.E.; et al. A Diet Mimicking Fasting Promotes Regeneration and Reduces Autoimmunity and Multiple Sclerosis Symptoms. Cell Rep. 2016, 15, 2136–2146. [Google Scholar] [CrossRef] [Green Version]
- Caffa, I.; Spagnolo, V.; Vernieri, C.; Valdemarin, F.; Becherini, P.; Wei, M.; Brandhorst, S.; Zucal, C.; Driehuis, E.; Ferrando, L.; et al. Fasting-mimicking diet and hormone therapy induce breast cancer regression. Nature 2020, 583, 620–624. [Google Scholar] [CrossRef]
- Fontana, L.; Partridge, L.; Longo, V.D. Extending healthy life span—From yeast to humans. Science 2010, 328, 321–326. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kwon, Y.Y.; Lee, S.K.; Lee, C.K. Caloric Restriction-Induced Extension of Chronological Lifespan Requires Intact Respiration in Budding Yeast. Mol. Cells 2017, 40, 307–313. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wierman, M.B.; Maqani, N.; Strickler, E.; Li, M.; Smith, J.S. Caloric Restriction Extends Yeast Chronological Life Span by Optimizing the Snf1 (AMPK) Signaling Pathway. Mol. Cell Biol. 2017, 37, e00562-16. [Google Scholar] [CrossRef] [Green Version]
- Leonov, A.; Feldman, R.; Piano, A.; Arlia-Ciommo, A.; Lutchman, V.; Ahmadi, M.; Elsaser, S.; Fakim, H.; Heshmati-Moghaddam, M.; Hussain, A.; et al. Caloric restriction extends yeast chronological lifespan via a mechanism linking cellular aging to cell cycle regulation, maintenance of a quiescent state, entry into a non-quiescent state and survival in the non-quiescent state. Oncotarget 2017, 8, 69328–69350. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Anson, R.M.; Guo, Z.; de Cabo, R.; Iyun, T.; Rios, M.; Hagepanos, A.; Ingram, D.K.; Lane, M.A.; Mattson, M.P. Intermittent fasting dissociates beneficial effects of dietary restriction on glucose metabolism and neuronal resistance to injury from calorie intake. Proc. Natl. Acad. Sci. USA 2003, 100, 6216–6220. [Google Scholar] [CrossRef] [Green Version]
- Gregosa, A.; Vinuesa, A.; Todero, M.F.; Pomilio, C.; Rossi, S.P.; Bentivegna, M.; Presa, J.; Wenker, S.; Saravia, F.; Beauquis, J. Periodic dietary restriction ameliorates amyloid pathology and cognitive impairment in PDAPP-J20 mice: Potential implication of glial autophagy. Neurobiol. Dis. 2019, 132, 104542. [Google Scholar] [CrossRef]
- Fann, D.Y.; Ng, G.Y.; Poh, L.; Arumugam, T.V. Positive effects of intermittent fasting in ischemic stroke. Exp. Gerontol. 2017, 89, 93–102. [Google Scholar] [CrossRef]
- Zouhal, H.; Bagheri, R.; Triki, R.; Saeidi, A.; Wong, A.; Hackney, A.C.; Laher, I.; Suzuki, K.; Ben Abderrahman, A. Effects of Ramadan Intermittent Fasting on Gut Hormones and Body Composition in Males with Obesity. Int. J. Environ. Res. Public Health 2020, 17, 5600. [Google Scholar] [CrossRef]
- Dorling, J.L.; van Vliet, S.; Huffman, K.M.; Kraus, W.E.; Bhapkar, M.; Pieper, C.F.; Stewart, T.; Das, S.K.; Racette, S.B.; Roberts, S.B.; et al. Effects of caloric restriction on human physiological, psychological, and behavioral outcomes: Highlights from CALERIE phase 2. Nutr. Rev. 2021, 79, 98–113. [Google Scholar] [CrossRef]
- Rochon, J.; Bales, C.W.; Ravussin, E.; Redman, L.M.; Holloszy, J.O.; Racette, S.B.; Roberts, S.B.; Das, S.K.; Romashkan, S.; Galan, K.M.; et al. Design and conduct of the CALERIE study: Comprehensive assessment of the long-term effects of reducing intake of energy. J. Gerontol. A Biol. Sci. Med. Sci. 2011, 66, 97–108. [Google Scholar] [CrossRef]
- Rhoads, T.W.; Anderson, R.M. Caloric restriction has a new player. Science 2022, 375, 620–621. [Google Scholar] [CrossRef] [PubMed]
- Bayliss, J.A.; Lemus, M.B.; Stark, R.; Santos, V.V.; Thompson, A.; Rees, D.J.; Galic, S.; Elsworth, J.D.; Kemp, B.E.; Davies, J.S.; et al. Ghrelin-AMPK Signaling Mediates the Neuroprotective Effects of Calorie Restriction in Parkinson’s Disease. J. Neurosci. 2016, 36, 3049–3063. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cantó, C.; Auwerx, J. Calorie restriction: Is AMPK a key sensor and effector? Physiology 2011, 26, 214–224. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bjedov, I.; Rallis, C. The Target of Rapamycin Signalling Pathway in Ageing and Lifespan Regulation. Genes 2020, 11, 1043. [Google Scholar] [CrossRef]
- Hu, Y.; Mai, W.; Chen, L.; Cao, K.; Zhang, B.; Zhang, Z.; Liu, Y.; Lou, H.; Duan, S.; Gao, Z. mTOR-mediated metabolic reprogramming shapes distinct microglia functions in response to lipopolysaccharide and ATP. Glia 2020, 68, 1031–1045. [Google Scholar] [CrossRef]
- de Cabo, R.; Mattson, M.P. Effects of Intermittent Fasting on Health, Aging, and Disease. N. Engl. J. Med. 2019, 381, 2541–2551. [Google Scholar] [CrossRef]
- Longo, V.D.; Cortellino, S. Fasting, dietary restriction, and immunosenescence. J. Allergy Clin. Immunol. 2020, 146, 1002–1004. [Google Scholar] [CrossRef]
- Goto, S. Health span extension by later-life caloric or dietary restriction: A view based on rodent studies. Biogerontology 2006, 7, 135–138. [Google Scholar] [CrossRef]
- Goodrick, C.L.; Ingram, D.K.; Reynolds, M.A.; Freeman, J.R.; Cider, N. Effects of intermittent feeding upon body weight and lifespan in inbred mice: Interaction of genotype and age. Mech. Ageing Dev. 1990, 55, 69–87. [Google Scholar] [CrossRef]
- Ishaq, A.; Dufour, D.; Cameron, K.; von Zglinicki, T.; Saretzki, G. Metabolic memory of dietary restriction ameliorates DNA damage and adipocyte size in mouse visceral adipose tissue. Exp. Gerontol. 2018, 113, 228–236. [Google Scholar] [CrossRef]
- Mitchell, S.J.; Madrigal-Matute, J.; Scheibye-Knudsen, M.; Fang, E.; Aon, M.; Gonzalez-Reyes, J.A.; Cortassa, S.; Kaushik, S.; Gonzalez-Freire, M.; Patel, B.; et al. Effects of Sex, Strain, and Energy Intake on Hallmarks of Aging in Mice. Cell Metab. 2016, 23, 1093–1112. [Google Scholar] [CrossRef] [Green Version]
- Kane, A.E.; Sinclair, D.A.; Mitchell, J.R.; Mitchell, S.J. Sex differences in the response to dietary restriction in rodents. Curr. Opin. Physiol. 2018, 6, 28–34. [Google Scholar] [CrossRef] [PubMed]
- Honjoh, S.; Ihara, A.; Kajiwara, Y.; Yamamoto, T.; Nishida, E. The Sexual Dimorphism of Dietary Restriction Responsiveness in Caenorhabditis elegans. Cell Rep. 2017, 21, 3646–3652. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Adler, M.I.; Cassidy, E.J.; Fricke, C.; Bonduriansky, R. The lifespan-reproduction trade-off under dietary restriction is sex-specific and context-dependent. Exp. Gerontol. 2013, 48, 539–548. [Google Scholar] [CrossRef]
- Cameron, K.M.; Miwa, S.; Walker, C.; von Zglinicki, T. Male mice retain a metabolic memory of improved glucose tolerance induced during adult onset, short-term dietary restriction. Longev. Healthspan 2012, 1, 3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Novelli, M.; De Tata, V.; Fierabracci, V.; Barbera, M.; Rossetti, R.; Masiello, P. Comparative study on the preventing effects of oral vanadyl sulfate and dietary restriction on the age-related glucose intolerance in rats. Aging Clin. Exp. Res. 2005, 17, 351–357. [Google Scholar] [CrossRef] [PubMed]
- Matyi, S.; Jackson, J.; Garrett, K.; Deepa, S.S.; Unnikrishnan, A. The effect of different levels of dietary restriction on glucose homeostasis and metabolic memory. Geroscience 2018, 40, 139–149. [Google Scholar] [CrossRef] [Green Version]
- Hahn, O.; Drews, L.F.; Nguyen, A.; Tatsuta, T.; Gkioni, L.; Hendrich, O.; Zhang, Q.; Langer, T.; Pletcher, S.; Wakelam, M.J.O.; et al. A nutritional memory effect counteracts benefits of dietary restriction in old mice. Nat. Metab. 2019, 1, 1059–1073. [Google Scholar] [CrossRef] [Green Version]
- Acosta-Rodríguez, V.; Rijo-Ferreira, F.; Izumo, M.; Xu, P.; Wight-Carter, M.; Green, C.B.; Takahashi, J.S. Circadian alignment of early onset caloric restriction promotes longevity in male C57BL/6J mice. Science 2022, 376, 1192–1202. [Google Scholar] [CrossRef]
- Anderson, R.M.; Shanmuganayagam, D.; Weindruch, R. Caloric restriction and aging: Studies in mice and monkeys. Toxicol. Pathol. 2009, 37, 47–51. [Google Scholar] [CrossRef]
- Zhao, Y.; Jia, M.; Chen, W.; Liu, Z. The neuroprotective effects of intermittent fasting on brain aging and neurodegenerative diseases via regulating mitochondrial function. Free Radic. Biol. Med. 2022, 182, 206–218. [Google Scholar] [CrossRef] [PubMed]
- Mattison, J.A.; Roth, G.S.; Lane, M.A.; Ingram, D.K. Dietary restriction in aging nonhuman primates. Interdiscip. Top. Gerontol. 2007, 35, 137–158. [Google Scholar] [CrossRef] [PubMed]
- Hou, Y.; Dan, X.; Babbar, M.; Wei, Y.; Hasselbalch, S.G.; Croteau, D.L.; Bohr, V.A. Ageing as a risk factor for neurodegenerative disease. Nat. Rev. Neurol. 2019, 15, 565–581. [Google Scholar] [CrossRef] [PubMed]
- Mattson, M.P.; Arumugam, T.V. Hallmarks of Brain Aging: Adaptive and Pathological Modification by Metabolic States. Cell Metab. 2018, 27, 1176–1199. [Google Scholar] [CrossRef] [Green Version]
- Pizza, V.; Agresta, A.; D’Acunto, C.W.; Festa, M.; Capasso, A. Neuroinflamm-aging and neurodegenerative diseases: An overview. CNS Neurol. Disord. Drug Targets 2011, 10, 621–634. [Google Scholar] [CrossRef]
- Martin, B.; Mattson, M.P.; Maudsley, S. Caloric restriction and intermittent fasting: Two potential diets for successful brain aging. Ageing Res. Rev. 2006, 5, 332–353. [Google Scholar] [CrossRef] [Green Version]
- Murphy, T.; Dias, G.P.; Thuret, S. Effects of diet on brain plasticity in animal and human studies: Mind the gap. Neural. Plast. 2014, 2014, 563160. [Google Scholar] [CrossRef] [Green Version]
- Singh, R.; Lakhanpal, D.; Kumar, S.; Sharma, S.; Kataria, H.; Kaur, M.; Kaur, G. Late-onset intermittent fasting dietary restriction as a potential intervention to retard age-associated brain function impairments in male rats. Age 2012, 34, 917–933. [Google Scholar] [CrossRef] [Green Version]
- Laudisio, A.; Antonelli Incalzi, R.; Gemma, A.; Giovannini, S.; Lo Monaco, M.R.; Vetrano, D.L.; Padua, L.; Bernabei, R.; Zuccala, G. Use of proton-pump inhibitors is associated with depression: A population-based study. Int. Psychogeriatr. 2018, 30, 153–159. [Google Scholar] [CrossRef]
- Alexopoulos, G.S. Mechanisms and treatment of late-life depression. Transl. Psychiatry 2019, 9, 188. [Google Scholar] [CrossRef]
- Morimoto, S.S.; Kanellopoulos, D.; Manning, K.J.; Alexopoulos, G.S. Diagnosis and treatment of depression and cognitive impairment in late life. Ann. N. Y. Acad. Sci. 2015, 1345, 36–46. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Alexopoulos, G.S. Depression in the elderly. Lancet 2005, 365, 1961–1970. [Google Scholar] [CrossRef]
- Giovannini, S.; Macchi, C.; Liperoti, R.; Laudisio, A.; Coraci, D.; Loreti, C.; Vannetti, F.; Onder, G.; Padua, L.; Mugello Study Working, G. Association of Body Fat with Health-Related Quality of Life and Depression in Nonagenarians: The Mugello Study. J. Am. Med. Dir. Assoc. 2019, 20, 564–568. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Y.; Liu, C.; Zhao, Y.; Zhang, X.; Li, B.; Cui, R. The Effects of Calorie Restriction in Depression and Potential Mechanisms. Curr. Neuropharmacol. 2015, 13, 536–542. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Thomson, R.L.; Buckley, J.D.; Lim, S.S.; Noakes, M.; Clifton, P.M.; Norman, R.J.; Brinkworth, G.D. Lifestyle management improves quality of life and depression in overweight and obese women with polycystic ovary syndrome. Fertil. Steril. 2010, 94, 1812–1816. [Google Scholar] [CrossRef]
- Lutter, M.; Krishnan, V.; Russo, S.J.; Jung, S.; McClung, C.A.; Nestler, E.J. Orexin signaling mediates the antidepressant-like effect of calorie restriction. J. Neurosci. 2008, 28, 3071–3075. [Google Scholar] [CrossRef] [Green Version]
- Giovannini, S.; Onder, G.; Leeuwenburgh, C.; Carter, C.; Marzetti, E.; Russo, A.; Capoluongo, E.; Pahor, M.; Bernabei, R.; Landi, F. Myeloperoxidase levels and mortality in frail community-living elderly individuals. J. Gerontol. A Biol. Sci. Med. Sci. 2010, 65, 369–376. [Google Scholar] [CrossRef] [Green Version]
- Son, T.G.; Zou, Y.; Yu, B.P.; Lee, J.; Chung, H.Y. Aging effect on myeloperoxidase in rat kidney and its modulation by calorie restriction. Free Radic. Res. 2005, 39, 283–289. [Google Scholar] [CrossRef]
- Arum, O.; Saleh, J.K.; Boparai, R.K.; Kopchick, J.J.; Khardori, R.K.; Bartke, A. Preservation of blood glucose homeostasis in slow-senescing somatotrophism-deficient mice subjected to intermittent fasting begun at middle or old age. Age 2014, 36, 9651. [Google Scholar] [CrossRef] [Green Version]
- Hadem, I.K.H.; Majaw, T.; Kharbuli, B.; Sharma, R. Beneficial effects of dietary restriction in aging brain. J. Chem. Neuroanat. 2019, 95, 123–133. [Google Scholar] [CrossRef]
- Sombric, C.J.; Torres-Oviedo, G. Cognitive and Motor Perseveration Are Associated in Older Adults. Front. Aging Neurosci. 2021, 13, 610359. [Google Scholar] [CrossRef] [PubMed]
- Roy, S.; Frndak, S.; Drake, A.S.; Irwin, L.; Zivadinov, R.; Weinstock-Guttman, B.; Benedict, R.H. Differential effects of aging on motor and cognitive functioning in multiple sclerosis. Mult. Scler. 2017, 23, 1385–1393. [Google Scholar] [CrossRef] [PubMed]
- Gudden, J.; Arias Vasquez, A.; Bloemendaal, M. The Effects of Intermittent Fasting on Brain and Cognitive Function. Nutrients 2021, 13, 3166. [Google Scholar] [CrossRef] [PubMed]
- Geng, Y.Q.; Guan, J.T.; Xu, M.Y.; Xu, X.H.; Fu, Y.C. Behavioral study of calorie-restricted rats from early old age. In Proceedings of the 29th Annual International Conference of the IEEE Engineering in Medicine and Biology Society, Lyon, France, 23–26 August 2007; pp. 2393–2395. [Google Scholar] [CrossRef]
- Maswood, N.; Young, J.; Tilmont, E.; Zhang, Z.; Gash, D.M.; Gerhardt, G.A.; Grondin, R.; Roth, G.S.; Mattison, J.; Lane, M.A.; et al. Caloric restriction increases neurotrophic factor levels and attenuates neurochemical and behavioral deficits in a primate model of Parkinson’s disease. Proc. Natl. Acad. Sci. USA 2004, 101, 18171–18176. [Google Scholar] [CrossRef] [Green Version]
- Kim, C.; Pinto, A.M.; Bordoli, C.; Buckner, L.P.; Kaplan, P.C.; Del Arenal, I.M.; Jeffcock, E.J.; Hall, W.L.; Thuret, S. Energy Restriction Enhances Adult Hippocampal Neurogenesis-Associated Memory after Four Weeks in an Adult Human Population with Central Obesity; a Randomized Controlled Trial. Nutrients 2020, 12, 638. [Google Scholar] [CrossRef] [Green Version]
- Fu, Y.; Chen, Y.; Li, L.; Wang, Y.; Kong, X.; Wang, J. Food restriction affects Y-maze spatial recognition memory in developing mice. Int. J. Dev. Neurosci. Off. J. Int. Soc. Dev. Neurosci. 2017, 60, 8–15. [Google Scholar] [CrossRef]
- Martin, C.K.; Anton, S.D.; Han, H.; York-Crowe, E.; Redman, L.M.; Ravussin, E.; Williamson, D.A. Examination of cognitive function during six months of calorie restriction: Results of a randomized controlled trial. Rejuvenation Res. 2007, 10, 179–190. [Google Scholar] [CrossRef] [Green Version]
- Xiao, N.; Le, Q.T. Neurotrophic Factors and Their Potential Applications in Tissue Regeneration. Arch. Immunol. Ther. Exp. 2016, 64, 89–99. [Google Scholar] [CrossRef]
- Gillespie, L.N. Regulation of axonal growth and guidance by the neurotrophin family of neurotrophic factors. Clin. Exp. Pharmacol. Physiol. 2003, 30, 724–733. [Google Scholar] [CrossRef]
- Di Benedetto, S.; Muller, L.; Wenger, E.; Duzel, S.; Pawelec, G. Contribution of neuroinflammation and immunity to brain aging and the mitigating effects of physical and cognitive interventions. Neurosci. Biobehav. Rev. 2017, 75, 114–128. [Google Scholar] [CrossRef] [Green Version]
- Duan, W.; Guo, Z.; Mattson, M.P. Brain-derived neurotrophic factor mediates an excitoprotective effect of dietary restriction in mice. J. Neurochem. 2001, 76, 619–626. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, J.; Duan, W.; Mattson, M.P. Evidence that brain-derived neurotrophic factor is required for basal neurogenesis and mediates, in part, the enhancement of neurogenesis by dietary restriction in the hippocampus of adult mice. J. Neurochem. 2002, 82, 1367–1375. [Google Scholar] [CrossRef] [PubMed]
- de Carvalho, T.S.; Sanchez-Mendoza, E.H.; Schultz Moreira, A.R.; Nascentes Melo, L.M.; Wang, C.; Sardari, M.; Hagemann, N.; Doeppner, T.R.; Kleinschnitz, C.; Hermann, D.M. Hypocaloric Diet Initiated Post-Ischemia Provides Long-Term Neuroprotection and Promotes Peri-Infarct Brain Remodeling by Regulating Metabolic and Survival-Promoting Proteins. Mol. Neurobiol. 2021, 58, 1491–1503. [Google Scholar] [CrossRef] [PubMed]
- Holtmaat, A.; Caroni, P. Functional and structural underpinnings of neuronal assembly formation in learning. Nat. Neurosci. 2016, 19, 1553–1562. [Google Scholar] [CrossRef]
- Katz, L.C.; Shatz, C.J. Synaptic activity and the construction of cortical circuits. Science 1996, 274, 1133–1138. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mattson, M.P.; Moehl, K.; Ghena, N.; Schmaedick, M.; Cheng, A. Intermittent metabolic switching, neuroplasticity and brain health. Nat. Rev. Neurosci. 2018, 19, 63–80. [Google Scholar] [CrossRef] [Green Version]
- Li, L.; Wang, Z.; Zuo, Z. Chronic intermittent fasting improves cognitive functions and brain structures in mice. PLoS ONE 2013, 8, e66069. [Google Scholar] [CrossRef] [Green Version]
- Serger, E.; Luengo-Gutierrez, L.; Chadwick, J.S.; Kong, G.; Zhou, L.; Crawford, G.; Danzi, M.C.; Myridakis, A.; Brandis, A.; Bello, A.T.; et al. The gut metabolite indole-3 propionate promotes nerve regeneration and repair. Nature 2022, 607, 585–592. [Google Scholar] [CrossRef]
- Castello, L.; Froio, T.; Maina, M.; Cavallini, G.; Biasi, F.; Leonarduzzi, G.; Donati, A.; Bergamini, E.; Poli, G.; Chiarpotto, E. Alternate-day fasting protects the rat heart against age-induced inflammation and fibrosis by inhibiting oxidative damage and NF-kB activation. Free Radic. Biol. Med. 2010, 48, 47–54. [Google Scholar] [CrossRef]
- Honjoh, S.; Yamamoto, T.; Uno, M.; Nishida, E. Signalling through RHEB-1 mediates intermittent fasting-induced longevity in C. elegans. Nature 2009, 457, 726–730. [Google Scholar] [CrossRef]
- Guo, Y.; Luo, S.; Ye, Y.; Yin, S.; Fan, J.; Xia, M. Intermittent Fasting Improves Cardiometabolic Risk Factors and Alters Gut Microbiota in Metabolic Syndrome Patients. J. Clin. Endocrinol. Metab. 2021, 106, 64–79. [Google Scholar] [CrossRef] [PubMed]
- Mindikoglu, A.L.; Abdulsada, M.M.; Jain, A.; Jalal, P.K.; Devaraj, S.; Wilhelm, Z.R.; Opekun, A.R.; Jung, S.Y. Intermittent fasting from dawn to sunset for four consecutive weeks induces anticancer serum proteome response and improves metabolic syndrome. Sci. Rep. 2020, 10, 18341. [Google Scholar] [CrossRef] [PubMed]
- Trepanowski, J.F.; Kroeger, C.M.; Barnosky, A.; Klempel, M.; Bhutani, S.; Hoddy, K.K.; Rood, J.; Ravussin, E.; Varady, K.A. Effects of alternate-day fasting or daily calorie restriction on body composition, fat distribution, and circulating adipokines: Secondary analysis of a randomized controlled trial. Clin. Nutr. 2018, 37, 1871–1878. [Google Scholar] [CrossRef] [PubMed]
- Sundfor, T.M.; Svendsen, M.; Tonstad, S. Effect of intermittent versus continuous energy restriction on weight loss, maintenance and cardiometabolic risk: A randomized 1-year trial. Nutr. Metab. Cardiovasc. Dis. 2018, 28, 698–706. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kim, K.H.; Kim, Y.H.; Son, J.E.; Lee, J.H.; Kim, S.; Choe, M.S.; Moon, J.H.; Zhong, J.; Fu, K.; Lenglin, F.; et al. Intermittent fasting promotes adipose thermogenesis and metabolic homeostasis via VEGF-mediated alternative activation of macrophage. Cell Res. 2017, 27, 1309–1326. [Google Scholar] [CrossRef]
- Bruss, M.D.; Khambatta, C.F.; Ruby, M.A.; Aggarwal, I.; Hellerstein, M.K. Calorie restriction increases fatty acid synthesis and whole body fat oxidation rates. Am. J. Physiol. Endocrinol. Metab. 2010, 298, E108–E116. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, G.; Xie, C.; Lu, S.; Nichols, R.G.; Tian, Y.; Li, L.; Patel, D.; Ma, Y.; Brocker, C.N.; Yan, T.; et al. Intermittent Fasting Promotes White Adipose Browning and Decreases Obesity by Shaping the Gut Microbiota. Cell Metab. 2017, 26, 672–685.e4. [Google Scholar] [CrossRef] [Green Version]
- Liu, H.; Javaheri, A.; Godar, R.J.; Murphy, J.; Ma, X.; Rohatgi, N.; Mahadevan, J.; Hyrc, K.; Saftig, P.; Marshall, C.; et al. Intermittent fasting preserves beta-cell mass in obesity-induced diabetes via the autophagy-lysosome pathway. Autophagy 2017, 13, 1952–1968. [Google Scholar] [CrossRef] [Green Version]
- Currenti, W.; Godos, J.; Castellano, S.; Caruso, G.; Ferri, R.; Caraci, F.; Grosso, G.; Galvano, F. Association between Time Restricted Feeding and Cognitive Status in Older Italian Adults. Nutrients 2021, 13, 191. [Google Scholar] [CrossRef]
- Baik, S.H.; Rajeev, V.; Fann, D.Y.; Jo, D.G.; Arumugam, T.V. Intermittent fasting increases adult hippocampal neurogenesis. Brain Behav. 2020, 10, e01444. [Google Scholar] [CrossRef] [Green Version]
- Zhou, Z.L.; Jia, X.B.; Sun, M.F.; Zhu, Y.L.; Qiao, C.M.; Zhang, B.P.; Zhao, L.P.; Yang, Q.; Cui, C.; Chen, X.; et al. Neuroprotection of Fasting Mimicking Diet on MPTP-Induced Parkinson’s Disease Mice via Gut Microbiota and Metabolites. Neurotherapeutics 2019, 16, 741–760. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cignarella, F.; Cantoni, C.; Ghezzi, L.; Salter, A.; Dorsett, Y.; Chen, L.; Phillips, D.; Weinstock, G.M.; Fontana, L.; Cross, A.H.; et al. Intermittent Fasting Confers Protection in CNS Autoimmunity by Altering the Gut Microbiota. Cell Metab. 2018, 27, 1222–1235.e6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Manzanero, S.; Erion, J.R.; Santro, T.; Steyn, F.J.; Chen, C.; Arumugam, T.V.; Stranahan, A.M. Intermittent fasting attenuates increases in neurogenesis after ischemia and reperfusion and improves recovery. J. Cereb. Blood Flow Metab. 2014, 34, 897–905. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Coupe, B.; Leloup, C.; Asiedu, K.; Maillard, J.; Penicaud, L.; Horvath, T.L.; Bouret, S.G. Defective autophagy in Sf1 neurons perturbs the metabolic response to fasting and causes mitochondrial dysfunction. Mol. Metab. 2021, 47, 101186. [Google Scholar] [CrossRef] [PubMed]
- Mladenovic, A.; Perovic, M.; Tanic, N.; Petanceska, S.; Ruzdijic, S.; Kanazir, S. Dietary restriction modulates alpha-synuclein expression in the aging rat cortex and hippocampus. Synapse 2007, 61, 790–794. [Google Scholar] [CrossRef] [PubMed]
- Lee, J.; Herman, J.P.; Mattson, M.P. Dietary restriction selectively decreases glucocorticoid receptor expression in the hippocampus and cerebral cortex of rats. Exp. Neurol. 2000, 166, 435–441. [Google Scholar] [CrossRef]
- Rajabi, A.; Parinejad, N.; Ahmadi, K.; Khorramizadeh, M.R.; Raza, M. Anti-inflammatory effects of serum isolated from animals on intermittent feeding in C6 glioma cell line. Neurosci. Lett. 2011, 487, 32–35. [Google Scholar] [CrossRef]
- Rubovitch, V.; Pharayra, A.; Har-Even, M.; Dvir, O.; Mattson, M.P.; Pick, C.G. Dietary Energy Restriction Ameliorates Cognitive Impairment in a Mouse Model of Traumatic Brain Injury. J. Mol. Neurosci. MN 2019, 67, 613–621. [Google Scholar] [CrossRef]
- Du, X.Y.; Xie, X.X.; Liu, R.T. The Role of alpha-Synuclein Oligomers in Parkinson’s Disease. Int. J. Mol. Sci. 2020, 21, 8645. [Google Scholar] [CrossRef]
- Jankovic, J.; Tan, E.K. Parkinson’s disease: Etiopathogenesis and treatment. J. Neurol. Neurosurg. Psychiatry 2020, 91, 795–808. [Google Scholar] [CrossRef]
- Glass, C.K.; Saijo, K.; Winner, B.; Marchetto, M.C.; Gage, F.H. Mechanisms underlying inflammation in neurodegeneration. Cell 2010, 140, 918–934. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Stephenson, J.; Nutma, E.; van der Valk, P.; Amor, S. Inflammation in CNS neurodegenerative diseases. Immunology 2018, 154, 204–219. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, Z.; Zhou, T.; Ziegler, A.C.; Dimitrion, P.; Zuo, L. Oxidative Stress in Neurodegenerative Diseases: From Molecular Mechanisms to Clinical Applications. Oxid. Med. Cell Longev. 2017, 2017, 2525967. [Google Scholar] [CrossRef] [PubMed]
- Dionisio, P.A.; Amaral, J.D.; Rodrigues, C.M.P. Oxidative stress and regulated cell death in Parkinson’s disease. Ageing Res. Rev. 2021, 67, 101263. [Google Scholar] [CrossRef]
- Qin, Y.; Qiu, J.; Wang, P.; Liu, J.; Zhao, Y.; Jiang, F.; Lou, H. Impaired autophagy in microglia aggravates dopaminergic neurodegeneration by regulating NLRP3 inflammasome activation in experimental models of Parkinson’s disease. Brain. Behav. Immun. 2021, 91, 324–338. [Google Scholar] [CrossRef]
- Tu, H.Y.; Yuan, B.S.; Hou, X.O.; Zhang, X.J.; Pei, C.S.; Ma, Y.T.; Yang, Y.P.; Fan, Y.; Qin, Z.H.; Liu, C.F.; et al. α-synuclein suppresses microglial autophagy and promotes neurodegeneration in a mouse model of Parkinson’s disease. Aging Cell 2021, 20, e13522. [Google Scholar] [CrossRef]
- Tang, M.; Liu, T.; Jiang, P.; Dang, R. The interaction between autophagy and neuroinflammation in major depressive disorder: From pathophysiology to therapeutic implications. Pharm. Res. 2021, 168, 105586. [Google Scholar] [CrossRef]
- Aho, V.T.E.; Houser, M.C.; Pereira, P.A.B.; Chang, J.; Rudi, K.; Paulin, L.; Hertzberg, V.; Auvinen, P.; Tansey, M.G.; Scheperjans, F. Relationships of gut microbiota, short-chain fatty acids, inflammation, and the gut barrier in Parkinson’s disease. Mol. Neurodegener. 2021, 16, 6. [Google Scholar] [CrossRef]
- Lubomski, M.; Tan, A.H.; Lim, S.Y.; Holmes, A.J.; Davis, R.L.; Sue, C.M. Parkinson’s disease and the gastrointestinal microbiome. J. Neurol. 2020, 267, 2507–2523. [Google Scholar] [CrossRef]
- Zhang, Q.S.; Heng, Y.; Mou, Z.; Huang, J.Y.; Yuan, Y.H.; Chen, N.H. Reassessment of subacute MPTP-treated mice as animal model of Parkinson’s disease. Acta Pharm. Sin 2017, 38, 1317–1328. [Google Scholar] [CrossRef] [Green Version]
- Lee, E.; Hwang, I.; Park, S.; Hong, S.; Hwang, B.; Cho, Y.; Son, J.; Yu, J.W. MPTP-driven NLRP3 inflammasome activation in microglia plays a central role in dopaminergic neurodegeneration. Cell Death Differ. 2019, 26, 213–228. [Google Scholar] [CrossRef] [PubMed]
- Blandini, F.; Armentero, M.T.; Martignoni, E. The 6-hydroxydopamine model: News from the past. Parkinsonism Relat. Disord. 2008, 14 (Suppl. 2), S124–S129. [Google Scholar] [CrossRef] [PubMed]
- Simola, N.; Morelli, M.; Carta, A.R. The 6-hydroxydopamine model of Parkinson’s disease. Neurotox. Res. 2007, 11, 151–167. [Google Scholar] [CrossRef] [PubMed]
- Braak, H.; de Vos, R.A.; Bohl, J.; Del Tredici, K. Gastric alpha-synuclein immunoreactive inclusions in Meissner’s and Auerbach’s plexuses in cases staged for Parkinson’s disease-related brain pathology. Neurosci. Lett. 2006, 396, 67–72. [Google Scholar] [CrossRef] [PubMed]
- Wood, H. New models show gut-brain transmission of Parkinson disease pathology. Nat. Rev. Neurol. 2019, 15, 491. [Google Scholar] [CrossRef] [PubMed]
- Uemura, N.; Yagi, H.; Uemura, M.T.; Hatanaka, Y.; Yamakado, H.; Takahashi, R. Inoculation of alpha-synuclein preformed fibrils into the mouse gastrointestinal tract induces Lewy body-like aggregates in the brainstem via the vagus nerve. Mol. Neurodegener. 2018, 13, 21. [Google Scholar] [CrossRef] [Green Version]
- Challis, C.; Hori, A.; Sampson, T.R.; Yoo, B.B.; Challis, R.C.; Hamilton, A.M.; Mazmanian, S.K.; Volpicelli-Daley, L.A.; Gradinaru, V. Gut-seeded alpha-synuclein fibrils promote gut dysfunction and brain pathology specifically in aged mice. Nat. Neurosci. 2020, 23, 327–336. [Google Scholar] [CrossRef]
- Volpicelli-Daley, L.A.; Luk, K.C.; Patel, T.P.; Tanik, S.A.; Riddle, D.M.; Stieber, A.; Meaney, D.F.; Trojanowski, J.Q.; Lee, V.M. Exogenous alpha-synuclein fibrils induce Lewy body pathology leading to synaptic dysfunction and neuron death. Neuron 2011, 72, 57–71. [Google Scholar] [CrossRef] [Green Version]
- Taguchi, T.; Ikuno, M.; Hondo, M.; Parajuli, L.K.; Taguchi, K.; Ueda, J.; Sawamura, M.; Okuda, S.; Nakanishi, E.; Hara, J.; et al. α-Synuclein BAC transgenic mice exhibit RBD-like behaviour and hyposmia: A prodromal Parkinson’s disease model. Brain 2020, 143, 249–265. [Google Scholar] [CrossRef] [Green Version]
- Duan, W.; Mattson, M.P. Dietary restriction and 2-deoxyglucose administration improve behavioral outcome and reduce degeneration of dopaminergic neurons in models of Parkinson’s disease. J. Neurosci. Res. 1999, 57, 195–206. [Google Scholar] [CrossRef]
- Guedes, A.; Ludovico, P.; Sampaio-Marques, B. Caloric restriction alleviates alpha-synuclein toxicity in aged yeast cells by controlling the opposite roles of Tor1 and Sir2 on autophagy. Mech. Ageing Dev. 2017, 161, 270–276. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Griffioen, K.J.; Rothman, S.M.; Ladenheim, B.; Wan, R.; Vranis, N.; Hutchison, E.; Okun, E.; Cadet, J.L.; Mattson, M.P. Dietary energy intake modifies brainstem autonomic dysfunction caused by mutant alpha-synuclein. Neurobiol. Aging 2013, 34, 928–935. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hallett, P.J.; McLean, J.R.; Kartunen, A.; Langston, J.W.; Isacson, O. alpha-Synuclein overexpressing transgenic mice show internal organ pathology and autonomic deficits. Neurobiol. Dis. 2012, 47, 258–267. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yang, Y.W.; Hsieh, T.F.; Li, C.I.; Liu, C.S.; Lin, W.Y.; Chiang, J.H.; Li, T.C.; Lin, C.C. Increased risk of Parkinson disease with diabetes mellitus in a population-based study. Medicine 2017, 96, e5921. [Google Scholar] [CrossRef] [PubMed]
- Jeong, S.M.; Han, K.; Kim, D.; Rhee, S.Y.; Jang, W.; Shin, D.W. Body mass index, diabetes, and the risk of Parkinson’s disease. Mov. Disord. 2020, 35, 236–244. [Google Scholar] [CrossRef]
- Cheong, J.L.Y.; de Pablo-Fernandez, E.; Foltynie, T.; Noyce, A.J. The Association Between Type 2 Diabetes Mellitus and Parkinson’s Disease. J. Parkinsons Dis. 2020, 10, 775–789. [Google Scholar] [CrossRef] [Green Version]
- Wu, H.; Xie, B.; Ke, M.; Deng, Y. High-fat diet causes increased endogenous neurotoxins and phenotype of Parkinson’s disease in mice. Acta Biochim. Biophys. Sin. 2019, 51, 969–971. [Google Scholar] [CrossRef]
- Lee, E.B.; Mattson, M.P. The neuropathology of obesity: Insights from human disease. Acta Neuropathol. 2014, 127, 3–28. [Google Scholar] [CrossRef]
- Wang, G.-J.; Volkow, N.D.; Logan, J.; Pappas, N.R.; Wong, C.T.; Zhu, W.; Netusll, N.; Fowler, J.S. Brain dopamine and obesity. Lancet 2001, 357, 354–357. [Google Scholar] [CrossRef]
- Borgundvaag, E.; Mak, J.; Kramer, C.K. Metabolic Impact of Intermittent Fasting in Patients with Type 2 Diabetes Mellitus: A Systematic Review and Meta-analysis of Interventional Studies. J. Clin. Endocrinol. Metab. 2021, 106, 902–911. [Google Scholar] [CrossRef]
- Stranahan, A.M.; Lee, K.; Martin, B.; Maudsley, S.; Golden, E.; Cutler, R.G.; Mattson, M.P. Voluntary exercise and caloric restriction enhance hippocampal dendritic spine density and BDNF levels in diabetic mice. Hippocampus 2009, 19, 951–961. [Google Scholar] [CrossRef] [PubMed]
- Grajower, M.M.; Horne, B.D. Clinical Management of Intermittent Fasting in Patients with Diabetes Mellitus. Nutrients 2019, 11, 873. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Brett, B.L.; Gardner, R.C.; Godbout, J.; Dams-O’Connor, K.; Keene, C.D. Traumatic Brain Injury and Risk of Neurodegenerative Disorder. Biol. Psychiatry 2022, 91, 498–507. [Google Scholar] [CrossRef] [PubMed]
- Liu, Y.; Wang, R.; Zhao, Z.; Dong, W.; Zhang, X.; Chen, X.; Ma, L. Short-term caloric restriction exerts neuroprotective effects following mild traumatic brain injury by promoting autophagy and inhibiting astrocyte activation. Behav. Brain Res. 2017, 331, 135–142. [Google Scholar] [CrossRef]
- Xu, Y.; Liu, Z.; Xu, S.; Li, C.; Li, M.; Cao, S.; Sun, Y.; Dai, H.; Guo, Y.; Chen, X.; et al. Scientific Evidences of Calorie Restriction and Intermittent Fasting for Neuroprotection in Traumatic Brain Injury Animal Models: A Review of the Literature. Nutrients 2022, 14, 1431. [Google Scholar] [CrossRef]
- Medzhitov, R. Origin and physiological roles of inflammation. Nature 2008, 454, 428–435. [Google Scholar] [CrossRef]
- Hickman, S.; Izzy, S.; Sen, P.; Morsett, L.; El Khoury, J. Microglia in neurodegeneration. Nat. Neurosci. 2018, 21, 1359–1369. [Google Scholar] [CrossRef]
- Nuber, S.; Selkoe, D.J. Caspase-1 clipping causes complications for alpha-synuclein. Proc. Natl. Acad. Sci. USA 2016, 113, 9958–9960. [Google Scholar] [CrossRef] [Green Version]
- Pott Godoy, M.C.; Tarelli, R.; Ferrari, C.C.; Sarchi, M.I.; Pitossi, F.J. Central and systemic IL-1 exacerbates neurodegeneration and motor symptoms in a model of Parkinson’s disease. Brain 2008, 131, 1880–1894. [Google Scholar] [CrossRef]
- Traba, J.; Geiger, S.S.; Kwarteng-Siaw, M.; Han, K.; Ra, O.H.; Siegel, R.M.; Gius, D.; Sack, M.N. Prolonged fasting suppresses mitochondrial NLRP3 inflammasome assembly and activation via SIRT3-mediated activation of superoxide dismutase 2. J. Biol. Chem. 2017, 292, 12153–12164. [Google Scholar] [CrossRef] [Green Version]
- Traba, J.; Kwarteng-Siaw, M.; Okoli, T.C.; Li, J.; Huffstutler, R.D.; Bray, A.; Waclawiw, M.A.; Han, K.; Pelletier, M.; Sauve, A.A.; et al. Fasting and refeeding differentially regulate NLRP3 inflammasome activation in human subjects. J. Clin. Investig. 2015, 125, 4592–4600. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Williams, G.P.; Schonhoff, A.M.; Jurkuvenaite, A.; Gallups, N.J.; Standaert, D.G.; Harms, A.S. CD4 T cells mediate brain inflammation and neurodegeneration in a mouse model of Parkinson’s disease. Brain 2021, 144, 2047–2059. [Google Scholar] [CrossRef] [PubMed]
- Valko, M.; Leibfritz, D.; Moncol, J.; Cronin, M.T.; Mazur, M.; Telser, J. Free radicals and antioxidants in normal physiological functions and human disease. Int. J. Biochem. Cell Biol. 2007, 39, 44–84. [Google Scholar] [CrossRef] [PubMed]
- Stadtman, E.R. Role of oxidant species in aging. Curr. Med. Chem. 2004, 11, 1105–1112. [Google Scholar] [CrossRef] [Green Version]
- Salim, S. Oxidative Stress and the Central Nervous System. J. Pharmacol. Exp. Ther. 2017, 360, 201–205. [Google Scholar] [CrossRef] [Green Version]
- Mladenovic Djordjevic, A.; Loncarevic-Vasiljkovic, N.; Gonos, E.S. Dietary Restriction and Oxidative Stress: Friends or Enemies? Antioxid. Redox Signal. 2021, 34, 421–438. [Google Scholar] [CrossRef]
- Chang, K.H.; Chen, C.M. The Role of Oxidative Stress in Parkinson’s Disease. Antioxidants 2020, 9, 597. [Google Scholar] [CrossRef]
- Trist, B.G.; Hare, D.J.; Double, K.L. Oxidative stress in the aging substantia nigra and the etiology of Parkinson’s disease. Aging Cell 2019, 18, e13031. [Google Scholar] [CrossRef] [Green Version]
- Duarte-Jurado, A.P.; Gopar-Cuevas, Y.; Saucedo-Cardenas, O.; Loera-Arias, M.J.; Montes-de-Oca-Luna, R.; Garcia-Garcia, A.; Rodriguez-Rocha, H. Antioxidant Therapeutics in Parkinson’s Disease: Current Challenges and Opportunities. Antioxidants 2021, 10, 453. [Google Scholar] [CrossRef]
- Singh, C.K.; Chhabra, G.; Ndiaye, M.A.; Garcia-Peterson, L.M.; Mack, N.J.; Ahmad, N. The Role of Sirtuins in Antioxidant and Redox Signaling. Antioxid. Redox Signal. 2018, 28, 643–661. [Google Scholar] [CrossRef]
- Srivastava, S.; Haigis, M.C. Role of sirtuins and calorie restriction in neuroprotection: Implications in Alzheimer’s and Parkinson’s diseases. Curr. Pharm. Des. 2011, 17, 3418–3433. [Google Scholar] [CrossRef] [PubMed]
- Han, S.H. Potential role of sirtuin as a therapeutic target for neurodegenerative diseases. J. Clin. Neurol. 2009, 5, 120–125. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lan, A.P.; Chen, J.; Zhao, Y.; Chai, Z.; Hu, Y. mTOR Signaling in Parkinson’s Disease. Neuromolecular. Med. 2017, 19, 1–10. [Google Scholar] [CrossRef]
- Celebi-Birand, D.; Ardic, N.I.; Karoglu-Eravsar, E.T.; Sengul, G.F.; Kafaligonul, H.; Adams, M.M. Dietary and Pharmacological Interventions That Inhibit Mammalian Target of Rapamycin Activity Alter the Brain Expression Levels of Neurogenic and Glial Markers in an Age-and Treatment-Dependent Manner. Rejuvenation Res. 2020, 23, 485–497. [Google Scholar] [CrossRef] [PubMed]
- Cheng, C.W.; Villani, V.; Buono, R.; Wei, M.; Kumar, S.; Yilmaz, O.H.; Cohen, P.; Sneddon, J.B.; Perin, L.; Longo, V.D. Fasting-Mimicking Diet Promotes Ngn3-Driven β-Cell Regeneration to Reverse Diabetes. Cell 2017, 168, 775–788.e12. [Google Scholar] [CrossRef] [Green Version]
- Li, N.; Guenancia, C.; Rigal, E.; Hachet, O.; Chollet, P.; Desmoulins, L.; Leloup, C.; Rochette, L.; Vergely, C. Short-term moderate diet restriction in adulthood can reverse oxidative, cardiovascular and metabolic alterations induced by postnatal overfeeding in mice. Sci. Rep. 2016, 6, 30817. [Google Scholar] [CrossRef]
- Daneshrad, Z.; Novel-Chaté, V.; Birot, O.; Serrurier, B.; Sanchez, H.; Bigard, A.X.; Rossi, A. Diet restriction plays an important role in the alterations of heart mitochondrial function following exposure of young rats to chronic hypoxia. Pflügers Arch. 2001, 442, 12–18. [Google Scholar] [CrossRef]
- Djajadikerta, A.; Keshri, S.; Pavel, M.; Prestil, R.; Ryan, L.; Rubinsztein, D.C. Autophagy Induction as a Therapeutic Strategy for Neurodegenerative Diseases. J. Mol. Biol. 2020, 432, 2799–2821. [Google Scholar] [CrossRef]
- Bagherniya, M.; Butler, A.E.; Barreto, G.E.; Sahebkar, A. The effect of fasting or calorie restriction on autophagy induction: A review of the literature. Ageing Res. Rev. 2018, 47, 183–197. [Google Scholar] [CrossRef]
- Tenreiro, S.; Reimão-Pinto, M.M.; Antas, P.; Rino, J.; Wawrzycka, D.; Macedo, D.; Rosado-Ramos, R.; Amen, T.; Waiss, M.; Magalhães, F.; et al. Phosphorylation modulates clearance of alpha-synuclein inclusions in a yeast model of Parkinson’s disease. PLoS Genet. 2014, 10, e1004302. [Google Scholar] [CrossRef] [Green Version]
- Choi, I.; Zhang, Y.; Seegobin, S.P.; Pruvost, M.; Wang, Q.; Purtell, K.; Zhang, B.; Yue, Z. Microglia clear neuron-released alpha-synuclein via selective autophagy and prevent neurodegeneration. Nat. Commun. 2020, 11, 1386. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sampaio-Marques, B.; Pereira, H.; Santos, A.R.; Teixeira, A.; Ludovico, P. Caloric restriction rescues yeast cells from alpha-synuclein toxicity through autophagic control of proteostasis. Aging 2018, 10, 3821–3833. [Google Scholar] [CrossRef] [PubMed]
- Chen, X.; Kondo, K.; Motoki, K.; Homma, H.; Okazawa, H. Fasting activates macroautophagy in neurons of Alzheimer’s disease mouse model but is insufficient to degrade amyloid-beta. Sci. Rep. 2015, 5, 12115. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ulgherait, M.; Midoun, A.M.; Park, S.J.; Gatto, J.A.; Tener, S.J.; Siewert, J.; Klickstein, N.; Canman, J.C.; Ja, W.W.; Shirasu-Hiza, M. Circadian autophagy drives iTRF-mediated longevity. Nature 2021, 598, 353–358. [Google Scholar] [CrossRef]
- Hsiao, E.Y.; McBride, S.W.; Hsien, S.; Sharon, G.; Hyde, E.R.; McCue, T.; Codelli, J.A.; Chow, J.; Reisman, S.E.; Petrosino, J.F.; et al. Microbiota modulate behavioral and physiological abnormalities associated with neurodevelopmental disorders. Cell 2013, 155, 1451–1463. [Google Scholar] [CrossRef] [Green Version]
- Mangiola, F.; Ianiro, G.; Franceschi, F.; Fagiuoli, S.; Gasbarrini, G.; Gasbarrini, A. Gut microbiota in autism and mood disorders. World J. Gastroenterol. 2016, 22, 361–368. [Google Scholar] [CrossRef]
- Ochoa-Repáraz, J.; Kirby, T.O.; Kasper, L.H. The Gut Microbiome and Multiple Sclerosis. Cold Spring Harb. Perspect. Med. 2018, 8, a029017. [Google Scholar] [CrossRef]
- Unger, M.M.; Spiegel, J.; Dillmann, K.U.; Grundmann, D.; Philippeit, H.; Burmann, J.; Fassbender, K.; Schwiertz, A.; Schafer, K.H. Short chain fatty acids and gut microbiota differ between patients with Parkinson’s disease and age-matched controls. Parkinsonism Relat. Disord. 2016, 32, 66–72. [Google Scholar] [CrossRef]
- Lai, F.; Jiang, R.; Xie, W.; Liu, X.; Tang, Y.; Xiao, H.; Gao, J.; Jia, Y.; Bai, Q. Intestinal Pathology and Gut Microbiota Alterations in a Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) Mouse Model of Parkinson’s Disease. Neurochem. Res. 2018, 43, 1986–1999. [Google Scholar] [CrossRef]
- Cryan, J.F.; O’Riordan, K.J.; Sandhu, K.; Peterson, V.; Dinan, T.G. The gut microbiome in neurological disorders. Lancet Neurol. 2020, 19, 179–194. [Google Scholar] [CrossRef]
- Nam, H.S. Gut Microbiota and Ischemic Stroke: The Role of Trimethylamine N-Oxide. J. Stroke 2019, 21, 151–159. [Google Scholar] [CrossRef] [PubMed]
- Needham, B.D.; Funabashi, M.; Adame, M.D.; Wang, Z.; Boktor, J.C.; Haney, J.; Wu, W.L.; Rabut, C.; Ladinsky, M.S.; Hwang, S.J.; et al. A gut-derived metabolite alters brain activity and anxiety behaviour in mice. Nature 2022, 602, 647–653. [Google Scholar] [CrossRef] [PubMed]
- Hawkes, C.H.; Del Tredici, K.; Braak, H. Parkinson’s disease: A dual-hit hypothesis. Neuropathol. Appl. Neurobiol. 2007, 33, 599–614. [Google Scholar] [CrossRef] [PubMed]
- Huang, H.K.; Wang, J.H.; Lei, W.Y.; Chen, C.L.; Chang, C.Y.; Liou, L.S. Helicobacter pylori infection is associated with an increased risk of Parkinson’s disease: A population-based retrospective cohort study. Parkinsonism Relat. Disord. 2018, 47, 26–31. [Google Scholar] [CrossRef]
- Lee, H.S.; Lobbestael, E.; Vermeire, S.; Sabino, J.; Cleynen, I. Inflammatory bowel disease and Parkinson’s disease: Common pathophysiological links. Gut 2021, 70, 408–417. [Google Scholar] [CrossRef] [PubMed]
- Dodiya, H.B.; Forsyth, C.B.; Voigt, R.M.; Engen, P.A.; Patel, J.; Shaikh, M.; Green, S.J.; Naqib, A.; Roy, A.; Kordower, J.H.; et al. Chronic stress-induced gut dysfunction exacerbates Parkinson’s disease phenotype and pathology in a rotenone-induced mouse model of Parkinson’s disease. Neurobiol. Dis. 2020, 135, 104352. [Google Scholar] [CrossRef]
- Tran, S.M.; Mohajeri, M.H. The Role of Gut Bacterial Metabolites in Brain Development, Aging and Disease. Nutrients 2021, 13, 732. [Google Scholar] [CrossRef]
- Chen, S.J.; Chen, C.C.; Liao, H.Y.; Lin, Y.T.; Wu, Y.W.; Liou, J.M.; Wu, M.S.; Kuo, C.H.; Lin, C.H. Association of Fecal and Plasma Levels of Short-Chain Fatty Acids with Gut Microbiota and Clinical Severity in Patients with Parkinson Disease. Neurology 2022, 98, e848–e858. [Google Scholar] [CrossRef]
- Hegelmaier, T.; Lebbing, M.; Duscha, A.; Tomaske, L.; Tönges, L.; Holm, J.B.; Bjørn Nielsen, H.; Gatermann, S.G.; Przuntek, H.; Haghikia, A. Interventional Influence of the Intestinal Microbiome Through Dietary Intervention and Bowel Cleansing Might Improve Motor Symptoms in Parkinson’s Disease. Cells 2020, 9, 376. [Google Scholar] [CrossRef]
- Morris, J.K.; Bomhoff, G.L.; Stanford, J.A.; Geiger, P.C. Neurodegeneration in an animal model of Parkinson’s disease is exacerbated by a high-fat diet. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2010, 299, R1082–R1090. [Google Scholar] [CrossRef] [Green Version]
- Tan, B.L.; Norhaizan, M.E. Effect of High-Fat Diets on Oxidative Stress, Cellular Inflammatory Response and Cognitive Function. Nutrients 2019, 11, 2579. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, X.L.; Li, L. Microglia Regulate Neuronal Circuits in Homeostatic and High-Fat Diet-Induced Inflammatory Conditions. Front. Cell Neurosci. 2021, 15, 722028. [Google Scholar] [CrossRef] [PubMed]
- Mendes, N.F.; Jara, C.P.; Zanesco, A.M.; de Araújo, E.P. Hypothalamic Microglial Heterogeneity and Signature under High Fat Diet-Induced Inflammation. Int. J. Mol. Sci. 2021, 22, 2256. [Google Scholar] [CrossRef] [PubMed]
- Baufeld, C.; Osterloh, A.; Prokop, S.; Miller, K.R.; Heppner, F.L. High-fat diet-induced brain region-specific phenotypic spectrum of CNS resident microglia. Acta Neuropathol. 2016, 132, 361–375. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Phillips, M.C.L.; Murtagh, D.K.J.; Gilbertson, L.J.; Asztely, F.J.S.; Lynch, C.D.P. Low-fat versus ketogenic diet in Parkinson’s disease: A pilot randomized controlled trial. Mov. Disord. 2018, 33, 1306–1314. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yin, Z.; Raj, D.D.; Schaafsma, W.; van der Heijden, R.A.; Kooistra, S.M.; Reijne, A.C.; Zhang, X.; Moser, J.; Brouwer, N.; Heeringa, P.; et al. Low-Fat Diet with Caloric Restriction Reduces White Matter Microglia Activation During Aging. Front. Mol. Neurosci. 2018, 11, 65. [Google Scholar] [CrossRef]
- Ma, D.; Shuler, J.M.; Raider, K.D.; Rogers, R.S.; Wheatley, J.L.; Geiger, P.C.; Stanford, J.A. Effects of discontinuing a high-fat diet on mitochondrial proteins and 6-hydroxydopamine-induced dopamine depletion in rats. Brain Res. 2015, 1613, 49–58. [Google Scholar] [CrossRef] [Green Version]
- Coughlin, S.S.; Pincus, J.H.; Karstaedt, P. An international comparison of dietary protein consumption and mortality from Parkinson’s disease. J. Neurol. 1992, 239, 236–237. [Google Scholar] [CrossRef]
- Pincus, J.H.; Barry, K.M. Dietary method for reducing fluctuations in Parkinson’s disease. Yale J. Biol. Med. 1987, 60, 133–137. [Google Scholar]
- Cereda, E.; Barichella, M.; Pedrolli, C.; Pezzoli, G. Low-protein and protein-redistribution diets for Parkinson’s disease patients with motor fluctuations: A systematic review. Mov. Disord. 2010, 25, 2021–2034. [Google Scholar] [CrossRef]
- Hirata, H.; Asanuma, M.; Kondo, Y.; Ogawa, N. Influence of protein-restricted diet on motor response fluctuations in Parkinson’s disease. Rinsho Shinkeigaku = Clin. Neurol. 1992, 32, 973–978. [Google Scholar]
- Pincus, J.H.; Barry, K.M. Plasma levels of amino acids correlate with motor fluctuations in parkinsonism. Arch. Neurol. 1987, 44, 1006–1009. [Google Scholar] [CrossRef] [PubMed]
- Contin, M.; Martinelli, P. Pharmacokinetics of levodopa. J. Neurol. 2010, 257, S253–S261. [Google Scholar] [CrossRef] [PubMed]
- Wade, L.A.; Katzman, R. Synthetic amino acids and the nature of L-DOPA transport at the blood-brain barrier. J. Neurochem. 1975, 25, 837–842. [Google Scholar] [CrossRef] [PubMed]
- Pincus, J.H.; Barry, K. Influence of dietary protein on motor fluctuations in Parkinson’s disease. Arch. Neurol. 1987, 44, 270–272. [Google Scholar] [CrossRef]
- Virmani, T.; Tazan, S.; Mazzoni, P.; Ford, B.; Greene, P.E. Motor fluctuations due to interaction between dietary protein and levodopa in Parkinson’s disease. J. Clin. Mov. Disord. 2016, 3, 8. [Google Scholar] [CrossRef] [Green Version]
- Anderson, J.G.; Hintze, K.; Marchant, E.D. Restricting branched-chain amino acids: An approach to improve metabolic health. J. Physiol. 2018, 596, 2469–2470. [Google Scholar] [CrossRef] [Green Version]
- Cummings, N.E.; Williams, E.M.; Kasza, I.; Konon, E.N.; Schaid, M.D.; Schmidt, B.A.; Poudel, C.; Sherman, D.S.; Yu, D.; Arriola Apelo, S.I.; et al. Restoration of metabolic health by decreased consumption of branched-chain amino acids. J. Physiol. 2018, 596, 623–645. [Google Scholar] [CrossRef] [Green Version]
- Hill, C.M.; Morrison, C.D. Dietary branched chain amino acids and metabolic health: When less is more. J. Physiol. 2018, 596, 555–556. [Google Scholar] [CrossRef] [Green Version]
- Asoudeh, F.; Salari-Moghaddam, A.; Keshteli, A.H.; Esmaillzadeh, A.; Adibi, P. Dietary intake of branched-chain amino acids in relation to general and abdominal obesity. Eat Weight Disord. 2022, 20, 1303–1311. [Google Scholar] [CrossRef]
Regimen | Start | Duration | Physiological Benefits | Reference | |
---|---|---|---|---|---|
Aging and Longevity | |||||
Humans | |||||
healthy humans | 14% CR | 2 years | ↑transcriptional reprogramming in adipose tissue-regulating mitochondrial bioenergetics, anti-inflammatory responses, longevity ↓age-related inflammation | [27] | |
healthy, non-obese adults | ADF | 35 to 65 years | 4 weeks | ↑cardiac health ↓body weight, fat-to-lean ratio, sICAM-1 (an age-associated inflammatory marker), low-density lipoprotein, metabolic regulator triiodothyronine | [42] |
Animals | |||||
Wistar strain male albino rats | ADF | 21 months | 3 months | ↓age-associated impairment in motor coordination and learning and memory function, age-related increase in protein carbonylation, age-related impairment of synaptic proteins | [88] |
rats | ADF | 2 months | 4 months/10 months/22 months | ↓age-related oxidative damage, age-related increase in lipid peroxidation markers, age-related increase in TGF-β1 and collagen | [120] |
C. elegans | ADF | ↑lifespan | [121] | ||
Drosophila melanogaster | TRF | 2 weeks | 5 weeks | ↑sleep, age-induced decline in cardiac function | [43] |
A/J mice | ADF | 1.5 months | ↑mean and maximum life span | [69] | |
male SD rats | 60% CR | 18 months | 6 months | ↑survival rate, spontaneous locomotor activity, spatial learning and reference memory, spatial cognition | [104] |
Metabolism | |||||
Humans | |||||
humans with metabolic syndrome | 69% CR 2 days a week | 30–50 years | 8 weeks | ↑vasodilatory parameters, production of short-chain fatty acids (SCFAs) ↓oxidative stress, inflammatory cytokines, circulating levels of lipopolysaccharides (LPS) | [122] |
humans with metabolic syndrome | TRF | 18 years old or older | 4 weeks | ↑levels of tumor suppressor and DNA repair gene protein products (GP)s (CALU, INTS6, KIT, CROCC, PIGR), key regulatory proteins of insulin signaling (VPS8, POLRMT, IGFBP-5) ↓levels of tumor promoter GPs (POLK, CD109, CAMP, NIFK, SRGN), body mass index, waist circumference, blood pressure | [123] |
males with obesity | TRF | 20–30 years | 30 days | ↑leptin ↓body mass, BMI, body fat percentage (BFP), fat-free mass (FFM) and waist-to-hip ratio (WHR), glucagon-like peptide-1 (GLP-1), peptide YY (PYY), cholecystokinin (CCK) | [58] |
humans | 40% CR | mean age 53.0 ± 11 years | average of 6.9 ± 5.5 years | ↑insulin sensitivity, serum adiponectin concentration, fructosamine, sRAGE, fasting serum free fatty acids ↓body fat and trunk fat, serum resistin concentration, serum IL-6, soluble TNF R-I and TNF R-II, plasma 2-h insulin and C-peptide concentrations | [26] |
overweight and obese humans | ADF/75% CR | 18 to 65 years | 24 weeks | ↑fat-free mass (FFM): total mass ratio ↓circulating leptin | [124] |
humans with BMI 30–45 | Intermittent/continuous CR | 21–70 years | 1 year | ↑weight loss ↓cardiovascular risk factors, waist circumference | [125] |
Animals | |||||
mice | 2:1 IF regimen (1 day of fasting followed by 2 days of feeding) | 8 weeks | 16 weeks | ↑adipose thermogenesis contributing to IF-mediated metabolic benefits, adipose vascular endothelial growth factor expression ↓diet-induced metabolic abnormalities | [126] |
mice | 30% CR | 10 weeks | ≥5 weeks | ↑endogenous fatty acid (FA) synthesis, FA oxidation, expression of FA synthase and acetyl-CoA carboxylase mRNA | [127] |
C57BL/6N mice | ADF | 7 to 8 weeks | 15 weeks | ↑energy expenditure, beiging of inguinal white adipose tissue ↓Ucp1 and Pgc1a mRNA | [128] |
mice with obesity-induced diabetes | ADF | 20 weeks | 6 weeks | ↑glucose-stimulated insulin secretion, beta cell survival, nuclear expression of NEUROG3 (a marker of pancreatic regeneration), autophagic flux in islets, glucose tolerance | [129] |
C57BL/6 mice | 40% DR | 3 months | 3/9/12 months | ↓DNA damage, adipocyte size (area and perimeter) in visceral adipocytes | [70] |
Nervous system | |||||
Humans | |||||
men and women | TRF | 50 years or older | 6 months | ↓cognitive impairment | [130] |
humans with central obesity | IER/CER | 35–75 years | 4 weeks | ↑cognitive function, pattern separation | [106] |
Animals | |||||
rhesus monkeys with PD | 30% CR | 9–17 years | 6 months | ↑locomotor activity, dopamine (DA) and DA metabolites in the striatal region, glial-cell-line-derived neurotrophic factor | [105] |
C57BL/6N mice | TRF/ADF | 3 months | 3 months | ↑activation of the Notch signaling pathway (Notch 1, NICD1, and HES5), BDNF, cAMP response element-binding protein (p-CREB), expression of postsynaptic marker, PSD95, neuronal stem cell marker, Nestin, in the hippocampus | [131] |
MPTP-induced PD mice | FMD | 7 weeks | 3 weeks | ↑motor function, levels of BDNF ↓loss of dopaminergic neurons in the SN, the number of glial cells, the release of TNF-α and IL-1β | [132] |
mice | ADF | 2 months | 3 months | ↑BDNF in the hippocampus, striatum, and cerebral cortex | [112] |
mice | ADF | 7 weeks | 11 months | ↑drebrin and expression of synaptophysin in the cerebral cortex and hippocampus ↓blood cholesterol, triglycerides, high-density lipoproteins (HDL) and low-density lipoproteins (LDL) in the blood, glutathione disulfide (GSSG), 4-hydroxy-2-nonenal (HNE) and nitrotyrosine-containing proteins in the cerebral cortex | [118] |
mice | ADF | 2 months | 3 months | ↑neurogenesis, BDNF protein levels | [113] |
mice | ADF | 8 weeks | 4 weeks | ↑diversity of the gut microbiome, adiponectin levels, corticosterone levels, β- hydroxybutyrate ↓EAE clinical course and pathology, production of pro-inflammatory T cell cytokines, serum leptin | [133] |
mice | TRF | 10 weeks | 3 months | ↑cell proliferation in the intact subventricular zone (SVZ) ↓serum levels of leptin, sensorimotor impairment and infarct size after ischemia and reperfusion, stroke-induced cell proliferation in the hippocampus | [134] |
mice | TRF | until 2 to 3 months | ↑autophagy in the ventromedial nucleus of the hypothalamus | [135] | |
rat | 50% DR every other day, and fed with vegetables on days in between | 6 months | 6/12/18 months | ↓age-related a-synuclein expression | [136] |
rat | ADF | 3 months | 3 months | ↓levels of glucocorticoid receptor mRNA and protein in the hippocampus and cerebral cortex | [137] |
PDAPP-J20 transgenic mice (AD model) | 60%CR | 6.5 months old | 6 weeks | ↑astroglial positive signal for LC3 ↓cognitive deficits, amyloid pathology and microglial reactivity | [56] |
rat MS model | ADF | 180–200 g | 8 weeks | ↑ TGF-β1 ↓IL-6, MMP-2 activity | [138] |
mouse with traumatic brain injury | ADF | 6–7 weeks | 30 days | ↑SIRT1 levels in the cortex and hippocampus ↓impaired hippocampus-dependent learning and memory | [139] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Wang, Z.; Cui, Y.; Wen, L.; Yu, H.; Feng, J.; Yuan, W.; He, X. Dietary Restriction against Parkinson’s Disease: What We Know So Far. Nutrients 2022, 14, 4108. https://doi.org/10.3390/nu14194108
Wang Z, Cui Y, Wen L, Yu H, Feng J, Yuan W, He X. Dietary Restriction against Parkinson’s Disease: What We Know So Far. Nutrients. 2022; 14(19):4108. https://doi.org/10.3390/nu14194108
Chicago/Turabian StyleWang, Zhonglei, Yueran Cui, Lulu Wen, Haiyang Yu, Juan Feng, Wei Yuan, and Xin He. 2022. "Dietary Restriction against Parkinson’s Disease: What We Know So Far" Nutrients 14, no. 19: 4108. https://doi.org/10.3390/nu14194108
APA StyleWang, Z., Cui, Y., Wen, L., Yu, H., Feng, J., Yuan, W., & He, X. (2022). Dietary Restriction against Parkinson’s Disease: What We Know So Far. Nutrients, 14(19), 4108. https://doi.org/10.3390/nu14194108