Targeting DNA Methylation in the Adult Brain through Diet
Abstract
:1. Introduction
1.1. The Role of Diet in DNA Methylation
1.2. The Microbiome as a Modulator of DNA Methylation
2. Role of Microbiota in Regulating This Relationship between Diet, DNA Methylation and Cognition in the Adulthood
2.1. The Symbiotic Association between Intestinal Microbiota and Their Host
2.2. The Microbiota–Gut–Brain Axis as a Modulator of Cognitive Behaviour
2.3. Microbiota-Derived Metabolites in Host Epigenetic Regulation
2.4. Interaction between Diet, Microbiota and Epigenetics in the Adult Brain
3. Impact of Diet on DNA Methylation on Adult Brain
3.1. Diet-Induced Methylation of the Dopaminergic System
3.2. Dietary Effects on BDNF Methylation
3.3. Diet and Methylation of Alzheimer’s Genes
4. Neuroprotection through the Diet
4.1. Vitamins (Deficit/Supplement)
4.2. Choline (Deficit/Supplement)
4.3. Methionine (Deficit/Supplement)
4.4. Probiotics
4.5. Diets
5. Conclusions and Future Directions
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
Abbreviations
AD | Alzheimer’s disease |
BBB | Blood–brain barrier |
BDNF | Brain-derived neurotrophic factor |
CNS | Central nervous system |
DNMT | DNA methyltransferase |
ENS | Enteric nervous system |
Hcy | Homocysteine |
HDAC | Histone deacetylase |
HF | High fat diet |
LTP | Long-term potentiation |
MCD/MCFD | Methionine-choline (folate) deficient diet |
MMSE | Mini-mental state examination |
PUFA | Poly-unsaturated fatty acids |
SAM | S-adenosyl methionine |
SAH | S-adenosyl homocysteine |
SCFA | Short-chain fatty acids |
References
- Turnbull, J.F.; Adams, R.L. DNA methylase: Purification from ascites cells and the effect of various DNA substrates on its activity. Nucleic Acids Res. 1976, 3, 677–695. [Google Scholar] [CrossRef] [Green Version]
- Cheng, X.; Kumar, S.; Klimasauskas, S.; Roberts, R.J. Crystal structure of the HhaI DNA methyltransferase. Cold Spring Harb. Symp. Quant. Biol. 1993, 58, 331–338. [Google Scholar] [CrossRef]
- Ho, D.K.; Wu, J.C.; Santi, D.V.; Floss, H.G. Stereochemical studies of the C-methylation of deoxycytidine catalyzed by HhaI methylase and the N-methylation of deoxyadenosine catalyzed by EcoRI methylase. Arch. Biochem. Biophys. 1991, 284, 264–269. [Google Scholar] [CrossRef]
- Wu, J.C.; Santi, D.V. On the mechanism and inhibition of DNA cytosine methyltransferases. Progress Clin. Biol. Res. 1985, 198, 119–129. [Google Scholar]
- Razin, A.; Riggs, A.D. DNA methylation and gene function. Science 1980, 210, 604–610. [Google Scholar] [CrossRef]
- Goto, K.; Numata, M.; Komura, J.I.; Ono, T.; Bestor, T.H.; Kondo, H. Expression of DNA methyltransferase gene in mature and immature neurons as well as proliferating cells in mice. Differentiation 1994, 56, 39–44. [Google Scholar] [CrossRef] [PubMed]
- Feng, J.; Chang, H.; Li, E.; Fan, G. Dynamic expression of de novo DNA methyltransferases Dnmt3a and Dnmt3b in the central nervous system. J. Neurosci. Res. 2005, 79, 734–746. [Google Scholar] [CrossRef] [PubMed]
- Numata, S.; Ye, T.; Hyde, T.M.; Guitart-Navarro, X.; Tao, R.; Wininger, M.; Colantuoni, C.; Weinberger, D.R.; Kleinman, J.E.; Lipska, B.K. DNA methylation signatures in development and aging of the human prefrontal cortex. Am. J. Hum. Genet. 2012, 90, 260–272. [Google Scholar] [CrossRef] [Green Version]
- Chouliaras, L.; Mastroeni, D.; Delvaux, E.; Grover, A.; Kenis, G.; Hof, P.R.; Steinbusch, H.W.; Coleman, P.D.; Rutten, B.P.; van den Hove, D.L. Consistent decrease in global DNA methylation and hydroxymethylation in the hippocampus of Alzheimer’s disease patients. Neurobiol. Aging 2013, 34, 2091–2099. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chouliaras, L.; van den Hove, D.L.; Kenis, G.; Keitel, S.; Hof, P.R.; van Os, J.; Steinbusch, H.W.; Schmitz, C.; Rutten, B.P. Age-related increase in levels of 5-hydroxymethylcytosine in mouse hippocampus is prevented by caloric restriction. Curr. Alzheimer Res. 2012, 9, 536–544. [Google Scholar] [CrossRef] [Green Version]
- Razin, A.; Szyf, M. DNA methylation patterns. Formation and function. Biochim. Biophys. Acta 1984, 782, 331–342. [Google Scholar] [CrossRef]
- Comb, M.; Goodman, H.M. CpG methylation inhibits proenkephalin gene expression and binding of the transcription factor AP-2. Nucleic Acids Res. 1990, 18, 3975–3982. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Inamdar, N.M.; Ehrlich, K.C.; Ehrlich, M. CpG methylation inhibits binding of several sequence-specific DNA-binding proteins from pea, wheat, soybean and cauliflower. Plant Mol. Biol. 1991, 17, 111–123. [Google Scholar] [CrossRef] [PubMed]
- Fujita, N.; Takebayashi, S.; Okumura, K.; Kudo, S.; Chiba, T.; Saya, H.; Nakao, M. Methylation-mediated transcriptional silencing in euchromatin by methyl-CpG binding protein MBD1 isoforms. Mol. Cell. Biol. 1999, 19, 6415–6426. [Google Scholar] [CrossRef] [Green Version]
- Hendrich, B.; Bird, A. Identification and characterization of a family of mammalian methyl-CpG binding proteins. Mol. Cell. Biol. 1998, 18, 6538–6547. [Google Scholar] [CrossRef] [Green Version]
- Nan, X.; Campoy, F.J.; Bird, A. MeCP2 is a transcriptional repressor with abundant binding sites in genomic chromatin. Cell 1997, 88, 471–481. [Google Scholar] [CrossRef] [Green Version]
- Ng, H.H.; Zhang, Y.; Hendrich, B.; Johnson, C.A.; Turner, B.M.; Erdjument-Bromage, H.; Tempst, P.; Reinberg, D.; Bird, A. MBD2 is a transcriptional repressor belonging to the MeCP1 histone deacetylase complex. Nat. Genet. 1999, 23, 58–61. [Google Scholar] [CrossRef]
- Amir, R.E.; Van den Veyver, I.B.; Wan, M.; Tran, C.Q.; Francke, U.; Zoghbi, H.Y. Rett syndrome is caused by mutations in X-linked MECP2, encoding methyl-CpG-binding protein 2. Nat. Genet. 1999, 23, 185–188. [Google Scholar] [CrossRef] [PubMed]
- Zhou, Z.; Hong, E.J.; Cohen, S.; Zhao, W.N.; Ho, H.Y.; Schmidt, L.; Chen, W.G.; Lin, Y.; Savner, E.; Griffith, E.C.; et al. Brain-specific phosphorylation of MeCP2 regulates activity-dependent Bdnf transcription, dendritic growth, and spine maturation. Neuron 2006, 52, 255–269. [Google Scholar] [CrossRef] [Green Version]
- Tao, J.; Hu, K.; Chang, Q.; Wu, H.; Sherman, N.E.; Martinowich, K.; Klose, R.J.; Schanen, C.; Jaenisch, R.; Wang, W.; et al. Phosphorylation of MeCP2 at Serine 80 regulates its chromatin association and neurological function. Proc. Natl. Acad. Sci. USA 2009, 106, 4882–4887. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, R.Z.; Akbarian, S.; Tudor, M.; Jaenisch, R. Deficiency of methyl-CpG binding protein-2 in CNS neurons results in a Rett-like phenotype in mice. Nat. Genet. 2001, 27, 327–331. [Google Scholar] [CrossRef] [PubMed]
- Moretti, P.; Levenson, J.M.; Battaglia, F.; Atkinson, R.; Teague, R.; Antalffy, B.; Armstrong, D.; Arancio, O.; Sweatt, J.D.; Zoghbi, H.Y. Learning and memory and synaptic plasticity are impaired in a mouse model of Rett syndrome. J. Neurosci. 2006, 26, 319–327. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Asaka, Y.; Jugloff, D.G.; Zhang, L.; Eubanks, J.H.; Fitzsimonds, R.M. Hippocampal synaptic plasticity is impaired in the Mecp2-null mouse model of Rett syndrome. Neurobiol. Dis. 2006, 21, 217–227. [Google Scholar] [CrossRef]
- Nelson, E.D.; Kavalali, E.T.; Monteggia, L.M. MeCP2-dependent transcriptional repression regulates excitatory neurotransmission. Curr. Biol. CB 2006, 16, 710–716. [Google Scholar] [CrossRef] [Green Version]
- Cohen, S.; Gabel, H.W.; Hemberg, M.; Hutchinson, A.N.; Sadacca, L.A.; Ebert, D.H.; Harmin, D.A.; Greenberg, R.S.; Verdine, V.K.; Zhou, Z.; et al. Genome-wide activity-dependent MeCP2 phosphorylation regulates nervous system development and function. Neuron 2011, 72, 72–85. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, H.; Zhong, X.; Chau, K.F.; Williams, E.C.; Chang, Q. Loss of activity-induced phosphorylation of MeCP2 enhances synaptogenesis, LTP and spatial memory. Nat. Neurosci. 2011, 14, 1001–1008. [Google Scholar] [CrossRef]
- Li, B.Z.; Huang, Z.; Cui, Q.Y.; Song, X.H.; Du, L.; Jeltsch, A.; Chen, P.; Li, G.; Li, E.; Xu, G.L. Histone tails regulate DNA methylation by allosterically activating de novo methyltransferase. Cell Res. 2011, 21, 1172–1181. [Google Scholar] [CrossRef] [Green Version]
- Lubin, F.D.; Roth, T.L.; Sweatt, J.D. Epigenetic regulation of BDNF gene transcription in the consolidation of fear memory. J. Neurosci. 2008, 28, 10576–10586. [Google Scholar] [CrossRef] [PubMed]
- Levenson, J.M.; Roth, T.L.; Lubin, F.D.; Miller, C.A.; Huang, I.C.; Desai, P.; Malone, L.M.; Sweatt, J.D. Evidence that DNA (cytosine-5) methyltransferase regulates synaptic plasticity in the hippocampus. J. Biol. Chem. 2006, 281, 15763–15773. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Miller, C.A.; Sweatt, J.D. Covalent modification of DNA regulates memory formation. Neuron 2007, 53, 857–869. [Google Scholar] [CrossRef] [Green Version]
- Weaver, I.C.; Cervoni, N.; Champagne, F.A.; D’Alessio, A.C.; Sharma, S.; Seckl, J.R.; Dymov, S.; Szyf, M.; Meaney, M.J. Epigenetic programming by maternal behavior. Nat. Neurosci. 2004, 7, 847–854. [Google Scholar] [CrossRef] [PubMed]
- Tomizawa, H.; Matsuzawa, D.; Ishii, D.; Matsuda, S.; Kawai, K.; Mashimo, Y.; Sutoh, C.; Shimizu, E. Methyl-donor deficiency in adolescence affects memory and epigenetic status in the mouse hippocampus. Genes Brain Behav. 2015, 14, 301–309. [Google Scholar] [CrossRef] [Green Version]
- Paternain, L.; Martisova, E.; Campión, J.; Martínez, J.A.; Ramírez, M.J.; Milagro, F.I. Methyl donor supplementation in rats reverses the deleterious effect of maternal separation on depression-like behaviour. Behav. Brain Res. 2016, 299, 51–58. [Google Scholar] [CrossRef] [PubMed]
- Kozuka, C.; Kaname, T.; Shimizu-Okabe, C.; Takayama, C.; Tsutsui, M.; Matsushita, M.; Abe, K.; Masuzaki, H. Impact of brown rice-specific γ-oryzanol on epigenetic modulation of dopamine D2 receptors in brain striatum in high-fat-diet-induced obesity in mice. Diabetologia 2017, 60, 1502–1511. [Google Scholar] [CrossRef] [PubMed]
- Heyward, F.D.; Gilliam, D.; Coleman, M.A.; Gavin, C.F.; Wang, J.; Kaas, G.; Trieu, R.; Lewis, J.; Moulden, J.; Sweatt, J.D. Obesity Weighs down Memory through a Mechanism Involving the Neuroepigenetic Dysregulation of Sirt1. J. Neurosci. 2016, 36, 1324–1335. [Google Scholar] [CrossRef] [Green Version]
- Hadad, N.; Unnikrishnan, A.; Jackson, J.A.; Masser, D.R.; Otalora, L.; Stanford, D.R.; Richardson, A.; Freeman, W.M. Caloric restriction mitigates age-associated hippocampal differential CG and non-CG methylation. Neurobiol. Aging 2018, 67, 53–66. [Google Scholar] [CrossRef] [PubMed]
- Nicholson, J.K.; Holmes, E.; Kinross, J.; Burcelin, R.; Gibson, G.; Jia, W.; Pettersson, S. Host-gut microbiota metabolic interactions. Science 2012, 336, 1262–1267. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Erny, D.; Hrabě de Angelis, A.L.; Jaitin, D.; Wieghofer, P.; Staszewski, O.; David, E.; Keren-Shaul, H.; Mahlakoiv, T.; Jakobshagen, K.; Buch, T.; et al. Host microbiota constantly control maturation and function of microglia in the CNS. Nat. Neurosci. 2015, 18, 965–977. [Google Scholar] [CrossRef] [PubMed]
- Silva, Y.P.; Bernardi, A.; Frozza, R.L. The Role of Short-Chain Fatty Acids From Gut Microbiota in Gut-Brain Communication. Front. Endocrinol. 2020, 11, 25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Spurling, C.C.; Suhl, J.A.; Boucher, N.; Nelson, C.E.; Rosenberg, D.W.; Giardina, C. The short chain fatty acid butyrate induces promoter demethylation and reactivation of RARbeta2 in colon cancer cells. Nutr. Cancer 2008, 60, 692–702. [Google Scholar] [CrossRef] [PubMed]
- Ursell, L.K.; Metcalf, J.L.; Parfrey, L.W.; Knight, R. Defining the human microbiome. Nutr. Rev. 2012, 70 (Suppl. S1), S38–S44. [Google Scholar] [CrossRef] [Green Version]
- Sender, R.; Fuchs, S.; Milo, R. Revised Estimates for the Number of Human and Bacteria Cells in the Body. PLoS Biol. 2016, 14, e1002533. [Google Scholar] [CrossRef] [Green Version]
- Microbiology by numbers. Nat. Rev. Microbiol. 2011, 9, 628. [CrossRef] [PubMed]
- Rinninella, E.; Raoul, P.; Cintoni, M.; Franceschi, F.; Miggiano, G.A.D.; Gasbarrini, A.; Mele, M.C. What is the Healthy Gut Microbiota Composition? A Changing Ecosystem across Age, Environment, Diet, and Diseases. Microorganisms 2019, 7, 14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Grice, E.A.; Segre, J.A. The human microbiome: Our second genome. Annu. Rev. Genom. Hum. Genet. 2012, 13, 151–170. [Google Scholar] [CrossRef] [Green Version]
- Rowland, I.; Gibson, G.; Heinken, A.; Scott, K.; Swann, J.; Thiele, I.; Tuohy, K. Gut microbiota functions: Metabolism of nutrients and other food components. Eur. J. Nutr. 2018, 57, 1–24. [Google Scholar] [CrossRef] [Green Version]
- Agus, A.; Clément, K.; Sokol, H. Gut microbiota-derived metabolites as central regulators in metabolic disorders. Gut 2021, 70, 1174–1182. [Google Scholar] [CrossRef]
- Ejtahed, H.S.; Hasani-Ranjbar, S. Neuromodulatory effect of microbiome on gut-brain axis; new target for obesity drugs. J. Diabetes Metab. Disord. 2019, 18, 263–265. [Google Scholar] [CrossRef] [PubMed]
- Chilloux, J.; Neves, A.L.; Boulangé, C.L.; Dumas, M.E. The microbial-mammalian metabolic axis: A critical symbiotic relationship. Curr. Opin. Clin. Nutr. Metab. Care 2016, 19, 250–256. [Google Scholar] [CrossRef]
- Mayer, E.A.; Knight, R.; Mazmanian, S.K.; Cryan, J.F.; Tillisch, K. Gut microbes and the brain: Paradigm shift in neuroscience. J. Neurosci. 2014, 34, 15490–15496. [Google Scholar] [CrossRef] [Green Version]
- Stadlbauer, V.; Engertsberger, L.; Komarova, I.; Feldbacher, N.; Leber, B.; Pichler, G.; Fink, N.; Scarpatetti, M.; Schippinger, W.; Schmidt, R.; et al. Dysbiosis, gut barrier dysfunction and inflammation in dementia: A pilot study. BMC Geriatr. 2020, 20, 248. [Google Scholar] [CrossRef]
- Kowalski, K.; Mulak, A. Brain-Gut-Microbiota Axis in Alzheimer’s Disease. J. Neurogastroenterol. Motil. 2019, 25, 48–60. [Google Scholar] [CrossRef] [Green Version]
- Saji, N.; Murotani, K.; Hisada, T.; Kunihiro, T.; Tsuduki, T.; Sugimoto, T.; Kimura, A.; Niida, S.; Toba, K.; Sakurai, T. Relationship between dementia and gut microbiome-associated metabolites: A cross-sectional study in Japan. Sci. Rep. 2020, 10, 8088. [Google Scholar] [CrossRef] [PubMed]
- Luca, M.; Chattipakorn, S.C.; Sriwichaiin, S.; Luca, A. Cognitive-Behavioural Correlates of Dysbiosis: A Review. Int. J. Mol. Sci. 2020, 21, 4834. [Google Scholar] [CrossRef]
- Fröhlich, E.E.; Farzi, A.; Mayerhofer, R.; Reichmann, F.; Jačan, A.; Wagner, B.; Zinser, E.; Bordag, N.; Magnes, C.; Fröhlich, E.; et al. Cognitive impairment by antibiotic-induced gut dysbiosis: Analysis of gut microbiota-brain communication. Brain Behav. Immun. 2016, 56, 140–155. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cryan, J.F.; O’Riordan, K.J.; Cowan, C.S.M.; Sandhu, K.V.; Bastiaanssen, T.F.S.; Boehme, M.; Codagnone, M.G.; Cussotto, S.; Fulling, C.; Golubeva, A.V.; et al. The Microbiota-Gut-Brain Axis. Physiol. Rev. 2019, 99, 1877–2013. [Google Scholar] [CrossRef]
- Breit, S.; Kupferberg, A.; Rogler, G.; Hasler, G. Vagus Nerve as Modulator of the Brain-Gut Axis in Psychiatric and Inflammatory Disorders. Front. Psychiatry 2018, 9, 44. [Google Scholar] [CrossRef] [Green Version]
- Badawy, A.A. Tryptophan availability for kynurenine pathway metabolism across the life span: Control mechanisms and focus on aging, exercise, diet and nutritional supplements. Neuropharmacology 2017, 112, 248–263. [Google Scholar] [CrossRef]
- Raybould, H.E. Gut chemosensing: Interactions between gut endocrine cells and visceral afferents. Auton. Neurosci. Basic Clin. 2010, 153, 41–46. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lal, S.; Kirkup, A.J.; Brunsden, A.M.; Thompson, D.G.; Grundy, D. Vagal afferent responses to fatty acids of different chain length in the rat. Am. J. Physiol. Gastrointest. Liver Physiol. 2001, 281, G907–G915. [Google Scholar] [CrossRef]
- Tanida, M.; Yamano, T.; Maeda, K.; Okumura, N.; Fukushima, Y.; Nagai, K. Effects of intraduodenal injection of Lactobacillus johnsonii La1 on renal sympathetic nerve activity and blood pressure in urethane-anesthetized rats. Neurosci. Lett. 2005, 389, 109–114. [Google Scholar] [CrossRef]
- Bravo, J.A.; Forsythe, P.; Chew, M.V.; Escaravage, E.; Savignac, H.M.; Dinan, T.G.; Bienenstock, J.; Cryan, J.F. Ingestion of Lactobacillus strain regulates emotional behavior and central GABA receptor expression in a mouse via the vagus nerve. Proc. Natl. Acad. Sci. USA 2011, 108, 16050–16055. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Catani, M.; Dell’acqua, F.; Thiebaut de Schotten, M. A revised limbic system model for memory, emotion and behaviour. Neurosci. Biobehav. Rev. 2013, 37, 1724–1737. [Google Scholar] [CrossRef] [PubMed]
- Li, B.; Wanka, L.; Blanchard, J.; Liu, F.; Chohan, M.O.; Iqbal, K.; Grundke-Iqbal, I. Neurotrophic peptides incorporating adamantane improve learning and memory, promote neurogenesis and synaptic plasticity in mice. FEBS Lett. 2010, 584, 3359–3365. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zheng, H.; Xu, P.; Jiang, Q.; Xu, Q.; Zheng, Y.; Yan, J.; Ji, H.; Ning, J.; Zhang, X.; Li, C.; et al. Depletion of acetate-producing bacteria from the gut microbiota facilitates cognitive impairment through the gut-brain neural mechanism in diabetic mice. Microbiome 2021, 9, 145. [Google Scholar] [CrossRef]
- Swann, J.R.; Spitzer, S.O.; Diaz Heijtz, R. Developmental Signatures of Microbiota-Derived Metabolites in the Mouse Brain. Metabolites 2020, 10, 172. [Google Scholar] [CrossRef]
- Koh, A.; Molinaro, A.; Ståhlman, M.; Khan, M.T.; Schmidt, C.; Mannerås-Holm, L.; Wu, H.; Carreras, A.; Jeong, H.; Olofsson, L.E.; et al. Microbially Produced Imidazole Propionate Impairs Insulin Signaling through mTORC1. Cell 2018, 175, 947–961.e17. [Google Scholar] [CrossRef] [Green Version]
- Stoica, L.; Zhu, P.J.; Huang, W.; Zhou, H.; Kozma, S.C.; Costa-Mattioli, M. Selective pharmacogenetic inhibition of mammalian target of Rapamycin complex I (mTORC1) blocks long-term synaptic plasticity and memory storage. Proc. Natl. Acad. Sci. USA 2011, 108, 3791–3796. [Google Scholar] [CrossRef] [Green Version]
- Kurella Tamura, M.; Chertow, G.M.; Depner, T.A.; Nissenson, A.R.; Schiller, B.; Mehta, R.L.; Liu, S.; Sirich, T.L. Metabolic Profiling of Impaired Cognitive Function in Patients Receiving Dialysis. J. Am. Soc. Nephrol. JASN 2016, 27, 3780–3787. [Google Scholar] [CrossRef]
- Krautkramer, K.A.; Kreznar, J.H.; Romano, K.A.; Vivas, E.I.; Barrett-Wilt, G.A.; Rabaglia, M.E.; Keller, M.P.; Attie, A.D.; Rey, F.E.; Denu, J.M. Diet-Microbiota Interactions Mediate Global Epigenetic Programming in Multiple Host Tissues. Mol. Cell 2016, 64, 982–992. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Scott, G.A.; Terstege, D.J.; Vu, A.P.; Law, S.; Evans, A.; Epp, J.R. Disrupted Neurogenesis in Germ-Free Mice: Effects of Age and Sex. Front. Cell Dev. Biol. 2020, 8, 407. [Google Scholar] [CrossRef] [PubMed]
- Luczynski, P.; McVey Neufeld, K.A.; Oriach, C.S.; Clarke, G.; Dinan, T.G.; Cryan, J.F. Growing up in a Bubble: Using Germ-Free Animals to Assess the Influence of the Gut Microbiota on Brain and Behavior. Int. J. Neuropsychopharmacol. 2016, 19, pyw020. [Google Scholar] [CrossRef]
- Diaz Heijtz, R.; Wang, S.; Anuar, F.; Qian, Y.; Björkholm, B.; Samuelsson, A.; Hibberd, M.L.; Forssberg, H.; Pettersson, S. Normal gut microbiota modulates brain development and behavior. Proc. Natl. Acad. Sci. USA 2011, 108, 3047–3052. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- De Palma, G.; Blennerhassett, P.; Lu, J.; Deng, Y.; Park, A.J.; Green, W.; Denou, E.; Silva, M.A.; Santacruz, A.; Sanz, Y.; et al. Microbiota and host determinants of behavioural phenotype in maternally separated mice. Nat. Commun. 2015, 6, 7735. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kundu, P.; Torres, E.R.S.; Stagaman, K.; Kasschau, K.; Okhovat, M.; Holden, S.; Ward, S.; Nevonen, K.A.; Davis, B.A.; Saito, T.; et al. Integrated analysis of behavioral, epigenetic, and gut microbiome analyses in App(NL-G-F), App(NL-F), and wild type mice. Sci. Rep. 2021, 11, 4678. [Google Scholar] [CrossRef] [PubMed]
- Cuomo, M.; Borrelli, L.; Della Monica, R.; Coretti, L.; De Riso, G.; D’Angelo Lancellotti di Durazzo, L.; Fioretti, A.; Lembo, F.; Dinan, T.G.; Cryan, J.F.; et al. DNA Methylation Profiles of Tph1A and BDNF in Gut and Brain of L. Rhamnosus-Treated Zebrafish. Biomolecules 2021, 11, 142. [Google Scholar] [CrossRef] [PubMed]
- Miranda, M.; Morici, J.F.; Zanoni, M.B.; Bekinschtein, P. Brain-Derived Neurotrophic Factor: A Key Molecule for Memory in the Healthy and the Pathological Brain. Front. Cell. Neurosci. 2019, 13, 363. [Google Scholar] [CrossRef] [PubMed]
- Höglund, E.; Øverli, Ø.; Winberg, S. Tryptophan Metabolic Pathways and Brain Serotonergic Activity: A Comparative Review. Front. Endocrinol. 2019, 10, 158. [Google Scholar] [CrossRef]
- Tooley, K.L. Effects of the Human Gut Microbiota on Cognitive Performance, Brain Structure and Function: A Narrative Review. Nutrients 2020, 12, 3009. [Google Scholar] [CrossRef]
- Marioni, R.E.; McRae, A.F.; Bressler, J.; Colicino, E.; Hannon, E.; Li, S.; Prada, D.; Smith, J.A.; Trevisi, L.; Tsai, P.C.; et al. Meta-analysis of epigenome-wide association studies of cognitive abilities. Mol. Psychiatry 2018, 23, 2133–2144. [Google Scholar] [CrossRef]
- Ramos-Molina, B.; Sánchez-Alcoholado, L.; Cabrera-Mulero, A.; Lopez-Dominguez, R.; Carmona-Saez, P.; Garcia-Fuentes, E.; Moreno-Indias, I.; Tinahones, F.J. Gut Microbiota Composition Is Associated with the Global DNA Methylation Pattern in Obesity. Front. Genet. 2019, 10, 613. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kumar, H.; Lund, R.; Laiho, A.; Lundelin, K.; Ley, R.E.; Isolauri, E.; Salminen, S. Gut microbiota as an epigenetic regulator: Pilot study based on whole-genome methylation analysis. mBio 2014, 5, e02113-14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, P.; Wu, L.; Peng, G.; Han, Y.; Tang, R.; Ge, J.; Zhang, L.; Jia, L.; Yue, S.; Zhou, K.; et al. Altered microbiomes distinguish Alzheimer’s disease from amnestic mild cognitive impairment and health in a Chinese cohort. Brain Behav. Immun. 2019, 80, 633–643. [Google Scholar] [CrossRef]
- Louwies, T.; Johnson, A.C.; Orock, A.; Yuan, T.; Greenwood-Van Meerveld, B. The microbiota-gut-brain axis: An emerging role for the epigenome. Exp. Biol. Med. 2020, 245, 138–145. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tan, J.; McKenzie, C.; Potamitis, M.; Thorburn, A.N.; Mackay, C.R.; Macia, L. The role of short-chain fatty acids in health and disease. Adv. Immunol. 2014, 121, 91–119. [Google Scholar] [CrossRef]
- Den Besten, G.; van Eunen, K.; Groen, A.K.; Venema, K.; Reijngoud, D.J.; Bakker, B.M. The role of short-chain fatty acids in the interplay between diet, gut microbiota, and host energy metabolism. J. Lipid Res. 2013, 54, 2325–2340. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dalile, B.; Van Oudenhove, L.; Vervliet, B.; Verbeke, K. The role of short-chain fatty acids in microbiota-gut-brain communication. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 461–478. [Google Scholar] [CrossRef]
- Ríos-Covián, D.; Ruas-Madiedo, P.; Margolles, A.; Gueimonde, M.; de Los Reyes-Gavilán, C.G.; Salazar, N. Intestinal Short Chain Fatty Acids and their Link with Diet and Human Health. Front. Microbiol. 2016, 7, 185. [Google Scholar] [CrossRef] [Green Version]
- Parada Venegas, D.; De la Fuente, M.K.; Landskron, G.; González, M.J.; Quera, R.; Dijkstra, G.; Harmsen, H.J.M.; Faber, K.N.; Hermoso, M.A. Short Chain Fatty Acids (SCFAs)-Mediated Gut Epithelial and Immune Regulation and Its Relevance for Inflammatory Bowel Diseases. Front. Immunol. 2019, 10, 277. [Google Scholar] [CrossRef] [Green Version]
- Champ, M.M. Physiological aspects of resistant starch and in vivo measurements. J. AOAC Int. 2004, 87, 749–755. [Google Scholar] [CrossRef] [Green Version]
- Louis, P.; Flint, H.J. Diversity, metabolism and microbial ecology of butyrate-producing bacteria from the human large intestine. FEMS Microbiol. Lett. 2009, 294, 1–8. [Google Scholar] [CrossRef] [Green Version]
- Hallert, C.; Björck, I.; Nyman, M.; Pousette, A.; Grännö, C.; Svensson, H. Increasing fecal butyrate in ulcerative colitis patients by diet: Controlled pilot study. Inflamm. Bowel Dis. 2003, 9, 116–121. [Google Scholar] [CrossRef] [PubMed]
- Boets, E.; Gomand, S.V.; Deroover, L.; Preston, T.; Vermeulen, K.; De Preter, V.; Hamer, H.M.; Van den Mooter, G.; De Vuyst, L.; Courtin, C.M.; et al. Systemic availability and metabolism of colonic-derived short-chain fatty acids in healthy subjects: A stable isotope study. J. Physiol. 2017, 595, 541–555. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rossi, M.; Amaretti, A.; Raimondi, S. Folate production by probiotic bacteria. Nutrients 2011, 3, 118–134. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Crider, K.S.; Yang, T.P.; Berry, R.J.; Bailey, L.B. Folate and DNA methylation: A review of molecular mechanisms and the evidence for folate’s role. Adv. Nutr. 2012, 3, 21–38. [Google Scholar] [CrossRef] [Green Version]
- Lawton, A.; Morgan, C.R.; Schreiner, C.R.; Schreiner, C.G.; Baumann, J.; Upchurch, B.; Xu, F.; Price, M.S.; Isaacs, G.D. Folate-Dependent Cognitive Impairment Associated with Specific Gene Networks in the Adult Mouse Hippocampus. Front. Nutr. 2020, 7, 574730. [Google Scholar] [CrossRef] [PubMed]
- Lyon, P.; Strippoli, V.; Fang, B.; Cimmino, L. B Vitamins and One-Carbon Metabolism: Implications in Human Health and Disease. Nutrients 2020, 12, 2867. [Google Scholar] [CrossRef] [PubMed]
- Yoshii, K.; Hosomi, K.; Sawane, K.; Kunisawa, J. Metabolism of Dietary and Microbial Vitamin B Family in the Regulation of Host Immunity. Front. Nutr. 2019, 6, 48. [Google Scholar] [CrossRef] [Green Version]
- Tao, L.; Liu, K.; Chen, S.; Yu, H.; An, Y.; Wang, Y.; Zhang, X.; Wang, Y.; Qin, Z.; Xiao, R. Dietary Intake of Riboflavin and Unsaturated Fatty Acid Can Improve the Multi-Domain Cognitive Function in Middle-Aged and Elderly Populations: A 2-Year Prospective Cohort Study. Front. Aging Neurosci. 2019, 11, 226. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Romano, K.A.; Martinez-Del Campo, A.; Kasahara, K.; Chittim, C.L.; Vivas, E.I.; Amador-Noguez, D.; Balskus, E.P.; Rey, F.E. Metabolic, Epigenetic, and Transgenerational Effects of Gut Bacterial Choline Consumption. Cell Host Microbe 2017, 22, 279–290.e7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tomova, A.; Bukovsky, I.; Rembert, E.; Yonas, W.; Alwarith, J.; Barnard, N.D.; Kahleova, H. The Effects of Vegetarian and Vegan Diets on Gut Microbiota. Front. Nutr. 2019, 6, 47. [Google Scholar] [CrossRef] [Green Version]
- Statovci, D.; Aguilera, M.; MacSharry, J.; Melgar, S. The Impact of Western Diet and Nutrients on the Microbiota and Immune Response at Mucosal Interfaces. Front. Immunol. 2017, 8, 838. [Google Scholar] [CrossRef] [Green Version]
- Ghosh, T.S.; Rampelli, S.; Jeffery, I.B.; Santoro, A.; Neto, M.; Capri, M.; Giampieri, E.; Jennings, A.; Candela, M.; Turroni, S.; et al. Mediterranean diet intervention alters the gut microbiome in older people reducing frailty and improving health status: The NU-AGE 1-year dietary intervention across five European countries. Gut 2020, 69, 1218–1228. [Google Scholar] [CrossRef] [Green Version]
- Cancello, R.; Turroni, S.; Rampelli, S.; Cattaldo, S.; Candela, M.; Cattani, L.; Mai, S.; Vietti, R.; Scacchi, M.; Brigidi, P.; et al. Effect of Short-Term Dietary Intervention and Probiotic Mix Supplementation on the Gut Microbiota of Elderly Obese Women. Nutrients 2019, 11, 3011. [Google Scholar] [CrossRef] [Green Version]
- Tian, T.; Zhang, X.; Luo, T.; Wang, D.; Sun, Y.; Dai, J. Effects of Short-Term Dietary Fiber Intervention on Gut Microbiota in Young Healthy People. Diabetes Metab. Syndr. Obes. Targets Ther. 2021, 14, 3507–3516. [Google Scholar] [CrossRef]
- Miro-Blanch, J.; Yanes, O. Epigenetic Regulation at the Interplay Between Gut Microbiota and Host Metabolism. Front. Genet. 2019, 10, 638. [Google Scholar] [CrossRef]
- De Filippo, C.; Cavalieri, D.; Di Paola, M.; Ramazzotti, M.; Poullet, J.B.; Massart, S.; Collini, S.; Pieraccini, G.; Lionetti, P. Impact of diet in shaping gut microbiota revealed by a comparative study in children from Europe and rural Africa. Proc. Natl. Acad. Sci. USA 2010, 107, 14691–14696. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ercolini, D.; Fogliano, V. Food Design To Feed the Human Gut Microbiota. J. Agric. Food Chem. 2018, 66, 3754–3758. [Google Scholar] [CrossRef] [Green Version]
- Arora, I.; Sharma, M.; Tollefsbol, T.O. Combinatorial Epigenetics Impact of Polyphenols and Phytochemicals in Cancer Prevention and Therapy. Int. J. Mol. Sci. 2019, 20, 4567. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gerhauser, C. Epigenetic impact of dietary isothiocyanates in cancer chemoprevention. Curr. Opin. Clin. Nutr. Metab. Care 2013, 16, 405–410. [Google Scholar] [CrossRef] [PubMed]
- Vamanu, E.; Gatea, F. Correlations between Microbiota Bioactivity and Bioavailability of Functional Compounds: A Mini-Review. Biomedicines 2020, 8, 39. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Oh, S.; Gwak, J.; Park, S.; Yang, C.S. Green tea polyphenol EGCG suppresses Wnt/β-catenin signaling by promoting GSK-3β- and PP2A-independent β-catenin phosphorylation/degradation. BioFactors 2014, 40, 586–595. [Google Scholar] [CrossRef] [PubMed]
- Braune, A.; Blaut, M. Bacterial species involved in the conversion of dietary flavonoids in the human gut. Gut Microbes 2016, 7, 216–234. [Google Scholar] [CrossRef] [Green Version]
- Paluszczak, J.; Krajka-Kuźniak, V.; Baer-Dubowska, W. The effect of dietary polyphenols on the epigenetic regulation of gene expression in MCF7 breast cancer cells. Toxicol. Lett. 2010, 192, 119–125. [Google Scholar] [CrossRef] [PubMed]
- Frolinger, T.; Herman, F.; Sharma, A.; Sims, S.; Wang, J.; Pasinetti, G.M. Epigenetic modifications by polyphenolic compounds alter gene expression in the hippocampus. Biol. Open 2018, 7, bio035196. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Engevik, M.A.; Morra, C.N.; Röth, D.; Engevik, K.; Spinler, J.K.; Devaraj, S.; Crawford, S.E.; Estes, M.K.; Kalkum, M.; Versalovic, J. Microbial Metabolic Capacity for Intestinal Folate Production and Modulation of Host Folate Receptors. Front. Microbiol. 2019, 10, 2305. [Google Scholar] [CrossRef]
- Ho, J.M.; Juurlink, D.N. Considerations when prescribing trimethoprim-sulfamethoxazole. CMAJ Can. Med. Assoc. J.—J. l’Assoc. Med. Can. 2011, 183, 1851–1858. [Google Scholar] [CrossRef] [Green Version]
- Partearroyo, T.; Samaniego-Vaesken, M.L.; Ruiz, E.; Olza, J.; Aranceta-Bartrina, J.; Gil, Á.; González-Gross, M.; Ortega, R.M.; Serra-Majem, L.; Varela-Moreiras, G. Dietary sources and intakes of folates and vitamin B12 in the Spanish population: Findings from the ANIBES study. PLoS ONE 2017, 12, e0189230. [Google Scholar] [CrossRef] [Green Version]
- An, Y.; Feng, L.; Zhang, X.; Wang, Y.; Wang, Y.; Tao, L.; Qin, Z.; Xiao, R. Dietary intakes and biomarker patterns of folate, vitamin B(6), and vitamin B(12) can be associated with cognitive impairment by hypermethylation of redox-related genes NUDT15 and TXNRD1. Clin. Epigenet. 2019, 11, 139. [Google Scholar] [CrossRef] [Green Version]
- Fact Sheets: Malnutrition. Available online: https://www.who.int/news-room/fact-sheets/detail/malnutrition (accessed on 29 September 2021).
- Choi, S.W.; Friso, S. Epigenetics: A New Bridge between Nutrition and Health. Adv. Nutr. 2010, 1, 8–16. [Google Scholar] [CrossRef] [PubMed]
- Niculescu, M.D.; Zeisel, S.H. Diet, methyl donors and DNA methylation: Interactions between dietary folate, methionine and choline. J. Nutr. 2002, 132, 2333s–2335s. [Google Scholar] [CrossRef] [PubMed]
- Pogribny, I.P.; Karpf, A.R.; James, S.R.; Melnyk, S.; Han, T.; Tryndyak, V.P. Epigenetic alterations in the brains of Fisher 344 rats induced by long-term administration of folate/methyl-deficient diet. Brain Res. 2008, 1237, 25–34. [Google Scholar] [CrossRef] [PubMed]
- Ishii, D.; Matsuzawa, D.; Matsuda, S.; Tomizawa, H.; Sutoh, C.; Shimizu, E. Methyl donor-deficient diet during development can affect fear and anxiety in adulthood in C57BL/6J mice. PLoS ONE 2014, 9, e105750. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vucetic, Z.; Carlin, J.L.; Totoki, K.; Reyes, T.M. Epigenetic dysregulation of the dopamine system in diet-induced obesity. J. Neurochem. 2012, 120, 891–898. [Google Scholar] [CrossRef] [Green Version]
- Gould, T.J. Addiction and cognition. Addict. Sci. Clin. Pract. 2010, 5, 4–14. [Google Scholar]
- Johnson, P.M.; Kenny, P.J. Dopamine D2 receptors in addiction-like reward dysfunction and compulsive eating in obese rats. Nat Neurosci 2010, 13, 635–641. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wiedholz, L.M.; Owens, W.A.; Horton, R.E.; Feyder, M.; Karlsson, R.M.; Hefner, K.; Sprengel, R.; Celikel, T.; Daws, L.C.; Holmes, A. Mice lacking the AMPA GluR1 receptor exhibit striatal hyperdopaminergia and ‘schizophrenia-related’ behaviors. Mol. Psychiatry 2008, 13, 631–640. [Google Scholar] [CrossRef]
- Reisel, D.; Bannerman, D.M.; Schmitt, W.B.; Deacon, R.M.; Flint, J.; Borchardt, T.; Seeburg, P.H.; Rawlins, J.N. Spatial memory dissociations in mice lacking GluR1. Nat. Neurosci. 2002, 5, 868–873. [Google Scholar] [CrossRef]
- Edlow, A.G. Maternal obesity and neurodevelopmental and psychiatric disorders in offspring. Prenat. Diagn. 2017, 37, 95–110. [Google Scholar] [CrossRef] [Green Version]
- Kordi-Tamandani, D.M.; Sahranavard, R.; Torkamanzehi, A. Analysis of association between dopamine receptor genes’ methylation and their expression profile with the risk of schizophrenia. Psychiatr. Genet. 2013, 23, 183–187. [Google Scholar] [CrossRef]
- Sørensen, H.J.; Gamborg, M.; Sørensen, T.I.A.; Baker, J.L.; Mortensen, E.L. Childhood body mass index and risk of schizophrenia in relation to childhood age, sex and age of first contact with schizophrenia. Eur. Psychiatry J. Assoc. Eur. Psychiatr. 2016, 34, 64–69. [Google Scholar] [CrossRef]
- Zhou, Y.; Zhu, H.; Wu, H.Y.; Jin, L.Y.; Chen, B.; Pang, H.Y.; Ming, Z.H.; Cheng, Y.; Zhou, C.L.; Guo, M.X.; et al. Diet-Induced Paternal Obesity Impairs Cognitive Function in Offspring by Mediating Epigenetic Modifications in Spermatozoa. Obesity 2018, 26, 1749–1757. [Google Scholar] [CrossRef] [PubMed]
- Arcego, D.M.; Krolow, R.; Lampert, C.; Toniazzo, A.P.; Berlitz, C.; Lazzaretti, C.; Schmitz, F.; Rodrigues, A.F.; Wyse, A.T.; Dalmaz, C. Early life adversities or high fat diet intake reduce cognitive function and alter BDNF signaling in adult rats: Interplay of these factors changes these effects. Int. J. Dev. Neurosci. Off. J. Int. Soc. Dev. Neurosci. 2016, 50, 16–25. [Google Scholar] [CrossRef] [PubMed]
- Kordi-Tamandani, D.M.; Sahranavard, R.; Torkamanzehi, A. DNA methylation and expression profiles of the brain-derived neurotrophic factor (BDNF) and dopamine transporter (DAT1) genes in patients with schizophrenia. Mol. Biol. Rep. 2012, 39, 10889–10893. [Google Scholar] [CrossRef] [PubMed]
- Gray, J.; Yeo, G.S.; Cox, J.J.; Morton, J.; Adlam, A.L.; Keogh, J.M.; Yanovski, J.A.; El Gharbawy, A.; Han, J.C.; Tung, Y.C.; et al. Hyperphagia, severe obesity, impaired cognitive function, and hyperactivity associated with functional loss of one copy of the brain-derived neurotrophic factor (BDNF) gene. Diabetes 2006, 55, 3366–3371. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Molteni, R.; Barnard, R.J.; Ying, Z.; Roberts, C.K.; Gómez-Pinilla, F. A high-fat, refined sugar diet reduces hippocampal brain-derived neurotrophic factor, neuronal plasticity, and learning. Neuroscience 2002, 112, 803–814. [Google Scholar] [CrossRef] [Green Version]
- Hu, Y.D.; Pang, W.; He, C.C.; Lu, H.; Liu, W.; Wang, Z.Y.; Liu, Y.Q.; Huang, C.Y.; Jiang, Y.G. The cognitive impairment induced by zinc deficiency in rats aged 0∼2 months related to BDNF DNA methylation changes in the hippocampus. Nutr. Neurosci. 2017, 20, 519–525. [Google Scholar] [CrossRef]
- Hsieh, M.T.; Lin, C.C.; Lee, C.T.; Huang, T.L. Abnormal Brain-Derived Neurotrophic Factor Exon IX Promoter Methylation, Protein, and mRNA Levels in Patients with Major Depressive Disorder. J. Clin. Med. 2019, 8, 568. [Google Scholar] [CrossRef] [Green Version]
- Perroud, N.; Salzmann, A.; Prada, P.; Nicastro, R.; Hoeppli, M.E.; Furrer, S.; Ardu, S.; Krejci, I.; Karege, F.; Malafosse, A. Response to psychotherapy in borderline personality disorder and methylation status of the BDNF gene. Transl. Psychiatry 2013, 3, e207. [Google Scholar] [CrossRef] [Green Version]
- Maes, M.; Vandoolaeghe, E.; Neels, H.; Demedts, P.; Wauters, A.; Meltzer, H.Y.; Altamura, C.; Desnyder, R. Lower serum zinc in major depression is a sensitive marker of treatment resistance and of the immune/inflammatory response in that illness. Biol. Psychiatry 1997, 42, 349–358. [Google Scholar] [CrossRef]
- Sales, A.J.; Biojone, C.; Terceti, M.S.; Guimarães, F.S.; Gomes, M.V.; Joca, S.R. Antidepressant-like effect induced by systemic and intra-hippocampal administration of DNA methylation inhibitors. Br. J. Pharmacol. 2011, 164, 1711–1721. [Google Scholar] [CrossRef] [Green Version]
- Tyagi, E.; Zhuang, Y.; Agrawal, R.; Ying, Z.; Gomez-Pinilla, F. Interactive actions of Bdnf methylation and cell metabolism for building neural resilience under the influence of diet. Neurobiol. Dis. 2015, 73, 307–318. [Google Scholar] [CrossRef] [Green Version]
- Fan, C.; Fu, H.; Dong, H.; Lu, Y.; Lu, Y.; Qi, K. Maternal n-3 polyunsaturated fatty acid deprivation during pregnancy and lactation affects neurogenesis and apoptosis in adult offspring: Associated with DNA methylation of brain-derived neurotrophic factor transcripts. Nutr. Res. 2016, 36, 1013–1021. [Google Scholar] [CrossRef] [PubMed]
- Wu, A.; Ying, Z.; Gomez-Pinilla, F. Docosahexaenoic acid dietary supplementation enhances the effects of exercise on synaptic plasticity and cognition. Neuroscience 2008, 155, 751–759. [Google Scholar] [CrossRef] [Green Version]
- Wu, A.; Ying, Z.; Gomez-Pinilla, F. Dietary omega-3 fatty acids normalize BDNF levels, reduce oxidative damage, and counteract learning disability after traumatic brain injury in rats. J. Neurotrauma 2004, 21, 1457–1467. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Haghighi, F.; Galfalvy, H.; Chen, S.; Huang, Y.Y.; Cooper, T.B.; Burke, A.K.; Oquendo, M.A.; Mann, J.J.; Sublette, M.E. DNA methylation perturbations in genes involved in polyunsaturated Fatty Acid biosynthesis associated with depression and suicide risk. Front. Neurol. 2015, 6, 92. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sona, C.; Kumar, A.; Dogra, S.; Kumar, B.A.; Umrao, D.; Yadav, P.N. Docosahexaenoic acid modulates brain-derived neurotrophic factor via GPR40 in the brain and alleviates diabesity-associated learning and memory deficits in mice. Neurobiol. Dis. 2018, 118, 94–107. [Google Scholar] [CrossRef]
- Wang, Z.; Zhu, M.; Wang, M.; Gao, Y.; Zhang, C.; Liu, S.; Qu, S.; Liu, Z.; Zhang, C. Integrated Multiomic Analysis Reveals the High-Fat Diet Induced Activation of the MAPK Signaling and Inflammation Associated Metabolic Cascades via Histone Modification in Adipose Tissues. Front. Genet. 2021, 12, 650863. [Google Scholar] [CrossRef]
- de Wit, L.; Luppino, F.; van Straten, A.; Penninx, B.; Zitman, F.; Cuijpers, P. Depression and obesity: A meta-analysis of community-based studies. Psychiatry Res. 2010, 178, 230–235. [Google Scholar] [CrossRef] [PubMed]
- Di Francesco, A.; Arosio, B.; Falconi, A.; Micioni Di Bonaventura, M.V.; Karimi, M.; Mari, D.; Casati, M.; Maccarrone, M.; D’Addario, C. Global changes in DNA methylation in Alzheimer’s disease peripheral blood mononuclear cells. Brain Behav. Immun. 2015, 45, 139–144. [Google Scholar] [CrossRef]
- Voisin, S.; Almén, M.S.; Moschonis, G.; Chrousos, G.P.; Manios, Y.; Schiöth, H.B. Dietary fat quality impacts genome-wide DNA methylation patterns in a cross-sectional study of Greek preadolescents. Eur. J. Hum. Genet. EJHG 2015, 23, 654–662. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cai, H.B.; Fan, Z.Z.; Tian, T.; Zhao, C.C.; Ge, Z.M. Epigenetic Control of CDK5 Promoter Regulates Diabetes-Associated Development of Alzheimer’s Disease. J. Alzheimer’s Dis. 2019, 69, 743–750. [Google Scholar] [CrossRef]
- Carone, B.R.; Fauquier, L.; Habib, N.; Shea, J.M.; Hart, C.E.; Li, R.; Bock, C.; Li, C.; Gu, H.; Zamore, P.D.; et al. Paternally induced transgenerational environmental reprogramming of metabolic gene expression in mammals. Cell 2010, 143, 1084–1096. [Google Scholar] [CrossRef] [Green Version]
- Umbaugh, D.S.; Schaller, M.L.; Harris, M.P.; Wooten, J.S.; Sandoval, K.E.; Witt, K.A. Western versus Omega-3 Based Diets Differentially Alter Key Brain Fatty Acid Transport Genes. FASEB J. 2018, 32, 922.1. [Google Scholar] [CrossRef]
- Hsieh, Y.-C.; Chiang, M.-C.; Huang, Y.-C.; Yeh, T.-H.; Shih, H.-Y.; Liu, H.-F.; Chen, H.-Y.; Wang, C.-P.; Cheng, Y.-C. Pparα deficiency inhibits the proliferation of neuronal and glial precursors in the zebrafish central nervous system. Dev. Dyn. 2018, 247, 1264–1275. [Google Scholar] [CrossRef] [Green Version]
- Luo, R.; Su, L.-Y.; Li, G.; Yang, J.; Liu, Q.; Yang, L.-X.; Zhang, D.-F.; Zhou, H.; Xu, M.; Fan, Y.; et al. Activation of PPARA-mediated autophagy reduces Alzheimer disease-like pathology and cognitive decline in a murine model. Autophagy 2020, 16, 52–69. [Google Scholar] [CrossRef] [PubMed]
- Zheng, M.; Zou, C.; Li, M.; Huang, G.; Gao, Y.; Liu, H. Folic Acid Reduces Tau Phosphorylation by Regulating PP2A Methylation in Streptozotocin-Induced Diabetic Mice. Int. J. Mol. Sci. 2017, 18, 861. [Google Scholar] [CrossRef] [Green Version]
- Yoon, S.Y.; Choi, H.I.; Choi, J.E.; Sul, C.A.; Choi, J.M.; Kim, D.H. Methotrexate decreases PP2A methylation and increases tau phosphorylation in neuron. Biochem. Biophys. Res. Commun. 2007, 363, 811–816. [Google Scholar] [CrossRef] [PubMed]
- Davinelli, S.; Calabrese, V.; Zella, D.; Scapagnini, G. Epigenetic nutraceutical diets in Alzheimer’s disease. J. Nutr. Health Aging 2014, 18, 800–805. [Google Scholar] [CrossRef]
- Mellott, T.J.; Huleatt, O.M.; Shade, B.N.; Pender, S.M.; Liu, Y.B.; Slack, B.E.; Blusztajn, J.K. Perinatal Choline Supplementation Reduces Amyloidosis and Increases Choline Acetyltransferase Expression in the Hippocampus of the APPswePS1dE9 Alzheimer’s Disease Model Mice. PLoS ONE 2017, 12, e0170450. [Google Scholar] [CrossRef]
- Nurk, E.; Refsum, H.; Bjelland, I.; Drevon, C.A.; Tell, G.S.; Ueland, P.M.; Vollset, S.E.; Engedal, K.; Nygaard, H.A.; Smith, D.A. Plasma free choline, betaine and cognitive performance: The Hordaland Health Study. Br. J. Nutr. 2013, 109, 511–519. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Botez, M.I.; Young, S.N.; Bachevalier, J.; Gauthier, S. Folate deficiency and decreased brain 5-hydroxytryptamine synthesis in man and rat. Nature 1979, 278, 182–183. [Google Scholar] [CrossRef] [PubMed]
- Goodwin, J.S.; Goodwin, J.M.; Garry, P.J. Association between nutritional status and cognitive functioning in a healthy elderly population. JAMA 1983, 249, 2917–2921. [Google Scholar] [CrossRef]
- Robins Wahlin, T.B.; Wahlin, A.; Winblad, B.; Bäckman, L. The influence of serum vitamin B12 and folate status on cognitive functioning in very old age. Biol. Psychol. 2001, 56, 247–265. [Google Scholar] [CrossRef]
- Wood, I.C. The Contribution and Therapeutic Potential of Epigenetic Modifications in Alzheimer’s Disease. Front. Neurosci. 2018, 12, 649. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hasegawa, T.; Mikoda, N.; Kitazawa, M.; LaFerla, F.M. Treatment of Alzheimer’s disease with anti-homocysteic acid antibody in 3xTg-AD male mice. PLoS ONE 2010, 5, e8593. [Google Scholar] [CrossRef] [PubMed]
- Van Asselt, D.Z.; Pasman, J.W.; van Lier, H.J.; Vingerhoets, D.M.; Poels, P.J.; Kuin, Y.; Blom, H.J.; Hoefnagels, W.H. Cobalamin supplementation improves cognitive and cerebral function in older, cobalamin-deficient persons. J. Gerontol. Ser. A Biol. Sci. Med. Sci. 2001, 56, M775–M779. [Google Scholar] [CrossRef] [Green Version]
- Martin, A.; Wiggs, C.L.; Lalonde, F.; Mack, C. Word retrieval to letter and semantic cues: A double dissociation in normal subjects using interference tasks. Neuropsychologia 1994, 32, 1487–1494. [Google Scholar] [CrossRef]
- Kapoor, A.; Baig, M.; Tunio, S.A.; Memon, A.S.; Karmani, H. Neuropsychiatric and neurological problems among Vitamin B12 deficient young vegetarians. Neurosci. J. 2017, 22, 228–232. [Google Scholar] [CrossRef] [Green Version]
- Khan, M.I.; Rath, S.; Adhami, V.M.; Mukhtar, H. Targeting epigenome with dietary nutrients in cancer: Current advances and future challenges. Pharmacol. Res. 2018, 129, 375–387. [Google Scholar] [CrossRef] [PubMed]
- Smulders, Y.M.; Blom, H.J. The homocysteine controversy. J. Inherit. Metab. Dis. 2011, 34, 93–99. [Google Scholar] [CrossRef] [Green Version]
- Lu, H.; Liu, X.; Deng, Y.; Qing, H. DNA methylation, a hand behind neurodegenerative diseases. Front. Aging Neurosci. 2013, 5, 85. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fuso, A.; Nicolia, V.; Ricceri, L.; Cavallaro, R.A.; Isopi, E.; Mangia, F.; Fiorenza, M.T.; Scarpa, S. S-adenosylmethionine reduces the progress of the Alzheimer-like features induced by B-vitamin deficiency in mice. Neurobiol. Aging 2012, 33, 1482.e1–1482.e16. [Google Scholar] [CrossRef]
- Gao, J.; Cahill, C.M.; Huang, X.; Roffman, J.L.; Lamon-Fava, S.; Fava, M.; Mischoulon, D.; Rogers, J.T. S-Adenosyl Methionine and Transmethylation Pathways in Neuropsychiatric Diseases Throughout Life. Neurother. J. Am. Soc. Exp. NeuroTher. 2018, 15, 156–175. [Google Scholar] [CrossRef] [Green Version]
- Collaboration, H.L.T. Lowering blood homocysteine with folic acid based supplements: Meta-analysis of randomised trials. Homocysteine Lowering Trialists’ Collaboration. BMJ 1998, 316, 894–898. [Google Scholar] [CrossRef] [Green Version]
- Clarke, M.; Ward, M.; Strain, J.J.; Hoey, L.; Dickey, W.; McNulty, H. B-vitamins and bone in health and disease: The current evidence. Proc. Nutr. Soc. 2014, 73, 330–339. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sai, X.; Kawamura, Y.; Kokame, K.; Yamaguchi, H.; Shiraishi, H.; Suzuki, R.; Suzuki, T.; Kawaichi, M.; Miyata, T.; Kitamura, T.; et al. Endoplasmic reticulum stress-inducible protein, Herp, enhances presenilin-mediated generation of amyloid beta-protein. J. Biol. Chem. 2002, 277, 12915–12920. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vafai, S.B.; Stock, J.B. Protein phosphatase 2A methylation: A link between elevated plasma homocysteine and Alzheimer’s Disease. FEBS Lett. 2002, 518, 1–4. [Google Scholar] [CrossRef] [Green Version]
- Seshadri, S.; Beiser, A.; Selhub, J.; Jacques, P.F.; Rosenberg, I.H.; D’Agostino, R.B.; Wilson, P.W.; Wolf, P.A. Plasma homocysteine as a risk factor for dementia and Alzheimer’s disease. N. Engl. J. Med. 2002, 346, 476–483. [Google Scholar] [CrossRef]
- McCormick, D.B.; Suttie, J.W.; Rucker, R.B.; Zempleni, J. Handbook of Vitamins, 4th ed.; CRC Press: Boca Raton, FL, USA, 2012. [Google Scholar]
- Mattson, M.P.; Shea, T.B. Folate and homocysteine metabolism in neural plasticity and neurodegenerative disorders. Trends Neurosci. 2003, 26, 137–146. [Google Scholar] [CrossRef]
- Licking, N.; Murchison, C.; Cholerton, B.; Zabetian, C.P.; Hu, S.C.; Montine, T.J.; Peterson-Hiller, A.L.; Chung, K.A.; Edwards, K.; Leverenz, J.B.; et al. Homocysteine and cognitive function in Parkinson’s disease. Parkinsonism Relat. Disord. 2017, 44, 1–5. [Google Scholar] [CrossRef]
- Lehmann, M.; Gottfries, C.G.; Regland, B. Identification of cognitive impairment in the elderly: Homocysteine is an early marker. Dement. Geriatr. Cogn. Disord. 1999, 10, 12–20. [Google Scholar] [CrossRef]
- Smith, A.D.; Refsum, H. Homocysteine, B Vitamins, and Cognitive Impairment. Annu. Rev. Nutr. 2016, 36, 211–239. [Google Scholar] [CrossRef] [PubMed]
- Eastley, R.; Wilcock, G.K.; Bucks, R.S. Vitamin B12 deficiency in dementia and cognitive impairment: The effects of treatment on neuropsychological function. Int. J. Geriatr. Psychiatry 2000, 15, 226–233. [Google Scholar] [CrossRef]
- Reynolds, E.H. The neurology of folic acid deficiency. Handb. Clin. Neurol. 2014, 120, 927–943. [Google Scholar] [CrossRef] [PubMed]
- Hama, Y.; Hamano, T.; Shirafuji, N.; Hayashi, K.; Ueno, A.; Enomoto, S.; Nagata, M.; Kimura, H.; Matsunaga, A.; Ikawa, M.; et al. Influences of Folate Supplementation on Homocysteine and Cognition in Patients with Folate Deficiency and Cognitive Impairment. Nutrients 2020, 12, 3138. [Google Scholar] [CrossRef]
- Aisen, P.S.; Schneider, L.S.; Sano, M.; Diaz-Arrastia, R.; van Dyck, C.H.; Weiner, M.F.; Bottiglieri, T.; Jin, S.; Stokes, K.T.; Thomas, R.G.; et al. High-dose B vitamin supplementation and cognitive decline in Alzheimer disease: A randomized controlled trial. JAMA 2008, 300, 1774–1783. [Google Scholar] [CrossRef] [PubMed]
- Craig, S.A. Betaine in human nutrition. Am. J. Clin. Nutr. 2004, 80, 539–549. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Institute of Medicine (US) Standing Committee on the Scientific Evaluation of Dietary Reference Intakes; Panel on Folate, Other B Vitamins, and Choline. The National Academies Collection: Reports funded by National Institutes of Health. In Dietary Reference Intakes for Thiamin, Riboflavin, Niacin, Vitamin B(6), Folate, Vitamin B(12), Pantothenic Acid, Biotin, and Choline; National Academy of Sciences: Washington, DC, USA, 1998. [Google Scholar]
- Da Costa, K.A.; Gaffney, C.E.; Fischer, L.M.; Zeisel, S.H. Choline deficiency in mice and humans is associated with increased plasma homocysteine concentration after a methionine load. Am. J. Clin. Nutr. 2005, 81, 440–444. [Google Scholar] [CrossRef] [Green Version]
- Velazquez, R.; Ash, J.A.; Powers, B.E.; Kelley, C.M.; Strawderman, M.; Luscher, Z.I.; Ginsberg, S.D.; Mufson, E.J.; Strupp, B.J. Maternal choline supplementation improves spatial learning and adult hippocampal neurogenesis in the Ts65Dn mouse model of Down syndrome. Neurobiol. Dis. 2013, 58, 92–101. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Powers, B.E.; Kelley, C.M.; Velazquez, R.; Ash, J.A.; Strawderman, M.S.; Alldred, M.J.; Ginsberg, S.D.; Mufson, E.J.; Strupp, B.J. Maternal choline supplementation in a mouse model of Down syndrome: Effects on attention and nucleus basalis/substantia innominata neuron morphology in adult offspring. Neuroscience 2017, 340, 501–514. [Google Scholar] [CrossRef] [Green Version]
- Moon, J.; Chen, M.; Gandhy, S.U.; Strawderman, M.; Levitsky, D.A.; Maclean, K.N.; Strupp, B.J. Perinatal choline supplementation improves cognitive functioning and emotion regulation in the Ts65Dn mouse model of Down syndrome. Behav. Neurosci. 2010, 124, 346–361. [Google Scholar] [CrossRef] [Green Version]
- Stevens, K.E.; Adams, C.E.; Yonchek, J.; Hickel, C.; Danielson, J.; Kisley, M.A. Permanent improvement in deficient sensory inhibition in DBA/2 mice with increased perinatal choline. Psychopharmacology 2008, 198, 413–420. [Google Scholar] [CrossRef]
- Haense, C.; Kalbe, E.; Herholz, K.; Hohmann, C.; Neumaier, B.; Krais, R.; Heiss, W.D. Cholinergic system function and cognition in mild cognitive impairment. Neurobiol. Aging 2012, 33, 867–877. [Google Scholar] [CrossRef] [PubMed]
- Mapstone, M.; Cheema, A.K.; Fiandaca, M.S.; Zhong, X.; Mhyre, T.R.; MacArthur, L.H.; Hall, W.J.; Fisher, S.G.; Peterson, D.R.; Haley, J.M.; et al. Plasma phospholipids identify antecedent memory impairment in older adults. Nat. Med. 2014, 20, 415–418. [Google Scholar] [CrossRef] [PubMed]
- Sanchez, C.J.; Hooper, E.; Garry, P.J.; Goodwin, J.M.; Goodwin, J.S. The relationship between dietary intake of choline, choline serum levels, and cognitive function in healthy elderly persons. J. Am. Geriatr. Soc. 1984, 32, 208–212. [Google Scholar] [CrossRef] [PubMed]
- Drachman, D.A.; Glosser, G.; Fleming, P.; Longenecker, G. Memory decline in the aged: Treatment with lecithin and physostigmine. Neurology 1982, 32, 944–950. [Google Scholar] [CrossRef] [PubMed]
- Spiers, P.A.; Myers, D.; Hochanadel, G.S.; Lieberman, H.R.; Wurtman, R.J. Citicoline improves verbal memory in aging. Arch. Neurol. 1996, 53, 441–448. [Google Scholar] [CrossRef]
- Moskovitz, J.; Bar-Noy, S.; Williams, W.M.; Requena, J.; Berlett, B.S.; Stadtman, E.R. Methionine sulfoxide reductase (MsrA) is a regulator of antioxidant defense and lifespan in mammals. Proc. Natl. Acad. Sci. USA 2001, 98, 12920–12925. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Richie, J.P., Jr.; Leutzinger, Y.; Parthasarathy, S.; Malloy, V.; Orentreich, N.; Zimmerman, J.A. Methionine restriction increases blood glutathione and longevity in F344 rats. FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol. 1994, 8, 1302–1307. [Google Scholar] [CrossRef] [Green Version]
- Noda, T.; Iwakiri, R.; Fujimoto, K.; Rhoads, C.A.; Aw, T.Y. Exogenous cysteine and cystine promote cell proliferation in CaCo-2 cells. Cell Prolif. 2002, 35, 117–129. [Google Scholar] [CrossRef]
- Zimmerman, J.A.; Malloy, V.; Krajcik, R.; Orentreich, N. Nutritional control of aging. Exp. Gerontol. 2003, 38, 47–52. [Google Scholar] [CrossRef]
- Miller, R.A.; Buehner, G.; Chang, Y.; Harper, J.M.; Sigler, R.; Smith-Wheelock, M. Methionine-deficient diet extends mouse lifespan, slows immune and lens aging, alters glucose, T4, IGF-I and insulin levels, and increases hepatocyte MIF levels and stress resistance. Aging Cell 2005, 4, 119–125. [Google Scholar] [CrossRef] [PubMed]
- Sun, L.; Sadighi Akha, A.A.; Miller, R.A.; Harper, J.M. Life-span extension in mice by preweaning food restriction and by methionine restriction in middle age. J. Gerontol. Ser. A Biol. Sci. Med. Sci. 2009, 64, 711–722. [Google Scholar] [CrossRef] [Green Version]
- Grandison, R.C.; Piper, M.D.; Partridge, L. Amino-acid imbalance explains extension of lifespan by dietary restriction in Drosophila. Nature 2009, 462, 1061–1064. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ruan, H.; Tang, X.D.; Chen, M.L.; Joiner, M.L.; Sun, G.; Brot, N.; Weissbach, H.; Heinemann, S.H.; Iverson, L.; Wu, C.F.; et al. High-quality life extension by the enzyme peptide methionine sulfoxide reductase. Proc. Natl. Acad. Sci. USA 2002, 99, 2748–2753. [Google Scholar] [CrossRef] [Green Version]
- Novotný, M.; Klimova, B.; Valis, M. Microbiome and Cognitive Impairment: Can Any Diets Influence Learning Processes in a Positive Way? Front. Aging Neurosci. 2019, 11, 170. [Google Scholar] [CrossRef]
- De Weerth, C. Do bacteria shape our development? Crosstalk between intestinal microbiota and HPA axis. Neurosci. Biobehav. Rev. 2017, 83, 458–471. [Google Scholar] [CrossRef] [PubMed]
- Farzi, A.; Fröhlich, E.E.; Holzer, P. Gut Microbiota and the Neuroendocrine System. Neurother. J. Am. Soc. Exp. NeuroTher. 2018, 15, 5–22. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Quigley, E.M.M. Microbiota-Brain-Gut Axis and Neurodegenerative Diseases. Curr. Neurol. Neurosci. Rep. 2017, 17, 94. [Google Scholar] [CrossRef] [PubMed]
- Silva, L.G.; Ferguson, B.S.; Avila, A.S.; Faciola, A.P. Sodium propionate and sodium butyrate effects on histone deacetylase (HDAC) activity, histone acetylation, and inflammatory gene expression in bovine mammary epithelial cells. J. Anim. Sci. 2018, 96, 5244–5252. [Google Scholar] [CrossRef] [PubMed]
- Patnala, R.; Arumugam, T.V.; Gupta, N.; Dheen, S.T. HDAC Inhibitor Sodium Butyrate-Mediated Epigenetic Regulation Enhances Neuroprotective Function of Microglia During Ischemic Stroke. Mol. Neurobiol. 2017, 54, 6391–6411. [Google Scholar] [CrossRef] [PubMed]
- Batra, V.; Sridhar, S.; Devasagayam, T.P. Enhanced one-carbon flux towards DNA methylation: Effect of dietary methyl supplements against gamma-radiation-induced epigenetic modifications. Chem. Biol. Interact. 2010, 183, 425–433. [Google Scholar] [CrossRef] [PubMed]
- Qin, Y.; Wade, P.A. Crosstalk between the microbiome and epigenome: Messages from bugs. J. Biochem. 2018, 163, 105–112. [Google Scholar] [CrossRef] [Green Version]
- Strozzi, G.P.; Mogna, L. Quantification of folic acid in human feces after administration of Bifidobacterium probiotic strains. J. Clin. Gastroenterol. 2008, 42 Pt 2 (Suppl. S3), S179–S184. [Google Scholar] [CrossRef]
- Savignac, H.M.; Kiely, B.; Dinan, T.G.; Cryan, J.F. Bifidobacteria exert strain-specific effects on stress-related behavior and physiology in BALB/c mice. Neurogastroenterol. Motil. Off. J. Eur. Gastrointest. Motil. Soc. 2014, 26, 1615–1627. [Google Scholar] [CrossRef] [PubMed]
- Messaoudi, M.; Lalonde, R.; Violle, N.; Javelot, H.; Desor, D.; Nejdi, A.; Bisson, J.F.; Rougeot, C.; Pichelin, M.; Cazaubiel, M.; et al. Assessment of psychotropic-like properties of a probiotic formulation (Lactobacillus helveticus R0052 and Bifidobacterium longum R0175) in rats and human subjects. Br. J. Nutr. 2011, 105, 755–764. [Google Scholar] [CrossRef] [Green Version]
- Allen, A.P.; Hutch, W.; Borre, Y.E.; Kennedy, P.J.; Temko, A.; Boylan, G.; Murphy, E.; Cryan, J.F.; Dinan, T.G.; Clarke, G. Bifidobacterium longum 1714 as a translational psychobiotic: Modulation of stress, electrophysiology and neurocognition in healthy volunteers. Transl. Psychiatry 2016, 6, e939. [Google Scholar] [CrossRef] [Green Version]
- Liang, S.; Wang, T.; Hu, X.; Luo, J.; Li, W.; Wu, X.; Duan, Y.; Jin, F. Administration of Lactobacillus helveticus NS8 improves behavioral, cognitive, and biochemical aberrations caused by chronic restraint stress. Neuroscience 2015, 310, 561–577. [Google Scholar] [CrossRef]
- Luo, J.; Wang, T.; Liang, S.; Hu, X.; Li, W.; Jin, F. Ingestion of Lactobacillus strain reduces anxiety and improves cognitive function in the hyperammonemia rat. Sci. China. Life Sci. 2014, 57, 327–335. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhuang, Z.Q.; Shen, L.L.; Li, W.W.; Fu, X.; Zeng, F.; Gui, L.; Lü, Y.; Cai, M.; Zhu, C.; Tan, Y.L.; et al. Gut Microbiota is Altered in Patients with Alzheimer’s Disease. J. Alzheimer’s Dis. 2018, 63, 1337–1346. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tse, J.K.Y. Gut Microbiota, Nitric Oxide, and Microglia as Prerequisites for Neurodegenerative Disorders. ACS Chem. Neurosci. 2017, 8, 1438–1447. [Google Scholar] [CrossRef] [PubMed]
- Kouchaki, E.; Tamtaji, O.R.; Salami, M.; Bahmani, F.; Daneshvar Kakhaki, R.; Akbari, E.; Tajabadi-Ebrahimi, M.; Jafari, P.; Asemi, Z. Clinical and metabolic response to probiotic supplementation in patients with multiple sclerosis: A randomized, double-blind, placebo-controlled trial. Clin. Nutr. 2017, 36, 1245–1249. [Google Scholar] [CrossRef]
- Tamtaji, O.R.; Kouchaki, E.; Salami, M.; Aghadavod, E.; Akbari, E.; Tajabadi-Ebrahimi, M.; Asemi, Z. The Effects of Probiotic Supplementation on Gene Expression Related to Inflammation, Insulin, and Lipids in Patients with Multiple Sclerosis: A Randomized, Double-Blind, Placebo-Controlled Trial. J. Am. Coll. Nutr. 2017, 36, 660–665. [Google Scholar] [CrossRef]
- Xie, C.; Prasad, A.A. Probiotics Treatment Improves Hippocampal Dependent Cognition in a Rodent Model of Parkinson’s Disease. Microorganisms 2020, 8, 1661. [Google Scholar] [CrossRef] [PubMed]
- Alipour Nosrani, E.; Tamtaji, O.R.; Alibolandi, Z.; Sarkar, P.; Ghazanfari, M.; Azami Tameh, A.; Taghizadeh, M.; Banikazemi, Z.; Hadavi, R.; Naderi Taheri, M. Neuroprotective effects of probiotics bacteria on animal model of Parkinson’s disease induced by 6-hydroxydopamine: A behavioral, biochemical, and histological study. J. Immunoass. Immunochem. 2021, 42, 106–120. [Google Scholar] [CrossRef]
- Shaaban, S.Y.; El Gendy, Y.G.; Mehanna, N.S.; El-Senousy, W.M.; El-Feki, H.S.A.; Saad, K.; El-Asheer, O.M. The role of probiotics in children with autism spectrum disorder: A prospective, open-label study. Nutr. Neurosci. 2018, 21, 676–681. [Google Scholar] [CrossRef]
- West, N.P.; Horn, P.L.; Pyne, D.B.; Gebski, V.J.; Lahtinen, S.J.; Fricker, P.A.; Cripps, A.W. Probiotic supplementation for respiratory and gastrointestinal illness symptoms in healthy physically active individuals. Clin. Nutr. 2014, 33, 581–587. [Google Scholar] [CrossRef] [Green Version]
- Borre, Y.E.; Moloney, R.D.; Clarke, G.; Dinan, T.G.; Cryan, J.F. The impact of microbiota on brain and behavior: Mechanisms & therapeutic potential. Adv. Exp. Med. Biol. 2014, 817, 373–403. [Google Scholar] [CrossRef]
- Naseribafrouei, A.; Hestad, K.; Avershina, E.; Sekelja, M.; Linløkken, A.; Wilson, R.; Rudi, K. Correlation between the human fecal microbiota and depression. Neurogastroenterol. Motil. Off. J. Eur. Gastrointest. Motil. Soc. 2014, 26, 1155–1162. [Google Scholar] [CrossRef]
- Mohammadi, A.A.; Jazayeri, S.; Khosravi-Darani, K.; Solati, Z.; Mohammadpour, N.; Asemi, Z.; Adab, Z.; Djalali, M.; Tehrani-Doost, M.; Hosseini, M.; et al. The effects of probiotics on mental health and hypothalamic-pituitary-adrenal axis: A randomized, double-blind, placebo-controlled trial in petrochemical workers. Nutr. Neurosci. 2016, 19, 387–395. [Google Scholar] [CrossRef] [PubMed]
- Benton, D.; Williams, C.; Brown, A. Impact of consuming a milk drink containing a probiotic on mood and cognition. Eur. J. Clin. Nutr. 2007, 61, 355–361. [Google Scholar] [CrossRef] [Green Version]
- Tamtaji, O.R.; Heidari-Soureshjani, R.; Mirhosseini, N.; Kouchaki, E.; Bahmani, F.; Aghadavod, E.; Tajabadi-Ebrahimi, M.; Asemi, Z. Probiotic and selenium co-supplementation, and the effects on clinical, metabolic and genetic status in Alzheimer’s disease: A randomized, double-blind, controlled trial. Clin. Nutr. 2019, 38, 2569–2575. [Google Scholar] [CrossRef]
- Akkasheh, G.; Kashani-Poor, Z.; Tajabadi-Ebrahimi, M.; Jafari, P.; Akbari, H.; Taghizadeh, M.; Memarzadeh, M.R.; Asemi, Z.; Esmaillzadeh, A. Clinical and metabolic response to probiotic administration in patients with major depressive disorder: A randomized, double-blind, placebo-controlled trial. Nutrition 2016, 32, 315–320. [Google Scholar] [CrossRef]
- Gilsing, A.M.; Crowe, F.L.; Lloyd-Wright, Z.; Sanders, T.A.; Appleby, P.N.; Allen, N.E.; Key, T.J. Serum concentrations of vitamin B12 and folate in British male omnivores, vegetarians and vegans: Results from a cross-sectional analysis of the EPIC-Oxford cohort study. Eur. J. Clin. Nutr. 2010, 64, 933–939. [Google Scholar] [CrossRef]
- Beezhold, B.L.; Johnston, C.S.; Daigle, D.R. Vegetarian diets are associated with healthy mood states: A cross-sectional study in seventh day adventist adults. Nutr. J. 2010, 9, 26. [Google Scholar] [CrossRef] [Green Version]
- Beezhold, B.L.; Johnston, C.S. Restriction of meat, fish, and poultry in omnivores improves mood: A pilot randomized controlled trial. Nutr. J. 2012, 11, 9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Beezhold, B.; Radnitz, C.; Rinne, A.; DiMatteo, J. Vegans report less stress and anxiety than omnivores. Nutr. Neurosci. 2015, 18, 289–296. [Google Scholar] [CrossRef] [PubMed]
- Baines, S.; Powers, J.; Brown, W.J. How does the health and well-being of young Australian vegetarian and semi-vegetarian women compare with non-vegetarians? Public Health Nutr. 2007, 10, 436–442. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Agarwal, U.; Mishra, S.; Xu, J.; Levin, S.; Gonzales, J.; Barnard, N.D. A multicenter randomized controlled trial of a nutrition intervention program in a multiethnic adult population in the corporate setting reduces depression and anxiety and improves quality of life: The GEICO study. Am. J. Health Promot. AJHP 2015, 29, 245–254. [Google Scholar] [CrossRef]
- Velten, J.; Bieda, A.; Scholten, S.; Wannemüller, A.; Margraf, J. Lifestyle choices and mental health: A longitudinal survey with German and Chinese students. BMC Public Health 2018, 18, 632. [Google Scholar] [CrossRef] [PubMed]
- Hibbeln, J.R.; Northstone, K.; Evans, J.; Golding, J. Vegetarian diets and depressive symptoms among men. J. Affect. Disord. 2018, 225, 13–17. [Google Scholar] [CrossRef] [PubMed]
- Trichopoulou, A.; Costacou, T.; Bamia, C.; Trichopoulos, D. Adherence to a Mediterranean diet and survival in a Greek population. N. Engl. J. Med. 2003, 348, 2599–2608. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Román, G.C.; Jackson, R.E.; Gadhia, R.; Román, A.N.; Reis, J. Mediterranean diet: The role of long-chain ω-3 fatty acids in fish; polyphenols in fruits, vegetables, cereals, coffee, tea, cacao and wine; probiotics and vitamins in prevention of stroke, age-related cognitive decline, and Alzheimer disease. Rev. Neurol. 2019, 175, 724–741. [Google Scholar] [CrossRef] [PubMed]
- Foscolou, A.; Rallidis, L.S.; Tsirebolos, G.; Critselis, E.; Katsimardos, A.; Drosatos, A.; Chrysohoou, C.; Tousoulis, D.; Pitsavos, C.; Panagiotakos, D.B. The association between homocysteine levels, Mediterranean diet and cardiovascular disease: A case-control study. Int. J. Food Sci. Nutr. 2019, 70, 603–611. [Google Scholar] [CrossRef] [PubMed]
- Lourida, I.; Soni, M.; Thompson-Coon, J.; Purandare, N.; Lang, I.A.; Ukoumunne, O.C.; Llewellyn, D.J. Mediterranean diet, cognitive function, and dementia: A systematic review. Epidemiology 2013, 24, 479–489. [Google Scholar] [CrossRef] [PubMed]
- Solfrizzi, V.; Frisardi, V.; Seripa, D.; Logroscino, G.; Imbimbo, B.P.; D’Onofrio, G.; Addante, F.; Sancarlo, D.; Cascavilla, L.; Pilotto, A.; et al. Mediterranean diet in predementia and dementia syndromes. Curr. Alzheimer Res. 2011, 8, 520–542. [Google Scholar] [CrossRef] [PubMed]
- Féart, C.; Samieri, C.; Barberger-Gateau, P. Mediterranean diet and cognitive function in older adults. Curr. Opin. Clin. Nutr. Metab. Care 2010, 13, 14–18. [Google Scholar] [CrossRef] [Green Version]
- Sofi, F.; Macchi, C.; Abbate, R.; Gensini, G.F.; Casini, A. Effectiveness of the Mediterranean diet: Can it help delay or prevent Alzheimer’s disease? J. Alzheimer’s Dis. 2010, 20, 795–801. [Google Scholar] [CrossRef]
- Panza, F.; D’Introno, A.; Colacicco, A.M.; Capurso, C.; Del Parigi, A.; Caselli, R.J.; Pilotto, A.; Argentieri, G.; Scapicchio, P.L.; Scafato, E.; et al. Current epidemiology of mild cognitive impairment and other predementia syndromes. Am. J. Geriatr. Psychiatry Off. J. Am. Assoc. Geriatr. Psychiatry 2005, 13, 633–644. [Google Scholar] [CrossRef]
- Winblad, B.; Palmer, K.; Kivipelto, M.; Jelic, V.; Fratiglioni, L.; Wahlund, L.O.; Nordberg, A.; Bäckman, L.; Albert, M.; Almkvist, O.; et al. Mild cognitive impairment—Beyond controversies, towards a consensus: Report of the International Working Group on Mild Cognitive Impairment. J. Intern. Med. 2004, 256, 240–246. [Google Scholar] [CrossRef] [PubMed]
- Solfrizzi, V.; D’Introno, A.; Colacicco, A.M.; Capurso, C.; Del Parigi, A.; Capurso, S.; Gadaleta, A.; Capurso, A.; Panza, F. Dietary fatty acids intake: Possible role in cognitive decline and dementia. Exp. Gerontol. 2005, 40, 257–270. [Google Scholar] [CrossRef]
- Solfrizzi, V.; Frisardi, V.; Capurso, C.; D’Introno, A.; Colacicco, A.M.; Vendemiale, G.; Capurso, A.; Panza, F. Dietary fatty acids in dementia and predementia syndromes: Epidemiological evidence and possible underlying mechanisms. Ageing Res. Rev. 2010, 9, 184–199. [Google Scholar] [CrossRef]
- Solfrizzi, V.; Colacicco, A.M.; D’Introno, A.; Capurso, C.; Torres, F.; Rizzo, C.; Capurso, A.; Panza, F. Dietary intake of unsaturated fatty acids and age-related cognitive decline: A 8.5-year follow-up of the Italian Longitudinal Study on Aging. Neurobiol. Aging 2006, 27, 1694–1704. [Google Scholar] [CrossRef] [PubMed]
- Psaltopoulou, T.; Kyrozis, A.; Stathopoulos, P.; Trichopoulos, D.; Vassilopoulos, D.; Trichopoulou, A. Diet, physical activity and cognitive impairment among elders: The EPIC-Greece cohort (European Prospective Investigation into Cancer and Nutrition). Public Health Nutr. 2008, 11, 1054–1062. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hervert-Hernández, D.; Goñi, I. Dietary Polyphenols and Human Gut Microbiota: A Review. Food Rev. Int. 2011, 27, 154–169. [Google Scholar] [CrossRef]
- Queipo-Ortuño, M.I.; Boto-Ordóñez, M.; Murri, M.; Gomez-Zumaquero, J.M.; Clemente-Postigo, M.; Estruch, R.; Cardona Diaz, F.; Andrés-Lacueva, C.; Tinahones, F.J. Influence of red wine polyphenols and ethanol on the gut microbiota ecology and biochemical biomarkers. Am. J. Clin. Nutr. 2012, 95, 1323–1334. [Google Scholar] [CrossRef] [PubMed]
Diet/Supplementation | Changes in Methylation Patterns | Physiological and Behavioural Effects |
---|---|---|
MCD/MCFD diet | Impaired fear extinction [124] and hippocampal-dependent memory [32]. Reduction in GluR1 expression, which increased DA levels in the striatum, resulting in schizophrenia-like behaviours. Reductions in LTP and deficits in synaptic plasticity [32,128,129]. | |
Supplementation of Methionine/Choline |
| Ameliorated depressive-like symptoms resulting from maternal deprivation in rats [33]. Choline improved visual attention tasks/affective neural functioning and hippocampal neurogenesis. Evidence to suggest it may mitigate the pathogenesis of schizophrenia/AD [161,162]. |
Choline Deficiency |
| Learning and memory deficits [100]. |
Folate Deficiency |
| Cognitive impairment [96], increased Hcy and oxidative stress [160]. Impaired abstract reasoning, visuospatial memory and attention [163,164,165] Hyperphosphorylated tau (associated with AD) and increased DNMT1 mRNA [158,159]. |
High Fat Diet | Hypermethylation: Downregulation of D2Rs in the striatum (rescued by oryzanol) [34] and the VTA, which was reported to give rise to anhedonia and addiction-like behaviours [125,127], as well as exacerbation of schizophrenia symptoms [135], hyperactivity and cognitive impairment [136,137]. Hypomethylation: increased CDK5 expression and resulting tau hyperphosphorylation. Followed by memory impairments in mice [153]. Increase of DRs in hypothalamus resulting in compulsive eating [125]. | |
SCFAs |
| Increased BDNF expression, and improvement in spatial learning and memory [145,146]. Dysregulation associated with suicide status via p38 mitogen kinase pathway [147,148]. |
Caloric Restriction |
| Improved cognition and neurogenesis in mice [36] |
Probiotics |
| Improved behaviour, mood and cognition [76,77,78] |
Low Protein |
| Reductions in PPARα increase amyloid fibrils (associated with AD) [154]. There is evidence to suggest that an increase in PPARα improves memory and anxiety in AD mice [157]. |
Zinc Deficiency |
| Decreased expression of BDNF and DMNT3a and upregulation of DNMT1 [138]. Implicated in depression, particularly treatment-resistant [139,142]. |
Vitamin B | ||
Pan-vitamin B Supplementation |
| Improved cognitive performance [119] |
Vitamin B2 Supplement (Riboflavin) |
| Improved global cognitive function in elderly human subjects [99] |
Vitamin B6 Deficiency |
| Increased brain levels of Hcy, exacerbating AD pathology and symptoms of cognitive impairment [167]. |
Vitamin B12 Supplement |
| Improved cognition, decreased Hcy [168,169] |
Vitamin B12 Deficiency |
| Correlates with paresthesia, peripheral neuropathy and psychosis in vegetarians [170]. |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 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
Allison, J.; Kaliszewska, A.; Uceda, S.; Reiriz, M.; Arias, N. Targeting DNA Methylation in the Adult Brain through Diet. Nutrients 2021, 13, 3979. https://doi.org/10.3390/nu13113979
Allison J, Kaliszewska A, Uceda S, Reiriz M, Arias N. Targeting DNA Methylation in the Adult Brain through Diet. Nutrients. 2021; 13(11):3979. https://doi.org/10.3390/nu13113979
Chicago/Turabian StyleAllison, Joseph, Aleksandra Kaliszewska, Sara Uceda, Manuel Reiriz, and Natalia Arias. 2021. "Targeting DNA Methylation in the Adult Brain through Diet" Nutrients 13, no. 11: 3979. https://doi.org/10.3390/nu13113979
APA StyleAllison, J., Kaliszewska, A., Uceda, S., Reiriz, M., & Arias, N. (2021). Targeting DNA Methylation in the Adult Brain through Diet. Nutrients, 13(11), 3979. https://doi.org/10.3390/nu13113979