Natural Compounds for Alzheimer’s Disease Therapy: A Systematic Review of Preclinical and Clinical Studies
"> Figure 1
<p>Schematic representation of the several mechanisms associated with Alzheimer’s Disease (AD) therapy. Down and up oriented arrows indicate the decrease and the increase of the fenomena, respectively.</p> "> Figure 2
<p>Chemical structures of: (<b>a</b>) vitamin C, (<b>b</b>) vitamin E, (<b>c</b>) vitamin D<sub>3</sub> and (<b>d</b>) vitamin D<sub>2</sub>.</p> "> Figure 3
<p>Chemical structure of docosahexaenoic acid (DHA).</p> "> Figure 4
<p>Chemical structure of homotaurine.</p> "> Figure 5
<p>Chemical structure of huperzine A.</p> "> Figure 6
<p>Chemical structure of bryostatin.</p> "> Figure 7
<p>Chemical structure of melatonin.</p> "> Figure 8
<p>Chemical structure of resveratrol.</p> "> Figure 9
<p>Chemical structure of nicotine.</p> "> Figure 10
<p>Chemical structure of curcumin.</p> "> Figure 11
<p>Number of natural products studied in different development phases.</p> ">
Abstract
:1. Introduction
2. Natural Compounds in Clinical Trials and Their Effects on AD
2.1. Bioactive Compounds
2.2. Natural Extracts and Other Natural Products
3. Preclinical In Vivo Studies of Natural Compounds and Their Effects on AD
3.1. Bioactive Compounds
3.2. Natural Extracts and Other Natural Products
4. Preclinical In Vitro Studies of Natural Compounds and Their Effects on AD
5. Discussion
6. Conclusions
Author Contributions
Funding
Conflicts of Interest
Abbreviations
Aβ | β-amyloid |
ACH | Amyloid cascade hypothesis |
AD | Alzheimer’s disease |
APP | Amyloid precursor protein |
BBB | Blood-brain barrier |
DDS | Drug delivery systems |
DHA | Docosahexanoic acid |
EGCG | Epigallocatechin gallate |
NDGA | Nordihydroguaiaretic acid |
NFTs | Neurofibrillary tangles |
PGG | 1,2,3,4,6-Penta-O-galloyl-β-d-glucopyranose |
ROS | Reactive oxygen species |
References
- Reitz, C.; Mayeux, R. Alzheimer disease: Epidemiology, diagnostic criteria, risk factors and biomarkers. Biochem. Pharmacol. 2014, 88, 640–651. [Google Scholar] [CrossRef] [Green Version]
- Kumar, A.; Singh, A.; Ekavali. A review on Alzheimer’s disease pathophysiology and its management: An update. Pharmacol. Rep. 2015, 67, 195–203. [Google Scholar] [CrossRef]
- Solomon, A.; Mangialasche, F.; Richard, E.; Andrieu, S.; Bennett, D.A.; Breteler, M.; Fratiglioni, L.; Hooshmand, B.; Khachaturian, A.S.; Schneider, L.S.; et al. Advances in the prevention of Alzheimer’s disease and dementia. J. Int. Med. 2014, 275, 229–250. [Google Scholar] [CrossRef] [Green Version]
- Selkoe, D.J.; Hardy, J. The amyloid hypothesis of Alzheimer’s disease at 25 years. EMBO Mol. Med. 2016, 8, 595–608. [Google Scholar] [CrossRef]
- Dawkins, E.; Small, D.H. Insights into the physiological function of the β-amyloid precursor protein: Beyond Alzheimer’s disease. J. Neurochem. 2014, 129, 756–769. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Z.; Song, M.; Liu, X.; Su Kang, S.; Duong, D.M.; Seyfried, N.T.; Cao, X.; Cheng, L.; Sun, Y.E.; Ping Yu, S.; et al. Delta-secretase cleaves amyloid precursor protein and regulates the pathogenesis in Alzheimer’s disease. Nat. Commun. 2015, 6, 8762. [Google Scholar] [CrossRef] [Green Version]
- Mohamed, T.; Shakeri, A.; Rao, P.P.N. Amyloid cascade in Alzheimer’s disease: Recent advances in medicinal chemistry. Eur. J. Med. Chem. 2016, 113, 258–272. [Google Scholar] [CrossRef] [PubMed]
- Sengupta, U.; Nilson, A.N.; Kayed, R. The Role of Amyloid-β Oligomers in Toxicity, Propagation, and Immunotherapy. EBioMedicine 2016, 6, 42–49. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Verma, M.; Vats, A.; Taneja, V. Toxic species in amyloid disorders: Oligomers or mature fibrils. Ann. Indian Acad. Neurol. 2015, 18, 138–145. [Google Scholar]
- Swerdlow, R.H.; Burns, J.M.; Khan, S.M. The Alzheimer’s disease mitochondrial cascade hypothesis: Progress and perspectives. Biochim. Biophys. Acta (BBA) Mol. Basis Dis. 2014, 1842, 1219–1231. [Google Scholar] [CrossRef]
- Barage, S.H.; Sonawane, K.D. Amyloid cascade hypothesis: Pathogenesis and therapeutic strategies in Alzheimer’s disease. Neuropeptides 2015, 52, 1–18. [Google Scholar] [CrossRef] [PubMed]
- Heneka, M.T.; Carson, M.J.; Khoury, J.E.; Landreth, G.E.; Brosseron, F.; Feinstein, D.L.; Jacobs, A.H.; Wyss-Coray, T.; Vitorica, J.; Ransohoff, R.M.; et al. Neuroinflammation in Alzheimer’s disease. Lancet Neurol. 2015, 14, 388–405. [Google Scholar] [CrossRef]
- Huang, W.J.; Zhang, X.; Chen, W.W. Role of oxidative stress in Alzheimer’s disease. Biomed. Rep. 2016, 4, 519–522. [Google Scholar] [CrossRef] [PubMed]
- Karch, C.M.; Cruchaga, C.; Goate, Alison M. Alzheimer’s Disease Genetics: From the Bench to the Clinic. Neuron 2014, 83, 11–26. [Google Scholar] [CrossRef]
- Chin-Chan, M.; Navarro-Yepes, J.; Quintanilla-Vega, B. Environmental pollutants as risk factors for neurodegenerative disorders: Alzheimer and Parkinson diseases. Front. Cell. Neurosci. 2015, 9, 124. [Google Scholar] [CrossRef]
- Karch, C.M.; Goate, A.M. Alzheimer’s Disease Risk Genes and Mechanisms of Disease Pathogenesis. Biol. Psychiatry 2015, 77, 43–51. [Google Scholar] [CrossRef] [PubMed]
- Barbara, G.; Joana, A.L.; Manuel, A.N.C.; Maria do Carmo, P. The Potential Effect of Fluorinated Compounds in the Treatment of Alzheimer’s Disease. Curr. Pharm. Des. 2015, 21, 5725–5735. [Google Scholar]
- Loureiro, J.A.; Crespo, R.; Börner, H.; Martins, P.M.; Rocha, F.A.; Coelho, M.; Pereira, M.C.; Rocha, S. Fluorinated beta-sheet breaker peptides. J. Mater. Chem. B 2014, 2, 2259–2264. [Google Scholar] [CrossRef] [Green Version]
- Folch, J.; Petrov, D.; Ettcheto, M.; Abad, S.; Sánchez-López, E.; García, M.L.; Olloquequi, J.; Beas-Zarate, C.; Auladell, C.; Camins, A. Current Research Therapeutic Strategies for Alzheimer’s Disease Treatment. Neural Plast. 2016, 2016, 15. [Google Scholar] [CrossRef] [PubMed]
- Loureiro, J.A.; Gomes, B.; Fricker, G.; Coelho, M.A.N.; Rocha, S.; Pereira, M.C. Cellular uptake of PLGA nanoparticles targeted with anti-amyloid and anti-transferrin receptor antibodies for Alzheimer’s disease treatment. Colloids Surf. B Biointerfaces 2016, 145, 8–13. [Google Scholar] [CrossRef] [PubMed]
- Loureiro, J.A.; Gomes, B.; Fricker, G.; Cardoso, I.; Ribeiro, C.A.; Gaiteiro, C.; Coelho, M.A.N.; Pereira, M.D.C.; Rocha, S. Dual ligand immunoliposomes for drug delivery to the brain. Colloids Surf. B Biointerfaces 2015, 134, 213–219. [Google Scholar] [CrossRef]
- Loureiro, J.A.; Gomes, B.; Coelho, M.A.; do Carmo Pereira, M.; Rocha, S. Targeting nanoparticles across the blood-brain barrier with monoclonal antibodies. Nanomedicine (Lond.) 2014, 9, 709–722. [Google Scholar] [CrossRef]
- Silva, T.; Reis, J.; Teixeira, J.; Borges, F. Alzheimer’s disease, enzyme targets and drug discovery struggles: From natural products to drug prototypes. Ageing Res. Rev. 2014, 15, 116–145. [Google Scholar] [CrossRef]
- Dey, A.; Bhattacharya, R.; Mukherjee, A.; Pandey, D.K. Natural products against Alzheimer’s disease: Pharmaco-therapeutics and biotechnological interventions. Biotechnol. Adv. 2017, 35, 178–216. [Google Scholar] [CrossRef]
- David, B.; Wolfender, J.-L.; Dias, D.A. The pharmaceutical industry and natural products: Historical status and new trends. Phytochem. Rev. 2015, 14, 299–315. [Google Scholar] [CrossRef]
- Asha, H. A Review: Natural Compounds as Anti-Alzheimer´s Disease Agents. Curr. Nutr. Food Sci. 2017, 13, 247–254. [Google Scholar]
- Rasool, M.; Malik, A.; Qureshi, M.S.; Manan, A.; Pushparaj, P.N.; Asif, M.; Qazi, M.H.; Qazi, A.M.; Kamal, M.A.; Gan, S.H.; et al. Recent Updates in the Treatment of Neurodegenerative Disorders Using Natural Compounds. Evid. Based Complement. Altern. Med. 2014, 2014, 979730. [Google Scholar] [CrossRef]
- Andrade, S.; Ramalho, M.J.; Loureiro, J.A.; Pereira, M.C. Interaction of natural compounds with biomembrane models: A biophysical approach for the Alzheimer’s disease therapy. Colloids Surf. B Biointerfaces 2019, 180, 83–92. [Google Scholar] [CrossRef]
- Butler, M.S.; Robertson, A.A.; Cooper, M.A. Natural product and natural product derived drugs in clinical trials. Nat. Prod. Rep. 2014, 31, 1612–1661. [Google Scholar] [CrossRef]
- Doig, A.J.; Derreumaux, P. Inhibition of protein aggregation and amyloid formation by small molecules. Curr. Opin. Struct. Biol. 2015, 30, 50–56. [Google Scholar] [CrossRef]
- Baranello, R.J.; Bharani, K.L.; Padmaraju, V.; Chopra, N.K.; Lahiri, D.H.; Greig, N.A.; Pappolla, M.; Sambamurti, K. Amyloid-Beta Protein Clearance and Degradation (ABCD) Pathways and their Role in Alzheimer’s Disease. Curr. Alzheimer Res. 2015, 12, 32–46. [Google Scholar] [CrossRef]
- Wischik, C.M.; Harrington, C.R.; Storey, J.M.D. Tau-aggregation inhibitor therapy for Alzheimer’s disease. Biochem. Pharmacol. 2014, 88, 529–539. [Google Scholar] [CrossRef]
- Morales, I.; Guzmán-Martínez, L.; Cerda-Troncoso, C.; Farías, G.A.; Maccioni, R.B. Neuroinflammation in the pathogenesis of Alzheimer’s disease. A rational framework for the search of novel therapeutic approaches. Front. Cell. Neurosci. 2014, 8, 112. [Google Scholar] [CrossRef]
- Ahmed, T.; Javed, S.; Javed, S.; Tariq, A.; Šamec, D.; Tejada, S.; Nabavi, S.F.; Braidy, N.; Nabavi, S.M. Resveratrol and Alzheimer’s Disease: Mechanistic Insights. Mol. Neurobiol. 2017, 54, 2622–2635. [Google Scholar] [CrossRef]
- Miller, B.J.; Whisner, C.M.; Johnston, C.S. Vitamin D Supplementation Appears to Increase Plasma Aβ 40 in Vitamin D Insufficient Older Adults: A Pilot Randomized Controlled Trial. J. Alzheimer’s Dis. 2016, 52, 843–847. [Google Scholar] [CrossRef]
- SanMartín, C.D.; Henriquez, M.; Chacon, C.; Ponce, D.P.; Salech, F.; Rogers, N.K.; Behrens, M.I. Vitamin D Increases Aβ140 Plasma Levels and Protects Lymphocytes from Oxidative Death in Mild Cognitive Impairment Patients. Curr. Alzheimer Res. 2018, 15, 561–569. [Google Scholar] [CrossRef]
- Annweiler, C.; Herrmann, F.R.; Fantino, B.; Brugg, B.; Beauchet, O. Effectiveness of the Combination of Memantine Plus Vitamin D on Cognition in Patients With Alzheimer Disease: A Pre-Post Pilot Study. Cogn. Behav. Neurol. 2012, 25, 121–127. [Google Scholar] [CrossRef]
- Galasko, D.R.; Peskind, E.; Clark, C.M.; et al. Antioxidants for alzheimer disease: A randomized clinical trial with cerebrospinal fluid biomarker measures. Arch. Neurol. 2012, 69, 836–841. [Google Scholar] [CrossRef]
- Kontush, A.; Mann, U.; Arlt, S.; Ujeyl, A.; Lührs, C.; Müller-Thomsen, T.; Beisiegel, U. Influence of vitamin E and C supplementation on lipoprotein oxidation in patients with Alzheimer’s disease. Free Radic. Biol. Med. 2001, 31, 345–354. [Google Scholar] [CrossRef]
- Sano, M.; Ernesto, C.; Thomas, R.G.; Klauber, M.R.; Schafer, K.; Grundman, M.; Woodbury, P.; Growdon, J.; Cotman, C.W.; Pfeiffer, E.; et al. A Controlled Trial of Selegiline, Alpha-Tocopherol, or Both as Treatment for Alzheimer’s Disease. N. Engl. J. Med. 1997, 336, 1216–1222. [Google Scholar] [CrossRef]
- Petersen, R.C.; Thomas, R.G.; Grundman, M.; Bennett, D.; Doody, R.; Ferris, S.; Galasko, D.; Jin, S.; Kaye, J.; Levey, A.; et al. Vitamin E and Donepezil for the Treatment of Mild Cognitive Impairment. N. Engl. J. Med. 2005, 352, 2379–2388. [Google Scholar] [CrossRef]
- Dysken, M.W.; Sano, M.; Asthana, S.; et al. Effect of vitamin e and memantine on functional decline in alzheimer disease: The team-ad va cooperative randomized trial. JAMA 2014, 311, 33–44. [Google Scholar] [CrossRef] [PubMed]
- Kryscio, R.J.; Abner, E.L.; Caban-Holt, A.; Lovell, M.; Goodman, P.; Darke, A.K.; Yee, M.; Crowley, J.; Schmitt, F.A. Association of Antioxidant Supplement Use and Dementia in the Prevention of Alzheimer’s Disease by Vitamin E and Selenium Trial (PREADViSE). JAMA Neurol. 2017, 74, 567–573. [Google Scholar] [CrossRef] [Green Version]
- Freund-Levi, Y.; Eriksdotter-Jönhagen, M.; Cederholm, T.; Basun, H.; Faxén-Irving, G.; Garlind, A.; Vedin, I.; Vessby, B.; Wahlund, L.-O.; Palmblad, J. ω-3 Fatty Acid Treatment in 174 Patients With Mild to Moderate Alzheimer Disease: OmegAD Study: A Randomized Double-blind Trial. Arch. Neurol. 2006, 63, 1402–1408. [Google Scholar] [CrossRef] [PubMed]
- Quinn, J.F.; Raman, R.; Thomas, R.G.; Yurko-Mauro, K.; Nelson, E.B.; Van Dyck, C.; Galvin, J.E.; Emond, J.; Jack, C.R.; Weiner, M.; et al. Docosahexaenoic Acid Supplementation and Cognitive Decline in Alzheimer Disease: A Randomized Trial. JAMA 2010, 304, 1903–1911. [Google Scholar] [CrossRef]
- Yurko-Mauro, K.; McCarthy, D.; Rom, D.; Nelson, E.B.; Ryan, A.S.; Blackwell, A.; Salem, N.; Stedman, M. Beneficial effects of docosahexaenoic acid on cognition in age-related cognitive decline. Alzheimer’s Dement. 2010, 6, 456–464. [Google Scholar] [CrossRef] [PubMed]
- Lee, L.K.; Shahar, S.; Chin, A.-V.; Yusoff, N.A.M. Docosahexaenoic acid-concentrated fish oil supplementation in subjects with mild cognitive impairment (MCI): A 12-month randomised, double-blind, placebo-controlled trial. Psychopharmacology 2013, 225, 605–612. [Google Scholar] [CrossRef] [PubMed]
- Gauthier, S.; Aisen, P.S.; Ferris, S.H.; Saumier, D.; Duong, A.; Haine, D.; Garceau, D.; Suhy, J.; Oh, J.; Lau, W.; et al. Effect of tramiprosate in patients with mild-to-moderate alzheimer’s disease: Exploratory analyses of the MRI sub-group of the alphase study. JNHA J. Nutr. Health Aging 2009, 13, 550–557. [Google Scholar] [CrossRef]
- Aisen, P.S.; Gauthier, S.; Ferris, S.H.; Saumier, D.; Haine, D.; Garceau, D.; Duong, A.; Suhy, J.; Oh, J.; Lau, W.C.; et al. Tramiprosate in mild-to-moderate Alzheimer’s disease—A randomized, double-blind, placebo-controlled, multi-centre study (the Alphase Study). Arch. Med. Sci. AMS 2011, 7, 102–111. [Google Scholar] [CrossRef] [PubMed]
- Aisen, P.S.; Saumier, D.; Briand, R.; Laurin, J.; Gervais, F.; Tremblay, P.; Garceau, D. A Phase II study targeting amyloid-β with 3APS in mild-to-moderate Alzheimer disease. Neurology 2006, 67, 1757–1763. [Google Scholar] [CrossRef]
- Martorana, A.; Di Lorenzo, F.; Manenti, G.; Semprini, R.; Koch, G. Homotaurine Induces Measurable Changes of Short Latency Afferent Inhibition in a Group of Mild Cognitive Impairment Individuals. Front. Aging Neurosci. 2014, 6, 254. [Google Scholar] [CrossRef]
- Xu, S.S.; Gao, Z.X.; Weng, Z.; Du, Z.M.; Xu, W.A.; Yang, J.S.; Zhang, M.L.; Tong, Z.H.; Fang, Y.S.; Chai, X.S. Efficacy of tablet huperzine-A on memory, cognition, and behavior in Alzheimer’s disease. Zhongguo Yao Li Xue Bao 1995, 16, 391–395. [Google Scholar]
- Xu, S.S.; Cai, Z.Y.; Qu, Z.W.; Yang, R.M.; Cai, Y.L.; Wang, G.Q.; Su, X.Q.; Zhong, X.S.; Cheng, R.Y.; Xu, W.A.; et al. Huperzine-A in capsules and tablets for treating patients with Alzheimer disease. Zhongguo Yao Li Xue Bao 1999, 20, 486–490. [Google Scholar]
- Rafii, M.S.; Walsh, S.; Little, J.T.; Behan, K.; Reynolds, B.; Ward, C.; Jin, S.; Thomas, R.; Aisen, P.S. A phase II trial of huperzine A in mild to moderate Alzheimer disease. Neurology 2011, 76, 1389–1394. [Google Scholar] [CrossRef] [Green Version]
- Nelson, T.J.; Sun, M.K.; Lim, C.; Sen, A.; Khan, T.; Chirila, F.V.; Alkon, D.L. Bryostatin Effects on Cognitive Function and PKCvarepsilon in Alzheimer’s Disease Phase IIa and Expanded Access Trials. J. Alzheimers Dis. 2017, 58, 521–535. [Google Scholar] [CrossRef]
- Farlow, M.R.; Thompson, R.E.; Wei, L.J.; Tuchman, A.J.; Grenier, E.; Crockford, D.; Wilke, S.; Benison, J.; Alkon, D.L. A Randomized, Double-Blind, Placebo-Controlled, Phase II Study Assessing Safety, Tolerability, and Efficacy of Bryostatin in the Treatment of Moderately Severe to Severe Alzheimer’s Disease. J. Alzheimers Dis. 2018. [Google Scholar] [CrossRef]
- Brusco, L.I.; Márquez, M.; Cardinali, D.P. Monozygotic twins with Alzheimer’s disease treated with melatonin: Case report. J. Pineal Res. 1998, 25, 260–263. [Google Scholar] [CrossRef]
- Brusco, L.I.; Marquez, M.; Cardinali, D.P. Melatonin treatment stabilizes chronobiologic and cognitive symptoms in Alzheimer’s disease. Neuro Endocrinol. Lett. 2000, 21, 39–42. [Google Scholar]
- Furio, A.M.; Brusco, L.I.; Cardinali, D.P. Possible therapeutic value of melatonin in mild cognitive impairment: A retrospective study. J. Pineal Res. 2007, 43, 404–409. [Google Scholar] [CrossRef]
- Wade, A.G.; Farmer, M.; Harari, G.; Fund, N.; Laudon, M.; Nir, T.; Frydman-Marom, A.; Zisapel, N. Add-on prolonged-release melatonin for cognitive function and sleep in mild to moderate Alzheimer’s disease: A 6-month, randomized, placebo-controlled, multicenter trial. Clin. Interv. Aging 2014, 9, 947–961. [Google Scholar]
- Turner, R.S.; Thomas, R.G.; Craft, S.; van Dyck, C.H.; Mintzer, J.; Reynolds, B.A.; Brewer, J.B.; Rissman, R.A.; Raman, R.; Aisen, P.S.; et al. A randomized, double-blind, placebo-controlled trial of resveratrol for Alzheimer disease. Neurology 2015, 85, 1383–1391. [Google Scholar] [CrossRef] [Green Version]
- Zhu, C.W.; Grossman, H.; Neugroschl, J.; Parker, S.; Burden, A.; Luo, X.; Sano, M. A randomized, double-blind, placebo-controlled trial of resveratrol with glucose and malate (RGM) to slow the progression of Alzheimer’s disease: A pilot study. Alzheimer’s Dement. Transl. Res. Clin. Interv. 2018, 4, 609–616. [Google Scholar] [CrossRef]
- Jones, G.M.; Sahakian, B.J.; Levy, R.; Warburton, D.M.; Gray, J.A. Effects of acute subcutaneous nicotine on attention, information processing and short-term memory in Alzheimer’s disease. Psychopharmacol 1992, 108, 485–494. [Google Scholar] [CrossRef]
- Wilson, A.L.; Langley, L.K.; Monley, J.; Bauer, T.; Rottunda, S.; McFalls, E.; Kovera, C.; McCarten, J.R. Nicotine patches in Alzheimer’s disease: Pilot study on learning, memory, and safety. Pharmacol. Biochem. Behav. 1995, 51, 509–514. [Google Scholar] [CrossRef]
- White, H.K.; Levin, E.D. Four-week nicotine skin patch treatment effects on cognitive performance in Alzheimer’s disease. Psychopharmacology (Berl) 1999, 143, 158–165. [Google Scholar] [CrossRef]
- Baum, L.; Lam, C.W.K.; Cheung, S.K.-K.; Kwok, T.; Lui, V.; Tsoh, J.; Lam, L.; Leung, V.; Hui, E.; Ng, C.; et al. Six-Month Randomized, Placebo-Controlled, Double-Blind, Pilot Clinical Trial of Curcumin in Patients With Alzheimer Disease. J. Clin. Psychopharmacol. 2008, 28, 110–113. [Google Scholar] [CrossRef]
- Bachinskaya, N.; Hoerr, R.; Ihl, R. Alleviating neuropsychiatric symptoms in dementia: The effects of Ginkgo biloba extract EGb 761. Findings from a randomized controlled trial. Neuropsychiatr. Dis. Treat. 2011, 7, 209–215. [Google Scholar]
- Ihl, R.; Bachinskaya, N.; Korczyn, A.D.; Vakhapova, V.; Tribanek, M.; Hoerr, R.; Napryeyenko, O.; Group, G.S. Efficacy and safety of a once-daily formulation of Ginkgo biloba extract EGb 761 in dementia with neuropsychiatric features: A randomized controlled trial. Int. J. Geriatr. Psychiatry 2011, 26, 1186–1194. [Google Scholar] [CrossRef]
- Herrschaft, H.; Nacu, A.; Likhachev, S.; Sholomov, I.; Hoerr, R.; Schlaefke, S. Ginkgo biloba extract EGb 761® in dementia with neuropsychiatric features: A randomised, placebo-controlled trial to confirm the efficacy and safety of a daily dose of 240 mg. J. Psychiatr. Res. 2012, 46, 716–723. [Google Scholar] [CrossRef]
- Ihl, R.; Tribanek, M.; Bachinskaya, N.; for the, G.S.G. Efficacy and Tolerability of a Once Daily Formulation of Ginkgo biloba Extract EGb 761® in Alzheimer’s Disease and Vascular Dementia: Results from a Randomised Controlled Trial. Pharmacopsychiatry 2012, 45, 41–46. [Google Scholar] [CrossRef]
- Gavrilova, S.I.; Preuss, U.W.; Wong, J.W.M.; Hoerr, R.; Kaschel, R.; Bachinskaya, N. Efficacy and safety of Ginkgo biloba extract EGb 761® in mild cognitive impairment with neuropsychiatric symptoms: A randomized, placebo-controlled, double-blind, multi-center trial. Int. J. Geriatr. Psychiatry 2014, 29, 1087–1095. [Google Scholar] [CrossRef] [PubMed]
- Akhondzadeh, S.; Shafiee Sabet, M.; Harirchian, M.H.; Togha, M.; Cheraghmakani, H.; Razeghi, S.; Hejazi, S.S.; Yousefi, M.H.; Alimardani, R.; Jamshidi, A.; et al. A 22-week, multicenter, randomized, double-blind controlled trial of Crocus sativus in the treatment of mild-to-moderate Alzheimer’s disease. Psychopharmacology 2010, 207, 637–643. [Google Scholar] [CrossRef] [PubMed]
- Akhondzadeh, S.; Noroozian, M.; Mohammadi, M.; Ohadinia, S.; Jamshidi, A.; Khani, M. Melissa officinalis extract in the treatment of patients with mild to moderate Alzheimer’s disease: A double blind, randomised, placebo controlled trial. J. Neurol. Neurosurg. Psychiatry 2003, 74, 863–866. [Google Scholar] [CrossRef] [PubMed]
- Arab, H.; Mahjoub, S.; Hajian-Tilaki, K.; Moghadasi, M. The effect of green tea consumption on oxidative stress markers and cognitive function in patients with Alzheimer’s disease: A prospective intervention study. CASP J. Int. Med. 2016, 7, 188–194. [Google Scholar]
- Barbagallo, M.; Marotta, F.; Dominguez, L.J. Oxidative stress in patients with Alzheimer’s disease: Effect of extracts of fermented papaya powder. Mediat. Inflamm. 2015. [Google Scholar] [CrossRef] [PubMed]
- Scholey, A.B.; Tildesley, N.T.; Ballard, C.G.; Wesnes, K.A.; Tasker, A.; Perry, E.K.; Kennedy, D.O. An extract of Salvia (sage) with anticholinesterase properties improves memory and attention in healthy older volunteers. Psychopharmacology (Berl) 2008, 198, 127–139. [Google Scholar] [CrossRef]
- De la Rubia Orti, J.E.; Garcia-Pardo, M.P.; Drehmer, E.; Sancho Cantus, D.; Julian Rochina, M.; Aguilar, M.A.; Hu Yang, I. Improvement of Main Cognitive Functions in Patients with Alzheimer’s Disease after Treatment with Coconut Oil Enriched Mediterranean Diet: A Pilot Study. J. Alzheimers Dis. 2018, 65, 577–587. [Google Scholar] [CrossRef]
- Remington, R.; Chan, A.; Lepore, A.; Kotlya, E.; Shea, T.B. Apple juice improved behavioral but not cognitive symptoms in moderate-to-late stage Alzheimer’s disease in an open-label pilot study. Am. J. Alzheimers Dis. Dement. 2010, 25, 367–371. [Google Scholar] [CrossRef]
- Krikorian, R.; Shidler, M.D.; Nash, T.A.; Kalt, W.; Vinqvist-Tymchuk, M.R.; Shukitt-Hale, B.; Joseph, J.A. Blueberry supplementation improves memory in older adults. J. Agric. Food Chem. 2010, 58, 3996–4000. [Google Scholar] [CrossRef]
- Szaniszlo, P.; German, P.; Hajas, G.; Saenz, D.N.; Kruzel, M.; Boldogh, I. New insights into clinical trial for Colostrinin in Alzheimer’s disease. J. Nutr. Health Aging 2009, 13, 235–241. [Google Scholar] [CrossRef]
- Ahmad, A.A.; Shah, S.; Badshah, H.J.; Kim, M.; Ali, T.H.; Yoon, G.H.; Kim, T.B.; Abid, N.; Ur Rehman, S.; Khan, S.O.; et al. Neuroprotection by vitamin C against ethanol -induced neuroinflammation associated neurodegeneration in developing rat brain. CNS Neurol. Disord. Drug Targets 2016, 15, 360–370. [Google Scholar] [CrossRef]
- Sil, S.; Ghosh, T.; Gupta, P.; Ghosh, R.; Kabir, S.N.; Roy, A. Dual Role of Vitamin C on the Neuroinflammation Mediated Neurodegeneration and Memory Impairments in Colchicine Induced Rat Model of Alzheimer Disease. J. Mol. Neurosci. 2016, 60, 421–435. [Google Scholar] [CrossRef]
- Murakami, K.; Murata, N.; Ozawa, Y.; Kinoshita, N.; Irie, K.; Shirasawa, T.; Shimizu, T. Vitamin C restores behavioral deficits and amyloid-beta oligomerization without affecting plaque formation in a mouse model of Alzheimer’s disease. J. Alzheimers Dis. 2011, 26, 7–18. [Google Scholar] [CrossRef]
- Kook, S.Y.; Lee, K.M.; Kim, Y.; Cha, M.Y.; Kang, S.; Baik, S.H.; Lee, H.; Park, R.; Mook-Jung, I. High-dose of vitamin C supplementation reduces amyloid plaque burden and ameliorates pathological changes in the brain of 5XFAD mice. Cell Death Dis. 2014, 5, e1083. [Google Scholar] [CrossRef]
- Rosales-Corral, S.; Tan, D.X.; Reiter, R.J.; Valdivia-Velazquez, M.; Martinez-Barboza, G.; Acosta-Martinez, J.P.; Ortiz, G.G. Orally administered melatonin reduces oxidative stress and proinflammatory cytokines induced by amyloid-beta peptide in rat brain: A comparative, in vivo study versus vitamin C and E. J. Pineal Res. 2003, 35, 80–84. [Google Scholar] [CrossRef]
- Sung, S.; Yao, Y.; Uryu, K.; Yang, H.; Lee, V.M.; Trojanowski, J.Q.; Pratico, D. Early vitamin E supplementation in young but not aged mice reduces Abeta levels and amyloid deposition in a transgenic model of Alzheimer’s disease. FASEB J. 2004, 18, 323–325. [Google Scholar]
- Ramalho, M.J.; Loureiro, J.A.; Gomes, B.; Frasco, M.F.; Coelho, M.A.N.; Pereira, M.C. PLGA nanoparticles for calcitriol delivery. In Proceedings of the 2015 IEEE 4th Portuguese Meeting on Bioengineering, ENBENG, Porto, Portugal, 26–28 Februray 2015. [Google Scholar]
- Ramalho, M.J.; Loureiro, J.A.; Gomes, B.; Frasco, M.F.; Coelho, M.A.N.; Pereira, M.C. PLGA nanoparticles as a platform for vitamin D-based cancer therapy. Beilstein J. Nanotechnol. 2015, 6, 1306–1318. [Google Scholar] [CrossRef] [Green Version]
- Ramalho, M.J.; Coelho, M.A.N.; Pereira, M.C. Nanoparticles for Delivery of Vitamin D: Challenges and Opportunities. In A Critical Evaluation of Vitamin D—Clinical Overview; Gowder, S., Ed.; InTech: Rijeka, Croatia, 2017; p. 11. [Google Scholar]
- Yu, J.; Gattoni-Celli, M.; Zhu, H.; Bhat, N.R.; Sambamurti, K.; Gattoni-Celli, S.; Kindy, M.S. Vitamin D3-enriched diet correlates with a decrease of amyloid plaques in the brain of AbetaPP transgenic mice. J. Alzheimers Dis. 2011, 25, 295–307. [Google Scholar] [CrossRef]
- Grimm, M.; Thiel, A.; Lauer, A.; Winkler, J.; Lehmann, J.; Regner, L.; Nelke, C.; Janitschke, D.; Benoist, C.; Streidenberger, O.; et al. Vitamin D and Its Analogues Decrease Amyloid-β (Aβ) Formation and Increase Aβ-Degradation. Int. J. Mol. Sci. 2017, 18, 2764. [Google Scholar] [CrossRef]
- Landel, V.; Millet, P.; Baranger, K.; Loriod, B.; Féron, F. Vitamin D interacts with Esr1 and Igf1 to regulate molecular pathways relevant to Alzheimer’s disease. Mol. Neurodegener. 2016, 11, 22. [Google Scholar] [CrossRef]
- Morello, M.; Landel, V.; Lacassagne, E.; Baranger, K.; Annweiler, C.; Féron, F.; Millet, P. Vitamin D Improves Neurogenesis and Cognition in a Mouse Model of Alzheimer’s Disease. Mol. Neurobiol. 2018, 55, 6463–6479. [Google Scholar] [CrossRef] [Green Version]
- Kishi, T.; Matsunaga, S.; Oya, K.; Nomura, I.; Ikuta, T.; Iwata, N. Memantine for Alzheimer’s disease: An updated systematic review and meta-analysis. J. Alzheimer’s Dis. 2017, 60, 401–425. [Google Scholar] [CrossRef]
- Youdim, M.B.H.; Edmondson, D.; Tipton, K.F. The therapeutic potential of monoamine oxidase inhibitors. Nat. Rev. Neurosci. 2006, 7, 295. [Google Scholar] [CrossRef]
- Varikasuvu, S.R.; Prasad, V.S.; Kothapalli, J.; Manne, M. Brain Selenium in Alzheimer’s Disease (BRAIN SEAD Study): A Systematic Review and Meta-Analysis. Biol. Trace Elem. Res. 2018. [Google Scholar] [CrossRef]
- Hixson, S.M.; Sharma, B.; Kainz, M.J.; Wacker, A.; Arts, M.T. Production, distribution, and abundance of long-chain omega-3 polyunsaturated fatty acids: A fundamental dichotomy between freshwater and terrestrial ecosystems. Environ. Rev. 2015, 23, 414–424. [Google Scholar] [CrossRef]
- Hashimoto, M.; Hossain, S.; Katakura, M.; Al Mamun, A.; Shido, O. The binding of Aβ1-42 to lipid rafts of RBC is enhanced by dietary docosahexaenoic acid in rats: Implicates to Alzheimer’s disease. Biochim. Et Biophys. Acta Biomembr. 2015, 1848, 1402–1409. [Google Scholar] [CrossRef]
- Lim, G.P.; Calon, F.; Morihara, T.; Yang, F.; Teter, B.; Ubeda, O.; Salem, N., Jr.; Frautschy, S.A.; Cole, G.M. A diet enriched with the omega-3 fatty acid docosahexaenoic acid reduces amyloid burden in an aged Alzheimer mouse model. J. Neurosci. 2005, 25, 3032–3040. [Google Scholar] [CrossRef]
- Grimm, M.O.; Kuchenbecker, J.; Grosgen, S.; Burg, V.K.; Hundsdorfer, B.; Rothhaar, T.L.; Friess, P.; de Wilde, M.C.; Broersen, L.M.; Penke, B.; et al. Docosahexaenoic acid reduces amyloid beta production via multiple pleiotropic mechanisms. J. Biol. Chem. 2011, 286, 14028–14039. [Google Scholar] [CrossRef]
- Hossain, S.; Hashimoto, M.; Katakura, M.; Miwa, K.; Shimada, T.; Shido, O. Mechanism of docosahexaenoic acid-induced inhibition of in vitro Aβ1-42 fibrillation and Aβ1-42-induced toxicity in SH-S5Y5 cells. J. Neurochem. 2009, 111, 568–579. [Google Scholar] [CrossRef]
- Grimm, M.O.W.; Mett, J.; Stahlmann, C.P.; Haupenthal, V.J.; Blümel, T.; Stötzel, H.; Grimm, H.S.; Hartmann, T. Eicosapentaenoic acid and docosahexaenoic acid increase the degradation of amyloid-β by affecting insulin-degrading enzyme1. Biochem. Cell Biol. 2016, 94, 534–542. [Google Scholar] [CrossRef]
- Hashimoto, M.; Shahdat, H.M.; Yamashita, S.; Katakura, M.; Tanabe, Y.; Fujiwara, H.; Gamoh, S.; Miyazawa, T.; Arai, H.; Shimada, T.; et al. Docosahexaenoic acid disrupts in vitro amyloid β1-40 fibrillation and concomitantly inhibits amyloid levels in cerebral cortex of Alzheimer’s disease model rats. J. Neurochem. 2008, 107, 1634–1646. [Google Scholar] [CrossRef]
- Mehdinia, A.; Rostami, S.; Dadkhah, S.; Fumani, N.S. Simultaneous screening of homotaurine and taurine in marine macro-algae using liquid chromatography–fluorescence detection. J. Iran. Chem. Soc. 2017, 14, 2135–2142. [Google Scholar] [CrossRef]
- Santa-Maria, I.; Hernández, F.; Del Rio, J.; Moreno, F.J.; Avila, J. Tramiprosate, a drug of potential interest for the treatment of Alzheimer’s disease, promotes an abnormal aggregation of tau. Mol. Neurodegener. 2007, 2, 17. [Google Scholar] [CrossRef]
- Gervais, F.; Paquette, J.; Morissette, C.; Krzywkowski, P.; Yu, M.; Azzi, M.; Lacombe, D.; Kong, X.; Aman, A.; Laurin, J.; et al. Targeting soluble Abeta peptide with Tramiprosate for the treatment of brain amyloidosis. Neurobiol. Aging 2007, 28, 537–547. [Google Scholar] [CrossRef]
- Kocis, P.; Tolar, M.; Yu, J.; Sinko, W.; Ray, S.; Blennow, K.; Fillit, H.; Hey, J.A. Elucidating the Aβ42 Anti-Aggregation Mechanism of Action of Tramiprosate in Alzheimer’s Disease: Integrating Molecular Analytical Methods, Pharmacokinetic and Clinical Data. Cns. Drugs 2017, 31, 495–509. [Google Scholar] [CrossRef] [Green Version]
- Marambaud, P.; Zhao, H.; Davies, P. Resveratrol promotes clearance of Alzheimer’s disease amyloid-beta peptides. J. Biol. Chem. 2005, 280, 37377–37382. [Google Scholar] [CrossRef]
- Ladiwala, A.R.A.; Lin, J.C.; Bale, S.S.; Marcelino-Cruz, A.M.; Bhattacharya, M.; Dordick, J.S.; Tessier, P.M. Resveratrol selectively remodels soluble oligomers and fibrils of amyloid Aβ into off-pathway conformers. J. Biol. Chem. 2010, 285, 24228–24237. [Google Scholar] [CrossRef]
- Xiao, X.Q.; Zhang, H.Y.; Tang, X.C. Huperzine A attenuates amyloid beta-peptide fragment 25–35-induced apoptosis in rat cortical neurons via inhibiting reactive oxygen species formation and caspase-3 activation. J. Neurosci. Res. 2002, 67, 30–36. [Google Scholar] [CrossRef]
- Peng, Y.; Jiang, L.; Lee, D.Y.; Schachter, S.C.; Ma, Z.; Lemere, C.A. Effects of huperzine A on amyloid precursor protein processing and beta-amyloid generation in human embryonic kidney 293 APP Swedish mutant cells. J. Neurosci. Res. 2006, 84, 903–911. [Google Scholar] [CrossRef]
- Tian, X.R.; Tang, H.F.; Tian, X.L.; Hu, J.J.; Huang, L.L.; Gustafson, K.R. Review of bioactive secondary metabolites from marine bryozoans in the progress of new drugs discovery. Future Med. Chem. 2018, 10, 1497–1514. [Google Scholar] [CrossRef]
- Etcheberrigaray, R.; Tan, M.; Dewachter, I.; Kuiperi, C.; Van der Auwera, I.; Wera, S.; Qiao, L.; Bank, B.; Nelson, T.J.; Kozikowski, A.P.; et al. Therapeutic effects of PKC activators in Alzheimer’s disease transgenic mice. Proc. Natl. Acad. Sci. USA 2004, 101, 11141–11146. [Google Scholar] [CrossRef]
- Schrott, L.M.; Jackson, K.; Yi, P.; Dietz, F.; Johnson, G.S.; Basting, T.F.; Purdum, G.; Tyler, T.; Rios, J.D.; Castor, T.P.; et al. Acute Oral Bryostatin-1 Administration Improves Learning Deficits in the APP/PS1 Transgenic Mouse Model of Alzheimer’s Disease. Curr. Alzheimer Res. 2015, 12, 22–31. [Google Scholar] [CrossRef]
- Shah, S.A.; Khan, M.; Jo, M.-H.; Jo, M.G.; Amin, F.U.; Kim, M.O. Melatonin Stimulates the SIRT1/Nrf2 Signaling Pathway Counteracting Lipopolysaccharide (LPS)-Induced Oxidative Stress to Rescue Postnatal Rat Brain. CNS Neurosci. Ther. 2017, 23, 33–44. [Google Scholar] [CrossRef]
- Ali, T.; Badshah, H.; Kim, T.H.; Kim, M.O. Melatonin attenuates D-galactose-induced memory impairment, neuroinflammation and neurodegeneration via RAGE/NF-KB/JNK signaling pathway in aging mouse model. J. Pineal Res. 2015, 58, 71–85. [Google Scholar] [CrossRef]
- Pappolla, M.; Bozner, P.; Soto, C.; Shao, H.; Robakis, N.K.; Zagorski, M.; Frangione, B.; Ghiso, J. Inhibition of Alzheimer beta-fibrillogenesis by melatonin. J. Biol. Chem. 1998, 273, 7185–7188. [Google Scholar] [CrossRef]
- Rudnitskaya, E.A.; Muraleva, N.A.; Maksimova, K.Y.; Kiseleva, E.; Kolosova, N.G.; Stefanova, N.A. Melatonin Attenuates Memory Impairment, Amyloid-beta Accumulation, and Neurodegeneration in a Rat Model of Sporadic Alzheimer’s Disease. J. Alzheimers Dis. 2015, 47, 103–116. [Google Scholar] [CrossRef]
- Panmanee, J.; Nopparat, C.; Chavanich, N.; Shukla, M.; Mukda, S.; Song, W.; Vincent, B.; Govitrapong, P. Melatonin regulates the transcription of betaAPP-cleaving secretases mediated through melatonin receptors in human neuroblastoma SH-SY5Y cells. J. Pineal Res. 2015, 59, 308–320. [Google Scholar] [CrossRef]
- Andrade, S.; Ramalho, M.J.; Pereira, M.D.C.; Loureiro, J.A. Resveratrol Brain Delivery for Neurological Disorders Prevention and Treatment. Front. Pharmacol. 2018, 9, 1261. [Google Scholar] [CrossRef]
- Andrade, S.; Loureiro, J.A.; Coelho, M.A.N.; Pereira, M.D.C. Interaction studies of amyloid beta-peptide with the natural compound resveratrol. In Proceedings of the 2015 IEEE 4th Portuguese Meeting on Bioengineering (ENBENG), Porto, Portugal, 26–28 Februray 2015; pp. 1–3. [Google Scholar]
- Zhao, H.F.; Li, N.; Wang, Q.; Cheng, X.J.; Li, X.M.; Liu, T.T. Resveratrol decreases the insoluble Aβ1-42 level in hippocampus and protects the integrity of the blood-brain barrier in AD rats. Neuroscience 2015, 310, 641–649. [Google Scholar] [CrossRef]
- Karuppagounder, S.S.; Pinto, J.T.; Xu, H.; Chen, H.L.; Beal, M.F.; Gibson, G.E. Dietary supplementation with resveratrol reduces plaque pathology in a transgenic model of Alzheimer’s disease. Neurochem. Int. 2009, 54, 111–118. [Google Scholar] [CrossRef]
- Ma, X.R.; Sun, Z.K.; Liu, Y.R.; Jia, Y.J.; Zhang, B.A.; Zhang, J.W. Resveratrol improves cognition and reduces oxidative stress in rats with vascular dementia. Neural Regen. Res. 2013, 8, 2050–2059. [Google Scholar]
- He, X.; Li, Z.; Rizak, J.D.; Wu, S.; Wang, Z.; He, R.; Su, M.; Qin, D.; Wang, J.; Hu, X. Resveratrol attenuates formaldehyde induced hyperphosphorylation of tau protein and cytotoxicity in N2a cells. Front. Neurosci. 2017, 10, 598. [Google Scholar] [CrossRef]
- Salomon, A.R.; Marcinowski, K.J.; Friedland, R.P.; Zagorski, M.G. Nicotine inhibits amyloid formation by the beta-peptide. Biochemistry 1996, 35, 13568–13578. [Google Scholar] [CrossRef]
- Srivareerat, M.; Tran, T.T.; Salim, S.; Aleisa, A.M.; Alkadhi, K.A. Chronic nicotine restores normal Aβ levels and prevents short-term memory and E-LTP impairment in Aβ rat model of Alzheimer’s disease. Neurobiol. Aging 2011, 32, 834–844. [Google Scholar] [CrossRef]
- Nordberg, A.; Hellstrom-Lindahl, E.; Lee, M.; Johnson, M.; Mousavi, M.; Hall, R.; Perry, E.; Bednar, I.; Court, J. Chronic nicotine treatment reduces beta-amyloidosis in the brain of a mouse model of Alzheimer’s disease (APPsw). J. Neurochem. 2002, 81, 655–658. [Google Scholar] [CrossRef]
- Ono, K.; Hasegawa, K.; Yamada, M.; Naiki, H. Nicotine breaks down preformed Alzheimer’s beta-amyloid fibrils in vitro. Biol. Psychiatry 2002, 52, 880–886. [Google Scholar] [CrossRef]
- Navarro, E.; Buendia, I.; Parada, E.; León, R.; Jansen-Duerr, P.; Pircher, H.; Egea, J.; Lopez, M.G. Alpha7 nicotinic receptor activation protects against oxidative stress via heme-oxygenase I induction. Biochem. Pharmacol. 2015, 97, 473–481. [Google Scholar] [CrossRef]
- Lahiri, D.K.; Utsuki, T.; Chen, D.; Farlow, M.R.; Shoaib, M.; Ingram, D.K.; Greig, N.H. Nicotine reduces the secretion of Alzheimer’s β-amyloid precursor protein containing β-amyloid peptide in the rat without altering synaptic proteins. Ann. N. Y. Acad. Sci. 2002, 965, 364–372. [Google Scholar] [CrossRef]
- Yang, F.; Lim, G.P.; Begum, A.N.; Ubeda, O.J.; Simmons, M.R.; Ambegaokar, S.S.; Chen, P.P.; Kayed, R.; Glabe, C.G.; Frautschy, S.A.; et al. Curcumin inhibits formation of amyloid beta oligomers and fibrils, binds plaques, and reduces amyloid in vivo. J. Biol. Chem. 2005, 280, 5892–5901. [Google Scholar] [CrossRef]
- Wang, P.; Su, C.; Li, R.; Wang, H.; Ren, Y.; Sun, H.; Yang, J.; Sun, J.; Shi, J.; Tian, J.; et al. Mechanisms and effects of curcumin on spatial learning and memory improvement in APPswe/PS1dE9 mice. J. Neurosci. Res. 2014, 92, 218–231. [Google Scholar] [CrossRef]
- Reddy, P.H.; Manczak, M.; Yin, X.; Grady, M.C.; Mitchell, A.; Kandimalla, R.; Kuruva, C.S. Protective effects of a natural product, curcumin, against amyloid beta induced mitochondrial and synaptic toxicities in Alzheimer’s disease. J. Investig. Med. 2016, 64, 1220–1234. [Google Scholar]
- Liu, Z.-J.; Li, Z.-H.; Liu, L.; Tang, W.-X.; Wang, Y.; Dong, M.-R.; Xiao, C. Curcumin Attenuates Beta-Amyloid-Induced Neuroinflammation via Activation of Peroxisome Proliferator-Activated Receptor-Gamma Function in a Rat Model of Alzheimer’s Disease. Front. Pharmacol. 2016, 7, 261. [Google Scholar] [CrossRef]
- Lin, R.; Chen, X.; Li, W.; Han, Y.; Liu, P.; Pi, R. Exposure to metal ions regulates mRNA levels of APP and BACE1 in PC12 cells: Blockage by curcumin. Neurosci. Lett. 2008, 440, 344–347. [Google Scholar] [CrossRef]
- Xiong, Z.; Hongmei, Z.; Lu, S.; Yu, L. Curcumin mediates presenilin-1 activity to reduce beta-amyloid production in a model of Alzheimer’s Disease. Pharmacol. Rep. PR 2011, 63, 1101–1108. [Google Scholar] [CrossRef]
- Xie, H.; Wang, J.R.; Yau, L.F.; Liu, Y.; Liu, L.; Han, Q.B.; Zhao, Z.; Jiang, Z.H. Catechins and procyanidins of Ginkgo biloba show potent activities towards the inhibition of beta-amyloid peptide aggregation and destabilization of preformed fibrils. Molecules 2014, 19, 5119–5134. [Google Scholar] [CrossRef]
- Belviranlı, M.; Okudan, N. The effects of Ginkgo biloba extract on cognitive functions in aged female rats: The role of oxidative stress and brain-derived neurotrophic factor. Behav. Brain Res. 2015, 278, 453–461. [Google Scholar] [CrossRef]
- Zhang, L.-D.; Ma, L.; Zhang, L.; Dai, J.-G.; Chang, L.-G.; Huang, P.-L.; Tian, X.-Q. Hyperbaric Oxygen and Ginkgo Biloba Extract Ameliorate Cognitive and Memory Impairment via Nuclear Factor Kappa-B Pathway in Rat Model of Alzheimer’s Disease. Chin. Med. J. 2015, 128, 3088–3093. [Google Scholar] [CrossRef]
- Colciaghi, F.; Borroni, B.; Zimmermann, M.; Bellone, C.; Longhi, A.; Padovani, A.; Cattabeni, F.; Christen, Y.; Di Luca, M. Amyloid precursor protein metabolism is regulated toward alpha-secretase pathway by Ginkgo biloba extracts. Neurobiol. Dis. 2004, 16, 454–460. [Google Scholar] [CrossRef]
- Yao, Z.X.; Han, Z.; Drieu, K.; Papadopoulos, V. Ginkgo biloba extract (Egb 761) inhibits β-amyloid production by lowering free cholesterol levels. J. Nutr. Biochem. 2004, 15, 749–756. [Google Scholar] [CrossRef]
- Samarghandian, S.; Azimi-Nezhad, M.; Samini, F.; Farkhondeh, T. The Role of Saffron in Attenuating Age-related Oxidative Damage in Rat Hippocampus. Recent Pat. Food Nutr. Agric. 2016, 8, 183–189. [Google Scholar] [CrossRef]
- Moallem, S.A.; Hariri, A.T.; Mahmoudi, M.; Hosseinzadeh, H. Effect of aqueous extract of Crocus sativus L. (saffron) stigma against subacute effect of diazinon on specific biomarkers in rats. Toxicol. Ind. Health 2012, 30, 141–146. [Google Scholar] [CrossRef] [PubMed]
- Papandreou, M.A.; Kanakis, C.D.; Polissiou, M.G.; Efthimiopoulos, S.; Cordopatis, P.; Margarity, M.; Lamari, F.N. Inhibitory activity on amyloid-beta aggregation and antioxidant properties of Crocus sativus stigmas extract and its crocin constituents. J. Agric. Food Chem. 2006, 54, 8762–8768. [Google Scholar] [CrossRef] [PubMed]
- Dastmalchi, K.; Damien Dorman, H.J.; Oinonen, P.P.; Darwis, Y.; Laakso, I.; Hiltunen, R. Chemical composition and in vitro antioxidative activity of a lemon balm (Melissa officinalis L.) extract. LWT Food Sci. Technol. 2008, 41, 391–400. [Google Scholar] [CrossRef]
- Ozarowski, M.; Mikolajczak, P.L.; Piasecka, A.; Kachlicki, P.; Kujawski, R.; Bogacz, A.; Bartkowiak-Wieczorek, J.; Szulc, M.; Kaminska, E.; Kujawska, M.; et al. Influence of the Melissa officinalis Leaf Extract on Long-Term Memory in Scopolamine Animal Model with Assessment of Mechanism of Action. Evid. Based Complement. Altern. Med. 2016, 2016, 9729818. [Google Scholar] [CrossRef] [PubMed]
- Schimidt, H.L.; Garcia, A.; Martins, A.; Mello-Carpes, P.B.; Carpes, F.P. Green tea supplementation produces better neuroprotective effects than red and black tea in Alzheimer-like rat model. Food Res. Int. 2017, 100, 442–448. [Google Scholar] [CrossRef] [PubMed]
- Li, Q.; Zhao, H.F.; Zhang, Z.F.; Liu, Z.G.; Pei, X.R.; Wang, J.B.; Li, Y. Long-term green tea catechin administration prevents spatial learning and memory impairment in senescence-accelerated mouse prone-8 mice by decreasing Abeta1-42 oligomers and upregulating synaptic plasticity-related proteins in the hippocampus. Neuroscience 2009, 163, 741–749. [Google Scholar] [CrossRef] [PubMed]
- Li, H.; Wu, X.; Wu, Q.; Gong, D.; Shi, M.; Guan, L.; Zhang, J.; Liu, J.; Yuan, B.; Han, G.; et al. Green tea polyphenols protect against okadaic acid-induced acute learning and memory impairments in rats. Nutrition 2014, 30, 337–342. [Google Scholar] [CrossRef] [PubMed]
- Kerem, K.U.; Cengiz, U.M.; Neslihan, T.; Derya, U.; Emine, C.; Emre, E. The Anti-Inflammatory and Antioxidant Effects of Salvia officinalis on Lipopolysaccharide-Induced Inflammation in Rats. J. Med. Food 2017, 20, 1193–1200. [Google Scholar]
- Manalo, R.V.; Silvestre, M.A.; Barbosa, A.L.A.; Medina, P.M. Coconut (Cocos nucifera) Ethanolic Leaf Extract Reduces Amyloid-β (1-42) Aggregation and Paralysis Prevalence in Transgenic Caenorhabditis elegans Independently of Free Radical Scavenging and Acetylcholinesterase Inhibition. Biomedicines 2017, 5, 17. [Google Scholar] [CrossRef]
- Rahim, N.S.; Lim, S.M.; Mani, V.; Abdul Majeed, A.B.; Ramasamy, K. Enhanced memory in Wistar rats by virgin coconut oil is associated with increased antioxidative, cholinergic activities and reduced oxidative stress. Pharm. Biol. 2017, 55, 825–832. [Google Scholar] [CrossRef] [Green Version]
- Bansal, A.; Kirschner, M.; Zu, L.; Cai, D.; Zhang, L. Coconut Oil Decreases Expression of Amyloid Precursor Protein (APP) and Secretion of Amyloid Peptides throughInhibitionof ADP-ribosylation factor 1 (ARF1). Brain Res. 2018. [Google Scholar] [CrossRef]
- Cheng, D.; Xi, Y.; Cao, J.; Cao, D.; Ma, Y.; Jiang, W. Protective effect of apple (Ralls) polyphenol extract against aluminum-induced cognitive impairment and oxidative damage in rat. NeuroToxicology 2014, 45, 111–120. [Google Scholar] [CrossRef]
- Chan, A.; Shea, T.B. Dietary Supplementation with Apple Juice Decreases Endogenous Amyloid-beta Levels in Murine Brain. J. Alzheimers Dis. 2009, 16, 167–171. [Google Scholar] [CrossRef]
- Fuentealba, J.; Dibarrart, A.J.; Fuentes-Fuentes, M.C.; Saez-Orellana, F.; Quiñones, K.; Guzmán, L.; Perez, C.; Becerra, J.; Aguayo, L.G. Synaptic failure and adenosine triphosphate imbalance induced by amyloid-β aggregates are prevented by blueberry-enriched polyphenols extract. J. Neurosci. Res. 2011, 89, 1499–1508. [Google Scholar] [CrossRef]
- Shukitt-Hale, B.; Lau, F.C.; Carey, A.N.; Galli, R.L.; Spangler, E.L.; Ingram, D.K.; Joseph, J.A. Blueberry polyphenols attenuate kainic acid-induced decrements in cognition and alter inflammatory gene expression in rat hippocampus. Nutr. Neurosci. 2008, 11, 172–182. [Google Scholar] [CrossRef] [Green Version]
- Zhu, Y.; Bickford, P.C.; Sanberg, P.; Giunta, B.; Tan, J. Blueberry opposes β-amyloid peptide-induced microglial activation via inhibition of p44/42 mitogen-activation protein kinase. Rejuvenation Res. 2008, 11, 891–901. [Google Scholar] [CrossRef]
- Tan, L.; Yang, H.; Pang, W.; Li, H.; Liu, W.; Sun, S.; Song, N.; Zhang, W.; Jiang, Y. Investigation on the Role of BDNF in the Benefits of Blueberry Extracts for the Improvement of Learning and Memory in Alzheimer’s Disease Mouse Model. J. Alzheimers Dis. 2017, 56, 629–640. [Google Scholar] [CrossRef]
- Kadakkuzha, B.M.; Liu, X.-A.; Swarnkar, S.; Chen, Y. Chapter 18—Genomic and Proteomic Mechanisms and Models in Toxicity and Safety Evaluation of Nutraceuticals. In Nutraceuticals; Gupta, R.C., Ed.; Academic Press: Boston, MA, USA, 2016; pp. 227–237. [Google Scholar]
- Schuster, D.; Rajendran, A.; Hui, S.W.; Nicotera, T.; Srikrishnan, T.; Kruzel, M.L. Protective effect of colostrinin on neuroblastoma cell survival is due to reduced aggregation of β-amyloid. Neuropeptides 2005, 39, 419–426. [Google Scholar] [CrossRef]
- Stewart, M.G. Colostrinin™: A naturally occurring compound derived from mammalian colostrum with efficacy in treatment of neurodegenerative diseases, including Alzheimer’s. Expert Opin. Pharmacother. 2008, 9, 2553–2559. [Google Scholar] [CrossRef]
- Walker, J.M.; Klakotskaia, D.; Ajit, D.; Weisman, G.A.; Wood, W.G.; Sun, G.Y.; Serfozo, P.; Simonyi, A.; Schachtman, T.R. Beneficial effects of dietary EGCG and voluntary exercise on behavior in an Alzheimer’s disease mouse model. J. Alzheimers Dis. 2015, 44, 561–572. [Google Scholar] [CrossRef]
- Harvey, B.S.; Musgrave, I.F.; Ohlsson, K.S.; Fransson, A.; Smid, S.D. The green tea polyphenol (-)-epigallocatechin-3-gallate inhibits amyloid-β evoked fibril formation and neuronal cell death in vitro. Food Chem. 2011, 129, 1729–1736. [Google Scholar] [CrossRef]
- Abbas, S.; Wink, M. Epigallocatechin gallate inhibits beta amyloid oligomerization in Caenorhabditis elegans and affects the daf-2/insulin-like signaling pathway. Phytomedicine Int. J. Phytother. Phytopharm. 2010, 17, 902–909. [Google Scholar] [CrossRef]
- Wobst, H.J.; Sharma, A.; Diamond, M.I.; Wanker, E.E.; Bieschke, J. The green tea polyphenol (-)-epigallocatechin gallate prevents the aggregation of tau protein into toxic oligomers at substoichiometric ratios. FEBS Lett. 2015, 589, 77–83. [Google Scholar] [CrossRef]
- Chesser, A.S.; Ganeshan, V.; Yang, J.; Johnson, G.V.W. Epigallocatechin-3-gallate enhances clearance of phosphorylated tau in primary neurons. Nutr. Neurosci. 2016, 19, 21–31. [Google Scholar] [CrossRef]
- 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]
- Lee, Y.J.; Choi, D.Y.; Yun, Y.P.; Han, S.B.; Oh, K.W.; Hong, J.T. Epigallocatechin-3-gallate prevents systemic inflammation-induced memory deficiency and amyloidogenesis via its anti-neuroinflammatory properties. J. Nutr. Biochem. 2013, 24, 298–310. [Google Scholar] [CrossRef]
- Ono, K.; Yoshiike, Y.; Takashima, A.; Hasegawa, K.; Naiki, H.; Yamada, M. Vitamin A exhibits potent antiamyloidogenic and fibril-destabilizing effects in vitro. Exp. Neurol. 2004, 189, 380–392. [Google Scholar] [CrossRef]
- Koryakina, A.; Aeberhard, J.; Kiefer, S.; Hamburger, M.; Kuenzi, P. Regulation of secretases by all-trans-retinoic acid. FEBS J. 2009, 276, 2645–2655. [Google Scholar] [CrossRef]
- Kapoor, A.; Wang, B.J.; Hsu, W.M.; Chang, M.Y.; Liang, S.M.; Liao, Y.F. Retinoic acid-elicited RARalpha/RXRalpha signaling attenuates Abeta production by directly inhibiting gamma-secretase-mediated cleavage of amyloid precursor protein. ACS Chem. Neurosci. 2013, 4, 1093–1100. [Google Scholar] [CrossRef]
- Ding, Y.; Qiao, A.; Wang, Z.; Goodwin, J.S.; Lee, E.-S.; Block, M.L.; Allsbrook, M.; McDonald, M.P.; Fan, G.-H. Retinoic Acid Attenuates β-Amyloid Deposition and Rescues Memory Deficits in an Alzheimer’s Disease Transgenic Mouse Model. J. Neurosci. 2008, 28, 11622–11634. [Google Scholar] [CrossRef]
- Arendash, G.W.; Mori, T.; Cao, C.; Mamcarz, M.; Runfeldt, M.; Dickson, A.; Rezai-Zadeh, K.; Tane, J.; Citron, B.A.; Lin, X.; et al. Caffeine reverses cognitive impairment and decreases brain amyloid-beta levels in aged Alzheimer’s disease mice. J. Alzheimers Dis. 2009, 17, 661–680. [Google Scholar] [CrossRef]
- Sharma, B.; Kalita, S.; Paul, A.; Mandal, B.; Paul, S. The role of caffeine as an inhibitor in the aggregation of amyloid forming peptides: A unified molecular dynamics simulation and experimental study. RSC Adv. 2016, 6, 78548–78558. [Google Scholar] [CrossRef]
- Qosa, H.; Abuznait, A.H.; Hill, R.A.; Kaddoumi, A. Enhanced brain amyloid-beta clearance by rifampicin and caffeine as a possible protective mechanism against Alzheimer’s disease. J. Alzheimers Dis. 2012, 31, 151–165. [Google Scholar] [CrossRef]
- Ullah, F.; Ali, T.; Ullah, N.; Kim, M.O. Caffeine prevents d-galactose-induced cognitive deficits, oxidative stress, neuroinflammation and neurodegeneration in the adult rat brain. Neurochem. Int. 2015, 90, 114–124. [Google Scholar] [CrossRef]
- Laurent, C.; Eddarkaoui, S.; Derisbourg, M.; Leboucher, A.; Demeyer, D.; Carrier, S.; Schneider, M.; Hamdane, M.; Muller, C.E.; Buee, L.; et al. Beneficial effects of caffeine in a transgenic model of Alzheimer’s disease-like tau pathology. Neurobiol. Aging 2014, 35, 2079–2090. [Google Scholar] [CrossRef]
- Bend, J.R.; Xia, X.Y.; Chen, D.; Awaysheh, A.; Lo, A.; Rieder, M.J.; Jane Rylett, R. Attenuation of oxidative stress in HEK 293 cells by the TCM constituents schisanhenol, baicalein, resveratrol or crocetin and two defined mixtures. J. Pharm. Pharm. Sci. 2015, 18, 661–682. [Google Scholar] [CrossRef]
- Lu, J.H.; Ardah, M.T.; Durairajan, S.S.; Liu, L.F.; Xie, L.X.; Fong, W.F.; Hasan, M.Y.; Huang, J.D.; El-Agnaf, O.M.; Li, M. Baicalein inhibits formation of alpha-synuclein oligomers within living cells and prevents Abeta peptide fibrillation and oligomerisation. Chembiochem. A Eur. J. Chem. Biol. 2011, 12, 615–624. [Google Scholar] [CrossRef]
- Zhang, S.Q.; Obregon, D.; Ehrhart, J.; Deng, J.; Tian, J.; Hou, H.; Giunta, B.; Sawmiller, D.; Tan, J. Baicalein reduces beta-amyloid and promotes nonamyloidogenic amyloid precursor protein processing in an Alzheimer’s disease transgenic mouse model. J. Neurosci. Res. 2013, 91, 1239–1246. [Google Scholar] [CrossRef]
- Manca, M.L.; Marongiu, F.; Castangia, I.; Catalán-Latorre, A.; Caddeo, C.; Bacchetta, G.; Ennas, G.; Zaru, M.; Fadda, A.M.; Manconi, M. Protective effect of grape extract phospholipid vesicles against oxidative stress skin damages. Ind. Crop. Prod. 2016, 83, 561–567. [Google Scholar] [CrossRef]
- Huang, M.; Jiang, X.; Liang, Y.; Liu, Q.; Chen, S.; Guo, Y. Berberine improves cognitive impairment by promoting autophagic clearance and inhibiting production of β-amyloid in APP/tau/PS1 mouse model of Alzheimer’s disease. Exp. Gerontol. 2017, 91, 25–33. [Google Scholar] [CrossRef]
- He, W.; Wang, C.; Chen, Y.; He, Y.; Cai, Z. Berberine attenuates cognitive impairment and ameliorates tau hyperphosphorylation by limiting the self-perpetuating pathogenic cycle between NF-κB signaling, oxidative stress and neuroinflammation. Pharmacol. Rep. 2017, 69, 1341–1348. [Google Scholar] [CrossRef] [PubMed]
- Beg, T.; Jyoti, S.; Naz, F.; Ali, F.; Ali, S.K.; Reyad, A.M.; Siddique, Y.H. Protective Effect of Kaempferol on the Transgenic Drosophila Model of Alzheimer’s Disease. CNS Neurol. Disord. Drug Targets 2018, 17, 421–429. [Google Scholar] [CrossRef] [PubMed]
- Garcia-Mediavilla, V.; Crespo, I.; Collado, P.S.; Esteller, A.; Sanchez-Campos, S.; Tunon, M.J.; Gonzalez-Gallego, J. The anti-inflammatory flavones quercetin and kaempferol cause inhibition of inducible nitric oxide synthase, cyclooxygenase-2 and reactive C-protein, and down-regulation of the nuclear factor kappaB pathway in Chang Liver cells. Eur. J. Pharmacol. 2007, 557, 221–229. [Google Scholar] [CrossRef]
- Akaishi, T.; Morimoto, T.; Shibao, M.; Watanabe, S.; Sakai-Kato, K.; Utsunomiya-Tate, N.; Abe, K. Structural requirements for the flavonoid fisetin in inhibiting fibril formation of amyloid beta protein. Neurosci. Lett. 2008, 444, 280–285. [Google Scholar] [CrossRef] [PubMed]
- Ono, K.; Yoshiike, Y.; Takashima, A.; Hasegawa, K.; Naiki, H.; Yamada, M. Potent anti-amyloidogenic and fibril-destabilizing effects of polyphenols in vitro: Implications for the prevention and therapeutics of Alzheimer’s disease. J. Neurochem. 2003, 87, 172–181. [Google Scholar] [CrossRef] [PubMed]
- Shimmyo, Y.; Kihara, T.; Akaike, A.; Niidome, T.; Sugimoto, H. Flavonols and flavones as BACE-1 inhibitors: Structure-activity relationship in cell-free, cell-based and in silico studies reveal novel pharmacophore features. Biochim. Biophys. Acta 2008, 1780, 819–825. [Google Scholar] [CrossRef] [PubMed]
- Kim, J.H.; Lee, J.; Lee, S.; Cho, E.J. Quercetin and quercetin-3-β-d-glucoside improve cognitive and memory function in Alzheimer’s disease mouse. Appl. Biol. Chem. 2016, 59, 721–728. [Google Scholar] [CrossRef]
- Regitz, C.; Dussling, L.M.; Wenzel, U. Amyloid-beta (Abeta(1)(-)(4)(2))-induced paralysis in Caenorhabditis elegans is inhibited by the polyphenol quercetin through activation of protein degradation pathways. Mol. Nutr. Food Res. 2014, 58, 1931–1940. [Google Scholar] [CrossRef]
- Jimenez-Aliaga, K.; Bermejo-Bescos, P.; Benedi, J.; Martin-Aragon, S. Quercetin and rutin exhibit antiamyloidogenic and fibril-disaggregating effects in vitro and potent antioxidant activity in APPswe cells. Life Sci. 2011, 89, 939–945. [Google Scholar] [CrossRef]
- Zhang, X.; Hu, J.; Zhong, L.; Wang, N.; Yang, L.; Liu, C.C.; Li, H.; Wang, X.; Zhou, Y.; Zhang, Y.; et al. Quercetin stabilizes apolipoprotein e and reduces brain Aβ levels in amyloid model mice. Neuropharmacology 2016, 108, 179–192. [Google Scholar] [CrossRef]
- Sabogal-Guáqueta, A.M.; Muñoz-Manco, J.I.; Ramírez-Pineda, J.R.; Lamprea-Rodriguez, M.; Osorio, E.; Cardona-Gómez, G.P. The flavonoid quercetin ameliorates Alzheimer’s disease pathology and protects cognitive and emotional function in aged triple transgenic Alzheimer’s disease model mice. Neuropharmacology 2015, 93, 134–145. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Prakash, D.; Sudhandiran, G. Dietary flavonoid fisetin regulates aluminium chloride-induced neuronal apoptosis in cortex and hippocampus of mice brain. J. Nutr. Biochem. 2015, 26, 1527–1539. [Google Scholar] [CrossRef] [PubMed]
- Ahmad, A.; Ali, T.; Park, H.Y.; Badshah, H.; Rehman, S.U.; Kim, M.O. Neuroprotective Effect of Fisetin Against Amyloid-Beta-Induced Cognitive/Synaptic Dysfunction, Neuroinflammation, and Neurodegeneration in Adult Mice. Mol. Neurobiol. 2017, 54, 2269–2285. [Google Scholar] [CrossRef] [PubMed]
- Kim, S.; Choi, K.J.; Cho, S.J.; Yun, S.M.; Jeon, J.P.; Koh, Y.H.; Song, J.; Johnson, G.V.W.; Jo, C. Fisetin stimulates autophagic degradation of phosphorylated tau via the activation of TFEB and Nrf2 transcription factors. Sci. Rep. 2016, 6, 24933. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pourkhodadad, S.; Alirezaei, M.; Moghaddasi, M.; Ahmadvand, H.; Karami, M.; Delfan, B.; Khanipour, Z. Neuroprotective effects of oleuropein against cognitive dysfunction induced by colchicine in hippocampal CA1 area in rats. J. Physiol. Sci. 2016, 66, 397–405. [Google Scholar] [CrossRef] [PubMed]
- Luccarini, I.; Ed Dami, T.; Grossi, C.; Rigacci, S.; Stefani, M.; Casamenti, F. Oleuropein aglycone counteracts Aβ42 toxicity in the rat brain. Neurosci. Lett. 2014, 558, 67–72. [Google Scholar] [CrossRef]
- Pantano, D.; Luccarini, I.; Nardiello, P.; Servili, M.; Stefani, M.; Casamenti, F. Oleuropein aglycone and polyphenols from olive mill waste water ameliorate cognitive deficits and neuropathology. Br. J. Clin. Pharmacol. 2017, 83, 54–62. [Google Scholar] [CrossRef]
- Stefania, R.; Valentina, G.; Monica, B.; Daniela, N.; Annalisa, R.; Andrea, B.; Massimo, S. Aβ(1-42) Aggregates into Non-Toxic Amyloid Assemblies in the Presence of the Natural Polyphenol Oleuropein Aglycon. Curr. Alzheimer Res. 2011, 8, 841–852. [Google Scholar]
- Kostomoiri, M.; Fragkouli, A.; Sagnou, M.; Skaltsounis, L.A.; Pelecanou, M.; Tsilibary, E.C.; Τzinia, A.K. Oleuropein, an Anti-oxidant Polyphenol Constituent of Olive Promotes α-Secretase Cleavage of the Amyloid Precursor Protein (AβPP). Cell. Mol. Neurobiol. 2013, 33, 147–154. [Google Scholar] [CrossRef]
- Daccache, A.; Lion, C.; Sibille, N.; Gerard, M.; Slomianny, C.; Lippens, G.; Cotelle, P. Oleuropein and derivatives from olives as Tau aggregation inhibitors. Neurochem. Int. 2011, 58, 700–707. [Google Scholar] [CrossRef]
- Mori, T.; Rezai-Zadeh, K.; Koyama, N.; Arendash, G.W.; Yamaguchi, H.; Kakuda, N.; Horikoshi-Sakuraba, Y.; Tan, J.; Town, T. Tannic acid is a natural beta-secretase inhibitor that prevents cognitive impairment and mitigates Alzheimer-like pathology in transgenic mice. J. Biol. Chem. 2012, 287, 6912–6927. [Google Scholar] [CrossRef]
- Ono, K.; Hasegawa, K.; Naiki, H.; Yamada, M. Anti-amyloidogenic activity of tannic acid and its activity to destabilize Alzheimer’s β-amyloid fibrils in vitro. Biochim. Biophys. Acta (BBA) Mol. Basis Dis. 2004, 1690, 193–202. [Google Scholar] [CrossRef]
- Yao, J.; Gao, X.; Sun, W.; Yao, T.; Shi, S.; Ji, L. Molecular hairpin: A possible model for inhibition of tau aggregation by tannic acid. Biochemistry 2013, 52, 1893–1902. [Google Scholar] [CrossRef]
- Ghahghaei, A.; Bathaie, S.Z.; Bahraminejad, E. Mechanisms of the Effects of Crocin on Aggregation and Deposition of Aβ1–40 Fibrils in Alzheimer’s Disease. Int. J. Pept. Res. 2012, 18, 347–351. [Google Scholar] [CrossRef]
- Ghahghaei, A.; Bathaie, S.Z.; Kheirkhah, H.; Bahraminejad, E. The protective effect of crocin on the amyloid fibril formation of Abeta42 peptide in vitro. Cell. Mol. Biol. Lett. 2013, 18, 328–339. [Google Scholar] [CrossRef]
- Naghizadeh, B.; Mansouri, M.T.; Ghorbanzadeh, B. Protective effects of crocin against streptozotocin-induced oxidative damage in rat striatum. Acta Med. Iran 2014, 52, 101–105. [Google Scholar]
- Heidari, S.; Mehri, S.; Hosseinzadeh, H. Memory enhancement and protective effects of crocin against d-galactose aging model in the hippocampus of Wistar rats. Iran. J. Basic Med. Sci. 2017, 20, 1250–1259. [Google Scholar]
- Zhang, Z.; Wu, H.; Huang, H. Epicatechin Plus Treadmill Exercise are Neuroprotective Against Moderate-stage Amyloid Precursor Protein/Presenilin 1 Mice. Pharmacogn. Mag. 2016, 12, S139–S146. [Google Scholar] [Green Version]
- Zeng, Y.Q.; Wang, Y.J.; Zhou, X.F. Effects of (-)epicatechin on the pathology of APP/PS1 transgenic mice. Front. Neurol. 2014, 5, 69. [Google Scholar] [CrossRef]
- Cox, C.J.; Choudhry, F.; Peacey, E.; Perkinton, M.S.; Richardson, J.C.; Howlett, D.R.; Lichtenthaler, S.F.; Francis, P.T.; Williams, R.J. Dietary (-)-epicatechin as a potent inhibitor of betagamma-secretase amyloid precursor protein processing. Neurobiol. Aging 2015, 36, 178–187. [Google Scholar] [CrossRef]
- George, R.C.; Lew, J.; Graves, D.J. Interaction of cinnamaldehyde and epicatechin with tau: Implications of beneficial effects in modulating alzheimer’s disease pathogenesis. J. Alzheimer’s Dis. 2013, 36, 21–40. [Google Scholar] [CrossRef]
- Carbonaro, M.; Di Venere, A.; Filabozzi, A.; Maselli, P.; Minicozzi, V.; Morante, S.; Nicolai, E.; Nucara, A.; Placidi, E.; Stellato, F. Role of dietary antioxidant (−)-epicatechin in the development of β-lactoglobulin fibrils. Biochim. Biophys. Acta (BBA) Proteins Proteom. 2016, 1864, 766–772. [Google Scholar] [CrossRef] [PubMed]
- Hajipour, S.; Sarkaki, A.; Farbood, Y.; Eidi, A.; Mortazavi, P.; Valizadeh, Z. Effect of gallic acid on dementia type of Alzheimer disease in rats: Electrophysiological and histological studies. Basic Clin. Neurosci. 2016, 7, 97–106. [Google Scholar] [CrossRef]
- Kim, M.J.; Seong, A.R.; Yoo, J.Y.; Jin, C.H.; Lee, Y.H.; Kim, Y.J.; Lee, J.; Jun, W.J.; Yoon, H.G. Gallic acid, a histone acetyltransferase inhibitor, suppresses beta-amyloid neurotoxicity by inhibiting microglial-mediated neuroinflammation. Mol. Nutr. Food Res. 2011, 55, 1798–1808. [Google Scholar] [CrossRef]
- Jayamani, J.; Shanmugam, G. Gallic acid, one of the components in many plant tissues, is a potential inhibitor for insulin amyloid fibril formation. Eur. J. Med. Chem. 2014, 85, 352–358. [Google Scholar] [CrossRef]
- Valizadeh, Z.; Eidi, A.; Sarkaki, A.; Farbood, Y.; Mortazavi, P. Dementia type of Alzheimer’s disease due to beta-amyloid was improved by Gallic acid in rats. HealthMED 2012, 6, 3648–3656. [Google Scholar]
- Tsai, F.-S.; Wu, L.-Y.; Yang, S.-E.; Cheng, H.-Y.; Tsai, C.-C.; Wu, C.-R.; Lin, L.-W. Ferulic Acid Reverses the Cognitive Dysfunction Caused by Amyloid β Peptide 1-40 Through Anti-Oxidant Activity and Cholinergic Activation in Rats. Am. J. Chin. Med. 2015, 43, 319–335. [Google Scholar] [CrossRef] [PubMed]
- Mori, T.; Koyama, N.; Guillot-Sestier, M.V.; Tan, J.; Town, T. Ferulic acid is a nutraceutical beta-secretase modulator that improves behavioral impairment and alzheimer-like pathology in transgenic mice. PLoS ONE 2013, 8, e55774. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Y.; Cui, L.; Zhang, Y.; Cao, H.; Wang, Y.; Teng, T.; Ma, G.; Li, Y.; Li, K. Ferulic acid inhibits the transition of amyloid-β42 monomers to oligomers but accelerates the transition from oligomers to fibrils. J. Alzheimer’s Dis. 2013, 37, 19–28. [Google Scholar]
- Yan, J.J.; Jung, J.S.; Kim, T.K.; Hasan, M.A.; Hong, C.W.; Nam, J.S.; Song, D.K. Protective effects of ferulic acid in amyloid precursor protein plus presenilin-1 transgenic mouse model of Alzheimer disease. Biol. Pharm. Bull. 2013, 36, 140–143. [Google Scholar] [CrossRef]
- Ono, K.; Hirohata, M.; Yamada, M. Ferulic acid destabilizes preformed beta-amyloid fibrils in vitro. Biochem. Biophys. Res. Commun. 2005, 336, 444–449. [Google Scholar] [CrossRef] [PubMed]
- Xu, P.-X.; Wang, S.-W.; Yu, X.-L.; Su, Y.-J.; Wang, T.; Zhou, W.-W.; Zhang, H.; Wang, Y.-J.; Liu, R.-T. Rutin improves spatial memory in Alzheimer’s disease transgenic mice by reducing Aβ oligomer level and attenuating oxidative stress and neuroinflammation. Behav. Brain Res. 2014, 264, 173–180. [Google Scholar] [CrossRef]
- Lee, Y.W.; Kim, D.H.; Jeon, S.J.; Park, S.J.; Kim, J.M.; Jung, J.M.; Lee, H.E.; Bae, S.G.; Oh, H.K.; Ho Son, K.H.; et al. Neuroprotective effects of salvianolic acid B on an Aβ25–35 peptide-induced mouse model of Alzheimer’s disease. Eur. J. Pharmacol. 2013, 704, 70–77. [Google Scholar] [CrossRef] [PubMed]
- Durairajan, S.S.; Yuan, Q.; Xie, L.; Chan, W.S.; Kum, W.F.; Koo, I.; Liu, C.; Song, Y.; Huang, J.D.; Klein, W.L.; et al. Salvianolic acid B inhibits Abeta fibril formation and disaggregates preformed fibrils and protects against Abeta-induced cytotoxicty. Neurochem. Int. 2008, 52, 741–750. [Google Scholar] [CrossRef] [PubMed]
- Tang, Y.; Huang, D.; Zhang, M.H.; Zhang, W.S.; Tang, Y.X.; Shi, Z.X.; Deng, L.; Zhou, D.H.; Lu, X.Y. Salvianolic acid B inhibits Aβ generation by modulating BACE1 activity in SH-SY5Y-APPsw cells. Nutrients 2016, 8, 333. [Google Scholar] [CrossRef] [PubMed]
- Fiori, J.; Naldi, M.; Bartolini, M.; Andrisano, V. Disclosure of a fundamental clue for the elucidation of the myricetin mechanism of action as amyloid aggregation inhibitor by mass spectrometry. Electrophoresis 2012, 33, 3380–3386. [Google Scholar] [CrossRef] [PubMed]
- Shimmyo, Y.; Kihara, T.; Akaike, A.; Niidome, T.; Sugimoto, H. Multifunction of myricetin on A beta: Neuroprotection via a conformational change of A beta and reduction of A beta via the interference of secretases. J. Neurosci. Res. 2008, 86, 368–377. [Google Scholar] [CrossRef] [PubMed]
- Ramezani, M.; Darbandi, N.; Khodagholi, F.; Hashemi, A. Myricetin protects hippocampal CA3 pyramidal neurons and improves learning and memory impairments in rats with Alzheimer’s disease. Neural Regen Res. 2016, 11, 1976–1980. [Google Scholar] [CrossRef] [PubMed]
- Md, S.; Gan, S.Y.; Haw, Y.H.; Ho, C.L.; Wong, S.; Choudhury, H. In vitro neuroprotective effects of naringenin nanoemulsion against β-amyloid toxicity through the regulation of amyloidogenesis and tau phosphorylation. Int. J. Biol. Macromol. 2018, 118, 1211–1219. [Google Scholar] [CrossRef] [PubMed]
- Yang, W.; Ma, J.; Liu, Z.; Lu, Y.; Hu, B.; Yu, H. Effect of naringenin on brain insulin signaling and cognitive functions in ICV-STZ induced dementia model of rats. Neurol. Sci. 2014, 35, 741–751. [Google Scholar] [CrossRef]
- Ghofrani, S.; Joghataei, M.-T.; Mohseni, S.; Baluchnejadmojarad, T.; Bagheri, M.; Khamse, S.; Roghani, M. Naringenin improves learning and memory in an Alzheimer’s disease rat model: Insights into the underlying mechanisms. Eur. J. Pharmacol. 2015, 764, 195–201. [Google Scholar] [CrossRef]
- Wu, L.-H.; Lin, C.; Lin, H.-Y.; Liu, Y.-S.; Wu, C.Y.-J.; Tsai, C.-F.; Chang, P.-C.; Yeh, W.-L.; Lu, D.-Y. Naringenin Suppresses Neuroinflammatory Responses Through Inducing Suppressor of Cytokine Signaling 3 Expression. Mol. Neurobiol. 2016, 53, 1080–1091. [Google Scholar] [CrossRef]
- Zhang, J.X.; Xing, J.G.; Wang, L.L.; Jiang, H.L.; Guo, S.L.; Liu, R. Luteolin inhibits fibrillary β-amyloid1-40 -induced inflammation in a human blood-brain barrier mode by suppressing the p38 MAPK-mediated NF-eκB signaling pathways. Molecules 2017, 22. [Google Scholar] [CrossRef]
- Liu, R.; Gao, M.; Qiang, G.F.; Zhang, T.T.; Lan, X.; Ying, J.; Du, G.H. The anti-amnesic effects of luteolin against amyloid β25–35 peptide-induced toxicity in mice involve the protection of neurovascular unit. Neuroscience 2009, 162, 1232–1243. [Google Scholar] [CrossRef]
- Wang, H.; Wang, H.; Cheng, H.; Che, Z. Ameliorating effect of luteolin on memory impairment in an Alzheimer’s disease model. Mol. Med. Rep. 2016, 13, 4215–4220. [Google Scholar] [CrossRef]
- Zheng, N.; Yuan, P.; Li, C.; Wu, J.; Huang, J. Luteolin Reduces BACE1 Expression through NF-κB and Estrogen Receptor Mediated Pathways in HEK293 and SH-SY5Y Cells. J. Alzheimer’s Dis. 2015, 45, 659–671. [Google Scholar] [CrossRef]
- Sawmiller, D.; Li, S.; Shahaduzzaman, M.; Smith, A.J.; Obregon, D.; Giunta, B.; Borlongan, C.V.; Sanberg, P.R.; Tan, J. Luteolin Reduces Alzheimer’s Disease Pathologies Induced by Traumatic Brain Injury. Int. J. Mol. Sci. 2014, 15, 895–904. [Google Scholar] [CrossRef] [Green Version]
- Patil, S.P.; Maki, S.; Khedkar, S.A.; Rigby, A.C.; Chan, C. Withanolide A and Asiatic Acid Modulate Multiple Targets Associated with Amyloid-beta Precursor Protein Processing and Amyloid-beta Protein Clearance. J. Nat. Prod. 2010, 73, 1196–1202. [Google Scholar] [CrossRef]
- Dhanasekaran, M.; Holcomb, L.A.; Hitt, A.R.; Tharakan, B.; Porter, J.W.; Young, K.A.; Manyam, B.V. Centella asiatica extract selectively decreases amyloid β levels in hippocampus of Alzheimer’s disease animal model. Phytother. Res. 2009, 23, 14–19. [Google Scholar] [CrossRef]
- Veerendra Kumar, M.H.; Gupta, Y.K. Effect of Centella asiatica on cognition and oxidative stress in an intracerebroventricular streptozotocin model of Alzheimer’s disease in rats. Clin. Exp. Pharmacol. Physiol. 2003, 30, 336–342. [Google Scholar] [CrossRef]
- Mei, Z.R.; Tan, X.P.; Liu, S.Z.; Huang, H.H. Puerarin alleviates cognitive impairment and tau hyperphosphorylation in APP/PS1 transgenic mice. Zhongguo Zhongyao Zazhi 2016, 41, 3285–3289. [Google Scholar]
- Zhao, S.S.; Yang, W.N.; Jin, H.; Ma, K.G.; Feng, G.F. Puerarin attenuates learning and memory impairments and inhibits oxidative stress in STZ-induced SAD mice. NeuroToxicol 2015, 51, 166–171. [Google Scholar] [CrossRef]
- Mahdy, H.M.; Mohamed, M.R.; Emam, M.A.; Karim, A.M.; Abdel-Naim, A.B.; Khalifa, A.E. The anti-apoptotic and anti-inflammatory properties of puerarin attenuate 3-nitropropionic-acid induced neurotoxicity in rats. Can. J. Physiol. Pharmacol. 2014, 92, 252–258. [Google Scholar] [CrossRef]
- Monti, M.C.; Margarucci, L.; Riccio, R.; Casapullo, A. Modulation of Tau Protein Fibrillization by Oleocanthal. J. Nat. Prod. 2012, 75, 1584–1588. [Google Scholar] [CrossRef]
- Li, W.; Sperry, J.B.; Crowe, A.; Trojanowski, J.Q.; Smith, A.B., III; Lee, V.M.-Y. Inhibition of tau fibrillization by oleocanthal via reaction with the amino groups of tau. J. Neurochem. 2009, 110, 1339–1351. [Google Scholar] [CrossRef] [Green Version]
- Qosa, H.; Batarseh, Y.S.; Mohyeldin, M.M.; El Sayed, K.A.; Keller, J.N.; Kaddoumi, A. Oleocanthal enhances amyloid-β clearance from the brains of TgSwDI mice and in vitro across a human blood-brain barrier model. ACS Chem. Neurosci. 2015, 6, 1849–1859. [Google Scholar] [CrossRef]
- Vion, E.; Page, G.; Bourdeaud, E.; Paccalin, M.; Guillard, J.; Rioux Bilan, A. Trans ε-viniferin is an amyloid-β disaggregating and anti-inflammatory drug in a mouse primary cellular model of Alzheimer’s disease. Mol. Cell. Neurosci. 2018, 88, 1–6. [Google Scholar] [CrossRef]
- Jeong, H.Y.; Kim, J.Y.; Lee, H.K.; Ha, D.T.; Song, K.-S.; Bae, K.; Seong, Y.H. Leaf and stem of Vitis amurensis and its active components protect against amyloid β protein (25–35)-induced neurotoxicity. Arch. Pharmacal Res. 2010, 33, 1655–1664. [Google Scholar] [CrossRef]
- Rivière, C.; Papastamoulis, Y.; Fortin, P.-Y.; Delchier, N.; Andriamanarivo, S.; Waffo-Teguo, P.; Kapche, G.D.W.F.; Amira-Guebalia, H.; Delaunay, J.-C.; Mérillon, J.-M.; et al. New stilbene dimers against amyloid fibril formation. Bioorg. Med. Chem. Lett. 2010, 20, 3441–3443. [Google Scholar] [CrossRef]
- McLaurin, J.; Kierstead, M.E.; Brown, M.E.; Hawkes, C.A.; Lambermon, M.H.; Phinney, A.L.; Darabie, A.A.; Cousins, J.E.; French, J.E.; Lan, M.F.; et al. Cyclohexanehexol inhibitors of Abeta aggregation prevent and reverse Alzheimer phenotype in a mouse model. Nat. Med. 2006, 12, 801–808. [Google Scholar] [CrossRef]
- McLaurin, J.; Golomb, R.; Jurewicz, A.; Antel, J.P.; Fraser, P.E. Inositol stereoisomers stabilize an oligomeric aggregate of Alzheimer amyloid beta peptide and inhibit abeta -induced toxicity. J. Biol. Chem. 2000, 275, 18495–18502. [Google Scholar] [CrossRef]
- Wang, M.; Li, Y.; Ni, C.; Song, G. Honokiol Attenuates Oligomeric Amyloid β1-42-Induced Alzheimer’s Disease in Mice Through Attenuating Mitochondrial Apoptosis and Inhibiting the Nuclear Factor Kappa-B Signaling Pathway. Cell. Physiol. Biochem. 2017, 43, 69–81. [Google Scholar] [CrossRef]
- Wang, D.; Dong, X.; Wang, C. Honokiol Ameliorates Amyloidosis and Neuroinflammation and Improves Cognitive Impairment in Alzheimer’s Disease Transgenic Mice. J. Pharmacol. Exp. Ther. 2018, 366, 470–478. [Google Scholar] [CrossRef]
- Balez, R.; Steiner, N.; Engel, M.; Munoz, S.S.; Lum, J.S.; Wu, Y.; Wang, D.; Vallotton, P.; Sachdev, P.; O’Connor, M.; et al. Neuroprotective effects of apigenin against inflammation, neuronal excitability and apoptosis in an induced pluripotent stem cell model of Alzheimer’s disease. Sci. Rep. 2016, 6, 31450. [Google Scholar] [CrossRef] [Green Version]
- Zhao, L.; Wang, J.-L.; Liu, R.; Li, X.-X.; Li, J.-F.; Zhang, L. Neuroprotective, Anti-Amyloidogenic and Neurotrophic Effects of Apigenin in an Alzheimer’s Disease Mouse Model. Molecules 2013, 18, 9949. [Google Scholar] [CrossRef]
- Wang, Y.; Wang, Y.; Li, J.; Hua, L.; Han, B.; Zhang, Y.; Yang, X.; Zeng, Z.; Bai, H.; Yin, H.; et al. Effects of caffeic acid on learning deficits in a model of Alzheimer’s disease. Int. J. Mol. Med. 2016, 38, 869–875. [Google Scholar] [CrossRef]
- Sul, D.; Kim, H.-S.; Lee, D.; Joo, S.S.; Hwang, K.W.; Park, S.-Y. Protective effect of caffeic acid against beta-amyloid-induced neurotoxicity by the inhibition of calcium influx and tau phosphorylation. Life Sci. 2009, 84, 257–262. [Google Scholar] [CrossRef]
- Burton, G.; Ingold, K. beta-Carotene: An unusual type of lipid antioxidant. Science 1984, 224, 569–573. [Google Scholar] [CrossRef]
- Alkam, T.; Nitta, A.; Mizoguchi, H.; Itoh, A.; Nabeshima, T. A natural scavenger of peroxynitrites, rosmarinic acid, protects against impairment of memory induced by Abeta(25-35). Behav. Brain Res. 2007, 180, 139–145. [Google Scholar] [CrossRef]
- Iuvone, T.; De Filippis, D.; Esposito, G.; D’Amico, A.; Izzo, A.A. The spice sage and its active ingredient rosmarinic acid protect PC12 cells from amyloid-beta peptide-induced neurotoxicity. J. Pharmacol. Exp. Ther. 2006, 317, 1143–1149. [Google Scholar] [CrossRef]
- Cornejo, A.; Aguilar Sandoval, F.; Caballero, L.; Machuca, L.; Muñoz, P.; Caballero, J.; Perry, G.; Ardiles, A.; Areche, C.; Melo, F. Rosmarinic acid prevents fibrillization and diminishes vibrational modes associated to β sheet in tau protein linked to Alzheimer’s disease. J. Enzym. Inhib. Med. Chem. 2017, 32, 945–953. [Google Scholar] [CrossRef] [Green Version]
- Siddique, Y.H.; Ali, F. Protective effect of nordihydroguaiaretic acid (NDGA) on the transgenic Drosophila model of Alzheimer’s disease. Chem. Biol. Interact. 2017, 269, 59–66. [Google Scholar] [CrossRef]
- Ono, K.; Hasegawa, K.; Yoshiike, Y.; Takashima, A.; Yamada, M.; Naiki, H. Nordihydroguaiaretic acid potently breaks down pre-formed Alzheimer’s beta-amyloid fibrils in vitro. J. Neurochem. 2002, 81, 434–440. [Google Scholar] [CrossRef]
- Dong, X.; Zhang, D.; Zhang, L.; Li, W.; Meng, X. Osthole improves synaptic plasticity in the hippocampus and cognitive function of Alzheimer’s disease rats via regulating glutamate. Neural Regen Res. 2012, 7, 2325–2332. [Google Scholar]
- Jiao, Y.; Kong, L.; Yao, Y.; Li, S.; Tao, Z.; Yan, Y.; Yang, J. Osthole decreases beta amyloid levels through up-regulation of miR-107 in Alzheimer’s disease. Neuropharmacology 2016, 108, 332–344. [Google Scholar] [CrossRef]
- Yao, Y.; Wang, Y.; Kong, L.; Chen, Y.; Yang, J. Osthole decreases tau protein phosphorylation via PI3K/AKT/GSK-3β signaling pathway in Alzheimer’s disease. Life Sci. 2019, 217, 16–24. [Google Scholar] [CrossRef]
- Kwak, H.M.; Jeon, S.Y.; Sohng, B.H.; Kim, J.G.; Lee, J.M.; Lee, K.B.; Jeong, H.H.; Hur, J.M.; Kang, Y.H.; Song, K.S. beta-Secretase (BACE1) inhibitors from pomegranate (Punica granatum) husk. Arch. Pharmacal Res. 2005, 28, 1328–1332. [Google Scholar] [CrossRef]
- Kiasalari, Z.; Heydarifard, R.; Khalili, M.; Afshin-Majd, S.; Baluchnejadmojarad, T.; Zahedi, E.; Sanaierad, A.; Roghani, M. Ellagic acid ameliorates learning and memory deficits in a rat model of Alzheimer’s disease: An exploration of underlying mechanisms. Psychopharmacology 2017. [Google Scholar] [CrossRef]
- Chai, G.S.; Jiang, X.; Ni, Z.F.; Ma, Z.W.; Xie, A.J.; Cheng, X.S.; Wang, Q.; Wang, J.Z.; Liu, G.P. Betaine attenuates Alzheimer-like pathological changes and memory deficits induced by homocysteine. J. Neurochem. 2013, 124, 388–396. [Google Scholar] [CrossRef] [Green Version]
- Liu, X.P.; Qian, X.; Xie, Y.; Qi, Y.; Peng, M.F.; Zhan, B.C.; Lou, Z.Q. Betaine suppressed Abeta generation by altering amyloid precursor protein processing. Neurol. Sci. 2014, 35, 1009–1013. [Google Scholar]
- Peng, Y.; Hou, C.; Yang, Z.; Li, C.; Jia, L.; Liu, J.; Tang, Y.; Shi, L.; Li, Y.; Long, J.; et al. Hydroxytyrosol mildly improve cognitive function independent of APP processing in APP/PS1 mice. Mol. Nutr. Food Res. 2016, 60, 2331–2342. [Google Scholar] [CrossRef]
- Nardiello, P.; Pantano, D.; Lapucci, A.; Stefani, M.; Casamenti, F. Diet Supplementation with Hydroxytyrosol Ameliorates Brain Pathology and Restores Cognitive Functions in a Mouse Model of Amyloid-beta Deposition. J. Alzheimers Dis. 2018, 63, 1161–1172. [Google Scholar] [CrossRef]
- Kim, T.I.; Lee, Y.K.; Park, S.G.; Choi, I.S.; Ban, J.O.; Park, H.K.; Nam, S.Y.; Yun, Y.W.; Han, S.B.; Oh, K.W.; et al. l-Theanine, an amino acid in green tea, attenuates beta-amyloid-induced cognitive dysfunction and neurotoxicity: Reduction in oxidative damage and inactivation of ERK/p38 kinase and NF-kappaB pathways. Free Radic. Biol. Med. 2009, 47, 1601–1610. [Google Scholar] [CrossRef]
- Ben, P.; Zhang, Z.; Zhu, Y.; Xiong, A.; Gao, Y.; Mu, J.; Yin, Z.; Luo, L. l-Theanine attenuates cadmium-induced neurotoxicity through the inhibition of oxidative damage and tau hyperphosphorylation. NeuroToxicology 2016, 57, 95–103. [Google Scholar] [CrossRef] [PubMed]
- Alonso, E.; Vale, C.; Vieytes, M.R.; Laferla, F.M.; Gimenez-Llort, L.; Botana, L.M. 13-Desmethyl spirolide-C is neuroprotective and reduces intracellular Abeta and hyperphosphorylated tau in vitro. Neurochem. Int. 2011, 59, 1056–1065. [Google Scholar] [CrossRef] [PubMed]
- Alonso, E.; Otero, P.; Vale, C.; Alfonso, A.; Antelo, A.; Giménez-Llort, L.; Chabaud, L.; Guillou, C.; Botana, L.M. Benefit of 13-desmethyl spirolide C treatment in triple transgenic mouse model of Alzheimer disease: Beta-amyloid and neuronal markers improvement. Curr. Alzheimer Res. 2013, 10, 279–289. [Google Scholar] [CrossRef]
- Ferrandiz, M.L.; Alcaraz, M.J. Anti-inflammatory activity and inhibition of arachidonic acid metabolism by flavonoids. Agents Actions 1991, 32, 283–288. [Google Scholar] [CrossRef] [PubMed]
- Thamizhiniyan, V.; Vijayaraghavan, K.; Subramanian, S.P. Gossypin, a flavonol glucoside protects pancreatic beta-cells from glucotoxicity in streptozotocin-induced experimental diabetes in rats. Biomed. Prev. Nutr. 2012, 2, 239–245. [Google Scholar] [CrossRef]
- Zhang, G.L.; Deng, J.P.; Wang, B.H.; Zhao, Z.W.; Li, J.; Gao, L.; Liu, B.L.; Xong, J.R.; Guo, X.D.; Yan, Z.Q.; et al. Gypenosides improve cognitive impairment induced by chronic cerebral hypoperfusion in rats by suppressing oxidative stress and astrocytic activation. Behav. Pharmacol. 2011, 22, 633–644. [Google Scholar] [CrossRef] [PubMed]
- Fujiwara, H.; Tabuchi, M.; Yamaguchi, T.; Iwasaki, K.; Furukawa, K.; Sekiguchi, K.; Ikarashi, Y.; Kudo, Y.; Higuchi, M.; Saido, T.C.; et al. A traditional medicinal herb Paeonia suffruticosa and its active constituent 1,2,3,4,6-penta-O-galloyl-beta-d-glucopyranose have potent anti-aggregation effects on Alzheimer’s amyloid beta proteins in vitro and in vivo. J. Neurochem. 2009, 109, 1648–1657. [Google Scholar] [CrossRef] [PubMed]
- Cui, H.; King, A.E.; Jacobson, G.A.; Small, D.H. Peripheral treatment with enoxaparin exacerbates amyloid plaque pathology in Tg2576 mice. J. Neurosci. Res. 2017, 95, 992–999. [Google Scholar] [CrossRef]
- Bergamaschini, L.; Rossi, E.; Storini, C.; Pizzimenti, S.; Distaso, M.; Perego, C.; De Luigi, A.; Vergani, C.; De Simoni, M.G. Peripheral treatment with enoxaparin, a low molecular weight heparin, reduces plaques and beta-amyloid accumulation in a mouse model of Alzheimer’s disease. J. Neurosci. 2004, 24, 4181–4186. [Google Scholar] [CrossRef]
- Timmer, N.M.; van Dijk, L.; der Zee, C.E.E.M.V.; Kiliaan, A.; de Waal, R.M.W.; Verbeek, M.M. Enoxaparin treatment administered at both early and late stages of amyloid β deposition improves cognition of APPswe/PS1dE9 mice with differential effects on brain Aβ levels. Neurobiol. Dis. 2010, 40, 340–347. [Google Scholar] [CrossRef]
- Gong, E.J.; Park, H.R.; Kim, M.E.; Piao, S.; Lee, E.; Jo, D.G.; Chung, H.Y.; Ha, N.C.; Mattson, M.P.; Lee, J. Morin attenuates tau hyperphosphorylation by inhibiting GSK3beta. Neurobiol. Dis. 2011, 44, 223–230. [Google Scholar] [CrossRef]
- Sachdeva, A.K.; Kuhad, A.; Chopra, K. Naringin ameliorates memory deficits in experimental paradigm of Alzheimer’s disease by attenuating mitochondrial dysfunction. Pharmacol. Biochem. Behav. 2014, 127, 101–110. [Google Scholar] [CrossRef]
- Ulamin, F.; Alishah, S.; Ok Kim, M. Vanillic Acid Attenuates Aβ1-42-Induced Oxidative Stress and Cognitive Impairment in Mice. Sci. Rep. 2017, 7, 40753. [Google Scholar]
- Kim, Y.E.; Hwang, C.J.; Lee, H.P.; Kim, C.S.; Son, D.J.; Ham, Y.W.; Hellström, M.; Han, S.B.; Kim, H.S.; Park, E.K.; et al. Inhibitory effect of punicalagin on lipopolysaccharide-induced neuroinflammation, oxidative stress and memory impairment via inhibition of nuclear factor-kappaB. Neuropharmacology 2017, 117, 21–32. [Google Scholar] [CrossRef]
- Chonpathompikunlert, P.; Wattanathorn, J.; Muchimapura, S. Piperine, the main alkaloid of Thai black pepper, protects against neurodegeneration and cognitive impairment in animal model of cognitive deficit like condition of Alzheimer’s disease. Food Chem. Toxicol. 2010, 48, 798–802. [Google Scholar] [CrossRef]
- Shrivastava, P.; Vaibhav, K.; Tabassum, R.; Khan, A.; Ishrat, T.; Khan, M.M.; Ahmad, A.; Islam, F.; Safhi, M.M.; Islam, F. Anti-apoptotic and Anti-inflammatory effect of Piperine on 6-OHDA induced Parkinson’s Rat model. J. Nutr. Biochem. 2013, 24, 680–687. [Google Scholar] [CrossRef]
- Gao, J.; Inagaki, Y.; Li, X.; Kokudo, N.; Tang, W. Research progress on natural products from traditional Chinese medicine in treatment of Alzheimer’s disease. Drug Discov. Ther. 2013, 7, 46–57. [Google Scholar]
- Jeong, J.H.; Jeong, H.R.; Jo, Y.N.; Kim, H.J.; Shin, J.H.; Heo, H.J. Ameliorating effects of aged garlic extracts against Aβ-induced neurotoxicity and cognitive impairment. BMC Complement. Altern. Med. 2013, 13, 268. [Google Scholar] [CrossRef]
- Gupta, V.B.; Indi, S.S.; Rao, K.S. Garlic extract exhibits antiamyloidogenic activity on amyloid-beta fibrillogenesis: Relevance to Alzheimer’s disease. Phytother. Res. PTR 2009, 23, 111–115. [Google Scholar] [CrossRef]
- Nillert, N.; Pannangrong, W.; Welbat, J.U.; Chaijaroonkhanarak, W.; Sripanidkulchai, K.; Sripanidkulchai, B. Neuroprotective effects of aged garlic extract on cognitive dysfunction and neuroinflammation induced by β-amyloid in rats. Nutrients 2017, 9, 24. [Google Scholar] [CrossRef]
- Chauhan, N.B. Effect of aged garlic extract on APP processing and tau phosphorylation in Alzheimer’s transgenic model Tg2576. J. Ethnopharmacol. 2006, 108, 385–394. [Google Scholar] [CrossRef]
- Ho, S.C.; Chang, K.S.; Chang, P.W. Inhibition of neuroinflammation by cinnamon and its main components. Food Chem. 2013, 138, 2275–2282. [Google Scholar] [CrossRef]
- Modi, K.K.; Roy, A.; Brahmachari, S.; Rangasamy, S.B.; Pahan, K. Cinnamon and Its Metabolite Sodium Benzoate Attenuate the Activation of p21rac and Protect Memory and Learning in an Animal Model of Alzheimer’s Disease. PLoS ONE 2015, 10, e0130398. [Google Scholar] [CrossRef]
- Frydman-Marom, A.; Levin, A.; Farfara, D.; Benromano, T.; Scherzer-Attali, R.; Peled, S.; Vassar, R.; Segal, D.; Gazit, E.; Frenkel, D.; et al. Orally administrated cinnamon extract reduces beta-amyloid oligomerization and corrects cognitive impairment in Alzheimer’s disease animal models. PLoS ONE 2011, 6, e16564. [Google Scholar] [CrossRef]
- Lauretti, E.; Iuliano, L.; Praticò, D. Extra-virgin olive oil ameliorates cognition and neuropathology of the 3xTg mice: Role of autophagy. Ann. Clin. Transl. Neurol. 2017, 4, 564–574. [Google Scholar] [CrossRef]
- Qosa, H.; Mohamed, L.A.; Batarseh, Y.S.; Alqahtani, S.; Ibrahim, B.; LeVine, H.; Keller, J.N.; Kaddoumi, A. Extra-virgin olive oil attenuates amyloid-β and tau pathologies in the brains of TgSwDI mice. J. Nutr. Biochem. 2015, 26, 1479–1490. [Google Scholar] [CrossRef]
- Amel, N.; Wafa, T.; Samia, D.; Yousra, B.; Issam, C.; Cheraif, I.; Attia, N.; Mohamed, H. Extra virgin olive oil modulates brain docosahexaenoic acid level and oxidative damage caused by 2,4-Dichlorophenoxyacetic acid in rats. J. Food Sci. Technol. 2016, 53, 1454–1464. [Google Scholar] [CrossRef] [Green Version]
- Chauhan, N.; Wang, K.C.; Wegiel, J.; Malik, M.N. Walnut extract inhibits the fibrillization of amyloid beta-protein, and also defibrillizes its preformed fibrils. Curr. Alzheimer Res. 2004, 1, 183–188. [Google Scholar] [CrossRef]
- Zou, J.; Cai, P.-S.; Xiong, C.-M.; Ruan, J.-L. Neuroprotective effect of peptides extracted from walnut (Juglans Sigilata Dode) proteins on Aβ25-35-induced memory impairment in mice. J. Huazhong Univ. Sci. Technol. Med. Sci. 2016, 36, 21–30. [Google Scholar] [CrossRef]
- Balu, M.; Sangeetha, P.; Murali, G.; Panneerselvam, C. Age-related oxidative protein damages in central nervous system of rats: Modulatory role of grape seed extract. Int. J. Dev. Neurosci. 2005, 23, 501–507. [Google Scholar] [CrossRef]
- Ono, K.; Condron, M.M.; Ho, L.; Wang, J.; Zhao, W.; Pasinetti, G.M.; Teplow, D.B. Effects of grape seed-derived polyphenols on amyloid beta-protein self-assembly and cytotoxicity. J. Biol. Chem. 2008, 283, 32176–32187. [Google Scholar] [CrossRef]
- Wang, J.; Ho, L.; Zhao, W.; Ono, K.; Rosensweig, C.; Chen, L.; Humala, N.; Teplow, D.B.; Pasinetti, G.M. Grape-derived polyphenolics prevent Abeta oligomerization and attenuate cognitive deterioration in a mouse model of Alzheimer’s disease. J. Neurosci. 2008, 28, 6388–6392. [Google Scholar] [CrossRef]
- Wang, Y.J.; Thomas, P.; Zhong, J.H.; Bi, F.F.; Kosaraju, S.; Pollard, A.; Fenech, M.; Zhou, X.F. Consumption of grape seed extract prevents amyloid-beta deposition and attenuates inflammation in brain of an Alzheimer’s disease mouse. Neurotox Res. 2009, 15, 3–14. [Google Scholar] [CrossRef]
- Pervin, M.; Hasnat, M.A.; Lee, Y.M.; Kim, D.H.; Jo, J.E.; Lim, B.O. Antioxidant activity and acetylcholinesterase inhibition of grape skin anthocyanin (GSA). Molecules 2014, 19, 9403–9418. [Google Scholar] [CrossRef]
- Loureiro, J.; Andrade, S.; Duarte, A.; Neves, A.; Queiroz, J.; Nunes, C.; Sevin, E.; Fenart, L.; Gosselet, F.; Coelho, M.; et al. Resveratrol and Grape Extract-loaded Solid Lipid Nanoparticles for the Treatment of Alzheimer’s Disease. Molecules 2017, 22, 277. [Google Scholar] [CrossRef]
- Hartman, R.E.; Shah, A.; Fagan, A.M.; Schwetye, K.E.; Parsadanian, M.; Schulman, R.N.; Finn, M.B.; Holtzman, D.M. Pomegranate juice decreases amyloid load and improves behavior in a mouse model of Alzheimer’s disease. Neurobiol. Dis. 2006, 24, 506–515. [Google Scholar] [CrossRef]
- Ahmed, A.H.; Subaiea, G.M.; Eid, A.; Li, L.; Seeram, N.P.; Zawia, N.H. Pomegranate extract modulates processing of amyloid-beta precursor protein in an aged Alzheimer’s disease animal model. Curr. Alzheimer Res. 2014, 11, 834–843. [Google Scholar] [CrossRef]
- Essa, M.M.; Subash, S.; Akbar, M.; Al-Adawi, S.; Guillemin, G.J. Long-term dietary supplementation of pomegranates, figs and dates alleviate neuroinflammation in a transgenic mouse model of Alzheimer’s disease. PLoS ONE 2015, 10, e0120964. [Google Scholar] [CrossRef]
- Subash, S.; Essa, M.M.; Al-Asmi, A.; Al-Adawi, S.; Vaishnav, R.; Braidy, N.; Manivasagam, T.; Guillemin, G.J. Pomegranate from Oman Alleviates the Brain Oxidative Damage in Transgenic Mouse Model of Alzheimer’s disease. J. Tradit. Complement. Med. 2014, 4, 232–238. [Google Scholar] [CrossRef]
- Jeong, K.; Shin, Y.C.; Park, S.; Park, J.S.; Kim, N.; Um, J.Y.; Go, H.; Sun, S.; Lee, S.; Park, W.; et al. Ethanol extract of Scutellaria baicalensis Georgi prevents oxidative damage and neuroinflammation and memorial impairments in artificial senescense mice. J. Biomed. Sci. 2011, 18, 14. [Google Scholar] [CrossRef]
- Ebenezer, P.J.; Wilson, C.B.; Wilson, L.D.; Nair, A.R.; J, F. The Anti-Inflammatory Effects of Blueberries in an Animal Model of Post-Traumatic Stress Disorder (PTSD). PLoS ONE 2016, 11, e0160923. [Google Scholar] [CrossRef]
- Ma, H.; Johnson, S.L.; Liu, W.; DaSilva, N.A.; Meschwitz, S.; Dain, J.A.; Seeram, N.P. Evaluation of Polyphenol Anthocyanin-Enriched Extracts of Blackberry, Black Raspberry, Blueberry, Cranberry, Red Raspberry, and Strawberry for Free Radical Scavenging, Reactive Carbonyl Species Trapping, Anti-Glycation, Anti-beta-Amyloid Aggregation, and Microglial Neuroprotective Effects. Int. J. Mol. Sci. 2018, 19, 461. [Google Scholar]
- Sutalangka, C.; Wattanathorn, J.; Muchimapura, S.; Thukham-mee, W. Moringa oleifera mitigates memory impairment and neurodegeneration in animal model of age-related dementia. Oxid. Med. Cell. Longev. 2013, 2013, 695936. [Google Scholar] [CrossRef]
- Forloni, G.; Colombo, L.; Girola, L.; Tagliavini, F.; Salmona, M. Anti-amyloidogenic activity of tetracyclines: Studies in vitro. FEBS Lett. 2001, 487, 404–407. [Google Scholar] [CrossRef]
- Airoldi, C.; Sironi, E.; Dias, C.; Marcelo, F.; Martins, A.; Rauter, A.P.; Nicotra, F.; Jimenez-Barbero, J. Natural compounds against Alzheimer’s disease: Molecular recognition of Abeta1-42 peptide by Salvia sclareoides extract and its major component, rosmarinic acid, as investigated by NMR. Chem. Asian J. 2013, 8, 596–602. [Google Scholar] [CrossRef]
- Fujiwara, H.; Iwasaki, K.; Furukawa, K.; Seki, T.; He, M.; Maruyama, M.; Tomita, N.; Kudo, Y.; Higuchi, M.; Saido, T.C.; et al. Uncaria rhynchophylla, a Chinese medicinal herb, has potent antiaggregation effects on Alzheimer’s beta-amyloid proteins. J. Neurosci. Res. 2006, 84, 427–433. [Google Scholar] [CrossRef]
- Luo, Q.; Lin, T.; Zhang, C.Y.; Zhu, T.; Wang, L.; Ji, Z.; Jia, B.; Ge, T.; Peng, D.; Chen, W. A novel glyceryl monoolein-bearing cubosomes for gambogenic acid: Preparation, cytotoxicity and intracellular uptake. Int. J. Pharm. 2015, 493, 30–39. [Google Scholar] [CrossRef]
- Williams, P.; Sorribas, A.; Liang, Z. New methods to explore marine resources for Alzheimer’s therapeutics. Curr. Alzheimer Res. 2010, 7, 210–213. [Google Scholar] [CrossRef]
- Zhang, H.; Conte, M.M.; Khalil, Z.; Huang, X.-C.; Capon, R.J. New dictyodendrins as BACE inhibitors from a southern Australian marine sponge, Ianthella sp. RSC Adv. 2012, 2, 4209–4214. [Google Scholar] [CrossRef]
- Jeon, S.Y.; Bae, K.; Seong, Y.H.; Song, K.S. Green tea catechins as a BACE1 (beta-secretase) inhibitor. Bioorg. Med. Chem. Lett. 2003, 13, 3905–3908. [Google Scholar] [CrossRef]
- Leiros, M.; Alonso, E.; Rateb, M.E.; Houssen, W.E.; Ebel, R.; Jaspars, M.; Alfonso, A.; Botana, L.M. Gracilins: Spongionella-derived promising compounds for Alzheimer disease. Neuropharmacology 2015, 93, 285–293. [Google Scholar] [CrossRef]
- Zhang, H.; Conte, M.M.; Huang, X.C.; Khalil, Z.; Capon, R.J. A search for BACE inhibitors reveals new biosynthetically related pyrrolidones, furanones and pyrroles from a southern Australian marine sponge, Ianthella sp. Org. Biomol. Chem. 2012, 10, 2656–2663. [Google Scholar] [CrossRef]
- Williams, P.; Sorribas, A.; Howes, M.J. Natural products as a source of Alzheimer’s drug leads. Nat. Prod. Rep. 2011, 28, 48–77. [Google Scholar] [CrossRef]
- Liu, J.; Chen, W.; Xu, Y.; Ren, S.; Zhang, W.; Li, Y. Design, synthesis and biological evaluation of tasiamide B derivatives as BACE1 inhibitors. Bioorg. Med. Chem. 2015, 23, 1963–1974. [Google Scholar] [CrossRef]
- Dai, J.; Sorribas, A.; Yoshida, W.Y.; Kelly, M.; Williams, P.G. Topsentinols, 24-isopropyl steroids from the marine sponge Topsentia sp. J. Nat. Prod. 2010, 73, 1597–1600. [Google Scholar] [CrossRef]
- Dai, J.; Sorribas, A.; Yoshida, W.Y.; Kelly, M.; Williams, P.G. Xestosaprols from the Indonesian marine sponge Xestospongia sp. J. Nat. Prod. 2010, 73, 1188–1191. [Google Scholar] [CrossRef]
- Alonso, E.; Vale, C.; Vieytes, M.R.; Botana, L.M. Translocation of PKC by yessotoxin in an in vitro model of Alzheimer’s disease with improvement of tau and beta-amyloid pathology. ACS Chem. Neurosci. 2013, 4, 1062–1070. [Google Scholar] [CrossRef]
- Alonso, E.; Fuwa, H.; Vale, C.; Suga, Y.; Goto, T.; Konno, Y.; Sasaki, M.; LaFerla, F.M.; Vieytes, M.R.; Gimenez-Llort, L.; et al. Design and synthesis of skeletal analogues of gambierol: Attenuation of amyloid-beta and tau pathology with voltage-gated potassium channel and N-methyl-d-aspartate receptor implications. J. Am. Chem. Soc. 2012, 134, 7467–7479. [Google Scholar] [CrossRef]
- Alonso, E.; Vale, C.; Vieytes, M.R.; Laferla, F.M.; Gimenez-Llort, L.; Botana, L.M. The cholinergic antagonist gymnodimine improves Abeta and tau neuropathology in an in vitro model of Alzheimer disease. Cell. Physiol. Biochem. Int. J. Exp. Cell. Physiol. Biochem. Pharmacol. 2011, 27, 783–794. [Google Scholar] [CrossRef]
- Bidon-Chanal, A.; Fuertes, A.; Alonso, D.; Perez, D.I.; Martinez, A.; Luque, F.J.; Medina, M. Evidence for a new binding mode to GSK-3: Allosteric regulation by the marine compound palinurin. Eur. J. Med. Chem. 2013, 60, 479–489. [Google Scholar] [CrossRef]
- Li, G.D.; Yan, W.H.; Xing, Y. Effects of schisandrone on Tau protein hyperphosphorylation in differentiation of neural stem cells from APP transgenic mice. J. Clin. Rehabil. Tissue Eng. Res. 2009, 13, 4490–4494. [Google Scholar]
- Choi, B.; Kim, S.; Jang, B.G.; Kim, M.J. Piceatannol, a natural analogue of resveratrol, effectively reduces beta-amyloid levels via activation of alpha-secretase and matrix metalloproteinase-9. J. Funct. Foods 2016, 23, 124–134. [Google Scholar] [CrossRef]
- Leiros, M.; Sanchez, J.A.; Alonso, E.; Rateb, M.E.; Houssen, W.E.; Ebel, R.; Jaspars, M.; Alfonso, A.; Botana, L.M. Spongionella secondary metabolites protect mitochondrial function in cortical neurons against oxidative stress. Mar. Drugs 2014, 12, 700–718. [Google Scholar] [CrossRef]
- Soininen, H.; Solomon, A.; Visser, P.J.; Hendrix, S.B.; Blennow, K.; Kivipelto, M.; Hartmann, T.; Hallikainen, I.; Hallikainen, M.; Helisalmi, S.; et al. 24-month intervention with a specific multinutrient in people with prodromal Alzheimer’s disease (LipiDiDiet): A randomised, double-blind, controlled trial. Lancet Neurol. 2017, 16, 965–975. [Google Scholar] [CrossRef]
- Ramalho, M.J.; Andrade, S.; Coelho, M.Á.N.; Loureiro, J.A.; Pereira, M.C. Biophysical interaction of temozolomide and its active metabolite with biomembrane models: The relevance of drug-membrane interaction for Glioblastoma Multiforme therapy. Eur. J. Pharm. Biopharm. 2019, 136, 156–163. [Google Scholar] [CrossRef]
- Ramalho, M.J.; Sevin, E.; Gosselet, F.; Lima, J.; Coelho, M.A.N.; Loureiro, J.A.; Pereira, M.C. Receptor-mediated PLGA nanoparticles for glioblastoma multiforme treatment. Int. J. Pharm. 2018, 545, 84–92. [Google Scholar] [CrossRef]
- Ramalho, M.J.; Pereira, M.C. Preparation and Characterization of Polymeric Nanoparticles: An Interdisciplinary Experiment. J. Chem. Educ. 2016, 93, 1446–1451. [Google Scholar] [CrossRef] [Green Version]
- Jia, T.; Sun, Z.; Lu, Y.; Gao, J.; Zou, H.; Xie, F.; Zhang, G.; Xu, H.; Sun, D.; Yu, Y.; et al. A dual brain-targeting curcumin-loaded polymersomes ameliorated cognitive dysfunction in intrahippocampal amyloid-beta1-42-injected mice. Int. J. Nanomed. 2016, 11, 3765–3775. [Google Scholar]
- Djiokeng Paka, G.; Doggui, S.; Zaghmi, A.; Safar, R.; Dao, L.; Reisch, A.; Klymchenko, A.; Roullin, V.G.; Joubert, O.; Ramassamy, C. Neuronal Uptake and Neuroprotective Properties of Curcumin-Loaded Nanoparticles on SK-N-SH Cell Line: Role of Poly(lactide-co-glycolide) Polymeric Matrix Composition. Mol. Pharm. 2016, 13, 391–403. [Google Scholar] [CrossRef]
- Mourtas, S.; Lazar, A.N.; Markoutsa, E.; Duyckaerts, C.; Antimisiaris, S.G. Multifunctional nanoliposomes with curcumin–lipid derivative and brain targeting functionality with potential applications for Alzheimer disease. Eur. J. Med. Chem. 2014, 80, 175–183. [Google Scholar] [CrossRef]
- Hoppe, J.B.; Coradini, K.; Frozza, R.L.; Oliveira, C.M.; Meneghetti, A.B.; Bernardi, A.; Pires, E.S.; Beck, R.C.R.; Salbego, C.G. Free and nanoencapsulated curcumin suppress β-amyloid-induced cognitive impairments in rats: Involvement of BDNF and Akt/GSK-3β signaling pathway. Neurobiol. Learn. Mem. 2013, 106, 134–144. [Google Scholar] [CrossRef]
- Zhang, J.; Zhou, X.; Yu, Q.; Yang, L.; Sun, D.; Zhou, Y.; Liu, J. Epigallocatechin-3-gallate (EGCG)-Stabilized Selenium Nanoparticles Coated with Tet-1 Peptide To Reduce Amyloid-β Aggregation and Cytotoxicity. ACS Appl. Mater. Interfaces 2014, 6, 8475–8487. [Google Scholar] [CrossRef]
- Debnath, K.; Shekhar, S.; Kumar, V.; Jana, N.R.; Jana, N.R. Efficient Inhibition of Protein Aggregation, Disintegration of Aggregates, and Lowering of Cytotoxicity by Green Tea Polyphenol-Based Self-Assembled Polymer Nanoparticles. ACS Appl. Mater. Interfaces 2016, 8, 20309–20318. [Google Scholar] [CrossRef]
- Meng, Q.; Wang, A.; Hua, H.; Jiang, Y.; Wang, Y.; Mu, H.; Wu, Z.; Sun, K. Intranasal delivery of Huperzine A to the brain using lactoferrin-conjugated N-trimethylated chitosan surface-modified PLGA nanoparticles for treatment of Alzheimer’s disease. Int. J. Nanomed. 2018, 13, 705–718. [Google Scholar] [CrossRef]
- Elnaggar, Y.S.; Etman, S.M.; Abdelmonsif, D.A.; Abdallah, O.Y. Novel piperine-loaded Tween-integrated monoolein cubosomes as brain-targeted oral nanomedicine in Alzheimer’s disease: Pharmaceutical, biological, and toxicological studies. Int. J. Nanomed. 2015, 10, 5459–5473. [Google Scholar] [CrossRef]
- Kuo, Y.-C.; Tsao, C.-W. Neuroprotection against apoptosis of SK-N-MC cells using RMP-7- and lactoferrin-grafted liposomes carrying quercetin. Int. J. Nanomed. 2017, 12, 2857–2869. [Google Scholar] [CrossRef] [Green Version]
Bioactive Compound | Condition of Participants | Number of Subjects | Duration | Outcomes | Ref. |
---|---|---|---|---|---|
Vitamin D | Mild cognitive impairment | 8 | 8 weeks | Reduction of Aβ level | [35] |
Mild cognitive impairment and early AD | 48 | 20 months | Reduction of Aβ level; Improvement of cognitive functions | [36] | |
Vitamin D and memantine | Moderate AD | 43 | 24 weeks | Improvement of cognitive functions | [37] |
Vitamin D and antioxidants | Mild to moderate AD | 78 | 16 weeks | Reduction of oxidative stress | [38] |
Vitamin E and vitamin C | AD | 20 | 1 month | Reduction of oxidative stress | [39] |
Vitamin E and selegiline | Moderate AD | 341 | 2 years | Delay of AD progression | [40] |
Vitamin E and donepezil | Mild cognitive impairment | 769 | 5 years | No effectiveness in delaying AD progression | [41] |
Vitamin E and memantine | Mild to moderate AD | 613 | 5 years | Delay of AD progression | [42] |
Vitamin E and selenium | Healthy patients | 3786 | 13 years | No prevention of dementia | [43] |
Docosahexaenoic acid (DHA) and eicosapentaenoic acid | AD | 204 | 12 months | Safe and well tolerated; No effectiveness in delaying cognitive decline | [44] |
DHA | AD | 295 | 18 months | No effectiveness in delaying cognitive decline | [45] |
Cognitive impairments | 485 | 24 weeks | Improvement of cognitive functions | [46] | |
Mild cognitive impairment | 36 | 1 year | Safe and well tolerated; Improvement of memory | [47] | |
Homotaurine | Mild to moderate AD | 1052 | 78 weeks | Improvement of cognitive functions | [48,49] |
58 | 3 months | No harmful effects on vital signs; Side effects | [50] | ||
10 | 4 weeks | Improvement of the central cholinergic transmission | [51] | ||
Huperzine A | AD | 103 | 8 weeks | Safe and well tolerated; Improvement of memory and behaviour | [52] |
60 | 60 days | Safe and well tolerated; Reduction of oxidative stress | [53] | ||
Mild to moderate AD | 177 | 16 weeks | Safe and well tolerated; Improvement of cognitive functions | [54] | |
Bryostatin | AD | 9 | 46 weeks | Safe and well tolerated: Improvement of cognitive functions | [55] |
150 | 12 weeks | Improvement of cognitive functions | [56] | ||
Melatonin | AD | 150 | 12 weeks | Improvement of memory | [57] |
14 | 22 to 35 months | Improvement of cognitive functions | [58] | ||
Mild cognitive impairment | 50 | 9 to 18 months | Improvement of cognitive functions | [59] | |
Mild to moderate AD | 80 | 24 weeks | Safe; Improvement of cognitive functions | [60] | |
Resveratrol | Mild to moderate AD | 119 | 52 weeks | Side effects; No effectiveness in reducing biomarkers levels | [61] |
39 | 1 year | Safe and well tolerated; No effectiveness in treat AD | [62] | ||
Nicotine | AD | 70 | 2 weeks | Improvement of perceptual and visual attentional deficits | [63] |
6 | 9 weeks | Safe; Improvement of learning | [64] | ||
8 | 10 weeks | Improvement of attentional performance | [65] | ||
Curcumin | AD | 34 | 6 months | Safe and well tolerated | [66] |
Natural Extracts and Other Products | Condition of Participants | Number of Subjects | Duration | Outcomes | Ref. |
---|---|---|---|---|---|
Ginkgo biloba | Mild to moderate dementia | 410 | 24 weeks | Safe; Improvement of neuropsychiatric symptoms | [67,68] |
410 | 24 weeks | Improvement of cognitive and functional functions | [69] | ||
AD or vascular dementia | 404 | 24 weeks | Improvement of cognitive functions and functional abilities; Improvement of neuropsychiatric symptoms | [70] | |
Mild cognitive impairment | 160 | 24 weeks | Safe and well tolerated; Improvement of cognitive functions | [71] | |
Saffron | Mild to moderate AD | 46 | 16 weeks | Safe; Improvement of cognitive functions and memory | [72] |
Lemon balm | Mild to moderate AD | 40 | 4 months | Improvement of cognition function and agitation | [73] |
Green tea | Severe AD | 30 | 2 months | Improvement of cognitive functions | [74] |
Papaya | AD | 20 | 6 months | Reduction of oxidative stress | [75] |
Sage | Mild to moderate AD | 20 | 4 months | Improvement of cognitive functions; No side effects except agitation | [76] |
Coconut | AD | 44 | 21 days | Improvement of cognitive functions | [77] |
Apple | Moderate to severe AD | 21 | 1 month | No improvement of cognitive functions; Improvement behavioural and psychotic symptoms; Reduction of anxiety, agitation and delusion | [78] |
Blueberry | Early memory failures | 9 | 12 weeks | Improvement of learning; Reduction of depressive symptoms | [79] |
Colostrinin | AD | n. d. | 15 weeks | Improvement of cognitive and daily functions | [80] |
© 2019 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 (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Andrade, S.; Ramalho, M.J.; Loureiro, J.A.; Pereira, M.d.C. Natural Compounds for Alzheimer’s Disease Therapy: A Systematic Review of Preclinical and Clinical Studies. Int. J. Mol. Sci. 2019, 20, 2313. https://doi.org/10.3390/ijms20092313
Andrade S, Ramalho MJ, Loureiro JA, Pereira MdC. Natural Compounds for Alzheimer’s Disease Therapy: A Systematic Review of Preclinical and Clinical Studies. International Journal of Molecular Sciences. 2019; 20(9):2313. https://doi.org/10.3390/ijms20092313
Chicago/Turabian StyleAndrade, Stephanie, Maria João Ramalho, Joana Angélica Loureiro, and Maria do Carmo Pereira. 2019. "Natural Compounds for Alzheimer’s Disease Therapy: A Systematic Review of Preclinical and Clinical Studies" International Journal of Molecular Sciences 20, no. 9: 2313. https://doi.org/10.3390/ijms20092313
APA StyleAndrade, S., Ramalho, M. J., Loureiro, J. A., & Pereira, M. d. C. (2019). Natural Compounds for Alzheimer’s Disease Therapy: A Systematic Review of Preclinical and Clinical Studies. International Journal of Molecular Sciences, 20(9), 2313. https://doi.org/10.3390/ijms20092313