New Possibilities in the Therapeutic Approach to Alzheimer’s Disease
Abstract
:1. Introduction
2. Pathology of AD
3. Existing Drug Therapies
3.1. Cholinesterase Inhibitors
3.2. Memantine
3.3. Aducanumab
3.4. Antidepressants
4. Alternative Approach
4.1. Vitamin E
4.2. Melatonin
4.3. Curcumin
4.4. Other Herbs
4.4.1. Ginkgo Biloba
4.4.2. Saffron
4.4.3. Ashwagandha and Flavonoids
4.5. Non-Pharmacological Treatment
4.5.1. Physical Activities
4.5.2. Social Activities
4.5.3. Music Therapy
5. New Approach
5.1. Anti-Inflammatory Treatment
5.2. miRNA Treatment
5.3. Gut-Microbiota Modulation
6. Summary
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Schneider, J.A.; Arvanitakis, Z.; Leurgans, S.E.; Bennett, D.A. The neuropathology of probable Alzheimer disease and mild cognitive impairment. Ann. Neurol. 2009, 66, 200–208. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Soria Lopez, J.A.; González, H.M.; Léger, G.C. Alzheimer’s disease. Handb. Clin. Neurol. 2019, 167, 231–255. [Google Scholar] [CrossRef] [PubMed]
- Lane, C.A.; Hardy, J.; Schott, J.M. Alzheimer’s disease. Eur. J. Neurol. 2018, 25, 59–70. [Google Scholar] [CrossRef] [PubMed]
- Briggs, R.; Kennelly, S.P.; O’Neill, D. Drug treatments in Alzheimer’s disease. Clin. Med. 2016, 16, 247. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mandelkow, E.M.; Mandelkow, E. Tau in Alzheimer’s disease. Trends Cell Biol. 1998, 8, 425–427. [Google Scholar] [CrossRef]
- Martin, L.; Latypova, X.; Wilson, C.M.; Magnaudeix, A.; Perrin, M.-L.; Yardin, C.; Terro, F. Tau protein kinases: Involvement in Alzheimer’s disease. Ageing Res. Rev. 2013, 12, 289–309. [Google Scholar] [CrossRef]
- Naseri, N.N.; Wang, H.; Guo, J.; Sharma, M.; Luo, W. The complexity of tau in Alzheimer’s disease. Neurosci. Lett. 2019, 705, 183–194. [Google Scholar] [CrossRef]
- Bloom, G.S. Amyloid-β and tau: The trigger and bullet in Alzheimer disease pathogenesis. JAMA Neurol. 2014, 71, 505–508. [Google Scholar] [CrossRef] [Green Version]
- Huang, H.C.; Jiang, Z.F. Accumulated amyloid-beta peptide and hyperphosphorylated tau protein: Relationship and links in Alzheimer’s disease. J. Alzheimers. Dis. 2009, 16, 15–27. [Google Scholar] [CrossRef]
- Wang, C.; Wang, Q.; Lou, Y.; Xu, J.; Feng, Z.; Chen, Y.; Tang, Q.; Zheng, G.; Zhang, Z.; Wu, Y.; et al. Salidroside attenuates neuroinflammation and improves functional recovery after spinal cord injury through microglia polarization regulation. J. Cell. Mol. Med. 2018, 22, 1148–1166. [Google Scholar] [CrossRef] [Green Version]
- Tang, Y.; Le, W. Differential Roles of M1 and M2 Microglia in Neurodegenerative Diseases. Mol. Neurobiol. 2016, 53, 1181–1194. [Google Scholar] [CrossRef] [PubMed]
- Jin, X.; Liu, M.-Y.Y.; Zhang, D.-F.F.; Zhong, X.; Du, K.; Qian, P.; Gao, H.; Wei, M.-J.J. Natural products as a potential modulator of microglial polarization in neurodegenerative diseases. Pharmacol. Res. 2019, 145, 104253. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Z.Z.; Zhang, Z.Z.; Lu, H.; Yang, Q.; Wu, H.; Wang, J. Microglial Polarization and Inflammatory Mediators After Intracerebral Hemorrhage. Mol. Neurobiol. 2017, 54, 1874–1886. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Frank, S.; Burbach, G.J.; Bonin, M.; Walter, M.; Streit, W.; Bechmann, I.; Deller, T. TREM2 is upregulated in amyloid plaque-associated microglia in aged APP23 transgenic mice. Glia 2008, 56, 1438–1447. [Google Scholar] [CrossRef]
- Hickman, S.E.; El Khoury, J. TREM2 and the neuroimmunology of Alzheimer’s disease. Biochem. Pharmacol. 2014, 88, 495–498. [Google Scholar] [CrossRef] [Green Version]
- Liddelow, S.A.; Guttenplan, K.A.; Clarke, L.E.; Bennett, F.C.; Bohlen, C.J.; Schirmer, L.; Bennett, M.L.; Münch, A.E.; Chung, W.S.; Peterson, T.C.; et al. Neurotoxic reactive astrocytes are induced by activated microglia. Nature 2017, 541, 481–487. [Google Scholar] [CrossRef]
- Medeiros, R.; LaFerla, F.M. Astrocytes: Conductors of the Alzheimer disease neuroinflammatory symphony. Exp. Neurol. 2013, 239, 133–138. [Google Scholar] [CrossRef]
- Olabarria, M.; Noristani, H.N.; Verkhratsky, A.; Rodríguez, J.J. Concomitant astroglial atrophy and astrogliosis in a triple transgenic animal model of Alzheimer’s disease. Glia 2010, 58, 831–838. [Google Scholar] [CrossRef]
- Fakhoury, M. Microglia and Astrocytes in Alzheimer’s Disease: Implications for Therapy. Curr. Neuropharmacol. 2018, 16, 508–518. [Google Scholar] [CrossRef]
- Wang, F.; Wang, J.; Shen, Y.; Li, H.; Rausch, W.D.; Huang, X. Iron Dyshomeostasis and Ferroptosis: A New Alzheimer’s Disease Hypothesis? Front. Aging Neurosci. 2022, 14, 235. [Google Scholar] [CrossRef]
- Loeffler, D.A.; Connor, J.R.; Juneau, P.L.; Snyder, B.S.; Kanaley, L.; DeMaggio, A.J.; Nguyen, H.; Brickman, C.M.; LeWitt, P.A. Transferrin and iron in normal, Alzheimer’s disease, and Parkinson’s disease brain regions. J. Neurochem. 1995, 65, 710–716. [Google Scholar] [CrossRef] [PubMed]
- Kim, A.C.; Lim, S.; Kim, Y.K. Metal Ion Effects on Aβ and Tau Aggregation. Int. J. Mol. Sci. 2018, 19, 128. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wan, W.; Cao, L.; Kalionis, B.; Murthi, P.; Xia, S.; Guan, Y. Iron Deposition Leads to Hyperphosphorylation of Tau and Disruption of Insulin Signaling. Front. Neurol. 2019, 10, 607. [Google Scholar] [CrossRef] [PubMed]
- Mesulam, M.; Guillozet, A.; Shaw, P.; Quinn, B. Widely Spread Butyrylcholinesterase Can Hydrolyze Acetylcholine in the Normal and Alzheimer Brain. Neurobiol. Dis. 2002, 9, 88–93. [Google Scholar] [CrossRef] [PubMed]
- Gu, X.; Wang, X. An overview of recent analysis and detection of acetylcholine. Anal. Biochem. 2021, 632, 114381. [Google Scholar] [CrossRef]
- Chen, Z.R.; Huang, J.B.; Yang, S.L.; Hong, F.F. Role of Cholinergic Signaling in Alzheimer’s Disease. Molecules 2022, 27, 1816. [Google Scholar] [CrossRef]
- Contestabile, A. The history of the cholinergic hypothesis. Behav. Brain Res. 2011, 221, 334–340. [Google Scholar] [CrossRef]
- Bowen, D.M.; Smith, C.B.; White, P.; Davison, A.N. Neurotransmitter-related enzymes and indices of hypoxia in senile dementia and other abiotrophies. Brain 1976, 99, 459–496. [Google Scholar] [CrossRef] [Green Version]
- Bucci, D.J.; Holland, P.C.; Gallagher, M. Removal of Cholinergic Input to Rat Posterior Parietal Cortex Disrupts Incremental Processing of Conditioned Stimuli. J. Neurosci. 1998, 18, 8038. [Google Scholar] [CrossRef]
- Bell, K.F.S.; Ducatenzeiler, A.; Ribeiro-da-Silva, A.; Duff, K.; Bennett, D.A.; Claudio Cuello, A. The amyloid pathology progresses in a neurotransmitter-specific manner. Neurobiol. Aging 2006, 27, 1644–1657. [Google Scholar] [CrossRef]
- Bracco, L.; Bessi, V.; Padiglioni, S.; Marini, S.; Pepeu, G. Do cholinesterase inhibitors act primarily on attention deficit? A naturalistic study in Alzheimer’s disease patients. J. Alzheimers. Dis. 2014, 40, 737–742. [Google Scholar] [CrossRef] [PubMed]
- Ferreira-Vieira, T.H.; Guimaraes, I.M.; Silva, F.R.; Ribeiro, F.M. Alzheimer’s Disease: Targeting the Cholinergic System. Curr. Neuropharmacol. 2016, 14, 101. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fine, A.; Hoyle, C.; Maclean, C.J.; LeVatte, T.L.; Baker, H.F.; Ridley, R.M. Learning impairments following injection of a selective cholinergic immunotoxin, ME20.4 IgG-saporin, into the basal nucleus of Meynert in monkeys. Neuroscience 1997, 81, 331–343. [Google Scholar] [CrossRef]
- Sarter, M.; Bruno, J.P. Cognitive functions of cortical acetylcholine: Toward a unifying hypothesis. Brain Res. Brain Res. Rev. 1997, 23, 28–46. [Google Scholar] [CrossRef]
- Rogers, S.L.; Friedhoff, L.T. The efficacy and safety of donepezil in patients with Alzheimer’s disease: Results of a US Multicentre, Randomized, Double-Blind, Placebo-Controlled Trial. The Donepezil Study Group. Dementia 1996, 7, 293–303. [Google Scholar] [CrossRef]
- Stage, E.; Svaldi, D.; Sokolow, S.; Risacher, S.L.; Marosi, K.; Rotter, J.I.; Saykin, A.J.; Apostolova, L.G. Prescribing cholinesterase inhibitors in mild cognitive impairment—Observations from the Alzheimer’s Disease Neuroimaging Initiative. Alzheimer’s Dement. Transl. Res. Clin. Interv. 2021, 7. [Google Scholar] [CrossRef] [PubMed]
- Sharma, K. Cholinesterase inhibitors as Alzheimer’s therapeutics. Mol. Med. Rep. 2019, 20, 1479. [Google Scholar] [CrossRef] [Green Version]
- Rogers, S.L.; Farlow, M.R.; Doody, R.S.; Mohs, R.; Friedhoff, L.T. A 24-week, double-blind, placebo-controlled trial of donepezil in patients with Alzheimer’s disease. Donepezil Study Group. Neurology 1998, 50, 136–145. [Google Scholar] [CrossRef] [Green Version]
- Erkinjuntti, T.; Kurz, A.; Gauthier, S.; Bullock, R.; Lilienfeld, S.; Damaraju, C.R.V. Efficacy of galantamine in probable vascular dementia and Alzheimer’s disease combined with cerebrovascular disease: A randomised trial. Lancet 2002, 359, 1283–1290. [Google Scholar] [CrossRef]
- Tariot, P.N.; Solomon, P.R.; Morris, J.C.; Kershaw, P.; Lilienfeld, S.; Ding, C. A 5-month, randomized, placebo-controlled trial of galantamine in AD. The Galantamine USA-10 Study Group. Neurology 2000, 54, 2269–2276. [Google Scholar] [CrossRef] [Green Version]
- Nguyen, K.; Hoffman, H.; Chakkamparambil, B.; Grossberg, G.T. Evaluation of rivastigmine in Alzheimer’s disease. Neurodegener. Dis. Manag. 2021, 11, 35–48. [Google Scholar] [CrossRef] [PubMed]
- Chu, L. Alzheimer’s disease: Early diagnosis and treatment. Hong Kong Med. J. 2012, 18, 228–237. [Google Scholar] [PubMed]
- Memantine (Oral Route) Side Effects-Mayo Clinic. Available online: https://www.mayoclinic.org/drugs-supplements/memantine-oral-route/side-effects/drg-20067012 (accessed on 3 August 2022).
- 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] [PubMed]
- Matsunaga, S.; Kishi, T.; Iwata, N. Memantine Monotherapy for Alzheimer’s Disease: A Systematic Review and Meta-Analysis. PLoS ONE 2015, 10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Matsunaga, S.; Kishi, T.; Nomura, I.; Sakuma, K.; Okuya, M.; Ikuta, T.; Iwata, N. The efficacy and safety of memantine for the treatment of Alzheimer’s disease. Expert Opin. Drug Saf. 2018, 17, 1053–1061. [Google Scholar] [CrossRef] [PubMed]
- Vaz, M.; Silva, V.; Monteiro, C.; Silvestre, S. Role of Aducanumab in the Treatment of Alzheimer’s Disease: Challenges and Opportunities. Clin. Interv. Aging 2022, 17, 797–810. [Google Scholar] [CrossRef]
- Alzheimer’s Association. Alzheimer’s Facts and Figures Report | Alzheimer’s Association. Alzheimer’s Assoc. 2021, 1. [Google Scholar]
- Sevigny, J.; Chiao, P.; Bussière, T.; Weinreb, P.H.; Williams, L.; Maier, M.; Dunstan, R.; Salloway, S.; Chen, T.; Ling, Y.; et al. The antibody aducanumab reduces Aβ plaques in Alzheimer’s disease. Nature 2016, 537, 50–56. [Google Scholar] [CrossRef]
- Linse, S.; Scheidt, T.; Bernfur, K.; Vendruscolo, M.; Dobson, C.M.; Cohen, S.I.A.; Sileikis, E.; Lundqvist, M.; Qian, F.; O’Malley, T.; et al. Kinetic fingerprints differentiate the mechanisms of action of anti-Aβ antibodies. Nat. Struct. Mol. Biol. 2020, 27, 1125–1133. [Google Scholar] [CrossRef]
- Knopman, D.S.; Jones, D.T.; Greicius, M.D. Failure to demonstrate efficacy of aducanumab: An analysis of the EMERGE and ENGAGE trials as reported by Biogen, December 2019. Alzheimers. Dement. 2021, 17, 696–701. [Google Scholar] [CrossRef]
- Fleck, L.M. Alzheimer’s and Aducanumab: Unjust Profits and False Hopes. Hastings Cent. Rep. 2021, 51, 9–11. [Google Scholar] [CrossRef] [PubMed]
- Sturchio, A.; Dwivedi, A.K.; Young, C.B.; Malm, T.; Marsili, L.; Sharma, J.S.; Mahajan, A.; Hill, E.J.; Andaloussi, S.E.L.; Poston, K.L.; et al. High cerebrospinal amyloid-β 42 is associated with normal cognition in individuals with brain amyloidosis. EClinicalMedicine 2021, 38. [Google Scholar] [CrossRef] [PubMed]
- La Joie, R.; Visani, A.V.; Baker, S.L.; Brown, J.A.; Bourakova, V.; Cha, J.; Chaudhary, K.; Edwards, L.; Iaccarino, L.; Janabi, M.; et al. Prospective longitudinal atrophy in Alzheimer’s disease correlates with the intensity and topography of baseline tau-PET. Sci. Transl. Med. 2020, 12. [Google Scholar] [CrossRef]
- Leuzy, A.; Smith, R.; Cullen, N.C.; Strandberg, O.; Vogel, J.W.; Binette, A.P.; Borroni, E.; Janelidze, S.; Ohlsson, T.; Jögi, J.; et al. Biomarker-Based Prediction of Longitudinal Tau Positron Emission Tomography in Alzheimer Disease. JAMA Neurol. 2022, 79, 1. [Google Scholar] [CrossRef]
- Walsh, S.; Merrick, R.; Milne, R.; Brayne, C. Aducanumab for Alzheimer’s disease? BMJ 2021, 374. [Google Scholar] [CrossRef]
- Salloway, S.; Chalkias, S.; Barkhof, F.; Burkett, P.; Barakos, J.; Purcell, D.; Suhy, J.; Forrestal, F.; Tian, Y.; Umans, K.; et al. Amyloid-Related Imaging Abnormalities in 2 Phase 3 Studies Evaluating Aducanumab in Patients With Early Alzheimer Disease. JAMA Neurol. 2022, 79, 13–21. [Google Scholar] [CrossRef]
- Aduhelm: Withdrawn application | European Medicines Agency. Available online: https://www.ema.europa.eu/en/medicines/human/withdrawn-applications/aduhelm (accessed on 25 June 2022).
- Bessey, L.J.; Walaszek, A. Management of Behavioral and Psychological Symptoms of Dementia. Curr. Psychiatry Rep. 2019, 21, 1–11. [Google Scholar] [CrossRef]
- Kales, H.C.; Gitlin, L.N.; Lyketsos, C.G. Assessment and management of behavioral and psychological symptoms of dementia. BMJ 2015, 350. [Google Scholar] [CrossRef] [Green Version]
- Cheng, S.T. Dementia Caregiver Burden: A Research Update and Critical Analysis. Curr. Psychiatry Rep. 2017, 19, 1–8. [Google Scholar] [CrossRef] [Green Version]
- Farina, N.; Morrell, L.; Banerjee, S. What is the therapeutic value of antidepressants in dementia? A narrative review. Int. J. Geriatr. Psychiatry 2017, 32, 32–49. [Google Scholar] [CrossRef]
- Ho, T.; Pollock, B.G.; Mulsant, B.H.; Schantz, O.; Devanand, D.P.; Mintzer, J.E.; Porsteinsson, A.P.; Schneider, L.S.; Weintraub, D.; Yesavage, J.; et al. R- and S-citalopram concentrations have differential effects on neuropsychiatric scores in elders with dementia and agitation. Br. J. Clin. Pharmacol. 2016, 82, 784–792. [Google Scholar] [CrossRef] [PubMed]
- Tampi, R.R.; Tampi, D.J.; Balachandran, S.; Srinivasan, S. Antipsychotic use in dementia: A systematic review of benefits and risks from meta-analyses. Ther. Adv. Chronic Dis. 2016, 7, 229–245. [Google Scholar] [CrossRef] [PubMed]
- Tampi, R.R.; Tampi, D.J. Efficacy and tolerability of benzodiazepines for the treatment of behavioral and psychological symptoms of dementia: A systematic review of randomized controlled trials. Am. J. Alzheimers. Dis. Other Demen. 2014, 29, 565–574. [Google Scholar] [CrossRef] [PubMed]
- Lloret, A.; Esteve, D.; Monllor, P.; Cervera-Ferri, A.; Lloret, A. The Effectiveness of Vitamin E Treatment in Alzheimer’s Disease. Int. J. Mol. Sci. 2019, 20, 879. [Google Scholar] [CrossRef] [Green Version]
- Jiang, Q. Natural forms of vitamin E: Metabolism, antioxidant and anti-inflammatory activities and the role in disease prevention and therapy. Free Radic. Biol. Med. 2014, 72, 76. [Google Scholar] [CrossRef] [Green Version]
- Lopes Da Silva, S.; Vellas, B.; Elemans, S.; Luchsinger, J.; Kamphuis, P.; Yaffe, K.; Sijben, J.; Groenendijk, M.; Stijnen, T. Plasma nutrient status of patients with Alzheimer’s disease: Systematic review and meta-analysis. Alzheimers. Dement. 2014, 10, 485–502. [Google Scholar] [CrossRef] [Green Version]
- Mullan, K.; Cardwell, C.R.; McGuinness, B.; Woodside, J.V.; McKay, G.J. Plasma Antioxidant Status in Patients with Alzheimer’s Disease and Cognitively Intact Elderly: A Meta-Analysis of Case-Control Studies. J. Alzheimers. Dis. 2018, 62, 305–317. [Google Scholar] [CrossRef] [Green Version]
- Mullan, K.; Williams, M.A.; Cardwell, C.R.; McGuinness, B.; Passmore, P.; Silvestri, G.; Woodside, J.V.; McKay, G.J. Serum concentrations of vitamin E and carotenoids are altered in Alzheimer’s disease: A case-control study. Alzheimer’s Dement. Transl. Res. Clin. Interv. 2017, 3, 432. [Google Scholar] [CrossRef] [Green Version]
- Morris, M.C.; Evans, D.A.; Bienias, J.L.; Tangney, C.C.; Bennett, D.A.; Aggarwal, N.; Wilson, R.S.; Scherr, P.A. Dietary intake of antioxidant nutrients and the risk of incident Alzheimer disease in a biracial community study. JAMA 2002, 287, 3230–3237. [Google Scholar] [CrossRef]
- Basambombo, L.L.; Carmichael, P.H.; Côté, S.; Laurin, D. Use of Vitamin E and C Supplements for the Prevention of Cognitive Decline. Ann. Pharmacother. 2017, 51, 118–124. [Google Scholar] [CrossRef]
- Luchsinger, J.A.; Tang, M.X.; Shea, S.; Mayeux, R. Antioxidant vitamin intake and risk of Alzheimer disease. Arch. Neurol. 2003, 60, 203–208. [Google Scholar] [CrossRef] [PubMed]
- Masaki, K.H.; Losonczy, K.G.; Izmirlian, G.; Foley, D.J.; Ross, G.W.; Petrovitch, H.; Havlik, R.; White, L.R. Association of vitamin E and C supplement use with cognitive function and dementia in elderly men. Neurology 2000, 54, 1265–1272. [Google Scholar] [CrossRef] [PubMed]
- Dysken, M.W.; Sano, M.; Asthana, S.; Vertrees, J.E.; Pallaki, M.; Llorente, M.; Love, S.; Schellenberg, G.D.; McCarten, J.R.; Malphurs, J.; 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. [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. [Google Scholar] [CrossRef] [PubMed]
- Farina, N.; Llewellyn, D.; Isaac, M.G.E.K.N.; Tabet, N. Vitamin E for Alzheimer’s dementia and mild cognitive impairment. Cochrane Database Syst. Rev. 2017, 2017, CD002854. [Google Scholar] [CrossRef]
- Li, Y.; Zhang, J.; Wan, J.; Liu, A.; Sun, J. Melatonin regulates Aβ production/clearance balance and Aβ neurotoxicity: A potential therapeutic molecule for Alzheimer’s disease. Biomed. Pharmacother. 2020, 132, 110887. [Google Scholar] [CrossRef] [PubMed]
- Hossain, M.F.; Uddin, M.S.; Uddin, G.M.S.; Sumsuzzman, D.M.; Islam, M.S.; Barreto, G.E.; Mathew, B.; Ashraf, G.M. Melatonin in Alzheimer’s Disease: A Latent Endogenous Regulator of Neurogenesis to Mitigate Alzheimer’s Neuropathology. Mol. Neurobiol. 2019 5612 2019, 56, 8255–8276. [Google Scholar] [CrossRef]
- Payne, J.K. The Trajectory of Biomarkers in Symptom Management for Older Adults With Cancer. Semin. Oncol. Nurs. 2006, 22, 31–35. [Google Scholar] [CrossRef]
- Rosales-Corral, S.; Tan, D.X.; Reiter, R.J.; Valdivia-Velázquez, M.; Martínez-Barboza, G.; Acosta-Martínez, J.P.; Ortiz, G.G. Orally administered melatonin reduces oxidative stress and proinflammatory cytokines induced by amyloid-β peptide in rat brain: A comparative, in vivo study versus vitamin C and E. J. Pineal Res. 2003, 35, 80–84. [Google Scholar] [CrossRef]
- Wu, Y.H.; Swaab, D.F. The human pineal gland and melatonin in aging and Alzheimer’s disease. J. Pineal Res. 2005, 38, 145–152. [Google Scholar] [CrossRef]
- Gunasingh Masilamoni, J.; Philip Jesudason, E.; Dhandayuthapani, S.; Ashok, B.S.; Vignesh, S.; Jebaraj, W.C.E.; Paul, S.F.D.; Jayakumar, R. The neuroprotective role of melatonin against amyloid β peptide injected mice. Free Radic. Res. 2009, 42, 661–673. [Google Scholar] [CrossRef] [PubMed]
- Lin, L.; Huang, Q.X.; Yang, S.S.; Chu, J.; Wang, J.Z.; Tian, Q. Melatonin in Alzheimer’s Disease. Int. J. Mol. Sci. 2013, 14, 14575–14593. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shukla, M.; Govitrapong, P.; Boontem, P.; Reiter, R.J.; Satayavivad, J. Mechanisms of Melatonin in Alleviating Alzheimer’s Disease. Curr. Neuropharmacol. 2017, 15, 1010. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rondanelli, M.; Opizzi, A.; Faliva, M.; Mozzoni, M.; Antoniello, N.; Cazzola, R.; Savarè, R.; Cerutti, R.; Grossi, E.; Cestaro, B. Effects of a diet integration with an oily emulsion of DHA-phospholipids containing melatonin and tryptophan in elderly patients suffering from mild cognitive impairment. Nutr. Neurosci. 2012, 15, 46–54. [Google Scholar] [CrossRef]
- Chainoglou, E.; Hadjipavlou-Litina, D. Curcumin in Health and Diseases: Alzheimer’s Disease and Curcumin Analogues, Derivatives, and Hybrids. Int. J. Mol. Sci. 2020, 21, 1975. [Google Scholar] [CrossRef] [Green Version]
- Fang, L.; Gou, S.; Liu, X.; Cao, F.; Cheng, L. Design, synthesis and anti-Alzheimer properties of dimethylaminomethyl-substituted curcumin derivatives. Bioorg. Med. Chem. Lett. 2014, 24, 40–43. [Google Scholar] [CrossRef]
- Okuda, M.; Fujita, Y.; Hijikuro, I.; Wada, M.; Uemura, T.; Kobayashi, Y.; Waku, T.; Tanaka, N.; Nishimoto, T.; Izumi, Y.; et al. PE859, A Novel Curcumin Derivative, Inhibits Amyloid-β and Tau Aggregation, and Ameliorates Cognitive Dysfunction in Senescence-Accelerated Mouse Prone 8. J. Alzheimer’s Dis. 2017, 59, 313–328. [Google Scholar] [CrossRef]
- Khanna, S.; Park, H.-A.; Sen, C.K.; Golakoti, T.; Sengupta, K.; Venkateswarlu, S.; Roy, S. Neuroprotective and Antiinflammatory Properties of a Novel Demethylated Curcuminoid. Antioxid. Redox Signal. 2009, 11, 449–468. [Google Scholar] [CrossRef]
- Ray, B.; Lahiri, D.K. Neuroinflammation in Alzheimer’s disease: Different molecular targets and potential therapeutic agents including curcumin. Curr. Opin. Pharmacol. 2009, 9, 434–444. [Google Scholar] [CrossRef]
- Konno, H.; Endo, H.; Ise, S.; Miyazaki, K.; Aoki, H.; Sanjoh, A.; Kobayashi, K.; Hattori, Y.; Akaji, K. Synthesis and evaluation of curcumin derivatives toward an inhibitor of beta-site amyloid precursor protein cleaving enzyme 1. Bioorg. Med. Chem. Lett. 2014, 24, 685–690. [Google Scholar] [CrossRef]
- Doroszkiewicz, J.; Mroczko, P.; Kulczyńska-Przybik, A. Inflammation in the CNS: Understanding Various Aspects of the Pathogenesis of Alzheimer’s Disease. Curr. Alzheimer Res. 2021, 19, 16–31. [Google Scholar] [CrossRef] [PubMed]
- Agrawal, R.; Mishra, B.; Tyagi, E.; Nath, C.; Shukla, R. Effect of curcumin on brain insulin receptors and memory functions in STZ (ICV) induced dementia model of rat. Pharmacol. Res. 2010, 61, 247–252. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed]
- Small, G.W.; Siddarth, P.; Li, Z.; Miller, K.J.; Ercoli, L.; Emerson, N.D.; Martinez, J.; Wong, K.P.; Liu, J.; Merrill, D.A.; et al. Memory and Brain Amyloid and Tau Effects of a Bioavailable Form of Curcumin in Non-Demented Adults: A Double-Blind, Placebo-Controlled 18-Month Trial. Am. J. Geriatr. Psychiatry 2018, 26, 266–277. [Google Scholar] [CrossRef] [PubMed]
- Rainey-Smith, S.R.; Brown, B.M.; Sohrabi, H.R.; Shah, T.; Goozee, K.G.; Gupta, V.B.; Martins, R.N. Curcumin and cognition: A randomised, placebo-controlled, double-blind study of community-dwelling older adults. Br. J. Nutr. 2016, 115, 2106–2113. [Google Scholar] [CrossRef]
- Voulgaropoulou, S.D.; van Amelsvoort, T.A.M.J.; Prickaerts, J.; Vingerhoets, C. The effect of curcumin on cognition in Alzheimer’s disease and healthy aging: A systematic review of pre-clinical and clinical studies. Brain Res. 2019, 1725, 146476. [Google Scholar] [CrossRef] [PubMed]
- Singh, S.K.; Srivastav, S.; Castellani, R.J.; Plascencia-Villa, G.; Perry, G. Neuroprotective and Antioxidant Effect of Ginkgo biloba Extract Against AD and Other Neurological Disorders. Neurotherapeutics 2019, 16, 666. [Google Scholar] [CrossRef]
- Ginkgo | NCCIH. Available online: https://www.nccih.nih.gov/health/ginkgo (accessed on 26 June 2022).
- Kellermann, A.J.; Kloft, C. Is there a risk of bleeding associated with standardized Ginkgo biloba extract therapy? A systematic review and meta-analysis. Pharmacotherapy 2011, 31, 490–502. [Google Scholar] [CrossRef]
- Shi, C.; Liu, J.; Wu, F.; Yew, D.T. Ginkgo biloba Extract in Alzheimer’s Disease: From Action Mechanisms to Medical Practice. Int. J. Mol. Sci. 2010, 11, 107. [Google Scholar] [CrossRef]
- Laws, K.R.; Sweetnam, H.; Kondel, T.K. Is Ginkgo biloba a cognitive enhancer in healthy individuals? A meta-analysis. Hum. Psychopharmacol. Clin. Exp. 2012, 27, 527–533. [Google Scholar] [CrossRef]
- Yuan, Q.; Wang, C.-W.; Shi, J.; Lin, Z.-X. Effects of Ginkgo biloba on dementia: An overview of systematic reviews. J. Ethnopharmacol. 2017, 195, 1–9. [Google Scholar] [CrossRef] [PubMed]
- Brondino, N.; Re, S.; Boldrini, A.; Cuccomarino, A.; Lanati, N.; Barale, F.; Politi, P. Curcumin as a Therapeutic Agent in Dementia: A Mini Systematic Review of Human Studies. Sci. World J. 2014, 2014, 1–6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hosseinzadeh, H.; Younesi, H.M. Antinociceptive and anti-inflammatory effects of Crocus sativus L. stigma and petal extracts in mice. BMC Pharmacol. 2002, 2, 7. [Google Scholar] [CrossRef] [Green Version]
- Razavi, B.M.; Hosseinzadeh, H. Saffron as an antidote or a protective agent against natural or chemical toxicities. DARU J. Pharm. Sci. 2015, 23. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Geromichalos, G.D.; Lamari, F.N.; Papandreou, M.A.; Trafalis, D.T.; Margarity, M.; Papageorgiou, A.; Sinakos, Z. Saffron as a source of novel acetylcholinesterase inhibitors: Molecular docking and in vitro enzymatic studies. J. Agric. Food Chem. 2012, 60, 6131–6138. [Google Scholar] [CrossRef]
- Adalier, N.; Parker, H. Vitamin E, Turmeric and Saffron in Treatment of Alzheimer’s Disease. Antioxidants 2016, 5, 40. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ghaffari, S.; Hatami, H.; Dehghan, G. Saffron ethanolic extract attenuates oxidative stress, spatial learning, and memory impairments induced by local injection of ethidium bromide. Res. Pharm. Sci. 2015, 10, 222. [Google Scholar]
- Tsolaki, M.; Karathanasi, E.; Lazarou, I.; Dovas, K.; Verykouki, E.; Karacostas, A.; Georgiadis, K.; Tsolaki, A.; Adam, K.; Kompatsiaris, I.; et al. Efficacy and Safety of Crocus sativus L. in Patients with Mild Cognitive Impairment: One Year Single-Blind Randomized, with Parallel Groups, Clinical Trial. J. Alzheimers. Dis. 2016, 54, 129–133. [Google Scholar] [CrossRef]
- 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]
- Farokhnia, M.; Shafiee Sabet, M.; Iranpour, N.; Gougol, A.; Yekehtaz, H.; Alimardani, R.; Farsad, F.; Kamalipour, M.; Akhondzadeh, S. Comparing the efficacy and safety of Crocus sativus L. with memantine in patients with moderate to severe Alzheimer’s disease: A double-blind randomized clinical trial. Hum. Psychopharmacol. 2014, 29, 351–359. [Google Scholar] [CrossRef]
- Ven Murthy, M.R.; Ranjekar, P.K.; Ramassamy, C.; Deshpande, M. Scientific Basis for the Use of Indian Ayurvedic Medicinal Plants in the Treatment of Neurodegenerative Disorders: 1. Ashwagandha. Cent. Nerv. Syst. Agents Med. Chem. 2012, 10, 238–246. [Google Scholar] [CrossRef]
- Gregory, J.; Vengalasetti, Y.V.; Bredesen, D.E.; Rao, R.V. Neuroprotective Herbs for the Management of Alzheimer’s Disease. Biomolecules 2021, 11, 543. [Google Scholar] [CrossRef] [PubMed]
- Russo, A.; Izzo, A.A.; Cardile, V.; Borrelli, F.; Vanella, A. Indian medicinal plants as antiradicals and DNA cleavage protectors. Phytomedicine 2001, 8, 125–132. [Google Scholar] [CrossRef] [PubMed]
- Sehgal, N.; Gupta, A.; Valli, R.K.; Joshi, S.D.; Mills, J.T.; Hamel, E.; Khanna, P.; Jain, S.C.; Thakur, S.S.; Ravindranath, V. Withania somnifera reverses Alzheimer’s disease pathology by enhancing low-density lipoprotein receptor-related protein in liver. Proc. Natl. Acad. Sci. USA 2012, 109, 3510–3515. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Halim, M.A.; Rosli, I.M.; Shafika, S.; Jaafar, M.; Ooi, H.-M.; Leong, P.-W.; Shamsuddin, S.; Najimudin, N.; Azzam, G. Withania somnifera showed neuroprotective effect and increase longevity in Drosophila Alzheimer’s disease model. bioRxiv 2020. [Google Scholar] [CrossRef]
- Ng, Q.X.; Loke, W.; Foo, N.X.; Tan, W.J.; Chan, H.W.; Lim, D.Y.; Yeo, W.S. A systematic review of the clinical use of Withania somnifera (Ashwagandha) to ameliorate cognitive dysfunction. Phytother. Res. 2020, 34, 583–590. [Google Scholar] [CrossRef]
- Zahiruddin, S.; Basist, P.; Parveen, A.; Parveen, R.; Khan, W.; Gaurav; Ahmad, S. Ashwagandha in brain disorders: A review of recent developments. J. Ethnopharmacol. 2020, 257, 112876. [Google Scholar] [CrossRef]
- Ullah, A.; Munir, S.; Badshah, S.L.; Khan, N.; Ghani, L.; Poulson, B.G.; Emwas, A.H.; Jaremko, M. Important Flavonoids and Their Role as a Therapeutic Agent. Molecules 2020, 25, 5243. [Google Scholar] [CrossRef]
- Lopez-Sanchez, C.; Poejo, J.; Garcia-Lopez, V.; Salazar, J.; Garcia-Martinez, V.; Gutierrez-Merino, C. Kaempferol prevents the activation of complement C3 protein and the generation of reactive A1 astrocytes that mediate rat brain degeneration induced by 3-nitropropionic acid. Food Chem. Toxicol. 2022, 164, 113017. [Google Scholar] [CrossRef]
- Petersen, R.C.; Lopez, O.; Armstrong, M.J.; Getchius, T.S.D.; Ganguli, M.; Gloss, D.; Gronseth, G.S.; Marson, D.; Pringsheim, T.; Day, G.S.; et al. Practice guideline update summary: Mild cognitive impairment: Report of the Guideline Development, Dissemination, and Implementation Subcommittee of the American Academy of Neurology. Neurology 2018, 90, 126. [Google Scholar] [CrossRef]
- Abbott, R.D.; White, L.R.; Ross, G.W.; Masaki, K.H.; Curb, J.D.; Petrovitch, H. Walking and dementia in physically capable elderly men. JAMA 2004, 292, 1447–1453. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Buchman, A.S.; Boyle, P.A.; Yu, L.; Shah, R.C.; Wilson, R.S.; Bennett, D.A. Total daily physical activity and the risk of AD and cognitive decline in older adults. Neurology 2012, 78, 1323–1329. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kouloutbani, K.; Karteroliotis, K.; Politis, A. [The effect of physical activity on dementia]. Psychiatrike 2019, 30, 142–155. [Google Scholar] [CrossRef] [PubMed]
- O’Keefe, J.; Nadel, L.; Keightley, S.; Kill, D. Fornix lesions selectively abolish place learning in the rat. Exp. Neurol. 1975, 48, 152–166. [Google Scholar] [CrossRef]
- Sahay, A.; Scobie, K.N.; Hill, A.S.; O’Carroll, C.M.; Kheirbek, M.A.; Burghardt, N.S.; Fenton, A.A.; Dranovsky, A.; Hen, R. Increasing adult hippocampal neurogenesis is sufficient to improve pattern separation. Nature 2011, 472, 466. [Google Scholar] [CrossRef] [Green Version]
- Cotman, C.W.; Berchtold, N.C. Exercise: A behavioral intervention to enhance brain health and plasticity. Trends Neurosci. 2002, 25, 295–301. [Google Scholar] [CrossRef]
- Belarbi, K.; Burnouf, S.; Fernandez-Gomez, F.J.; Laurent, C.; Lestavel, S.; Figeac, M.; Sultan, A.; Troquier, L.; Leboucher, A.; Caillierez, R.; et al. Beneficial effects of exercise in a transgenic mouse model of Alzheimer’s disease-like Tau pathology. Neurobiol. Dis. 2011, 43, 486–494. [Google Scholar] [CrossRef]
- Du, Z.; Li, Y.; Li, J.; Zhou, C.; Li, F.; Yang, X. Physical activity can improve cognition in patients with Alzheimer’s disease: A systematic review and meta-analysis of randomized controlled trials. Clin. Interv. Aging 2018, 13, 1593. [Google Scholar] [CrossRef] [Green Version]
- Farina, N.; Rusted, J.; Tabet, N. The effect of exercise interventions on cognitive outcome in Alzheimer’s disease: A systematic review. Int. psychogeriatrics 2014, 26, 9–18. [Google Scholar] [CrossRef] [Green Version]
- Heyn, P.; Abreu, B.C.; Ottenbacher, K.J. The effects of exercise training on elderly persons with cognitive impairment and dementia: A meta-analysis. Arch. Phys. Med. Rehabil. 2004, 85, 1694–1704. [Google Scholar] [CrossRef]
- Friedler, B.; Crapser, J.; McCullough, L. One is the Deadliest Number: The Detrimental Effects of Social Isolation on Cerebrovascular Diseases and Cognition. Acta Neuropathol. 2015, 129, 493. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- O’Keefe, L.M.; Doran, S.J.; Mwilambwe-Tshilobo, L.; Conti, L.H.; Venna, V.R.; McCullough, L.D. Social isolation after stroke leads to depressive-like behavior and decreased BDNF levels in mice. Behav. Brain Res. 2014, 260, 162. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Leser, N.; Wagner, S. The effects of acute social isolation on long-term social recognition memory. Neurobiol. Learn. Mem. 2015, 124, 97–103. [Google Scholar] [CrossRef] [PubMed]
- Ali, A.A.; Khalil, M.G.; Elariny, H.A.; Abu-Elfotuh, K. Study on Social Isolation as a Risk Factor in Development of Alzheimer’s Disease in Rats. Brain Disord. Ther. 2017, 06. [Google Scholar] [CrossRef]
- Huang, H.; Wang, L.; Cao, M.; Marshall, C.; Gao, J.; Xiao, N.; Hu, G.; Xiao, M. Isolation Housing Exacerbates Alzheimer’s Disease-Like Pathophysiology in Aged APP/PS1 Mice. Int. J. Neuropsychopharmacol. 2015, 18, 1–10. [Google Scholar] [CrossRef] [Green Version]
- Powell, N.D.; Sloan, E.K.; Bailey, M.T.; Arevalo, J.M.G.; Miller, G.E.; Chen, E.; Kobor, M.S.; Reader, B.F.; Sheridan, J.F.; Cole, S.W. Social stress up-regulates inflammatory gene expression in the leukocyte transcriptome via β-adrenergic induction of myelopoiesis. Proc. Natl. Acad. Sci. 2013, 110, 16574–16579. [Google Scholar] [CrossRef] [Green Version]
- Azzinnari, D.; Sigrist, H.; Staehli, S.; Palme, R.; Hildebrandt, T.; Leparc, G.; Hengerer, B.; Seifritz, E.; Pryce, C.R. Mouse social stress induces increased fear conditioning, helplessness and fatigue to physical challenge together with markers of altered immune and dopamine function. Neuropharmacology 2014, 85, 328–341. [Google Scholar] [CrossRef] [Green Version]
- Murínová, J.; Hlaváčová, N.; Chmelová, M.; Riečanský, I. The Evidence for Altered BDNF Expression in the Brain of Rats Reared or Housed in Social Isolation: A Systematic Review. Front. Behav. Neurosci. 2017, 11, 101. [Google Scholar] [CrossRef] [Green Version]
- Liu, J.; Dietz, K.; Deloyht, J.M.; Pedre, X.; Kelkar, D.; Kaur, J.; Vialou, V.; Lobo, M.K.; Dietz, D.M.; Nestler, E.J.; et al. Impaired adult myelination in the prefrontal cortex of socially isolated mice. Nat. Neurosci. 2012, 15, 1621. [Google Scholar] [CrossRef] [Green Version]
- Hsiao, Y.H.; Chang, C.H.; Gean, P.W. Impact of social relationships on Alzheimer’s memory impairment: Mechanistic studies. J. Biomed. Sci. 2018, 25, 3. [Google Scholar] [CrossRef] [Green Version]
- Schinder, A.F.; Poo, M. ming The neurotrophin hypothesis for synaptic plasticity. Trends Neurosci. 2000, 23, 639–645. [Google Scholar] [CrossRef]
- Peng, S.; Wuu, J.; Mufson, E.J.; Fahnestock, M. Precursor form of brain-derived neurotrophic factor and mature brain-derived neurotrophic factor are decreased in the pre-clinical stages of Alzheimer’s disease. J. Neurochem. 2005, 93, 1412–1421. [Google Scholar] [CrossRef] [PubMed]
- Devi, L.; Ohno, M. TrkB reduction exacerbates Alzheimer’s disease-like signaling aberrations and memory deficits without affecting β-amyloidosis in 5XFAD mice. Transl. Psychiatry 2015, 5, e562. [Google Scholar] [CrossRef]
- O’Bryant, S.E.; Hobson, V.; Hall, J.R.; Waring, S.C.; Chan, W.; Massman, P.; Lacritz, L.; Cullum, C.M.; Diaz-Arrastia, R. Brain-Derived Neurotrophic Factor Levels in Alzheimer’s Disease. J. Alzheimers. Dis. 2009, 17, 337. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nagahara, A.H.; Merrill, D.A.; Coppola, G.; Tsukada, S.; Schroeder, B.E.; Shaked, G.M.; Wang, L.; Blesch, A.; Kim, A.; Conner, J.M.; et al. Neuroprotective effects of brain-derived neurotrophic factor in rodent and primate models of Alzheimer’s disease. Nat. Med. 2009, 15, 331. [Google Scholar] [CrossRef] [Green Version]
- Szekely, C.A.; Breitner, J.C.S.; Zandi, P.P. Prevention of Alzheimer’s disease. Int. Rev. Psychiatry 2007, 19, 693–706. [Google Scholar] [CrossRef]
- Paradise, M.; Cooper, C.; Livingston, G. Systematic review of the effect of education on survival in Alzheimer’s disease. Int. psychogeriatrics 2009, 21, 25–32. [Google Scholar] [CrossRef]
- Peck, K.J.; Girard, T.A.; Russo, F.A.; Fiocco, A.J. Music and Memory in Alzheimer’s Disease and The Potential Underlying Mechanisms. J. Alzheimer’s Dis. 2016, 51, 949–959. [Google Scholar] [CrossRef]
- Wang, Y.; Zheng, T.; Liao, Y.; Li, L.; Zhang, Y. A meta-analysis of the effect of music therapy on Alzheimer’s disease. Int J Clin Exp Med 2020, 13, 317–329. [Google Scholar]
- García-Casares, N.; Moreno-Leiva, R.M.; García-Arnés, J.A. Music therapy as a non-pharmacological treatment in alzheimer’s disease. A systematic review. Rev. Neurol. 2017, 65, 529–538. [Google Scholar] [CrossRef]
- Gómez Gallego, M.; Gómez García, J. Musicoterapia en la enfermedad de Alzheimer: Efectos cognitivos, psicológicos y conductuales. Neurología 2017, 32, 300–308. [Google Scholar] [CrossRef] [PubMed]
- Ozben, T.; Ozben, S. Neuro-inflammation and anti-inflammatory treatment options for Alzheimer’s disease. Clin. Biochem. 2019, 72, 87–89. [Google Scholar] [CrossRef] [PubMed]
- Miguel-Álvarez, M.; Santos-Lozano, A.; Sanchis-Gomar, F.; Fiuza-Luces, C.; Pareja-Galeano, H.; Garatachea, N.; Lucia, A. Non-steroidal anti-inflammatory drugs as a treatment for Alzheimer’s disease: A systematic review and meta-analysis of treatment effect. Drugs Aging 2015, 32, 139–147. [Google Scholar] [CrossRef]
- Steeland, S.; Gorlé, N.; Vandendriessche, C.; Balusu, S.; Brkic, M.; Van Cauwenberghe, C.; Van Imschoot, G.; Van Wonterghem, E.; De Rycke, R.; Kremer, A.; et al. Counteracting the effects of TNF receptor-1 has therapeutic potential in Alzheimer’s disease. EMBO Mol. Med. 2018, 10, e8300. [Google Scholar] [CrossRef]
- Leng, F.; Edison, P. Neuroinflammation and microglial activation in Alzheimer disease: Where do we go from here? Nat. Rev. Neurol. 2020, 17, 157–172. [Google Scholar] [CrossRef] [PubMed]
- Tobinick, E.L.; Gross, H. Rapid improvement in verbal fluency and aphasia following perispinal etanercept in Alzheimer’s disease. BMC Neurol. 2008, 8, 27. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kitazawa, M.; Cheng, D.; Tsukamoto, M.R.; Koike, M.A.; Wes, P.D.; Vasilevko, V.; Cribbs, D.H.; LaFerla, F.M. Blocking IL-1 signaling rescues cognition, attenuates tau pathology, and restores neuronal β-catenin pathway function in an Alzheimer’s disease model. J. Immunol. 2011, 187, 6539–6549. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Alves, S.; Churlaud, G.; Audrain, M.; Michaelsen-Preusse, K.; Fol, R.; Souchet, B.; Braudeau, J.; Korte, M.; Klatzmann, D.; Cartier, N. Interleukin-2 improves amyloid pathology, synaptic failure and memory in Alzheimer’s disease mice. Brain 2017, 140, 826–842. [Google Scholar] [CrossRef]
- Kiyota, T.; Okuyama, S.; Swan, R.J.; Jacobsen, M.T.; Gendelman, H.E.; Ikezu, T. CNS expression of anti-inflammatory cytokine interleukin-4 attenuates Alzheimer’s disease-like pathogenesis in APP+PS1 bigenic mice. FASEB J. 2010, 24, 3093–3102. [Google Scholar] [CrossRef] [Green Version]
- Fu, A.K.Y.; Hung, K.W.; Yuen, M.Y.F.; Zhou, X.; Mak, D.S.Y.; Chan, I.C.W.; Cheung, T.H.; Zhang, B.; Fu, W.Y.; Liew, F.Y.; et al. IL-33 ameliorates Alzheimer’s disease-like pathology and cognitive decline. Proc. Natl. Acad. Sci. USA 2016, 113, E2705–E2713. [Google Scholar] [CrossRef] [Green Version]
- Zheng, C.; Zhou, X.-W.W.; Wang, J.-Z.Z. The dual roles of cytokines in Alzheimer’s disease: Update on interleukins, TNF-α, TGF-β and IFN-γ. Transl. Neurodegener. 2016, 5, 7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, C.Y.; Ryu, I.S.; Ryu, J.H.; Cho, H.J. miRNAs as Therapeutic Tools in Alzheimer’s Disease. Int. J. Mol. Sci. 2021, 22, 13012. [Google Scholar] [CrossRef] [PubMed]
- Lee, C.Y.; Shin, S.; Lee, J.; Seo, H.H.; Lim, K.H.; Kim, H.; Choi, J.W.; Kim, S.W.; Lee, S.; Lim, S.; et al. MicroRNA-Mediated Down-Regulation of Apoptosis Signal-Regulating Kinase 1 (ASK1) Attenuates the Apoptosis of Human Mesenchymal Stem Cells (MSCs) Transplanted into Infarcted Heart. Int. J. Mol. Sci. 2016, 17, 1752. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fu, G.; Brkić, J.; Hayder, H.; Peng, C. MicroRNAs in Human Placental Development and Pregnancy Complications. Int. J. Mol. Sci. 2013, 14, 5519–5544. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Grasso, M.; Piscopo, P.; Confaloni, A.; Denti, M.A. Circulating miRNAs as Biomarkers for Neurodegenerative Disorders. Molecular 2014, 19, 6891–6910. [Google Scholar] [CrossRef]
- Paul, P.; Chakraborty, A.; Sarkar, D.; Langthasa, M.; Rahman, M.; Bari, M.; Singha, R.K.S.; Malakar, A.K.; Chakraborty, S. Interplay between miRNAs and human diseases. J. Cell. Physiol. 2018, 233, 2007–2018. [Google Scholar] [CrossRef]
- Wang, M.; Qin, L.; Tang, B. MicroRNAs in Alzheimer’s disease. Front. Genet. 2019, 10, 153. [Google Scholar] [CrossRef] [Green Version]
- Sproviero, D.; Gagliardi, S.; Zucca, S.; Arigoni, M.; Giannini, M.; Garofalo, M.; Olivero, M.; Dell’orco, M.; Pansarasa, O.; Bernuzzi, S.; et al. Different miRNA Profiles in Plasma Derived Small and Large Extracellular Vesicles from Patients with Neurodegenerative Diseases. Int. J. Mol. Sci. 2021, 22, 2737. [Google Scholar] [CrossRef]
- Doroszkiewicz, J.; Groblewska, M.; Mroczko, B. The role of gut microbiota and gut–brain interplay in selected diseases of the central nervous system. Int. J. Mol. Sci. 2021, 22, 28. [Google Scholar] [CrossRef]
- Swarbrick, S.; Wragg, N.; Ghosh, S.; Stolzing, A. Systematic Review of miRNA as Biomarkers in Alzheimer’s Disease. Mol. Neurobiol. 2019, 56, 6156–6167. [Google Scholar] [CrossRef] [Green Version]
- Hébert, S.S.; Horré, K.; Nicolaï, L.; Papadopoulou, A.S.; Mandemakers, W.; Silahtaroglu, A.N.; Kauppinen, S.; Delacourte, A.; De Strooper, B. Loss of microRNA cluster miR-29a/b-1 in sporadic Alzheimer’s disease correlates with increased BACE1/β-secretase expression. Proc. Natl. Acad. Sci. USA 2008, 105, 6415–6420. [Google Scholar] [CrossRef] [Green Version]
- Kiko, T.; Nakagawa, K.; Tsuduki, T.; Furukawa, K.; Arai, H.; Miyazawa, T. MicroRNAs in plasma and cerebrospinal fluid as potential markers for Alzheimer’s disease. J. Alzheimer’s Dis. 2014, 39, 253–259. [Google Scholar] [CrossRef] [PubMed]
- Kowalski, K.; Mulak, A. Brain-gut-microbiota axis in Alzheimer’s disease. J. Neurogastroenterol. Motil. 2019, 25, 48–60. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kim, M.-S.; Kim, Y.; Choi, H.H.; Kim, W.; Park, S.; Lee, D.D.-S.; Kim, D.K.; Kim, H.J.; Choi, H.H.; Hyun, D.-W.; et al. Transfer of a healthy microbiota reduces amyloid and tau pathology in an Alzheimer’s disease animal model. Gut 2020, 69, 283–294. [Google Scholar] [CrossRef] [PubMed]
- Park, J.-C.; Han, S.-H.; Mook-Jung, I. Peripheral inflammatory biomarkers in Alzheimer’s disease: A brief review. BMB Rep. 2020, 53, 10–19. [Google Scholar] [CrossRef]
- Megur, A.; Baltriukienė, D.; Bukelskienė, V.; Burokas, A. The Microbiota–Gut–Brain Axis and Alzheimer’s Disease: Neuroinflammation Is to Blame? Nutrients 2021, 13, 37. [Google Scholar] [CrossRef]
- Friedland, R.P.; Chapman, M.R. The role of microbial amyloid in neurodegeneration. PLOS Pathog. 2017, 13, e1006654. [Google Scholar] [CrossRef]
- Cattaneo, A.; Cattane, N.; Galluzzi, S.; Provasi, S.; Lopizzo, N.; Festari, C.; Ferrari, C.; Guerra, U.P.; Paghera, B.; Muscio, C.; et al. Association of brain amyloidosis with pro-inflammatory gut bacterial taxa and peripheral inflammation markers in cognitively impaired elderly. Neurobiol. Aging 2017, 49, 60–68. [Google Scholar] [CrossRef] [Green Version]
- Bonfili, L.; Cecarini, V.; Berardi, S.; Scarpona, S.; Suchodolski, J.S.; Nasuti, C.; Fiorini, D.; Boarelli, M.C.; Rossi, G.; Eleuteri, A.M. Microbiota modulation counteracts Alzheimer’s disease progression influencing neuronal proteolysis and gut hormones plasma levels. Sci. Rep. 2017, 7, 2426. [Google Scholar] [CrossRef]
Type of Therapy | Influence | Author | |||||||||
---|---|---|---|---|---|---|---|---|---|---|---|
Cognitive Functions | Attention | Learning | Memory | Aβ Plaques | Agitation | Proinflammatory Response | Progression of AD | Emotions | Side Effects | ||
Cholinesterase inhibitors | ↑ | ↑ | ↑ | ↑ | insomnia, gastrointestinal symptoms (nausea, loss of appetite, diarrhea), muscle cramping and weakness | [31,32,33,34] | |||||
Memantine | ↑ | ↑ | ↑ | Confusion, aniety | [42,43,44,45] | ||||||
Aducanumab | ↑ | Removal, disturbing accumulation | micro-bleeds, swelling in the brain, dizziness, headaches nausea | [49] | |||||||
Antidepressants | ↓ | ↑ | Citalopram: risk of QT prolongation | [62,63] | |||||||
Vitamin E | ↑ | Overdosage: fatigue, gastrointestinal cramps, diarrhea | [72] | ||||||||
Melatonin | Prevent accumulation | ↓ | ND | [83,85,86] | |||||||
Curcumin | ↑ | Prevent accumulation | ND | [89,94,95] | |||||||
Ginkgo biloba | ↑ | ↓ | ND | [102] | |||||||
Saffron | ↑ | ↑ | ↑ | ND | [109,110] | ||||||
Ashwaganda | ↓toxicity | ND | [118] | ||||||||
Physical activities | ↑ | ND | [124,125] | ||||||||
Social activities | ↑ | ND | [144,148] | ||||||||
Music therapy | ↑ | ↑ | ↑ | ND | [151,153] | ||||||
Anti-inflammatory treatment | ↑ | ↓pathology | ND | [157,158,160] | |||||||
Gut microbiota alterations | Stopping development | ND | [177] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Doroszkiewicz, J.; Mroczko, B. New Possibilities in the Therapeutic Approach to Alzheimer’s Disease. Int. J. Mol. Sci. 2022, 23, 8902. https://doi.org/10.3390/ijms23168902
Doroszkiewicz J, Mroczko B. New Possibilities in the Therapeutic Approach to Alzheimer’s Disease. International Journal of Molecular Sciences. 2022; 23(16):8902. https://doi.org/10.3390/ijms23168902
Chicago/Turabian StyleDoroszkiewicz, Julia, and Barbara Mroczko. 2022. "New Possibilities in the Therapeutic Approach to Alzheimer’s Disease" International Journal of Molecular Sciences 23, no. 16: 8902. https://doi.org/10.3390/ijms23168902
APA StyleDoroszkiewicz, J., & Mroczko, B. (2022). New Possibilities in the Therapeutic Approach to Alzheimer’s Disease. International Journal of Molecular Sciences, 23(16), 8902. https://doi.org/10.3390/ijms23168902