Therapeutic Effects of Catechins in Less Common Neurological and Neurodegenerative Disorders
Abstract
:1. Introduction
2. Materials and Methods
3. Results
3.1. Molecular Mechanisms Associated to Catechin Use in Neurodegenerative Disorders
3.1.1. Antioxidant and Anti-Inflammatory Activity
3.1.2. Neuritogenic and Autophagic Activity
3.1.3. Inhibition of Dual-Specificity Tyrosine Phosphorylation Regulated Kinase 1A
3.2. Catechin Use in Neurodegenerative Disorders
3.2.1. Huntington Disease (HD)
3.2.2. Multiple Sclerosis (MS)
3.3. Catechins in Fetal Alcohol Spectrum Disorders (FASD)
3.4. Effects of Catechins on Down Syndrome (DS) Neurocognitive Disorders
3.5. Neurologic Effects of Catechins on Healthy Populations
Age-Related Cognitive Decline and Catechins
4. Discussion
5. Conclusions
Supplementary Materials
Author Contributions
Funding
Conflicts of Interest
References
- Sutherland, B.A.; Rahman, R.M.; Appleton, I. Mechanisms of action of green tea catechins, with a focus on ischemia-induced neurodegeneration. J. Nutr. Biochem. 2006, 17, 291–306. [Google Scholar] [CrossRef] [PubMed]
- Cardoso, R.R.; Neto, R.O.; D’Almeida, C.T.D.S.; Nascimento, T.P.D.; Pressete, C.G.; Azevedo, L.; Martino, H.S.D.; Cameron, L.C.; Ferreira, M.S.L.; de Barros, F.A.R. Kombuchas from green and black teas have different phenolic profile, which impacts their antioxidant capacities, antibacterial and antiproliferative activities. Food Res. Int. 2020, 128, 108782. [Google Scholar] [CrossRef] [PubMed]
- Bernatoniene, J.; Kopustinskiene, D.M. The Role of Catechins in Cellular Responses to Oxidative Stress. Molecules 2018, 23, 965. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Reygaert, W.C. Green Tea Catechins: Their Use in Treating and Preventing Infectious Diseases. BioMed Res. Int. 2018, 2018, 1–9. [Google Scholar] [CrossRef]
- Ide, K.; Yamada, H.; Takuma, N.; Kawasaki, Y.; Harada, S.; Nakase, J.; Ukawa, Y.; Sagesaka, Y.M. Effects of green tea consumption on cognitive dysfunction in an elderly population: A randomized placebo-controlled study. Nutr. J. 2015, 15, 1–9. [Google Scholar] [CrossRef] [Green Version]
- Lim, H.J.; Shim, S.B.; Jee, S.W.; Lee, S.H.; Lim, C.J.; Hong, J.T.; Sheen, Y.Y.; Hwang, D.Y. Green tea catechin leads to global improvement among Alzheimer’s disease-related phenotypes in NSE/hAPP-C105 Tg mice. J. Nutr. Biochem. 2013, 24, 1302–1313. [Google Scholar] [CrossRef] [PubMed]
- Pervin, M.; Unno, K.; Ohishi, T.; Tanabe, H.; Miyoshi, N.; Nakamura, Y. Beneficial Effects of Green Tea Catechins on Neurodegenerative Diseases. Molecules 2018, 23, 1297. [Google Scholar] [CrossRef] [Green Version]
- Mandel, S.A.; Avramovich-Tirosh, Y.; Reznichenko, L.; Zheng, H.; Weinreb, O.; Amit, T.; Youdim, M.B.H. Multifunctional activities of green tea catechins in neuroprotection: Modulation of cell survival genes, iron-dependent oxidative stress and PKC signaling pathway. NeuroSignals 2005, 14, 46–60. [Google Scholar] [CrossRef]
- Pringsheim, T.; Wiltshire, K.; Day, L.; Dykeman, J.; Steeves, T.; Jette, N. The incidence and prevalence of Huntington’s disease: A systematic review and meta-analysis. Mov. Disord. 2012, 27, 1083–1091. [Google Scholar] [CrossRef]
- Ramagopalan, S.V.; Sadovnick, A.D. Epidemiology of Multiple Sclerosis. Neurol. Clin. 2011, 29, 207–217. [Google Scholar] [CrossRef] [PubMed]
- Glaser, A.; Stahmann, A.; Meissner, T.; Flachenecker, P.; Horáková, D.; Zaratin, P.; Brichetto, G.; Pugliatti, M.; Rienhoff, O.; Vukusic, S.; et al. Multiple sclerosis registries in Europe—An updated mapping survey. Mult. Scler. Relat. Disord. 2019, 27, 171–178. [Google Scholar] [CrossRef] [Green Version]
- Wallin, M.T.; Culpepper, W.J.; Nichols, E.; Bhutta, Z.A.; Gebrehiwot, T.T.; Hay, S.I.; Khalil, I.A.; Krohn, K.J.; Liang, X.; Naghavi, M.; et al. Global, regional, and national burden of multiple sclerosis 1990–2016: A systematic analysis for the Global Burden of Disease Study 2016. Lancet Neurol. 2019, 18, 269–285. [Google Scholar] [CrossRef] [Green Version]
- Lanzoni, M.; Kinsner-Ovaskainen, A.; Morris, J. Socio-Economic Regional Microscope Series EUROCAT-Surveillance of Congenital Anomalies in Europe: Epidemiology of Down syndrome 1990–2014. Available online: https://www.researchgate.net/profile/Monica-Lanzoni-2/publication/332030915_Socio-economic_regional_microscope_series_EUROCAT_-Surveillance_of_congenital_anomalies_in_Europe_epidemiology_of_Down_syndrome_1990-2014/links/5ca1d831a6fdccd46047feed/Socio-economic-regional-microscope-series-EUROCAT-Surveillance-of-congenital-anomalies-in-Europe-epidemiology-of-Down-syndrome-1990-2014.pdf (accessed on 22 March 2021).
- Lanzillotta, C.; Di Domenico, F. Stress Responses in Down Syndrome Neurodegeneration: State of the Art and Therapeutic Molecules. Biomolecules 2021, 11, 266. [Google Scholar] [CrossRef]
- Kobor, M.S.; Weinberg, J. Focus on: Epigenetics and Fetal Alcohol Spectrum Disorders. Alcohol Res. Health J. Natl. Inst. Alcohol Abus. Alcohol. 2011, 34, 29–37. [Google Scholar]
- Brocardo, P.S.; Gil-Mohapel, J.; Christie, B.R. The role of oxidative stress in fetal alcohol spectrum disorders. Brain Res. Rev. 2011, 67, 209–225. [Google Scholar] [CrossRef]
- Solzak, J.P.; Liang, Y.; Zhou, F.C.; Roper, R.J. Commonality in Down and fetal alcohol syndromes. Birth Defects Res. Part A Clin. Mol. Teratol. 2013, 97, 187–197. [Google Scholar] [CrossRef] [Green Version]
- Shamseer, L.; Moher, D.; Clarke, M.; Ghersi, D.; Liberati, A.; Petticrew, M.; Shekelle, P.; Stewart, L.A.; The PRISMA-P Group. Preferred reporting items for systematic review and meta-analysis protocols (PRISMA-P) 2015: Elaboration and explanation. Br. Med. J. 2015, 349, g7647. [Google Scholar] [CrossRef] [Green Version]
- Liberati, A.; Altman, D.; Tetzlaff, J. The PRISMA statement for reporting systematic reviews and meta-analyses of studies that evaluate health care interventions: Explanation and elaboration. PLoS Med. 2009, 6. [Google Scholar] [CrossRef]
- Guyatt, G.; Oxman, A.D.; Akl, E.A.; Kunz, R.; Vist, G.; Brozek, J.; Norris, S.; Falck-Ytter, Y.; Glasziou, P.; Debeer, H. GRADE guidelines: 1. Introduction—GRADE evidence profiles and summary of findings tables. J. Clin. Epidemiol. 2011, 64, 383–394. [Google Scholar] [CrossRef]
- Hooijmans, C.R.; De Vries, R.B.M.; Ritskes-Hoitinga, M.; Rovers, M.M.; Leeflang, M.M.; IntHout, J.; Wever, K.E.; Hooft, L.; De Beer, H.; Kuijpers, T.; et al. Facilitating healthcare decisions by assessing the certainty in the evidence from preclinical animal studies. PLoS ONE 2018, 13, e0187271. [Google Scholar] [CrossRef] [Green Version]
- Fraga, C.G.; Croft, K.D.; Kennedy, D.O.; Tomás-Barberán, F.A. The effects of polyphenols and other bioactives on human health. Food Funct. 2019, 10, 514–528. [Google Scholar] [CrossRef] [Green Version]
- Botten, D.; Fugallo, G.; Fraternali, F.; Molteni, C. Structural Properties of Green Tea Catechins. J. Phys. Chem. B 2015, 119, 12860–12867. [Google Scholar] [CrossRef] [Green Version]
- Weinreb, O.; Mandel, S.; Amit, T.; Youdim, M.B. Neurological mechanisms of green tea polyphenols in Alzheimer’s and Parkinson’s diseases. J. Nutr. Biochem. 2004, 15, 506–516. [Google Scholar] [CrossRef]
- Van Acker, S.A.; Berg, D.-J.V.D.; Tromp, M.N.; Griffioen, D.H.; Van Bennekom, W.P.; Van Der Vijgh, W.J.; Bast, A. Structural aspects of antioxidant activity of flavonoids. Free Radic. Biol. Med. 1996, 20, 331–342. [Google Scholar] [CrossRef]
- Fang, E.F.; Hou, Y.; Palikaras, K.; Adriaanse, B.A.; Kerr, J.S.; Yang, B.; Lautrup, S.; Hasan-Olive, M.; Caponio, D.; Dan, X.; et al. Mitophagy inhibits amyloid-β and tau pathology and reverses cognitive deficits in models of Alzheimer’s disease. Nat. Neurosci. 2019, 22, 401–412. [Google Scholar] [CrossRef]
- Andrade, S.; Loureiro, J.A.; Pereira, M.C. Green tea extract-biomembrane interaction study: The role of its two major components, (−)-epigallocatechin gallate and (−)-epigallocatechin. Biochim. Biophys. Acta Biomembr. 2021, 1863, 183476. [Google Scholar] [CrossRef]
- Yang, G.-Z.; Wang, Z.-J.; Bai, F.; Qin, X.-J.; Cao, J.; Lv, J.-Y.; Zhang, M.-S. Epigallocatechin-3-Gallate Protects HUVECs from PM2.5-Induced Oxidative Stress Injury by Activating Critical Antioxidant Pathways. Molecules 2015, 20, 6626–6639. [Google Scholar] [CrossRef] [Green Version]
- Ramsey, C.P.; Glass, C.A.; Montgomery, M.B.; Lindl, K.A.; Ritson, G.P.; Chia, L.A.; Hamilton, R.L.; Chu, C.T.; Jordan-Sciutto, K.L. Expression of Nrf2 in Neurodegenerative Diseases. J. Neuropathol. Exp. Neurol. 2007, 66, 75–85. [Google Scholar] [CrossRef]
- Romeo, L.; Intrieri, M.; D’Agata, V.; Mangano, N.G.; Oriani, G.; Ontario, M.L.; Scapagnini, G. The major green tea polyphenol, (-)-epigallocatechin-3-gallate, induces heme oxygenase in rat neurons and acts as an effective neuroprotective agent against oxidative stress. J. Am. Coll. Nutr. 2009, 28, 492S–499S. [Google Scholar] [CrossRef]
- Degan, D.; Ornello, R.; Tiseo, C.; Carolei, A.; Sacco, S.; Pistoia, F. The Role of Inflammation in Neurological Disorders. Curr. Pharm. Des. 2018, 24, 1485–1501. [Google Scholar] [CrossRef]
- Marinovic, M.; Morandi, A.; Otton, R. Green tea catechins alone or in combination alter functional parameters of human neutrophils via suppressing the activation of TLR-4/NFκB p65 signal pathway. Toxicol. Vitr. 2015, 29, 1766–1778. [Google Scholar] [CrossRef]
- Cheng, C.-Y.; Barro, L.; Tsai, S.-T.; Feng, T.-W.; Wu, X.-Y.; Chao, C.-W.; Yu, R.-S.; Chin, T.-Y.; Hsieh, M. Epigallocatechin-3-Gallate-Loaded Liposomes Favor Anti-Inflammation of Microglia Cells and Promote Neuroprotection. Int. J. Mol. Sci. 2021, 22, 3037. [Google Scholar] [CrossRef]
- Bao, J.; Liu, W.; Zhou, H.-Y.; Gui, Y.-R.; Yang, Y.-H.; Wu, M.-J.; Xiao, Y.-F.; Shang, J.-T.; Long, G.-F.; Shu, X.-J. Epigallocatechin-3-gallate Alleviates Cognitive Deficits in APP/PS1 Mice. Curr. Med. Sci. 2020, 40, 18–27. [Google Scholar] [CrossRef]
- Urrutia, P.; Aguirre, P.; Esparza, A.; Tapia, V.; Mena, N.P.; Arredondo, M.; González-Billault, C.; Núñez, M.T. Inflammation alters the expression of DMT1, FPN1 and hepcidin, and it causes iron accumulation in central nervous system cells. J. Neurochem. 2013, 126, 541–549. [Google Scholar] [CrossRef]
- Sheelakumari, R.; Kesavadas, C.; Varghese, T.; Sreedharan, R.M.; Thomas, B.; Verghese, J.; Mathuranath, P. Assessment of Iron Deposition in the Brain in Frontotemporal Dementia and Its Correlation with Behavioral Traits. Am. J. Neuroradiol. 2017, 38, 1953–1958. [Google Scholar] [CrossRef]
- Unno, K.; Pervin, M.; Nakagawa, A.; Iguchi, K.; Hara, A.; Takagaki, A.; Nanjo, F.; Minami, A.; Nakamura, Y. Blood–Brain Barrier Permeability of Green Tea Catechin Metabolites and their Neuritogenic Activity in Human Neuroblastoma SH-SY5Y Cells. Mol. Nutr. Food Res. 2017, 61. [Google Scholar] [CrossRef]
- Gundimeda, U.; McNeill, T.H.; Fan, T.K.; Deng, R.; Rayudu, D.; Chen, Z.; Cadenas, E.; Gopalakrishna, R. Green tea catechins potentiate the neuritogenic action of brain-derived neurotrophic factor: Role of 67-kDa laminin receptor and hydrogen peroxide. Biochem. Biophys. Res. Commun. 2014, 445, 218–224. [Google Scholar] [CrossRef]
- Andrade, V.; Cortés, N.; Pastor, G.; Gonzalez, A.; Ramos-Escobar, N.; Pastene, E.; Rojo, L.E.; Maccioni, R.B. N-Acetyl Cysteine and Catechin-Derived Polyphenols: A Path toward Multi-Target Compounds against Alzheimer’s Disease. J. Alzheimer’s Dis. 2020, 75, 1219–1227. [Google Scholar] [CrossRef]
- Wang, Y.; Li, C.; Zhang, X.; Kang, X.; Li, Y.; Zhang, W.; Chen, Y.; Liu, Y.; Wang, W.; Ge, M.; et al. Exposure to PM2.5 aggravates Parkinson’s disease via inhibition of autophagy and mitophagy pathway. Toxicology 2021, 456, 152770. [Google Scholar] [CrossRef]
- Sato, S.; Uchihara, T.; Fukuda, T.; Noda, S.; Kondo, H.; Saiki, S.; Komatsu, M.; Uchiyama, Y.; Tanaka, K.; Hattori, N. Loss of autophagy in dopaminergic neurons causes Lewy pathology and motor dysfunction in aged mice. Sci. Rep. 2018, 8, 1–10. [Google Scholar] [CrossRef] [Green Version]
- Prasanth, M.I.; Sivamaruthi, B.S.; Chaiyasut, C.; Tencomnao, T. A Review of the Role of Green Tea (Camellia sinensis) in Antiphotoaging, Stress Resistance, Neuroprotection, and Autophagy. Nutrients 2019, 11, 474. [Google Scholar] [CrossRef] [Green Version]
- Holczer, M.; Besze, B.; Zámbó, V.; Csala, M.; Bánhegyi, G.; Kapuy, O. Epigallocatechin-3-Gallate (EGCG) Promotes Autophagy-Dependent Survival via Influencing the Balance of mTOR-AMPK Pathways upon Endoplasmic Reticulum Stress. Oxidative Med. Cell. Longev. 2018, 2018, 1–15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Khalil, H.; Tazi, M.; Caution, K.; Ahmed, A.; Kanneganti, A.; Assani, K.; Kopp, B.; Marsh, C.; Dakhlallah, D.; Amer, A.O. Aging is associated with hypermethylation of autophagy genes in macrophages. Epigenetics 2016, 11, 381–388. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gu, H.-F.; Nie, Y.-X.; Tong, Q.-Z.; Tang, Y.-L.; Zeng, Y.; Jing, K.-Q.; Zheng, X.-L.; Liao, D.-F. Epigallocatechin-3-Gallate Attenuates Impairment of Learning and Memory in Chronic Unpredictable Mild Stress-Treated Rats by Restoring Hippocampal Autophagic Flux. PLoS ONE 2014, 9, e112683. [Google Scholar] [CrossRef]
- Choi, C.; Song, H.-D.; Son, Y.; Cho, Y.K.; Ahn, S.-Y.; Jung, Y.-S.; Yoon, Y.C.; Kwon, S.W.; Lee, Y.-H. Epigallocatechin-3-Gallate Reduces Visceral Adiposity Partly through the Regulation of Beclin1-Dependent Autophagy in White Adipose Tissues. Nutrients 2020, 12, 3072. [Google Scholar] [CrossRef]
- Reznichenko, L.; Amit, T.; Youdim, M.B.H.; Mandel, S. Green tea polyphenol (–)-epigallocatechin-3-gallate induces neurorescue of long-term serum-deprived PC12 cells and promotes neurite outgrowth. J. Neurochem. 2005, 93, 1157–1167. [Google Scholar] [CrossRef]
- Mandel, S.A.; Amit, T.; Weinreb, O.; Reznichenko, L.; Youdim, M.B.H. Simultaneous Manipulation of Multiple Brain Targets by Green Tea Catechins: A Potential Neuroprotective Strategy for Alzheimer and Parkinson Diseases. CNS Neurosci. Ther. 2008, 14, 352–365. [Google Scholar] [CrossRef] [PubMed]
- Tejedor, F.J.; Hämmerle, B. MNB/DYRK1A as a multiple regulator of neuronal development. FEBS J. 2010, 278, 223–235. [Google Scholar] [CrossRef] [Green Version]
- Gu, Y.; Moroy, G.; Paul, J.-L.; Rebillat, A.-S.; Dierssen, M.; De La Torre, R.; Cieuta-Walti, C.; Dairou, J.; Janel, N. Molecular Rescue of Dyrk1A Overexpression Alterations in Mice with Fontup® Dietary Supplement: Role of Green Tea Catechins. Int. J. Mol. Sci. 2020, 21, 1404. [Google Scholar] [CrossRef] [Green Version]
- Beasley, M.; Stonebraker, A.; Hasan, I.; Kapp, K.L.; Liang, B.J.; Agarwal, G.; Groover, S.; Sedighi, F.; Legleiter, J. Lipid Membranes Influence the Ability of Small Molecules to Inhibit Huntingtin Fibrillization. Biochemistry 2019, 58, 4361–4373. [Google Scholar] [CrossRef]
- Ehrnhoefer, D.E.; Duennwald, M.; Markovic, P.; Wacker, J.L.; Engemann, S.; Roark, M.; Legleiter, J.; Marsh, J.L.; Thompson, L.M.; Lindquist, S.; et al. Green tea (−)-epigallocatechin-gallate modulates early events in huntingtin misfolding and reduces toxicity in Huntington’s disease models. Hum. Mol. Genet. 2006, 15, 2743–2751. [Google Scholar] [CrossRef] [PubMed]
- Varga, J.; Dér, N.P.; Zsindely, N.; Bodai, L. Green tea infusion alleviates neurodegeneration induced by mutant Huntingtin in Drosophila. Nutr. Neurosci. 2018, 23, 183–189. [Google Scholar] [CrossRef] [Green Version]
- Bellmann-Strobl, J.; Paul, F.; Wuerfel, J.; Dörr, J.; Infante-Duarte, C.; Heidrich, E.; Körtgen, B.; Brandt, A.; Pfüller, C.; Radbruch, H.; et al. Epigallocatechin Gallate in Relapsing-Remitting Multiple Sclerosis. Neurol. Neuroimmunol. Neuroinflamm. 2021, 8, e981. [Google Scholar] [CrossRef]
- Rust, R.; Chien, C.; Scheel, M.; Brandt, A.U.; Dörr, J.; Wuerfel, J.; Klumbies, K.; Zimmermann, H.; Lorenz, M.; Wernecke, K.-D.; et al. Epigallocatechin Gallate in Progressive MS. Neurol. Neuroimmunol. Neuroinflamm. 2021, 8, e964. [Google Scholar] [CrossRef]
- Mossakowski, A.A.; Pohlan, J.; Bremer, D.; Lindquist, R.; Millward, J.M.; Bock, M.; Pollok, K.; Mothes, R.; Viohl, L.; Radbruch, M.; et al. Tracking CNS and systemic sources of oxidative stress during the course of chronic neuroinflammation. Acta Neuropathol. 2015, 130, 799–814. [Google Scholar] [CrossRef] [Green Version]
- Mähler, A.; Steiniger, J.; Bock, M.; Klug, L.; Parreidt, N.; Lorenz, M.; Zimmermann, B.F.; Krannich, A.; Paul, F.; Boschmann, M. Metabolic response to epigallocatechin-3-gallate in relapsing-remitting multiple sclerosis: A randomized clinical trial. Am. J. Clin. Nutr. 2015, 101, 487–495. [Google Scholar] [CrossRef]
- Lovera, J.; Ramos, A.; Devier, D.; Garrison, V.; Kovner, B.; Reza, T.; Koop, D.; Rooney, W.; Foundas, A.; Bourdette, D. Polyphenon E, Non-futile at Neuroprotection in Multiple Sclerosis but Unpredictably Hepatotoxic: Phase I Double-blind Group and Phase II Randomized Placebo-Controlled Studies. J. Neurol. Sci. 2015, 358, 46–52. [Google Scholar] [CrossRef] [Green Version]
- Wang, J.; Ren, Z.; Xu, Y.; Xiao, S.; Meydani, S.N.; Wu, D. Epigallocatechin-3-Gallate Ameliorates Experimental Autoimmune Encephalomyelitis by Altering Balance among CD4+ T-Cell Subsets. Am. J. Pathol. 2012, 180, 221–234. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sun, Q.; Zheng, Y.; Zhang, X.; Hu, X.; Wang, Y.; Zhang, S.; Zhang, N.; Nie, H. Novel immunoregulatory properties of EGCG on reducing inflammation in EAE. Front. Biosci. 2013, 18, 332–342. [Google Scholar]
- Herges, K.; Millward, J.M.; Hentschel, N.; Infante-Duarte, C.; Aktas, O.; Zipp, F. Neuroprotective Effect of Combination Therapy of Glatiramer Acetate and Epigallocatechin-3-Gallate in Neuroinflammation. PLoS ONE 2011, 6, e25456. [Google Scholar] [CrossRef] [PubMed]
- Mohammad-Reza, S.; Farhad, M.; Mahnaz, A.; Salehi, Z. Effects of Green Tea Epigallocatechin-3-Gallate (EGCG) On Proteolipid Protein (PLP) and Oligodendrocyte Transcription Factor 1 (Olig1) Expression in the Cerebral Cortex of Cuprizone Induced Multiple Sclerosis Mice; A Western Blot Study. Casp. J. Neurol. Sci. 2016, 1, 1–9. [Google Scholar]
- Semnani, M.; Mashayekhi, F.; Azarnia, M.; Salehi, Z. Effects of green tea epigallocatechin-3-gallate on the proteolipid protein and oligodendrocyte transcription factor 1 messenger RNA gene expression in a mouse model of multiple sclerosis. Folia Neuropathol. 2017, 3, 199–205. [Google Scholar] [CrossRef] [PubMed]
- Harjes, P.; E Wanker, E. The hunt for huntingtin function: Interaction partners tell many different stories. Trends Biochem. Sci. 2003, 28, 425–433. [Google Scholar] [CrossRef]
- Rosenblatt, A. Neuropsychiatry of Huntington’s disease. Dialog Clin. Neurosci. 2007, 9, 191–197. [Google Scholar] [CrossRef]
- Savani, A.A.; Login, I.S.; Marshall, F.J.; Fahn, S.; Clarence-Smith, K. Tetrabenazine as antichorea therapy in Huntington disease: A randomized controlled trial. Neurology 2007, 68, 797. [Google Scholar] [CrossRef] [PubMed]
- Wyant, K.J.; Ridder, A.J.; Dayalu, P. Huntington’s Disease—Update on Treatments. Curr. Neurol. Neurosci. Rep. 2017, 17, 33. [Google Scholar] [CrossRef]
- Priller, J. Effects of EGCG (Epigallocatechin Gallate) in Huntington’s Disease The ETON-Study—A Randomized, Double-blind, Stratified, Placebo-controlled Prospective Investigator Initiated Multicenter Trial-Charite University, Berlin, Germany ClinicalTrials.gov Identifier: NCT01357681. Available online: https://clinicaltrials.gov/ct2/show/NCT01357681 (accessed on 29 April 2021).
- Lublin, F.D. New Multiple Sclerosis Phenotypic Classification. Eur. Neurol. 2014, 72, 1–5. [Google Scholar] [CrossRef]
- Rae-Grant, A.; Day, G.; Marrie, R.A.; Rabinstein, A.; Cree, B.; Gronseth, G.S.; Haboubi, M.; Halper, J.; Hosey, J.P.; Jones, D.E.; et al. Practice guideline recommendations summary: Disease-modifying therapies for adults with multiple sclerosis. Neurology 2018, 90, 777–788. [Google Scholar] [CrossRef] [Green Version]
- Hoyme, H.E.; Kalberg, W.O.; Elliott, A.J.; Blankenship, J.; Buckley, D.; Marais, A.-S.; Manning, M.A.; Robinson, L.K.; Adam, M.P.; Abdul-Rahman, O.; et al. Updated Clinical Guidelines for Diagnosing Fetal Alcohol Spectrum Disorders. Pediatrics 2016, 138, e20154256. [Google Scholar] [CrossRef] [Green Version]
- Almeida, L.; Andreu-Fernández, V.; Navarro-Tapia, E.; Aras-López, R.; Serra-Delgado, M.; Martínez, L.; García-Algar, O.; Gómez-Roig, M.D. Murine Models for the Study of Fetal Alcohol Spectrum Disorders: An Overview. Front. Pediatr. 2020, 8, 359. [Google Scholar] [CrossRef]
- Heaton, M.B.; Paiva, M.; Madorsky, I.; Mayer, J.; Moore, D. Effects of ethanol on neurotrophic factors, apoptosis-related proteins, endogenous antioxidants, and reactive oxygen species in neonatal striatum: Relationship to periods of vulnerability. Dev. Brain Res. 2003, 140, 237–252. [Google Scholar] [CrossRef]
- Joya, X.; Garcia-Algar, O.; Salat-Batlle, J.; Pujades, C.; Vall, O. Advances in the development of novel antioxidant therapies as an approach for fetal alcohol syndrome prevention. Birth Defects Res. Part A Clin. Mol. Teratol. 2015, 103, 163–177. [Google Scholar] [CrossRef] [PubMed]
- Pei, J.; Baugh, L.; Andrew, G.; Rasmussen, C. Intervention recommendations and subsequent access to services following clinical assessment for fetal alcohol spectrum disorders. Res. Dev. Disabil. 2017, 60, 176–186. [Google Scholar] [CrossRef] [PubMed]
- Murawski, N.J.; Moore, E.M.; Thomas, J.D.; Riley, E.P. Advances in diagnosis and treatment of fetal alcohol spectrum disorders: From animal models to human studies. Alcohol Res. Curr. Rev. 2015, 37, 97. [Google Scholar]
- Chu, K.; Wang, C.; Chu, C.; Choy, K.W.; Pang, C.; Rogers, M. Uptake and distribution of catechins in fetal organs following in utero exposure in rats. Hum. Reprod. 2007, 22, 280–287. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Antonio, A.M.; Druse, M.J. Antioxidants prevent ethanol-associated apoptosis in fetal rhombencephalic neurons. Brain Res. 2008, 1204, 16–23. [Google Scholar] [CrossRef] [Green Version]
- Long, L.; Li, Y.; Wang, Y.D.; He, Q.Y.; Li, M.; Cai, X.D.; Peng, K.; Li, X.P.; Xie, D.; Wen, Y.L.; et al. The Preventive Effect of Oral EGCG in a Fetal Alcohol Spectrum Disorder Mouse Model. Alcohol. Clin. Exp. Res. 2010, 34, 1929–1936. [Google Scholar] [CrossRef] [PubMed]
- Tiwari, V.; Kuhad, A.; Chopra, K. Epigallocatechin-3-gallate ameliorates alcohol-induced cognitive dysfunctions and apoptotic neurodegeneration in the developing rat brain. Int. J. Neuropsychopharmacol. 2010, 13, 1053–1066. [Google Scholar] [CrossRef] [Green Version]
- Almeida-Toledano, L.; Andreu-Fernández, V.; Aras-López, R.; García-Algar, Ó.; Martínez, L.; Gómez-Roig, M.D. Epigallocatechin Gallate Ameliorates the Effects of Prenatal Alcohol Exposure in a Fetal Alcohol Spectrum Disorder-Like Mouse Model. Int. J. Mol. Sci. 2021, 22, 715. [Google Scholar] [CrossRef]
- Portales-Casamar, E.; Lussier, A.A.; Jones, M.J.; MacIsaac, J.L.; Edgar, R.D.; Mah, S.M.; Barhdadi, A.; Provost, S.; Lemieux-Perreault, L.-P.; Cynader, M.S.; et al. DNA methylation signature of human fetal alcohol spectrum disorder. Epigenetics Chromatin 2016, 9, 1–20. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tunc-Ozcan, E.; Wert, S.L.; Lim, P.H.; Ferreira, A.; Redei, E.E. Hippocampus-dependent memory and allele-specific gene expression in adult offspring of alcohol-consuming dams after neonatal treatment with thyroxin or metformin. Mol. Psychiatry 2017, 23, 1643–1651. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, W.J.; Shim, J.-Y.; Zhu, B.T. Mechanisms for the Inhibition of DNA Methyltransferases by Tea Catechins and Bioflavonoids. Mol. Pharmacol. 2005, 68, 1018–1030. [Google Scholar] [CrossRef] [Green Version]
- de la Torre, R.; de Sola, S.; Hernandez, G.; Farré, M.; Pujol, J.; Rodriguez, J.; Espadaler, J.M.; Langohr, K.; Cuenca-Royo, A.; Principe, A.; et al. Safety and efficacy of cognitive training plus epigallocatechin-3-gallate in young adults with Down’s syndrome (TESDAD): A double-blind, randomised, placebo-controlled, phase 2 trial. Lancet Neurol. 2016, 15, 801–810. [Google Scholar] [CrossRef]
- Algar, O.G.; de Salut Mar, P. Epigallocatechin Gallate (EGCG) to Improve Cognitive Performance in Foetal Alcohol Syndrome (FAS) Children (Neuro-SAF) Full Text View ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ct2/show/NCT02558933 (accessed on 8 April 2021).
- Dierssen, M. Down syndrome: The brain in trisomic mode. Nat. Rev. Neurosci. 2012, 13, 844–858. [Google Scholar] [CrossRef] [PubMed]
- Cué, C.M.; Dierssen, M. Plasticity as a therapeutic target for improving cognition and behavior in Down syndrome. Chang. Brains Appl. Brain Plast. Adv. Recover Hum. Abil. 2020, 251, 269–302. [Google Scholar] [CrossRef]
- Lott, I.T.; Dierssen, M. Cognitive deficits and associated neurological complications in individuals with Down’s syndrome. Lancet Neurol. 2010, 9, 623–633. [Google Scholar] [CrossRef]
- Vicari, S.; Pontillo, M.; Armando, M. Neurodevelopmental and psychiatric issues in Down’s syndrome. Psychiatr. Genet. 2013, 23, 95–107. [Google Scholar] [CrossRef]
- Vaughan, R.M.; McGee, C.; Guerin, S.; Tyrrell, J.; Dodd, P. The challenges of diagnosis and treatment of dementia in Down’s syndrome. Ir. J. Psychol. Med. 2016, 33, 151–158. [Google Scholar] [CrossRef]
- Hanney, M.; Prasher, V.; Williams, N.; Jones, E.L.; Aarsland, D.; Corbett, A.; Lawrence, D.; Yu, L.-M.; Tyrer, S.; Francis, P.T.; et al. Memantine for dementia in adults older than 40 years with Down’s syndrome (MEADOWS): A randomised, double-blind, placebo-controlled trial. Lancet 2012, 379, 528–536. [Google Scholar] [CrossRef]
- Singh, B.N.; Shankar, S.; Srivastava, R.K. Green tea catechin, epigallocatechin-3-gallate (EGCG): Mechanisms, perspectives and clinical applications. Biochem. Pharmacol. 2011, 82, 1807–1821. [Google Scholar] [CrossRef] [Green Version]
- Wyganowska-Swiatkowska, M.; Matthews-Kozanecka, M.; Matthews-Brzozowska, T.; Skrzypczak-Jankun, E.; Jankun, J. Can EGCG Alleviate Symptoms of Down Syndrome by Altering Proteolytic Activity? Int. J. Mol. Sci. 2018, 19, 248. [Google Scholar] [CrossRef] [Green Version]
- Souchet, B.; Guedj, F.; Penke-Verdier, Z.; Daubigney, F.; Duchon, A.; Herault, Y.; Bizot, J.-C.; Janel, N.; Créau, N.; Delatour, B.; et al. Pharmacological correction of excitation/inhibition imbalance in Down syndrome mouse models. Front. Behav. Neurosci. 2015, 9, 267. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Catuara-Solarz, S.; Espinosa-Carrasco, J.; Erb, I.; Langohr, K.; Gonzalez, J.R.; Notredame, C.; Dierssen, M. Combined Treatment With Environmental Enrichment and (–)-Epigallocatechin-3-Gallate Ameliorates Learning Deficits and Hippocampal Alterations in a Mouse Model of Down Syndrome. eNeuro 2016, 3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Goodlett, C.R.; Stringer, M.; Lacombe, J.; Patel, R.; Wallace, J.M.; Roper, R.J. Evaluation of the therapeutic potential of Epigallocatechin-3-gallate (EGCG) via oral gavage in young adult Down syndrome mice. Sci. Rep. 2020, 10, 1–17. [Google Scholar] [CrossRef] [PubMed]
- Stringer, M.; Abeysekera, I.; Dria, K.J.; Roper, R.J.; Goodlett, C.R. Low dose EGCG treatment beginning in adolescence does not improve cognitive impairment in a Down syndrome mouse model. Pharmacol. Biochem. Behav. 2015, 138, 70–79. [Google Scholar] [CrossRef] [Green Version]
- Stringer, M.; Abeysekera, I.; Thomas, J.; LaCombe, J.; Stancombe, K.; Stewart, R.J.; Dria, K.J.; Wallace, J.; Goodlett, C.R.; Roper, R.J. Epigallocatechin-3-gallate (EGCG) consumption in the Ts65Dn model of Down syndrome fails to improve behavioral deficits and is detrimental to skeletal phenotypes. Physiol. Behav. 2017, 177, 230–241. [Google Scholar] [CrossRef] [PubMed]
- Ecatuara-Solarz, S.; Espinosa-Carrasco, J.; Eerb, I.; Langohr, K.; Enotredame, C.; Egonzález, J.R.; Edierssen, M. Principal Component Analysis of the Effects of Environmental Enrichment and (–)-epigallocatechin-3-gallate on Age-Associated Learning Deficits in a Mouse Model of Down Syndrome. Front. Behav. Neurosci. 2015, 9, 330. [Google Scholar] [CrossRef]
- Valenti, D.; De Rasmo, D.; Signorile, A.; Rossi, L.; de Bari, L.; Scala, I.; Granese, B.; Papa, S.; Vacca, R.A. Epigallocatechin-3-gallate prevents oxidative phosphorylation deficit and promotes mitochondrial biogenesis in human cells from subjects with Down’s syndrome. Biochim. Biophys. Acta Mol. Basis Dis. 2013, 1832, 542–552. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Valenti, D.; de Bari, L.; de Rasmo, D.; Signorile, A.; Henrion-Caude, A.; Contestabile, A.; Vacca, R.A. The polyphenols resveratrol and epigallocatechin-3-gallate restore the severe impairment of mitochondria in hippocampal progenitor cells from a Down syndrome mouse model. Biochim. Biophys. Acta Mol. Basis Dis. 2016, 1862, 1093–1104. [Google Scholar] [CrossRef]
- Stagni, F.; Giacomini, A.; Emili, M.; Trazzi, S.; Guidi, S.; Sassi, M.; Ciani, E.; Rimondini, R.; Bartesaghi, R. Short- and long-term effects of neonatal pharmacotherapy with epigallocatechin-3-gallate on hippocampal development in the Ts65Dn mouse model of Down syndrome. Neuroscience 2016, 333, 277–301. [Google Scholar] [CrossRef]
- De La Torre, R.; De Sola, S.; Pons, M.; Duchon, A.; de Lagran, M.M.; Farré, M.; Fitó, M.; Benejam, B.; Langohr, K.; Rodriguez, J.; et al. Epigallocatechin-3-gallate, a DYRK1A inhibitor, rescues cognitive deficits in Down syndrome mouse models and in humans. Mol. Nutr. Food Res. 2014, 58, 278–288. [Google Scholar] [CrossRef]
- De Toma, I.; Ortega, M.; Aloy, P.; Sabidó, E.; Dierssen, M. DYRK1A Overexpression Alters Cognition and Neural-Related Proteomic Pathways in the Hippocampus That Are Rescued by Green Tea Extract and/or Environmental Enrichment. Front. Mol. Neurosci. 2019, 12, 272. [Google Scholar] [CrossRef] [PubMed]
- De Toma, I.; Ortega, M.; Catuara-Solarz, S.; Sierra, C.; Sabidó, E.; Dierssen, M. Re-establishment of the epigenetic state and rescue of kinome deregulation in Ts65Dn mice upon treatment with green tea extract and environmental enrichment. Sci. Rep. 2020, 10, 1–18. [Google Scholar] [CrossRef] [PubMed]
- Starbuck, J.M.; Llambrich, S.; Gonzàlez, R.; Albaigès, J.; Sarlé, A.; Wouters, J.; González, A.; Sevillano, X.; Sharpe, J.; De La Torre, R.; et al. Green tea extracts containing epigallocatechin-3-gallate modulate facial development in Down syndrome. Sci. Rep. 2021, 11, 1–13. [Google Scholar] [CrossRef] [PubMed]
- Kesse-Guyot, E.; Fézeu, L.; Andreeva, V.A.; Touvier, M.; Scalbert, A.; Hercberg, S.; Galan, P. Total and Specific Polyphenol Intakes in Midlife Are Associated with Cognitive Function Measured 13 Years Later. J. Nutr. 2011, 142, 76–83. [Google Scholar] [CrossRef] [PubMed]
- Biasibetti, R.; Tramontina, A.C.; Costa, A.P.; Dutra, M.F.; Quincozes-Santos, A.; Nardin, P.; Bernardi, C.L.; Wartchow, K.M.; Lunardi, P.S.; Gonçalves, C.-A. Green tea (−)epigallocatechin-3-gallate reverses oxidative stress and reduces acetylcholinesterase activity in a streptozotocin-induced model of dementia. Behav. Brain Res. 2013, 236, 186–193. [Google Scholar] [CrossRef]
- Scholey, A.; Downey, L.A.; Ciorciari, J.; Pipingas, A.; Nolidin, K.; Finn, M.; Wines, M.; Catchlove, S.; Terrens, A.; Barlow, E.; et al. Acute neurocognitive effects of epigallocatechin gallate (EGCG). Appetite 2012, 58, 767–770. [Google Scholar] [CrossRef]
- Wightman, E.L.; Haskell, C.F.; Forster, J.S.; Veasey, R.C.; Kennedy, D.O. Epigallocatechin gallate, cerebral blood flow parameters, cognitive performance and mood in healthy humans: A double-blind, placebo-controlled, crossover investigation. Hum. Psychopharmacol. Clin. Exp. 2012, 27, 177–186. [Google Scholar] [CrossRef]
- Liu, Y.; Fly, A.D.; Wang, Z.; Klaunig, J.E. The Effects of Green Tea Extract on Working Memory in Healthy Women. J. Nutr. Health Aging 2017, 22, 446–450. [Google Scholar] [CrossRef]
- Dietz, C.; Dekker, M.; Piqueras-Fiszman, B. An intervention study on the effect of matcha tea, in drink and snack bar formats, on mood and cognitive performance. Food Res. Int. 2017, 99, 72–83. [Google Scholar] [CrossRef]
- Mohamed, S.; Ming, T.L.; Jaffri, J.M. Cognitive enhancement and neuroprotection by catechin-rich oil palm leaf extract supplement. J. Sci. Food Agric. 2012, 93, 819–827. [Google Scholar] [CrossRef]
- Baba, Y.; Inagaki, S.; Nakagawa, S.; Kaneko, T.; Kobayashi, M.; Takihara, T. Effect of Daily Intake of Green Tea Catechins on Cognitive Function in Middle-Aged and Older Subjects: A Randomized, Placebo-Controlled Study. Molecules 2020, 25, 4265. [Google Scholar] [CrossRef] [PubMed]
- Sakurai, K.; Shen, C.; Ezaki, Y.; Inamura, N.; Fukushima, Y.; Masuoka, N.; Hisatsune, T. Effects of Matcha Green Tea Powder on Cognitive Functions of Community-Dwelling Elderly Individuals. Nutrients 2020, 12, 3639. [Google Scholar] [CrossRef]
- Unno, K.; Pervin, M.; Taguchi, K.; Konishi, T.; Nakamura, Y. Green Tea Catechins Trigger Immediate-Early Genes in the Hippocampus and Prevent Cognitive Decline and Lifespan Shortening. Molecules 2020, 25, 1484. [Google Scholar] [CrossRef] [Green Version]
- Ramis, M.R.; Sarubbo, F.; Tejada, S.; Jiménez, M.; Esteban, S.; Miralles, A.; Moranta, D. Chronic Polyphenon-60 or Catechin Treatments Increase Brain Monoamines Syntheses and Hippocampal SIRT1 LEVELS Improving Cognition in Aged Rats. Nutrients 2020, 12, 326. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Deary, I.J.; Brett, C.E. Predicting and retrodicting intelligence between childhood and old age in the 6-Day Sample of the Scottish Mental Survey 1947. Intelligence 2015, 50, 1–9. [Google Scholar] [CrossRef] [Green Version]
- Tucker-Drob, E.M.; Briley, D.A. Continuity of genetic and environmental influences on cognition across the life span: A meta-analysis of longitudinal twin and adoption studies. Psychol. Bull. 2014, 140, 949–979. [Google Scholar] [CrossRef] [Green Version]
- Legdeur, N.; Heymans, M.W.; Comijs, H.C.; Huisman, M.; Maier, A.B.; Visser, P.J. Age dependency of risk factors for cognitive decline. BMC Geriatr. 2018, 18, 1–10. [Google Scholar] [CrossRef]
- Forcano, L.; Fauria, K.; Soldevila-Domenech, N.; Minguillón, C.; Lorenzo, T.; Cuenca-Royo, A.; Menezes-Cabral, S.; Pizarro, N.; Boronat, A.; Molinuevo, J.L.; et al. Prevention of cognitive decline in subjective cognitive decline APOE ε4 carriers after EGCG and a multimodal intervention (PENSA): Study design. Alzheimer’s Dement. Transl. Res. Clin. Interv. 2021, 7, e12155. [Google Scholar] [CrossRef]
- Zhong, Y.; Ma, C.-M.; Shahidi, F. Antioxidant and antiviral activities of lipophilic epigallocatechin gallate (EGCG) derivatives. J. Funct. Foods 2012, 4, 87–93. [Google Scholar] [CrossRef]
- Dierssen, M.; De Lagrán, M.M. DYRK1A (Dual-Specificity Tyrosine-Phosphorylated and -Regulated Kinase 1A): A Gene with Dosage Effect During Development and Neurogenesis. Sci. World J. 2006, 6, 1911–1922. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- López-Otín, C.; Blasco, M.A.; Partridge, L.; Serrano, M.; Kroemer, G. The Hallmarks of Aging. Cell 2013, 153, 1194–1217. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Author (Year) | Objective | Type of Study and Sample Size | Inclusion and Exclusion Criteria | Interventions | Methodology | Main Outcomes | Conclusions | Quality of Evidence |
---|---|---|---|---|---|---|---|---|
Huntington disease | ||||||||
Beasley et al., 2019 [51] | To investigate the ability of EGCG to inhibit aggregation in the presence of lipid vesicles (POPC or TBLE) | In-vitro model | Glutathione S-transferase (GST)-htt-exon1(46Q) fusion protein purified from E. coli | EGCG co-incubation with -exon1(46Q) | POPC or TBLE lipid vesicles preparation. ThT aggregation assays Image acquisition by atomic force microscopy Vesicle-binding assay | ↓ exon 1(46Q) aggregation (p < 0.01) ↓ exon 1(46Q) fibril formation and aggregation in the presence of lipid vesicles (POPC or TBLE) (p < 0.01) | EGCG inhibitory effect on htt aggregation process persists in the presence of lipid vesicles | ++ |
Ehrnhoefer et al., 2006 [52] | To analyze the dose-dependent effect of EGCG on mutant htt exon 1 protein aggregation | In-vitro model | Yeast cultures and transgenic HD flies overexpressing a pathogenic htt exon 1 protein | Mutant GST-tagged htt exon 1 fusion protein with 51 glutamines (GST-HDQ51) was incubated with green tea polyphenols (GCG, GC, EGC, and EGCG) | Dot blot assays Atomic force microscopy studies | ↓ mutant htt exon 1 protein aggregation, polyQ-mediated htt protein aggregation and cytotoxicity ↓ photoreceptor degeneration ↑ motor function | EGCG acts as modulator of htt exon 1 misfolding and oligomerization reducing polyQ-mediated toxicity in vivo | ++ |
Varga et al., 2018 [53] | To study the effects of green tea on HD pathogenesis | Experimental transgenic Drosophila model of HD n = 356 (HttQ93on CM), n = 345 (HttQ93 on GTM), n = 122 (HttQ20on CM), n = 118 (HttQ20 on GTM) | Inclusion: Neuron-specific GAL4 driver strain w P{GawB}elavC155 males and w; UAS-Httex1p-Q93 or control w; UAS-Httex1p-Q20 females | Drosophila strain exposure to green tea medium | Eclosion, survival, climbing assay and pseudopupil tests Polyphenol content determination by Folin-Ciocalteau method Huntingtin protein level measurement by Immunoblot | Mutant huntingtin expressing Drosophila exposed to green tea presented: =Viability (p < 0.001) ↓ Neurodegeneration (p < 0.001) ↑ Longevity (p < 0.001) | Green tea consumption might modulate symptoms of HD. | ++ |
Multiple sclerosis | ||||||||
Bellmann-Strobl et al., 2021 [54] | To evaluate the safety and efficacy of EGCG + GA in RRMS patients | Prospective, double blind, Ph II, randomized controlled trial. n = 122 | Inclusion: age: 18–60, EDSS score 0–6.5, stable with GA. Exclusion: other forms of MS, major diseases, laboratory abnormalities, other medication | 800 mg oral EGCG/day or placebo for 18 months | Neurologic assessments, safety monitoring, laboratory exams and MRI at baseline and every three months | No differences with EGCG + GA treatment on brain MRI and on SAE and AE in comparison to placebo + GA. | No superiority of EGCG + GA compared to placebo + GA in MRI changes or clinical disease activity. EGCG at 800 mg/day was safe | ++++ |
Rust et al., 2021 [55] | To evaluate if treatment with EGCG affects progression of brain atrophy and its safety on primary and SPMS | Prospective, double blind, phase II, randomized controlled trial. n = 61 | Inclusion: age: 18–65, EDSS score: 3–8, relapse-free period of minimum 30 days. No MS modifying therapy. Exclusion: RRMS, major diseases, laboratory abnormalities, hepatotoxic medications. | Increasing doses of EGCG until reaching 1200 mg or placebo, for 36 months | Neurologic assessments at baseline and every six months. Safety monitoring and laboratory exams every 2–3 months. MRI at baseline and every year evaluating PBF | No differences on PBF decrease, SAE and AE between groups. | No differences on EGCG group didn’t on brain atrophy compared with placebo. EGCG 1200 mg/day was safe | ++++ |
Mossakowski et al., 2015 [56] | To investigate the role of oxidative stress on neuronal degeneration and on antioxidant therapy | Murine and human: n = 6. Groups: RRMS GA, RRMS GA + EGCG, RRMS untreated, CIS, controls, SPMS) | Patients with RRMS, SPMS, or CIS | EGCG 600 mg or placebo in groups of RRMS + GA. EAE murine model of MS | Examine the oxidation of NADH and NADPH in mononuclear cells with a two-photon laser-scanning microscopy to see activation of NOX enzymes | EGCG + GA ↓ NOX in CD11b + monocytes in MS and EAE. | EGCG counteracted NOX overactivation in MS patients | ++ |
Mähler et al., 2015 [57] | To investigate if EGCG improves energy metabolism and substrate utilization in MS | Randomized, double blind, placebo-controlled, crossover trial n = 18 (eight men) | Inclusion: RRMS, treated with GA for six months. EDSS score: 4.5. Age: 20–60. BMI: 18.5–30.0. Exclusion: other MS forms. Relapses three months before or during the study. Other diseases. Regular caffeine or green tea intake, social drugs | EGCG (600 mg/day) and placebo 12 weeks (four weeks of washout). | Measurements from blood samples, microdialysates from adipose tissue and skeletal muscle, fasting and postprandial EE, FAOx and CHOx rates, at rest or during 40-min of exercise | Working efficiency: placebo: 20 +/− 3 EGCG: 25 +/− 6 (p = 0.004) Postprandial FAOx: Placebo: 8.3 +/− 4.3 EGCG: 8.6 +/− 5.0 (sex differences) | EGCG given to MS patients over 12 weeks improves muscle metabolism during moderate exercise, predominantly in men | ++ |
Lovera et al., 2015 [58] | To evaluate the safety and futility of Polyphenon E treatment | PhI: single group. PhII: randomized double blind placebo-controlled study. n = 10 on phI n = 13 on phII | Inclusion: MS with RRMS or SPMS. six-month stability. Treatment with GA or no treatment in PhI, GA or Interferon β in PhII. Exclusion: Bone marrow ablation or alemtuzumab use. Mitoxantrone, cyclophosphamide, natalizumab or fingolimod use during the past nine months. Other diseases | PhI: Polyphenon E capsules (400 mg of EGCG) twice daily for six months. PhII: Polyphenon E or placebo, same dose for one year | Measure plasma levels. Evaluate adverse events monthly. Evaluate NAA levels at 0, 6, and 12 months using MRI | Polyphenon E: ↑ NAA adjusted creatinine. 5/7 participants had elevated liver enzymes in Ph 2 | 400 mg of EGCG/12 h olyphenon E increased NAA brain levels. Polyphenon E may increase the risk of hepatotoxicity | +++ |
Wang et al., 2012 [59] | Determine the effect and mechanisms of EGCG on EAE development. | Experimental animal model. Four groups, n = 12/group | Specific pathogen-free C57BL/6 female mice. | Diet supplement with 0%, 0.15%, 0.3%, or 0.6% EGCG 30 days, after produce EAE. | Signs were daily scored from day 0 to 30 after EAE induction. Euthanasia and histology and molecular evaluation. | EGCG ↓ symptoms and pathological features in the central nervous system. | EGCG may attenuate EAE autoimmune response. | + |
Sun et al., 2013 [60] | Investigate the mechanism of EGCG on amelioration of EAE | Experimental animal model n = 10/group. | Male C57BL/6 mice, 7 weeks old | EAE induction. When clinical signs start: 300 μg EGCG in 100 μL PBS daily or PBS alone. | Clinical signs evaluation. After death histopathology and molecular evaluation. | EGCG ↓ disease severity in EAE, ↓ brain inflammation and ↓ demyelination damage. | EGCG may be useful for the MS treatment. | + |
Herges et al., 2011 [61] | Evaluate the effect of GA and EGCG in vitro and in a EAE model. | Experimental animal model and in vitro. n = 8/group | 6–8 week old female SJL/L mice | Prevention and treatment with EGCG 300 μg/12 h or vehicle and 50–150 μg GA/24 h from day 9 before EAE production. | Valuation of clinical signs and histological examination. | EGCG + GA ↓ disease onset, ↓ clinical severity and ↓ inflammatory infiltrates. | GA + EGCG may be useful and safe approach for MS. | + |
Semnani et al., 2016 [62]; Semnani et al., 2017 [63] | Study EGCG effects on the PLP and Olig1 expression. | Experimental animal model. n = 60 (6 groups, n = 10) | C57BL/6 male mice, 8 weeks old. | Induction demyielinization with cuprizone. Injection of EGCG 50 mg/Kg/day, PBS, or nothing. | After 2 or 4 weeks, euthanasia and cerebral exam Western Blot or Real-time PCR | EGCG: ↑ PLP and Olig1 expression | EGCG increases PLP and Olig1 expression in the cerebral cortex of this mouse model of MS. | + |
Author (Year) | Objective | Type of Study and Sample Size | Inclusion and Exclusion Criteria | Interventions | Methodology | Main Outcomes | Conclusion | Quality of Evidence |
---|---|---|---|---|---|---|---|---|
Antonio and Druse, 2008 [78] | To investigate the protective effect of EGCG against ethanol-associated apoptosis in rhombencephalic neurons | In-vitro model of rhombencephalic neurons in culture | Culture of rhombencephalic tissue from GD 14 rat fetuses | 24-h ethanol treatment of rhombencephalic neurons with 75 mM ethanol. Co-treatment with 75 mM ethanol and 1 µM EGCG | TUNEL for detection and quantification of apoptotic nuclei | Ethanol ↑ apoptosis in fetal rhombencephalic neurons (p < 0.01). EGCG ↓ apoptotic rhombencephalic neurons (p < 0.01) | The treatment with EGCG provides neuroprotection to the ethanol-treated neurons | + |
Long et al., 2010 [79] | To evaluate the role of oxidative stress in FASD and the effect of EGCG in the prevention of ethanol-induced embryonic damage | Mouse model. Five experimental groups (n = 6 per group) | C57BL/6J pregnant female mice. Five experimental groups: (1) control, (2) ethanol 0.005 mL/g, (3) ethanol 0.01 mL/g, (4) ethanol 0.015 mL/g, (5) ethanol 0.02 mL/g +/−EGCG | Ethanol ip (25%, 0.005–0.02 mL/g) on GD8. EGCG intragastric gavage (200, 300 or 400 mg/Kg/day) on GD7–8 in 0.02 mL/g ethanol exposed group | Morphology assessment (GD10.25): HL, HW and CRL. Analysis of neural marker genes and proteins (GD9.25) by RT-PCR and western blot Study of neural marker genes and proteins. Measure H2O2 and MDA on GD9.25 | Ethanol ↑ growth restriction (HL, HW, and CRL). Ethanol ↓ Otx1 and Sox2. Ethanol ↑ H2O2 and MDA. EGCG ↓ embryo growth restriction EGCG ↑ Otx1 and Sox2 | Embryo growth restriction in FASD is mediated by overproduction of ROS. Ethanol affects neural marker genes and proteins involved in brain development and neural differentiation. EGCG has a protective effect against FASD | + |
Tiwari et al., 2010 [80] | To assess the effect of EGCG on ethanol-induced biochemical alterations and behavioral disorders | Rat model. Five experimental groups (n = 5–8 per group). | Wistar male rat pups (five-day-old neonates). Five experimental groups: (1) control, (2) ethanol administration, (3) ethanol + EGCG administration (50 mg/Kg), (4) ethanol + EGCG administration (100 mg/Kg), (5) EGCG administration (100 mg/Kg) | Randomization of the pups into five experimental groups. Ethanol administration (5 g/Kg, 12% v/v) by intragastric gavage. EGCG (50 and 100 mg/kg). | Behavioral tests: MWM test, memory consolidation test, elevated plus maze task. BAC quantification. Acetylcholinesterase activity measurement. Lipid peroxidation, glutathione, superoxide dismutase, catalase and nitrite, TNF-α and IL-1β estimation by ELISA. NF-κB p65 unit quantification. Caspase-3 colorimetric assay | Ethanol ↓ scores in MWM elevated plus maze task. Ethanol ↑ acetylcholinesterase activity, oxidative-nitrosative stress, cytokines (TNF-α and IL-1β), NF- κB and caspase-3 levels. EGCG ↓ behavioral and biochemical ethanol-induced alterations | Cognitive disorders in FASD are associated with oxidative-nitrosative stress-mediated apoptotic signaling. EGCG is useful in the prevention of FASD-related cognitive impairment | ++ |
Almeida et a., 2021 [81] | To analyze the effects of PAE according two human drinking patterns (Mediterranean vs. binge) on placenta and brain development and evaluate the effect of EGCG treatment on FASD development | Mouse model. Six experimental groups (n = 6 per group) | C57BL/6 pregnant mice. Six experimental groups: (1) control med, (2) EtOH med, (3) EtOH med + EGCG, (4) control bin, (5) EtOH bin, (6) EtOH bin + EGCG | Ethanol administration by intragastric gavage from GD0 to GD19, 10% v/v, 1.5 g/Kg/day or 3 g/Kg/day. EGCG 30 mg/Kg/day by intragastric gavage. Cesarean section at GD19 | Fetal and placental weights. Western blot (VEGF-A, PLGF, VEGF-R1, Nrf2, NeuN, DCX, GFAP and BDNF). Immunohistochemistry (VEGF-A, PLGF, VEGF-R1, NeuN, DCX and BDNF). Immunofluorescence (Nrf2 and GFAP) | Ethanol ↓ fetal growth (growth was negatively correlated with ethanol dose). Ethanol ↓ VEGF-A and ↓ VEGF-R1. Ethanol ↓ Nrf2. Ethanol ↓ NeuN, ↑ DCX and ↓ GFAP. EGCG ↓ ethanol-induced oxidative stress ameliorating FASD manifestations | Any drinking pattern may produce fetal growth, loss of mature neurons, delay in neuronal maturation and disorders in astrocyte differentiation (the highest doses cause to the most severe disturbances) EGCG ameliorates FASD alterations | + |
Author (Year) | Objective | Type of Study and Sample Size | Interventions | Methodology | Main Outcomes | Conclusions | Quality of Evidence |
---|---|---|---|---|---|---|---|
Gu et al., 2020 [50] | To evaluate the effect of EGCG on DYRK1A kinase activity | C57Bl/6J mice overexpressing Dyrk1A (TgBACDyrk1A) model (n = 30) (n = 30) | FontUp administration by oral gavage (25 mg/kg, 50 mg/kg or 75 mg/kg). | Three experimental groups (FontUp administration of 25 mg/kg, 50 mg/kg, or 75 mg/kg) Protein extraction and analysis. Truncated DYRK1A (DYRK1A-ΔC) purification. DYRK1A inhibition analysis HPLC analysis for FontUp polyphenols quantification. Computational molecular docking (ECG) and epicatechin (EC) on DYRK1A kinase activity. | EGCG and ECG ↓ DYRK1A activity EGC and EC = DYRK1A activity. FontUp = liver and cardiac function EGCG crosses blood-brain barrier | Oral FontUp® normalized brain and plasma biomarkers altered in TgBACDyrk1A, without damaging liver and cardiac performances | ++ |
Catuara-Solarz et al., 2016 [96] | To explore the effects of a combined therapy with EE and EGCG on neurological disorders of DS at young age | Experimental Ts65Dn 1–2-month-female mouse models of DS. (WT = 8; TS = 7; WT-EE-EGCG = 7; TS-EE-EGCG = 7) | Green tea extract containing 45% EGCG administrations by oral feeding (EGCG dosage: 0.326 mg/mL, 0.65 mg per day; 30 mg/kg per day during 30 days) | Four experimental groups (WT, TS, WT-EE-EGCG, TS-EE-EGCG). Morris Water Maze Novel object recognition test Quantification of dendritic spine density. Immunohistochemistry for synaptic studies Immunohistochemistry for synaptic modifications studies | EE-EGCG treatment ↑ corticohippocampal-dependent learning and memory. ↑ cornu ammonis 1 (CA1) dendritic spine density. Improvement of excitatory and inhibitory synaptic markers in CA1 and dentate gyrus | EE-EGCG treatment derived cognitive improvements are linked to modulation of neural synapsis at the hippocampus and normalization in dendritic spine density | ++ |
Goodlett et al., 2020 [97] | To test the effects of EGCG on neurobehavioral and skeletal phenotypes in a mouse model | Experimental Ts65Dn mouse models of DS (Euploid PB = 13 Euploid EGCG = 11 Ts65Dn PB = 12 Ts65Dn EGCG = 8 | Three-week EGCG oral gavage therapy (200 mg/kg/day) | Multivariate concentric square field maze. BB and MWM. Morris water maze CT imaging HPLC kinase activity assay WB for DYIRK1A protein quantification | ↓ growth in both euploid and trisomic mice =results in on conductual assessment of Ts65Dn mice. Ts65Dn mice. ↓ cortical bone formation and potency in Ts65Dn mice | EGCG has no effects on behavior. High-dose EGCG caused deleterious effects on growth and skeletal phenotypes | ++ |
Stringer et al., 2015 [98] | To evaluate the therapeutic effects of EGCG on locomotor activity and learning and memory on a mice model of DS | Experimental Ts65Dn mouse models of DS and euploid treatments: Ts65Dn—EGCG n = 8, water n = 9; Euploid—EGCG n = 12, water n = 13 | Ts65Dn or euploid mouse models were randomized to receive EGCG + H3PO4 (n = 10/n = 8), EGCG (n = 14/n = 8), water + H3PO4 (n = 9/n = 13) or water (n = 9/n = 17) for three months | 20 mg/kg/day EGCG HPLC/MS degradation analysis Locomotor activity assessment, NOR, DNMP, BB and MWM. Dyrk1A kinase activity assay | Ts65Dn ↑ locomotive performance Ts65Dn↓ novel object detection, balance beam and spatial learning and memory. EGCG did not ameliorate performance of the Ts65Dn mice on these tasks | Oral EGCG treatment up to 20 mg/kg/day did not improve learning and memory performance in adolescent Ts65Dn mice | ++ |
Souchet et al., 2015 [95] | To investigate the consequences of one-month therapy with EGCG-containing products on excitation/inhibition balance in DS adults. | Experimental adult mBACtgDyrk1a mice Transgenic (TG) = 10 Wild-type (WT) = 10 Treated transgenic (TG*) = 10 | Administration of 225 mg/kg/day of Polyphenon 60 for four months in adult mice mBACtgDyrk1a or for six weeks before and during behavioral analysis in Ts65Dn | Indicators of GABAergic and glutaminergic synaptic routes were evaluated by immunoblot Y-maze paradigm to assess working memory | ↓ GABA in cortex, hippocampus and cerebellum ↑ GLUR1, NR1, NR2a, VGLUT1 in cortex ↑ Ratio of PCAMKII/CAMKII in the hippocampus ↑ Short term memory | EGCG therapy restores excitation/inhibition balance disorders in DS adults | ++ |
Stringer et al., 2017 [99] | To investigate if an EGCG would yield improvements in either cognitive or skeletal deficits. | Experimental Ts65Dn mouse models of DS Eup + water = 19 Eup + EGCG = 18 Tsg + water = 13 Tsg + EGCG = 15 | Oral administration of EGCG from PD24 to PD68 | MCSF, NOR, BB, and MWM. CT, HPLC kinase activity assay and WB for Dyrk1A quantification | =growth ↓ Kinase activity (cerebellum) =cognitive deficits ↑ Adverse changes in skeleton | No beneficial therapeutic effects were seen with EGCG intake on behavior. Caused detrimental skeletal effects in Ts65Dn mice. | ++ |
Catuara et al., 2015 [100] | To investigate the effect of coadjuvant treatment with EGCG and EE on the cognitive decline in DS. | Experimental Ts65Dn mouse models of DS 5–6 months old female mice WT = 10; TS = 11; WT-EE = 14; TS-EE = 11; WT-EGCG = 11; TS-EGCG = 9; WT-EE-EGCG = 12; TS-EE-EGCG = 8. | Administration of green tea extract including 45% EGCG (0.326 mg/mL, 0.9 mg per day; 30 mg/kg per day) during 30 days | MWM for hippocampal-dependent learning and memory evaluation. | EGCG or EE = spatial learning EGCG + EE ↓ learning alterations of middle age Ts65Dn mice and this stratification continued upon treatments | Combining EE and EGCG ameliorates age-related cognitive degeneration in DS | ++ |
Valenti 2013 Italy [101] | To test the capability of EGCG to reestablish the energy in mitochondria and reduce oxidative stress in DS cells | Experimental Cultured lymphoblasts and fibroblasts from DS patients | Lymphoblastoid and fibroblast cells were treated with 20 μM EGCG joined to the cell culture for 24 h | Assessment of mitochondrial ATP production rate, cellular ATP and ROS detection | ↑ mitochondrial complex I and ATP synthase catalytic action ↓ oxidative stress. ↑ mitochondrial biogenesis ↑ Sirt1-dependent PGC-1α deacetylation, NRF-1 and T-FAM protein levels | EGCG antioxidant effects rescues mitochondrial energy and ROS production impairment, prevents overproduction of reactive oxygen species (ROS) and peroxidation of lipid membranes and increases mitochondrial biogenesis | ++ |
Valenti et al., 2016 [102] | To establish possible function of mitochondria in DS cognitive disability | Experimental Ts65Dn mouse models of DS Ts65Dn = 12 WT = 12 | NPCs, cultured for 48 h were complemented with EGCG and RSV, at a concentration of 20 μM and 10 μM, respectively | Measurements of oxygen consumption, mitochondrial ATP production and L-lactate, mitochondrial chain complex activities, ROS production. Immunoblot analysis, quantitative analysis of mtDNA content. | ↑ Oxidative phosphorylation ↑ Mitochondrial biogenesis ↑ Proliferation of NCPs ↑ PGC-1α/Sirt1/AMK axis mitochondrial energy, and improved growth of NPCs | EGCG and RSV reactivates mitochondria bioenergetics and biogenesis and promotes NPCs in DS | ++ |
Stagni et al., 2016 [103] | To study the effect of EGCG in hippocampal development and memory performance | Experimental Ts65Dn mouse models of DS EGCG: Euploid n = 40 and Ts65Dn n = 25 NT:Age-matched euploid (n = 53) and Ts65Dn (n = 25) | EGCG daily subcutaneous injection from PN3 to PN15 (25 mg/kg). | Nissl staining Immunohistochemistry (ki67, cleaved caspase-3, BrdU, NeuN, GFAP, Ayn, PSD-95) Western Blot (GSK3β) MWM and Y-maze | =brain and body weight =locomotor activity and learning and memory Short term effects: ↑ Ki67, BrdU, granular cells in hippocampus, SYN, PSD-95 ↓ GSK3β Long term effects: ↓ BrdU/NeuN, BrdU/GFAP, GSK3β =Ki67, SYN, PSD-95 | EGCG rescues hippocampal neurogenesis and synaptic processes but these effects do not persist for a long time. | ++ |
de La Torre et al., 2014 [104] | To explore if EGCG rescues the intellectual disabilities in adult DS | -Experimental Ts65Dn or TgDyrk1A mouse models of DS: WT untreated n = 13/n = 19; TG/Ts65Dn–untreated n = 16/n = 14; WT-EGCG n = 16/n = 22; TG-EGCG/Ts65Dn n = 14/n = 14.14. -A randomized, double blind, placebo-controlled study in DS humans: 13 EGCG group 16 placebo group | Mice were administered EGCG in drinking water for one month (90 mg/mL for a dose of 2–3 mg per day) Pilot study: groups were randomized to receive oral EGCG at dosage of 9 mg/kg/day or placebo over six months | Mouse model: -Water maze for hippocampal-dependent spatial recognition -NOR for learning and memory assessment deficits. -Dyrk1A kinase activity and homocysteine evaluation Pilot study: -Neurophysiological testing -Blood test and ALT, AST, glucose, cholesterol, TG, GSH-Px analysis -HPLC/MS for EGCG determinations | Mouse model: ↓ DYRK1A ↑ homocysteine ↑ Learning and memory Pilot study: =ALT, AST, TG, glucose, GSH-Px ↓ cholesterol ↑ Episodic and learning memory, visual memory recognition | EGCG improved learning and memory disorders in DS, blocking Dyrk1A expression Plasmatic homocysteine are a biomarker of hippocampal DYRK1A activity in human study | ++++ |
de La Torre et al., 2016 [85] | To test if the administration of EGCG would enhance the outcomes of intellectual rehabilitation in young adults with DS | Double blind, placebo-controlled, phase 2, single center trial (TESDAD) in DS humans 43: EGCG and intellectual treatment group 41: placebo and intellectual treatment group Aged 16–34 years | Randomization and EGCG (9 mg/kg per day) or placebo and cognitive rehabilitation for 12 months. Follow- up of 6 months after intervention discontinuation | Intellectual assessment for working memory, executive performance. Homocysteine, Dyrk1A kinase activity, ALT, AST, cholesterol, TG measurements. fMRI and TMS for functional connectivity patterns studies. | ↓ Cholesterol ↑ Homocysteine ↑ Inhibitory control, recognition memory, adaptive behavior =BMI ↑ Brain connectivity | EGCG joined to intellectual rehabilitation for 12 months had greater results than placebo and intellectual rehabilitation at improving visual memorial perception conduct control, and compliant behavior | ++++ |
De Toma et al., 2019 [105] | To compare proteomic changes in DS EE or GTE treated DS individuals | Experimental Ts65Dn male mouse models of DS at thee months 144 animals: 38 NT mice (18 TG, 20 WT); 38 EGCG (18 TG, 21 WT); 36 EE (16 TG, 18 WT); 33 EGCG + EE (16 TG, 17 WT) | EGCG 326.25 mg per capsule mixed with drinking water at 0.33 mg/mL at medium dose of 42 mg/kg per day for one month. | Western Blot for Dyrk1A quantification Mass-spectrometry-based proteomics Liquid chromatography tandem-mass spectrometry NOR | -Dyrk1A overexpression impacted the phosphoproteome in TG hippocampus (mainly proteins plasticity and cognitive-related proteins) -These (phospho-) proteomic changes were rescued by green tea and/or EE | -t Dyrk1A overexpression causes changes in the proteome and phosphoproteome of the hippocampus of transgenic mice -The cognitive enhancer treatments rescued these alterations | ++ |
De Toma et al., 2020 [106] | To study the effects of EGCG, EE and their mixture using proteome, and phosphoproteome analysis in the hippocampi of DS | Experimental Ts65Dn mouse models of DS Five mice per group, randomly chosen (40 total mice) | EGCG: 326.25 mg per capsule. mixed with water at 0.33 mg/mL equivalent to a medium dose of 42 mg/kg per day for one month. | Mass-spectrometry-based proteomics Liquid chromatography tandem-mass spectrometry Western blot. | -Neurocognitive-related GTPase/kinase activity and chromatin proteins were impaired. -EGCG, EE, and their mixture rescued higher than 70% of the phosphoprotein impairment in Ts65Dn, and induced probable beneficial effects | Green tea extracts may restore an appropriate epigenetic profile and reverse the kinome deregulation promoting the cognitive rescue | ++ |
Starbuck et al., 2021 [107] | To investigate the effect of GTE-EGCG for ameliorating facial dysmorphologies associated with DS | Experimental mouse models 55 Ts65Dn Cross sectional study in children 0–18 years old with DS 63 DS 4 mosaics 13 treated with EGCG 207 euploids | High (100 mg/kg/day) or low doses (30 mg/kg/day) of GTE-EGCG, were administered from embryonic Day 9 to Day 29 post-delivery, in mouse models. Children with DS received low doses of EGCG | Morphometric facial analysis evaluation by CT 3D quantitative morphometric measures of the face in mice and photogrammetry in humans | -The smallest GTE-EGCG dose ameliorated facial skeleton characteristics in a mouse model of DS. -In humans, GTE-EGCG administration restored facial dysmorphic features in children with DS if therapy was given over the first 3 years of life. -Greater GTE-EGCG dosing disrupted normal growth and augmented facial dysmorphic features in trisomy and euploid mice | GTE-EGCG modulates facial development with dose-dependent effects, but high doses have potentially detrimental effects observed in mice | +++ |
Author (Year) | Objective | Type of Study and Sample Size | Inclusion and Exclusion Criteria | Interventions | Methodology | Main Outcomes | Conclusion | Quality of Evidence |
---|---|---|---|---|---|---|---|---|
Kesse-Guyot et al., 2012 [108] | To evaluate the association between polyphenol intake and cognitive function, 13 years later | Prospective Observational n = 2574 | 45–60 years old | - | 24 h dietary records every two months for two years. Evaluation of episodic memory, lexical-semantic memory, working memory and mental flexibility | Catechins ↑ scores on language and verbal memory score, and ↓ scores on executive functioning factor | Catechins may be associated with dual effects on the brain | ++ |
Biasibetti et al., 2013 [109] | To explore the association between flavonoidintake and cognitive health | Observational. n = 808 | Inclusion: Individuals living on Catania, Italy. >50 years old. Exclusion: FFQ with unreliable intakes | - | Administration of two FFQ | Higher dietary intake of catechins ↓ impaired cognitive status. (Q4 vs. Q1: OR = 0.24, 95% CI: 0.08, 0.72) | Individuals with higher flavonoid intake may have better cognitive health | ++ |
Mohamed et al., 2013 [114] | To evaluate the cognitive effects of OPLE in humans and rats | Human: placebo -controlled trial. n = 15. Animal (n = 8): normal rat groups: control, OPLE, captopril. NO-deficient rat groups: vehicle, OPLE, captopril | Young, healthy, adult human volunteers. Rats: 16-week-old male Wistar Kyoto | Human: 500 mg OPLE/day or placebo. Rat: 500 mg OPLE/kg day or L-NAME (60 mg/L), captopril (100 mg/kg day) | Human: Cognitive tests at 0, 1, and 2 months evaluating short-term memory, processing speed, spatial visualization ability, and language skills. Rat: evaluation of hippocampal neuron damage. Evaluation of SOD activity | OPLE: ↑ Short term memory, ↑ spatial visualization ability, ↑ processing speed. OPLE ↑ hippocampal pyramidal and granule viable cells in NO-deficient rats | OPLE showed neuroprotection in NO-deficient rats, and a cognitive amelioration in humans | ++ |
Scholey et al., 2012 [110] | To investigate if EGCG modulates brain activity and self-reported mood | Double blind, placebo controlled crossover study. n = 31 | Inclusion: normal BMI, right-handed, no drugs or natural therapies, English speaking, healthy population | Administration of Teavigo (94% EGCG) 300 mg | Mood and an EEG before and 120 min after EGCG administration or placebo | EGCG ↑ alpha, beta and theta activity on the EEG, ↑ self-rated calmness, ↓ self-rated stress | EGCG group may have been in a morerelaxed and attentive state | +++ |
Emma et al., 2012 [111] | To elucidate the effects of EGCG on cognitive performance, mood and CBF in humans | Double blind, placebo-controlled, crossover study. n = 27 | Exclusion: drugs and herbal extracts/food supplements, head injury, neurological disorder, allergies or intolerances, more than 600 mg/day of caffeine | Administration of placebo and EGCG at two doses (135 and 270 mg) | After 45 min, performed cognitive tasks and measured the CBF and hemodynamics in the frontal cortex using near-infrared spectroscopy | No differences on cognitive performance and mood with EGCG use. EGCG 135 mg ↓ CBF in the frontal cortex. No differences with 270 mg EGCG | EGCG 135 mg modulated CBF | +++ |
Dietz et al., 2017 [113] | To investigate the effects of matcha on mood and cognitive performance | Randomized, placebo-controlled, single blind, crossover study. n = 27 | Inclusion: 100–400 mg caffeine per day. Exclusion: health problems, pregnancy, lactation, allergies/intolerances, drugs, night shift work, abnormal sleep pattern | Administration of matcha tea or bar with 4.0 g of matcha tea (67 mg L-theanine, 280 mg EGCG, 136 mg caffeine) or placebo | Preformation of CDR test battery and POMS test before and 60 min after administration | With matcha products ↑ basic attention abilities, ↑ psychomotor speed response. No mood changes | Matcha tea induce slight effects on episodic secondary memory and speed of attention | ++ |
Liu et al., 2018 [112] | To evaluate if green tea extract affects working memory in young and older women | Single blind, placebo-controlled, crossover design. n = 20 | Inclusion: Age: 18–30 or 50–70, Caucasian, healthy, normal BMI, non-smoking, waist circumference < 35 inches. Exclusion: pregnancy or lactating, use of green tea products, medications, laboratory test alterations | Decaffeinated GTE 5.4g and placebo, one-week washout period | Working memory tests. Blood extraction and serum analysis of MDA concentration and total TEAC | GTE ↑ reading span performance in older women, no differences on younger group. No differences on plasma concentration of TEAC and MDA | Acute administration of GTE may improve 50–63 aged woman working memory | ++ |
Ide et al., 2016 [5] | To examine if green tea intake modifies cognitive dysfunction | Double blind, randomized controlled study. n= 33 | Inclusion: age: ≥ 50, ability to orally ingest powders, no supplements, MMSE-J score < 28. Exclusion: tea allergy, severe diseases | Consumption of 2.0 g/day of green tea (220.2 mg of catechins) or placebo powder for one year | MMSE-J performance every 3 months. Laboratory tests | No changes of MMSE-J score after o ne year comparted with placebo. Green tea group: ↓ MDA-modified low-density lipoprotein | Green tea consumption may not affect cognitive function but prevent oxidative stress increase in elderly population | ++++ |
Baba et al., 2020 [115] | To investigate the effect of decaffeinated GTC on cognitive function | Double blind, randomized, controlled study. n = 52 | Inclusion: age: 50–69, self-assessed cognitive decline, MMSE-J score > 24, non-smokers. Exclusion: serious diseases, medication or supplements, extremely unbalanced diets, irregular habits, psychiatric problems or alcoholism | Placebo or GTC capsules (336.4 mg) per day for 12 weeks | MMSE-J and blood biomarkers measured initially and 12 weeks after ingestion. Cognitrax test battery before starting, after first dose, and after 12 weeks of daily administration | Catechin group after 12 weeks: ↑ Working Memory Tasks. No differences on memory tasks, Facial Expression Recognition Tasks, Visual Information Processing Tasks, Motor Function Tasks and blood Biomarkers | GTC may improve working memory | ++++ |
Sakurai et al., 2020 [116] | To evaluate if matcha tea extract affect cognitive function or impulsivity in the elderly | Randomized, double blind, placebo-controlled trial, n = 54 | Inclusion: age: < 60. No diagnosis of dementia and MCI | 3 g Matcha or placebo powder per day, for 12 weeks | Evaluation of cognitive (MoCA and MMSE) and memory function tests (WMS-DR) and assessment of Impulsivity (BIS-11) | No changes on cognitive test, memory and impulsivity scores. On the women subgroup ↑ MoCA in the matcha group | Daily Matcha Green Tea consumption may have protect against cognitive decline in elderly women | ++++ |
Unno et al., 2020 [117] | To examine which GTCdose is necessary to prevent age-related cognitive decline | Experimental animal. n = 180 mice/six groups | Four-week-old male SAMP10 mice | Oral administration of GTC intake of 0, 1, 5, 15, 30, 60 mg/kg depending on the group | Memory and working memory tests. Euthanasia at 2, 6, 12 month and prefrontal cortex, hippocampus and blood samples. Exam of qRT-PCR evaluating Egr2, Nr4a1, Fos, Arc, Egr1, Npas4, and Cyr61 genes | ↑ Long-term memory on the group with 60 mg/kg of GTC. ↑ Working memory on the groups with 30 and 60mg/kg. ↑Nr4a, Fos, Egr1, Npas4 and Cyr61 | GTC suppressed age-related cognitive decline | + |
Ramis et al., 2020 [118] | To study the effect of GTE on brain and cognitive status of old rats | Experimental animal. n = 16 | Old male rats (18 months) | 20 mg/kg/day of IP poliphenon-60 or catechin or vehicle for 28 days | Behavioral tests. Exam of the activity of TPH and TH. Euthanasia and Western Blot Analysis | ↑ working memory and episodic memory. ↑ noradrenergic, serotonergic and dopaminergic system. ↓ age associated neuroinflammation by modulating SIRT | Consumption of green tea delays cerebral senesce on rats | + |
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
Sebastiani, G.; Almeida-Toledano, L.; Serra-Delgado, M.; Navarro-Tapia, E.; Sailer, S.; Valverde, O.; Garcia-Algar, O.; Andreu-Fernández, V. Therapeutic Effects of Catechins in Less Common Neurological and Neurodegenerative Disorders. Nutrients 2021, 13, 2232. https://doi.org/10.3390/nu13072232
Sebastiani G, Almeida-Toledano L, Serra-Delgado M, Navarro-Tapia E, Sailer S, Valverde O, Garcia-Algar O, Andreu-Fernández V. Therapeutic Effects of Catechins in Less Common Neurological and Neurodegenerative Disorders. Nutrients. 2021; 13(7):2232. https://doi.org/10.3390/nu13072232
Chicago/Turabian StyleSebastiani, Giorgia, Laura Almeida-Toledano, Mariona Serra-Delgado, Elisabet Navarro-Tapia, Sebastian Sailer, Olga Valverde, Oscar Garcia-Algar, and Vicente Andreu-Fernández. 2021. "Therapeutic Effects of Catechins in Less Common Neurological and Neurodegenerative Disorders" Nutrients 13, no. 7: 2232. https://doi.org/10.3390/nu13072232
APA StyleSebastiani, G., Almeida-Toledano, L., Serra-Delgado, M., Navarro-Tapia, E., Sailer, S., Valverde, O., Garcia-Algar, O., & Andreu-Fernández, V. (2021). Therapeutic Effects of Catechins in Less Common Neurological and Neurodegenerative Disorders. Nutrients, 13(7), 2232. https://doi.org/10.3390/nu13072232