Journey on Naphthoquinone and Anthraquinone Derivatives: New Insights in Alzheimer’s Disease
Abstract
1. Introduction
1.1. Role of Aβ in AD
1.2. Role of Cholinesterase Enzymes (ChEs) in AD
1.3. Role of Tau Protein in AD
2. Quinone-Based Scaffolds for the Development of Novel Agents against AD
3. Naphthoquinones
3.1. NQs from Natural Sources
3.2. Synthetic NQ Derivatives
3.3. NQ-Based Hybrids
4. Anthraquinones
4.1. AQs from Natural Sources: Cholinesterase Inhibitors
4.2. AQs from Natural Sources: Tau Aggregation Inhibitors
4.3. AQs from Natural Sources: BACE1 Inhibitors and Antioxidants
4.4. Synthetic AQ Derivatives
4.5. AQ-Based Hybrids
5. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Adelina, C. The costs of dementia: Advocacy, media and stigma. In World Alzheimer Report 2019: Attitudes to Dementia; Alzheimer’s Disease International: London, UK, 2019; Chapter 3; pp. 100–101. Available online: https://www.alzint.org/u/WorldAlzheimerReport2019.pdf (accessed on 20 September 2019).
- Glenner, G.G.; Murphy, M.A. Amyloidosis of the nervous system. J. Neurol. Sci. 1989, 94, 1–28. [Google Scholar] [CrossRef]
- Nakhate, K.T.; Bharne, A.P.; Verma, V.S.; Aru, D.N.; Kokare, D.M. Plumbagin ameliorates memory dysfunction in streptozotocin induced Alzheimer’s disease via activation of Nrf2/ARE pathway and inhibition of β-secretase. Biomed. Pharmacother. 2018, 101, 379–390. [Google Scholar] [CrossRef] [PubMed]
- Grundke-Iqbal, I.; Iqbal, K.; Tung, Y.C.; Quinlan, M.; Wisniewski, H.M.; Binder, L.I. Abnormal phosphorylation of the microtubule-associated protein tau (tau) in Alzheimer cytoskeletal pathology. Proc. Natl. Acad. Sci. USA 1986, 83, 4913–4917. [Google Scholar] [CrossRef] [PubMed]
- Terry, R.D.; Gonatas, N.K.; Weiss, M. Ultrastructural studies in Alzheimer’s presenile dementia. Am. J. Pathol. 1964, 44, 269–297. [Google Scholar]
- Coyle, J.; Puttfarcken, P. Oxidative stress, glutamate, and neurodegenerative disorders. Science 1993, 262, 689–695. [Google Scholar] [CrossRef]
- Huang, X.; Moir, R.D.; Tanzi, R.E.; Bush, A.I.; Rogers, J.T. Redox-active metals, oxidative stress, and Alzheimer’s disease pathology. Ann. N. Y. Acad. Sci. 2004, 1012, 153–163. [Google Scholar] [CrossRef]
- Alam, J.; Sharma, L. Potential Enzymatic Targets in Alzheimer’s: A Comprehensive Review. Curr. Drug Targets 2018, 20, 316–339. [Google Scholar] [CrossRef]
- Blaikie, L.; Kay, G.; Lin, P.K.T. Current and emerging therapeutic targets of alzheimer’s disease for the design of multi-target directed ligands. Med. Chem. Comm. 2019, 10, 2052–2072. [Google Scholar] [CrossRef]
- Guo, J.; Wang, Z.; Liu, R.; Huang, Y.; Zhang, N.; Zhang, R. Memantine, Donepezil, or Combination Therapy—What is the best therapy for Alzheimer’s Disease? A Network Meta-Analysis. Brain Behav. 2020, 10, e01831. [Google Scholar] [CrossRef]
- Selkoe, D. Normal and Abnormal Biology of the β-Amyloid Precursor Protein. Annu. Rev. Neurosci. 1994, 17, 489–517. [Google Scholar] [CrossRef]
- Nhan, H.S.; Chiang, K.; Koo, E.H. The multifaceted nature of amyloid precursor protein and its proteolytic fragments: Friends and foes. Acta Neuropathol. 2015, 129, 1–19. [Google Scholar] [CrossRef] [PubMed]
- Multhaup, G.; Huber, O.; Buée, L.; Galas, M.C. Amyloid Precursor Protein (APP) Metabolites APP Intracellular Fragment (AICD), Aβ42, and Tau in Nuclear Roles. J. Biol. Chem. 2015, 290, 23515–23522. [Google Scholar] [CrossRef] [PubMed]
- Goate, A.; Chartier-Harlin, M.C.; Mullan, M.; Brown, J.; Crawford, F.; Fidani, L.; Giuffra, L.; Haynes, A.; Irving, N.; James, L.; et al. Segregation of a missense mutation in the amyloid precursor protein gene with familial Alzheimer’s disease. Nature 1991, 349, 704–706. [Google Scholar] [CrossRef] [PubMed]
- De Jonghe, C. Pathogenic APP mutations near the gamma-secretase cleavage site differentially affect Aβ secretion and APP C-terminal fragment stability. Hum. Mol. Genet. 2001, 10, 1665–1671. [Google Scholar] [CrossRef] [PubMed]
- Levin, O.S.; Vasenina, E.E. 25 Years of the Amyloid Hypothesis of the Origin of Alzheimer’s Disease: Advances, Failures, and New Perspectives. Neurosci. Behav. Physiol. 2017, 47, 1065–1070. [Google Scholar] [CrossRef]
- Glenner, G.G.; Wong, C.W. Alzheimer’s disease: Initial report of the purification and characterization of a novel cerebrovascular amyloid protein. Biochem. Biophys. Res. Commun. 1984, 120, 885–890. [Google Scholar] [CrossRef]
- Agrawal, N.; Skelton, A.A. Structure and Function of Alzheimer’s Amyloid βeta Proteins from Monomer to Fibrils: A Mini Review. Protein J. 2019, 38, 425–434. [Google Scholar] [CrossRef]
- Yoshiike, Y.; Chui, D.-H.; Akagi, T.; Tanaka, N.; Takashima, A. Specific Compositions of Amyloid-β Peptides as the Determinant of Toxic β-Aggregation. J. Biol. Chem. 2003, 278, 23648–23655. [Google Scholar] [CrossRef]
- Kirkitadze, M.D.; Condron, M.M.; Teplow, D.B. Identification and characterization of key kinetic intermediates in amyloid β-protein fibrillogenesis. J. Mol. Biol. 2001, 312, 1103–1119. [Google Scholar] [CrossRef]
- Walsh, D.M.; Lomakin, A.; Benedek, G.B.; Condron, M.M.; Teplow, D.B. Amyloid β-protein fibrillogenesis: Detection of a protofibrillar intermediate. J. Biol. Chem. 1997, 272, 22364–22372. [Google Scholar] [CrossRef]
- Marina, G.B.; Kirkitadze, D.; Lomakin, A.; Vollers, S.S.; Benedek, G.B.; Teplow, D.B. Amyloid β-protein (Aβ) assembly: Aβ40 and Aβ42 oligomerize through distinct pathways. Proc. Natl. Acad. Sci. USA 2003, 100, 330–335. [Google Scholar] [CrossRef]
- Kirkitadze, M.D.; Kowalska, A. Molecular mechanisms initiating amyloid β-fibril formation in Alzheimer’s disease. Acta Biochim. Pol. 2005, 52, 417–423. [Google Scholar] [CrossRef] [PubMed]
- Hardy, J.; Higgins, G. Alzheimer’s disease: The amyloid cascade hypothesis. Science 1992, 256, 184–185. [Google Scholar] [CrossRef] [PubMed]
- Folch, J.; Ettcheto, M.; Petrov, D.; Abad, S.; Pedrós, I.; Marin, M.; Olloquequi, J.; Camins, A. Review of the advances in treatment for Alzheimer disease: Strategies for combating β-amyloid protein. Neurol. Engl. Ed. 2018, 33, 47–58. [Google Scholar] [CrossRef]
- Hsiao, C.C.; Rombouts, F.; Gijsen, H.J.M. New evolutions in the BACE1 inhibitor field from 2014 to 2018. Bioorg. Med. Chem. Lett. 2019, 29, 761–777. [Google Scholar] [CrossRef] [PubMed]
- Moussa-Pacha, N.M.; Abdin, S.M.; Omar, H.A.; Alniss, H.; Al-Tel, T.H. BACE1 inhibitors: Current status and future directions in treating Alzheimer’s disease. Med. Res. Rev. 2020, 40, 339–384. [Google Scholar] [CrossRef]
- Nalivaeva, N.N.; Turner, A.J. Targeting amyloid clearance in Alzheimer’s disease as a therapeutic strategy. Br. J. Pharmacol. 2019, 176, 3447–3463. [Google Scholar] [CrossRef] [PubMed]
- Ittner, L.M.; Götz, J. Amyloid-β and tau—A toxic pas de deux in Alzheimer’s disease. Nat. Rev. Neurosci. 2011, 12, 67–72. [Google Scholar] [CrossRef]
- Guo, J.P.; Arai, T.; Miklossy, J.; McGeer, P.L. Aβ and tau form soluble complexes that may promote self aggregation of both into the insoluble forms in Alzheimer’s diseases. Proc. Natl. Acad. Sci. USA 2006, 103, 1953–1958. [Google Scholar] [CrossRef]
- Kaur, D.; Sharma, V.; Deshmukh, R. Activation of microglia and astrocytes: A roadway to neuroinflammation and Alzheimer’s disease. Inflammopharmacology 2019, 27, 663–677. [Google Scholar] [CrossRef]
- Blurton-Jones, M.; LaFerla, F. Pathways by Which Aβ Facilitates Tau Pathology. Curr. Alzheimer Res. 2006, 3, 437–448. [Google Scholar] [CrossRef] [PubMed]
- Roher, A.E.; Baudry, J.; Chaney, M.O.; Kuo, Y.M.; Stine, W.B.; Emmerling, M.R. Oligomerization and fibril assembly of the amyloid-β protein. Biochim. Biophys. Acta 2000, 1502, 31–43. [Google Scholar] [CrossRef]
- Bartus, R.T.; Dean, R.L., III; Beer, B.; Lippa, A.S. The cholinergic hypothesis of geriatric memory dysfunction. Science 1982, 217, 408–417. [Google Scholar] [CrossRef] [PubMed]
- Davies, P.; Maloney, A.J. Selective loss of central cholinergic neurons in Alzheimer’s disease. Lancet 1976, 2, 1403. [Google Scholar] [CrossRef]
- Picciotto, M.R.; Higley, M.J.; Mineur, Y.S. Acetylcholine as a Neuromodulator: Cholinergic Signaling Shapes Nervous System Function and Behavior. Neuron 2012, 76, 116–129. [Google Scholar] [CrossRef]
- Chen, X.-Q.; Mobley, W.C. Exploring the Pathogenesis of Alzheimer Disease in Basal Forebrain Cholinergic Neurons: Converging Insights from Alternative Hypotheses. Front. Neurosci. 2019, 13, 446. [Google Scholar] [CrossRef]
- Francis, P.T.; Palmer, A.M.; Snape, M.; Wilcock, G.K. The cholinergic hypothesis of Alzheimer’s disease: A review of progress. J. Neurol. Neurosurg. Psychiatry 1999, 66, 137–147. [Google Scholar] [CrossRef]
- Ferreira-Vieira, T.H.; Guimaraes, I.M.; Silva, F.R.; Ribeiro, F.M. Alzheimer’s disease: Targeting the Cholinergic System. Curr. Neuropharmacol. 2016, 14, 101–115. [Google Scholar] [CrossRef]
- David, L.; Cheah, E.; Cygler, M.; Dijkstra, B.; Frolow, F.; Sybille, M.; Harel, M.; James Remington, S.; Silman, I.; Schrag, J.; et al. The α/β hydrolase fold. Protein Eng. Des. Sel. 1992, 5, 197–211. [Google Scholar] [CrossRef]
- Mehta, M.; Adem, A.; Sabbagh, M. New Acetylcholinesterase Inhibitors for Alzheimer’s Disease. Int. J. Alzheimers Dis. 2012, 2012, 1–8. [Google Scholar] [CrossRef]
- Deture, M.A.; Dickson, D.W. The neuropathological diagnosis of Alzheimer’s disease. Mol. Neurodegener. 2019, 14, 1–18. [Google Scholar] [CrossRef] [PubMed]
- Giacobini, E. Cholinesterases: New roles in brain function and in Alzheimer’s disease. Neurochem. Res. 2003, 28, 515–522. [Google Scholar] [CrossRef] [PubMed]
- Stanciu, G.D.; Luca, A.; Rusu, R.N.; Bild, V.; Chiriac, S.I.B.; Solcan, C.; Bild, W.; Ababei, D.C. Alzheimer’s disease pharmacotherapy in relation to cholinergic system involvement. Biomolecules 2020, 10, 40. [Google Scholar] [CrossRef] [PubMed]
- Tougu, V. Acetylcholinesterase: Mechanism of catalysis and inhibition. Curr. Med. Chem. CNS Agents 2001, 1, 155–170. [Google Scholar] [CrossRef]
- Zhang, Y.; Kua, J.; McCammon, J.A. Role of the Catalytic Triad and Oxyanion Hole in Acetylcholinesterase Catalysis: An ab initio QM/MM Study. J. Am. Chem. Soc. 2002, 124, 10572–10577. [Google Scholar] [CrossRef] [PubMed]
- Silman, I.; Sussman, J.L. Acetylcholinesterase: How is structure related to function? Chem. Biol. Interact. 2008, 175, 3–10. [Google Scholar] [CrossRef]
- Rees, T.M.; Brimijoin, S. The role of acetylcholinesterase in the pathogenesis of Alzheimer’s disease. Drugs Today 2003, 39, 75–83. [Google Scholar] [CrossRef]
- Augustin, N.; Nuthakki, V.K.; Abdullaha, M.; Hassan, Q.P.; Gandhi, S.G.; Bharate, S.B. Discovery of Helminthosporin, an Anthraquinone Isolated from Rumex abyssinicus Jacq as a Dual Cholinesterase Inhibitor. ACS Omega 2020, 5, 1616–1624. [Google Scholar] [CrossRef]
- Inestrosa, N.C.; Dinamarca, M.C.; Alvarez, A. Amyloid-cholinesterase interactions: Implications for Alzheimer’s disease. FEBS J. 2008, 275, 625–632. [Google Scholar] [CrossRef]
- VandeVrede, L.; Boxer, A.L.; Polydoro, M. Targeting tau: Clinical trials and novel therapeutic approaches. Neurosci. Lett. 2020, 731, 134919. [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] [PubMed]
- Chen, D.; Drombosky, K.; Hou, Z.; Sari, L.; Kashmer, O.; Ryder, B.; Perez, V.; Woodard, D.; Lin, M.; Diamond, M.; et al. Tau local structure shields amyloid motif and controls aggregation propensity. bioRxiv 2018, 330266. [Google Scholar] [CrossRef] [PubMed]
- Noble, W.; Hanger, D.P.; Miller, C.C.J.; Lovestone, S. The importance of tau phosphorylation for neurodegenerative diseases. Front. Neurol. 2013, 4, 1–11. [Google Scholar] [CrossRef]
- López López, L.I.; Nery Flores, S.D.; Silva Belmares, S.Y.; Sáenz Galindo, A. Naphthoquinones: Biological properties and synthesis of lawsone and derivatives—A structured review. Vitae 2014, 21, 248–258. [Google Scholar]
- Vukic, M.D.; Vukovic, N.L.; Djelic, G.T.; Popovic, S.L.; Zaric, M.M.; Baskic, D.D.; Krstic, G.B.; Tesevic, V.V.; Kacaniova, M.M. Antibacterial and cytotoxic activities of naphthoquinone pigments from onosma Visianii clem. EXCLI J. 2017, 16, 73–88. [Google Scholar] [CrossRef] [PubMed]
- Tandon, V.K.; Chhor, R.B.; Singh, R.V.; Rai, S.; Yadav, D.B. Design, synthesis and evaluation of novel 1,4-naphthoquinone derivatives as antifungal and anticancer agents. Bioorganic Med. Chem. Lett. 2004, 14, 1079–1083. [Google Scholar] [CrossRef] [PubMed]
- Kim, B.H.; Yoo, J.; Park, S.H.; Jung, J.K.; Cho, H.; Chung, Y. Synthesis and evaluation of antitumor activity of novel 1,4-naphthoquinone derivatives (IV). Arch. Pharm. Res. 2006, 29, 123–130. [Google Scholar] [CrossRef] [PubMed]
- Alemán, J.; Jacobsen, C.B.; Frisch, K.; Overgaard, J.; Jørgensen, K.A. Organocatalytic asymmetric vinylogous addition to quinones—Formation of optically active α-aryl ketones. Chem. Commun. 2002, 8, 632–634. [Google Scholar] [CrossRef]
- Sun, J.W.; Wang, X.S.; Liu, Y. Copper(II)-catalyzed sequential C,N-difunctionalization of 1,4-naphthoquinone for the synthesis of benzo[f]indole-4,9-diones under base-free condition. J. Org. Chem. 2013, 78, 10560–10566. [Google Scholar] [CrossRef]
- Lee, J.; Panek, J.S. Synthesis of Isochromene-Type Scaffolds via Single-Flask Diels–Alder-[4 + 2]-Annulation Sequence of a Silyl-Substituted Diene with Menadione. Org. Lett. 2014, 16, 3320–3323. [Google Scholar] [CrossRef]
- Albrecht, Ł.; Gómez, C.V.; Jacobsen, C.B.; Jørgensen, K.A. 1,4-naphthoquinones in H-bond-directed trienamine-mediated strategies. Org. Lett. 2013, 15, 3010–3013. [Google Scholar] [CrossRef] [PubMed]
- He, Z.; Liu, T.; Tao, H.; Wang, C.J. A facile access to enantioenriched isoindolines via one-pot sequential Cu(I)-catalyzed asymmetric 1,3-dipolar cycloaddition/aromatization. Org. Lett. 2012, 14, 6230–6233. [Google Scholar] [CrossRef] [PubMed]
- Murphy, B.; Goodrich, P.; Hardacre, C.; Oelgemöller, M. Green photochemistry: Photo-Friedel–Crafts acylations of 1,4-naphthoquinone in room temperature ionic liquids. Green Chem. 2009, 11, 1867–1870. [Google Scholar] [CrossRef]
- Yu, J.-S. 1,4-Naphthoquinone. Synlett 2014, 25, 2377–2378. [Google Scholar] [CrossRef][Green Version]
- Berkessel, A.; Guixà, M.; Schmidt, F.; Neudörfl, J.M.; Lex, J. Highly enantioselective epoxidation of 2-methylnaphthoquinone (vitamin K3) mediated by new cinchona alkaloid phase-transfer catalysts. Chem. A Eur. J. 2007, 13, 4483–4498. [Google Scholar] [CrossRef]
- Scherzer-Attali, R.; Pellarin, R.; Convertino, M.; Frydman-Marom, A.; Egoz-Matia, N.; Peled, S.; Levy-Sakin, M.; Shalev, D.E.; Caflisch, A.; Gazit, E.; et al. Complete phenotypic recovery of an Alzheimer’s disease model by a quinone-tryptophan hybrid aggregation inhibitor. PLoS ONE 2010, 5, e11101. [Google Scholar] [CrossRef]
- Nam, K.N.; Son, M.S.; Park, J.H.; Lee, E.H. Shikonins attenuate microglial inflammatory responses by inhibition of ERK, Akt, and NF-κB: Neuroprotective implications. Neuropharmacology 2008, 55, 819–825. [Google Scholar] [CrossRef]
- Castagnoli, K.P.; Steyn, S.J.; Petzer, J.P.; Van Der Schyf, C.J.; Castagnoli, N. Neuroprotection in the MPTP parkinsonian C57BL/6 mouse model by a compound isolated from tobacco. Chem. Res. Toxicol. 2001, 14, 523–527. [Google Scholar] [CrossRef]
- Bermejo-Bescós, P.; Martín-Aragón, S.; Jiménez-Aliaga, K.L.; Ortega, A.; Molina, M.T.; Buxaderas, E.; Orellana, G.; Csákÿ, A.G. In vitro antiamyloidogenic properties of 1,4-naphthoquinones. Biochem. Biophys. Res. Commun. 2010, 400, 169–174. [Google Scholar] [CrossRef]
- Dutra, R.C.; Campos, M.M.; Santos, A.R.S.; Calixto, J.B. Medicinal plants in Brazil: Pharmacological studies, drug discovery, challenges and perspectives. Pharmacol. Res. 2016, 112, 4–29. [Google Scholar] [CrossRef]
- Raskin, I.; Ribnicky, D.M.; Komarnytsky, S.; Ilic, N.; Poulev, A.; Borisjuk, N.; Brinker, A.; Moreno, D.A.; Ripoll, C.; Yakoby, N.; et al. Plants and human health in the twenty-first century. Trends Biotechnol. 2002, 20, 522–531. [Google Scholar] [CrossRef]
- Soejarto, D.D.; Farnsworth, N.R. Tropical rain forests: Potential source of new drugs? Perspect. Biol. Med. 1989, 32, 244–256. [Google Scholar] [CrossRef] [PubMed]
- Tshisikhawe, M.P.; van Rooyen, M.W.; Bhat, R.B. An evaluation of the extent and threat of bark harvesting of medicinal plant species in the venda region, limpopo province, south africa. Phyton (B. Aires) 2012, 81, 89–100. [Google Scholar]
- Gordaliza, M. Natural products as leads to anticancer drugs. Clin. Transl. Oncol. 2007, 9, 767–776. [Google Scholar] [CrossRef] [PubMed]
- Coseri, S. Natural Products and their Analogues as Efficient Anticancer Drugs. Mini-Rev. Med. Chem. 2009, 9, 560–571. [Google Scholar] [CrossRef]
- de Carvalho da Silva, F.; Francisco Ferreira, V. Natural Naphthoquinones with Great Importance in Medicinal Chemistry. Curr. Org. Synth. 2016, 13, 334–371. [Google Scholar] [CrossRef]
- Thomson, R.H.; Thomson, R.H. Benzoquinones. In Naturally Occurring Quinones IV; Springer Netherlands: Heidelberg, Germany, 1997; pp. 1–111. ISBN 978-94-009-1551-0. [Google Scholar]
- Patai, S.; Rappoport, Z. The Chemistry of the Quinonoid Compounds. In The Chemistry of the Quinonoid Compounds; John Wiley & Sons, Inc.: Hoboken, NJ, USA, 1988; Volume 2, pp. 1–878. ISBN 9780470772119. [Google Scholar]
- Babula, P.; Adam, V.; Havel, L.; Kizek, R. Noteworthy secondary metabolites naphthoquinone—Their occurence, pharmacological properties and analysis. Curr. Pharm. Anal. 2009, 5, 47–68. [Google Scholar] [CrossRef]
- Van der Vijver, L.M. Distribution of plumbagin in the Plumbaginaceae. Phytochemistry 1972, 11, 3247–3248. [Google Scholar] [CrossRef]
- Hazra, B.; Das Sarma, M.; Sanyal, U. Separation methods of quinonoid constituents of plants used in Oriental traditional medicines. J. Chromatogr. B Analyt. Technol. Biomed. Life Sci. 2004, 812, 259–275. [Google Scholar] [CrossRef]
- Padhye, S.; Dandawate, P.; Yusufi, M.; Ahmad, A.; Sarkar, F.H. Perspectives on medicinal properties of plumbagin and its analogs. Med. Res. Rev. 2012, 32, 1131–1158. [Google Scholar] [CrossRef]
- Ahmad, A.; Syed, F.A.; Singh, S.; Hadi, S.M. Prooxidant activity of resveratrol in the presence of copper ions: Mutagenicity in plasmid DNA. Toxicol. Lett. 2005, 159, 1–12. [Google Scholar] [CrossRef] [PubMed]
- Khan, N.S.; Ahmad, A.; Hadi, S.M. Anti-oxidant, pro-oxidant properties of tannic acid and its binding to DNA. Chem. Biol. Interact. 2000, 125, 177–189. [Google Scholar] [CrossRef]
- Checker, R.; Sharma, D.; Sandur, S.K.; Khanam, S.; Poduval, T.B. Anti-inflammatory effects of plumbagin are mediated by inhibition of NF-kappaB activation in lymphocytes. Int. Immunopharmacol. 2009, 9, 949–958. [Google Scholar] [CrossRef] [PubMed]
- Lukiw, W.J. NF-κB-regulated, proinflammatory miRNAs in Alzheimer’s disease. Alzheimer’s Res. Ther. 2012, 4, 1–11. [Google Scholar] [CrossRef]
- Son, T.G.; Camandola, S.; Arumugam, T.V.; Cutler, R.G.; Telljohann, R.S.; Mughal, M.R.; Moore, T.A.; Luo, W.; Yu, Q.S.; Johnson, D.A.; et al. Plumbagin, a novel Nrf2/ARE activator, protects against cerebral ischemia. J. Neurochem. 2010, 112, 1316–1326. [Google Scholar] [CrossRef]
- Colaric, M.; Veberic, R.; Solar, A.; Hudina, M.; Stampar, F. Phenolic acids, syringaldehyde, and juglone in fruits of different cultivars of Juglans regia L. J. Agric. Food Chem. 2005, 53, 6390–6396. [Google Scholar] [CrossRef]
- Inbaraj, J.J.; Chignell, C.F. Cytotoxic Action of Juglone and Plumbagin: A Mechanistic Study Using HaCaT Keratinocytes. Chem. Res. Toxicol. 2004, 17, 55–62. [Google Scholar] [CrossRef]
- Ahmad, T.; Suzuki, Y.J. Juglone in Oxidative Stress and Cell Signaling. Antioxidants 2019, 8, 91. [Google Scholar] [CrossRef]
- Clark, A.M.; Jurgens, T.M.; Hufford, C.D. Antimicrobial activity of juglone. Phyther. Res. 1990, 4, 11–14. [Google Scholar] [CrossRef]
- Zhang, W.; Liu, A.; Li, Y.; Zhao, X.; Lv, S.; Zhu, W.; Jin, Y. Anticancer activity and mechanism of juglone on human cervical carcinoma hela cells. Can. J. Physiol. Pharmacol. 2012, 90, 1553–1558. [Google Scholar] [CrossRef]
- Fang, F.; Chen, S.; Ma, J.; Cui, J.; Li, Q.; Meng, G.; Wang, L. Juglone suppresses epithelial-mesenchymal transition in prostate cancer cells via the protein kinase b/glycogen synthase kinase-3β/snail signaling pathway. Oncol. Lett. 2018, 16, 2579–2584. [Google Scholar] [CrossRef] [PubMed]
- Sugie, S.; Okamoto, K.; Rahman, K.M.W.; Tanaka, T.; Kawai, K.; Yamahara, J.; Mori, H. Inhibitory effects of plumbagin and juglone on azoxymethane-induced intestinal carcinogenesis in rats. Cancer Lett. 1998, 127, 177–183. [Google Scholar] [CrossRef]
- Chobot, V.; Hadacek, F. Milieu-dependent pro- and antioxidant activity of juglone may explain linear and nonlinear effects on seedling development. J. Chem. Ecol. 2009, 35, 383–390. [Google Scholar] [CrossRef] [PubMed]
- Liu, X.; Chen, Y.; Zhang, Y.; Du, J.; Lv, Y.; Mo, S.; Liu, Y.; Ding, F.; Wu, J.; Li, J. Juglone potentiates TRAIL-induced apoptosis in human melanoma cells via activating the ROS-p38-p53 pathway. Mol. Med. Rep. 2017, 16, 9645–9651. [Google Scholar] [CrossRef] [PubMed]
- Wang, J.; Liu, K.; Wang, X.F.; Sun, D.J. Juglone reduces growth and migration of U251 glioblastoma cells and disrupts angiogenesis. Oncol. Rep. 2017, 38, 1959–1966. [Google Scholar] [CrossRef]
- Jin, R. A DFT study on the radical scavenging activity of juglone and its derivatives. J. Mol. Struct. Theochem. 2010, 939, 9–13. [Google Scholar] [CrossRef]
- De Castro, E.; De Castro, S.H.; Johnson, T.E. Isolation of long-lived mutants in Caenorhabditis elegans using selection for resistance to juglone. Free Radic. Biol. Med. 2004, 37, 139–145. [Google Scholar] [CrossRef]
- Tamafo Fouegue, A.D.; Ghogomu, J.N.; Bikélé Mama, D.; Nkungli, N.K.; Younang, E. Structural and Antioxidant Properties of Compounds Obtained from Fe2+ Chelation by Juglone and Two of Its Derivatives: DFT, QTAIM, and NBO Studies. Bioinorg. Chem. Appl. 2016, 2016, 1–13. [Google Scholar] [CrossRef]
- Galano, A.; Mazzone, G.; Alvarez-Diduk, R.; Marino, T.; Alvarez-Idaboy, J.R.; Russo, N. Food Antioxidants: Chemical Insights at the Molecular Level. Annu. Rev. Food Sci. Technol. 2016, 7, 335–352. [Google Scholar] [CrossRef]
- Leopoldini, M.; Russo, N.; Toscano, M. The molecular basis of working mechanism of natural polyphenolic antioxidants. Food Chem. 2011, 125, 288–306. [Google Scholar] [CrossRef]
- Bentes, A.L.A.; Borges, R.S.; Monteiro, W.R.; De Macedo, L.G.M.; Alves, C.N. Structure of dihydrochalcones and related derivatives and their scavenging and antioxidant activity against oxygen and nitrogen radical species. Molecules 2011, 16, 1749–1760. [Google Scholar] [CrossRef] [PubMed]
- Leopoldini, M.; Marino, T.; Russo, N.; Toscano, M. Antioxidant properties of phenolic compounds: H-atom versus electron transfer mechanism. J. Phys. Chem. A 2004, 108, 4916–4922. [Google Scholar] [CrossRef]
- Gülçin, I.; Huyut, Z.; Elmastaş, M.; Aboul-Enein, H.Y. Radical scavenging and antioxidant activity of tannic acid. Arab. J. Chem. 2010, 3, 43–53. [Google Scholar] [CrossRef]
- Galas, M.C.; Dourlen, P.; Bégard, S.; Ando, K.; Blum, D.; Hamdane, M.; Buée, L. The peptidylprolyl cis/trans-isomerase Pin1 modulates stress-induced dephosphorylation of Tau in neurons: Implication in a pathological mechanism related to Alzheimer disease. J. Biol. Chem. 2006, 281, 19296–19304. [Google Scholar] [CrossRef]
- Pandareesh, M.D.; Chauhan, V.; Chauhan, A. Walnut Supplementation in the Diet Reduces Oxidative Damage and Improves Antioxidant Status in Transgenic Mouse Model of Alzheimer’s Disease. J. Alzheimers. Dis. 2018, 64, 1295–1305. [Google Scholar] [CrossRef]
- Hennig, L.; Christner, C.; Kipping, M.; Schelbert, B.; Rücknagel, K.P.; Grabley, S.; Küllertz, G.; Fischer, G. Selective inactivation of parvulin-like peptidyl-prolyl cis/trans isomerases by juglone. Biochemistry 1998, 37, 5953–5960. [Google Scholar] [CrossRef]
- Jordens, J.; Janssens, V.; Longin, S.; Stevens, I.; Martens, E.; Bultynck, G.; Engelborghs, Y.; Lescrinier, E.; Waelkens, E.; Goris, J.; et al. The protein phosphatase 2A phosphatase activator is a novel peptidyl-prolyl cis/trans-isomerase. J. Biol. Chem. 2006, 281, 6349–6357. [Google Scholar] [CrossRef]
- Hamdane, M.; Smet, C.; Sambo, A.V.; Leroy, A.; Wieruszeski, J.M.; Delobel, P.; Maurage, C.A.; Ghestem, A.; Wintjens, R.; Bégard, S.; et al. Pin1: A therapeutic target in Alzheimer neurodegeneration. J. Mol. Neurosci. 2002, 19, 275–287. [Google Scholar] [CrossRef]
- Khelifi, I.; Tourrette, A.; Dhouafli, Z.; Bouajila, J.; Efferth, T.; Abdelfatah, S.; Ksouri, R.; Hayouni, E.A. The antioxidant 2,3-dichloro,5,8-dihydroxy,1,4-naphthoquinone inhibits acetyl-cholinesterase activity and amyloid β 42 aggregation: A dual target therapeutic candidate compound for the treatment of Alzheimer’s disease. Biotechnol. Appl. Biochem. 2020, bab.1870. [Google Scholar] [CrossRef]
- Chung, Y.C.; Chien, C.T.; Teng, K.Y.; Chou, S.T. Antioxidative and mutagenic properties of Zanthoxylum ailanthoides Sieb & zucc. Food Chem. 2006, 97, 418–425. [Google Scholar] [CrossRef]
- Woo, H.C.; Seong, S.H.; Lee, S.A.; Xiang, H.H.; Kyong, S.L.; Myung, K.L.; Bang, Y.H.; Jai, S.R. Monoamine oxidase inhibitory naphthoquinones from the roots of Lithospermum erythrorhizon. Arch. Pharm. Res. 2005, 28, 400–404. [Google Scholar] [CrossRef]
- Mostert, S.; Petzer, A.; Petzer, J.P. Evaluation of Natural and Synthetic 1,4-naphthoquinones as Inhibitors of Monoamine Oxidase. Chem. Biol. Drug Des. 2016, 87, 737–746. [Google Scholar] [CrossRef] [PubMed]
- Cai, Z. Monoamine oxidase inhibitors: Promising therapeutic agents for Alzheimer’s disease (Review). Mol. Med. Rep. 2014, 9, 1533–1541. [Google Scholar] [CrossRef] [PubMed]
- Calabrese, V.; Cornelius, C.; Mancuso, C.; Pennisi, G.; Calafato, S.; Bellia, F.; Bates, T.E.; Giuffrida Stella, A.M.; Schapira, T.; Dinkova Kostova, A.T.; et al. Cellular stress response: A novel target for chemoprevention and nutritional neuroprotection in aging, neurodegenerative disorders and longevity. Neurochem. Res. 2008, 33, 2444–2471. [Google Scholar] [CrossRef] [PubMed]
- Neo Shin, N.; Jeon, H.; Jung, Y.; Baek, S.; Lee, S.; Yoo, H.C.; Bae, G.H.; Park, K.; Yang, S.H.; Han, J.M.; et al. Fluorescent 1,4-Naphthoquinones to Visualize Diffuse and Dense-Core Amyloid Plaques in APP/PS1 Transgenic Mouse Brains. ACS Chem. Neurosci. 2019, 10, 3031–3044. [Google Scholar] [CrossRef] [PubMed]
- Bolognesi, M.L.; Chiriano, G.P.; Bartolini, M.; Mancini, F.; Bottegoni, G.; Maestri, V.; Czvitkovich, S.; Windisch, M.; Cavalli, A.; Minarini, A.; et al. Synthesis of monomeric derivatives to probe memoquin’s bivalent interactions. J. Med. Chem. 2011, 54, 8299–8304. [Google Scholar] [CrossRef]
- Bolognesi, M.L.; Bartolini, M.; Tarozzi, A.; Morroni, F.; Lizzi, F.; Milelli, A.; Minarini, A.; Rosini, M.; Hrelia, P.; Andrisano, V.; et al. Multitargeted drugs discovery: Balancing anti-amyloid and anticholinesterase capacity in a single chemical entity. Bioorg. Med. Chem. Lett. 2011, 21, 2655–2658. [Google Scholar] [CrossRef]
- Bolognesi, M.L.; Banzi, R.; Bartolini, M.; Cavalli, A.; Tarozzi, A.; Andrisano, V.; Minarini, A.; Rosini, M.; Tumiatti, V.; Bergamini, C.; et al. Novel Class of Quinone-Bearing Polyamines as Multi-Target-Directed Ligands to Combat Alzheimer’s Disease. J. Med. Chem. 2007, 50, 4882–4897. [Google Scholar] [CrossRef]
- Bartolini, M.; Bertucci, C.; Cavrini, V.; Andrisano, V. β-Amyloid aggregation induced by human acetylcholinesterase: Inhibition studies. Biochem. Pharmacol. 2003, 65, 407–416. [Google Scholar] [CrossRef]
- Inestrosa, N.C.; Alvarez, A.; Pérez, C.A.; Moreno, R.D.; Vicente, M.; Linker, C.; Casanueva, O.I.; Soto, C.; Garrido, J. Acetylcholinesterase accelerates assembly of amyloid-β-peptides into Alzheimer’s fibrils: Possible role of the peripheral site of the enzyme. Neuron 1996, 16, 881–891. [Google Scholar] [CrossRef]
- Czvitkovich, S.; Duller, S.; Mathiesen, E.; Lorenzoni, K.; Imbimbo, B.P.; Hutter-Paier, B.; Windisch, M.; Wronski, R. Comparison of pharmacological modulation of APP metabolism in primary chicken telencephalic neurons and in a human neuroglioma cell line. J. Mol. Neurosci. 2011, 43, 257–267. [Google Scholar] [CrossRef] [PubMed][Green Version]
- Tonelli, M.; Catto, M.; Tasso, B.; Novelli, F.; Canu, C.; Iusco, G.; Pisani, L.; De Stradis, A.; Denora, N.; Sparatore, A.; et al. Multitarget therapeutic leads for Alzheimer’s disease: Quinolizidinyl derivatives of Bi- and tricyclic systems as dual inhibitors of cholinesterases and β-amyloid (Aβ) aggregation. ChemMedChem 2015, 10, 1040–1053. [Google Scholar] [CrossRef] [PubMed]
- Puglielli, L. Two endoplasmic reticulum (ER)/ER Golgi intermediate compartment-based lysine acetyltransferases posttranslationally regulate BACE1 levels. J. Biol. Chem. 2009, 284, 2482–2492. [Google Scholar]
- Ding, Y.; Ko, M.H.; Pehar, M.; Kotch, F.; Peters, N.R.; Luo, Y.; Salamat, S.M.; Puglielli, L. Biochemical inhibition of the acetyltransferases ATase1 and ATase2 reduces β-secretase (BACE1) levels and Aβ generation. J. Biol. Chem. 2012, 287, 8424–8433. [Google Scholar] [CrossRef]
- Morphy, R.; Kay, C.; Rankovic, Z. From magic bullets to designed multiple ligands. Drug Discov. Today 2004, 9, 641–651. [Google Scholar] [CrossRef]
- Watkins, P.B.; Zimmerman, H.J.; Knapp, M.J.; Gracon, S.I.; Lewis, K.W. Hepatotoxic Effects of Tacrine Administration in Patients with Alzheimer’s Disease. JAMA 1994, 271, 992–998. [Google Scholar] [CrossRef]
- Romero, A.; Cacabelos, R.; Oset-Gasque, M.J.; Samadi, A.; Marco-Contelles, J. Novel tacrine-related drugs as potential candidates for the treatment of Alzheimer’s disease. Bioorganic Med. Chem. Lett. 2013, 23, 1916–1922. [Google Scholar] [CrossRef]
- Chioua, M.; Serrano, E.; Dgachi, Y.; Martin, H.; Jun, D.; Janockova, J.; Sepsova, V.; Soukup, O.; Moraleda, I.; Chabchoub, F.; et al. Synthesis, Biological Assessment and Molecular Modeling of Racemic QuinoPyranoTacrines for Alzheimer’s Disease Therapy. ChemistrySelect 2018, 3, 461–466. [Google Scholar] [CrossRef]
- Nepovimova, E.; Uliassi, E.; Korabecny, J.; Peña-Altamira, L.E.; Samez, S.; Pesaresi, A.; Garcia, G.E.; Bartolini, M.; Andrisano, V.; Bergamini, C.; et al. Multitarget drug design strategy: Quinone-tacrine hybrids designed to block amyloid-β aggregation and to exert anticholinesterase and antioxidant effects. J. Med. Chem. 2014, 57, 8576–8589. [Google Scholar] [CrossRef]
- Galdeano, C.; Viayna, E.; Sola, I.; Formosa, X.; Camps, P.; Badia, A.; Clos, M.V.; Relat, J.; Ratia, M.; Bartolini, M.; et al. Huprine-tacrine heterodimers as anti-amyloidogenic compounds of potential interest against Alzheimer’s and prion diseases. J. Med. Chem. 2012, 55, 661–669. [Google Scholar] [CrossRef]
- Soukup, O.; Jun, D.; Zdarova-Karasova, J.; Patocka, J.; Musilek, K.; Korabecny, J.; Krusek, J.; Kaniakova, M.; Sepsova, V.; Mandikova, J.; et al. A Resurrection of 7-MEOTA: A Comparison with Tacrine. Curr. Alzheimer Res. 2013, 10, 893–906. [Google Scholar] [CrossRef] [PubMed]
- Pawar, A.P.; DuBay, K.F.; Zurdo, J.; Chiti, F.; Vendruscolo, M.; Dobson, C.M. Prediction of “aggregation-prone” and “aggregation- susceptible” regions in proteins associated with neurodegenerative diseases. J. Mol. Biol. 2005, 350, 379–392. [Google Scholar] [CrossRef] [PubMed]
- Zhang, T.; Xu, W.; Mu, Y.; Derreumaux, P. Atomic and dynamic insights into the beneficial effect of the 1,4-naphthoquinon-2-yl- l -tryptophan inhibitor on Alzheimer’s Aβ1-42 dimer in terms of aggregation and toxicity. ACS Chem. Neurosci. 2014, 5, 148–159. [Google Scholar] [CrossRef] [PubMed]
- Scherzer-Attali, R.; Farfara, D.; Cooper, I.; Levin, A.; Ben-Romano, T.; Trudler, D.; Vientrov, M.; Shaltiel-Karyo, R.; Shalev, D.E.; Segev-Amzaleg, N.; et al. Naphthoquinone-tyrptophan reduces neurotoxic Aβ*56 levels and improves cognition in Alzheimer’s disease animal model. Neurobiol. Dis. 2012, 46, 663–672. [Google Scholar] [CrossRef] [PubMed]
- Scherzer-Attali, R.; Shaltiel-Karyo, R.; Adalist, Y.H.; Segal, D.; Gazit, E. Generic inhibition of amyloidogenic proteins by two naphthoquinone-tryptophan hybrid molecules. Proteins Struct. Funct. Bioinforma. 2012, 80, 1962–1973. [Google Scholar] [CrossRef] [PubMed]
- Frenkel-Pinter, M.; Tal, S.; Scherzer-Attali, R.; Abu-Hussien, M.; Alyagor, I.; Eisenbaum, T.; Gazit, E.; Segal, D. Cl-NQTrp Alleviates Tauopathy Symptoms in a Model Organism through the Inhibition of Tau Aggregation-Engendered Toxicity. Neurodegener. Dis. 2017, 17, 73–82. [Google Scholar] [CrossRef]
- Frenkel-Pinter, M.; Tal, S.; Scherzer-Attali, R.; Abu-Hussien, M.; Alyagor, I.; Eisenbaum, T.; Gazit, E.; Segal, D. Naphthoquinone-Tryptophan Hybrid Inhibits Aggregation of the Tau-Derived Peptide PHF6 and Reduces Neurotoxicity. J. Alzheimer’s Dis. 2016, 51, 165–178. [Google Scholar] [CrossRef]
- Dammers, C.; Yolcu, D.; Kukuk, L.; Willbold, D.; Pickhardt, M.; Mandelkow, E.; Horn, A.H.C.; Sticht, H.; Malhis, M.N.; Will, N.; et al. Selection and characterization of tau binding D-enantiomeric peptides with potential for therapy of Alzheimer disease. PLoS ONE 2016, 11, e0167432. [Google Scholar] [CrossRef]
- Friedhoff, P.; Von Bergen, M.; Mandelkow, E.M.; Mandelkow, E. Structure of tau protein and assembly into paired helical filaments. Biochim. Biophys. Acta—Mol. Basis Dis. 2000, 1502, 122–132. [Google Scholar] [CrossRef]
- Scherzer-Attali, R.; Convertino, M.; Pellarin, R.; Gazit, E.; Segal, D.; Caflisch, A. Methylations of tryptophan-modified naphthoquinone affect its inhibitory potential toward aβ aggregation. J. Phys. Chem. B 2013, 117, 1780–1789. [Google Scholar] [CrossRef]
- Paul, A.; Viswanathan, G.K.; Mahapatra, S.; Balboni, G.; Pacifico, S.; Gazit, E.; Segal, D. Antagonistic Activity of Naphthoquinone-Based Hybrids toward Amyloids Associated with Alzheimer’s Disease and Type-2 Diabetes. ACS Chem. Neurosci. 2019, 10, 3510–3520. [Google Scholar] [CrossRef] [PubMed]
- Linse, S. Mechanism of amyloid protein aggregation and the role of inhibitors. Pure Appl. Chem. 2019, 91, 211–229. [Google Scholar] [CrossRef]
- Arosio, P.; Vendruscolo, M.; Dobson, C.M.; Knowles, T.P.J. Chemical kinetics for drug discovery to combat protein aggregation diseases. Trends Pharmacol. Sci. 2014, 35, 127–135. [Google Scholar] [CrossRef] [PubMed]
- Gazit, E. A possible role for π-stacking in the self-assembly of amyloid fibrils. FASEB J. 2002, 16, 77–83. [Google Scholar] [CrossRef]
- Bertolani, A.; Pirrie, L.; Stefan, L.; Houbenov, N.; Haataja, J.S.; Catalano, L.; Terraneo, G.; Giancane, G.; Valli, L.; Milani, R.; et al. Supramolecular amplification of amyloid self-assembly by iodination. Nat. Commun. 2015, 6, 1–9. [Google Scholar] [CrossRef]
- Makin, O.S.; Atkins, E.; Sikorski, P.; Johansson, J.; Serpell, L.C. Molecular basis for amyloid fibril formation and stability. Proc. Natl. Acad. Sci. USA 2005, 102, 315–320. [Google Scholar] [CrossRef]
- KrishnaKumar, V.G.; Paul, A.; Gazit, E.; Segal, D. Mechanistic insights into remodeled Tau-derived PHF6 peptide fibrils by Naphthoquinone-Tryptophan hybrids. Sci. Rep. 2018, 8, 1–12. [Google Scholar] [CrossRef]
- Convertino, M.; Pellarin, R.; Catto, M.; Carotti, A.; Caflisch, A. 9,10-Anthraquinone hinders β-aggregation: How does a small molecule interfere with Aβ-peptide amyloid fibrillation? Protein Sci. 2009, 18, 792–800. [Google Scholar] [CrossRef]
- Churches, Q.I.; Caine, J.; Cavanagh, K.; Epa, V.C.; Waddington, L.; Tranberg, C.E.; Meyer, A.G.; Varghese, J.N.; Streltsov, V.; Duggan, P.J. Naturally occurring polyphenolic inhibitors of amyloid β aggregation. Bioorg. Med. Chem. Lett. 2014, 24, 3108–3112. [Google Scholar] [CrossRef]
- Chaturvedi, S.K.; Zaidi, N.; Alam, P.; Khan, J.M.; Qadeer, A.; Siddique, I.A.; Asmat, S.; Zaidi, Y.; Khan, R.H. Unraveling Comparative Anti-Amyloidogenic Behavior of Pyrazinamide and D-Cycloserine: A Mechanistic Biophysical Insight. PLoS ONE 2015, 10, e0136528. [Google Scholar] [CrossRef]
- Tjernberg, L.O.; Näslundt, J.; Lindqvist, F.; Johansson, J.; Karlström, A.R.; Thyberg, J.; Tereniust, L.; Nordstedt, C. Arrest of β-amyloid fibril formation by a pentapeptide ligand. J. Biol. Chem. 1996, 271, 8545–8548. [Google Scholar] [CrossRef] [PubMed]
- Diaz-Muñoz, G.; Miranda, I.L.; Sartori, S.K.; de Rezende, D.C.; Diaz, M.A.N. Anthraquinones: An Overview. Stud. Nat. Prod. Chem. 2018, 58, 313–338. [Google Scholar] [CrossRef]
- Li, Y.; Jiang, J.G. Health functions and structure-activity relationships of natural anthraquinones from plants. Food Funct. 2018, 9, 6063–6080. [Google Scholar] [CrossRef] [PubMed]
- Malik, E.M.; Müller, C.E. Anthraquinones As Pharmacological Tools and Drugs. Med. Res. Rev. 2016, 36, 705–748. [Google Scholar] [CrossRef]
- Jung, H.A.; Ali, M.Y.; Jung, H.J.; Jeong, H.O.; Chung, H.Y.; Choi, J.S. Inhibitory activities of major anthraquinones and other constituents from Cassia obtusifolia against β-secretase and cholinesterases. J. Ethnopharmacol. 2016, 191, 152–160. [Google Scholar] [CrossRef]
- Orhan, I.; Tosun, F.; Şener, B. Coumarin, anthroquinone and stilbene derivatives with anticholinesterase activity. Z. Naturforsch Sect. C J. Biosci. 2008, 63, 366–370. [Google Scholar] [CrossRef]
- Zhang, J.H.; Xin, H.L.; Xu, Y.M.; Shen, Y.; He, Y.Q.; Lin, B.; Song, H.T.; Yang, H.Y.; Qin, L.P.; Zhang, Q.Y.; et al. Morinda officinalis How—A comprehensive review of traditional uses, phytochemistry and pharmacology. J. Ethnopharmacol. 2018, 213, 230–255. [Google Scholar] [CrossRef]
- Lee, Y.K.; Bang, H.J.; Oh, J. Bin; Whang, W.K. Bioassay-Guided Isolated Compounds from Morinda officinalis Inhibit Alzheimer’s Disease Pathologies. Molecules 2017, 22, 1638. [Google Scholar] [CrossRef]
- Pickhardt, M.; Gazova, Z.; Von Bergen, M.; Khlistunova, I.; Wang, Y.; Hascher, A.; Mandelkow, E.M.; Biernat, J.; Mandelkow, E. Anthraquinones inhibit tau aggregation and dissolve Alzheimer’s paired helical filaments in vitro and in cells. J. Biol. Chem. 2005, 280, 3628–3635. [Google Scholar] [CrossRef]
- Paranjape, S.R.; Chiang, Y.M.; Sanchez, J.F.; Entwistle, R.; Wang, C.C.C.; Oakley, B.R.; Gamblin, T.C. Inhibition of tau aggregation by three aspergillus nidulans secondary metabolites: 2, ω -dihydroxyemodin, asperthecin, and asperbenzaldehyde. Planta Med. 2014, 80, 77–85. [Google Scholar] [CrossRef]
- Viswanathan, G.K.; Shwartz, D.; Losev, Y.; Arad, E.; Shemesh, C.; Pichinuk, E.; Engel, H.; Raveh, A.; Jelinek, R.; Cooper, I.; et al. Purpurin modulates Tau-derived VQIVYK fibrillization and ameliorates Alzheimer’s disease-like symptoms in animal model. Cell. Mol. Life Sci. 2020, 77, 2795–2813. [Google Scholar] [CrossRef] [PubMed]
- Bhattacharya, K.; Rank, K.B.; Evans, D.B.; Sharma, S.K. Role of cysteine-291 and cysteine-322 in the polymerization of human tau into Alzheimer-like filaments. Biochem. Biophys. Res. Commun. 2001, 285, 20–26. [Google Scholar] [CrossRef] [PubMed]
- Areche, C.; Zapata, F.; González, M.; Díaz, E.; Montecinos, R.; Hernández, M.; Melo, F.; Cornejo, A. Anthraquinone Derivative Reduces Tau Oligomer Progression by Inhibiting Cysteine-Cysteine Interaction. ChemistryOpen 2019, 8, 554–559. [Google Scholar] [CrossRef] [PubMed]
- Bandiera, T.; Lansen, J.; Post, C.; Varasi, M. Inhibitors of Aβ Peptide Aggregation as Potential anti-Alzheimer Agents. Curr. Med. Chem. 1997, 4, 159–170. [Google Scholar]
- Colombo, R.; Carotti, A.; Catto, M.; Racchi, M.; Lanni, C.; Verga, L.; Caccialanza, G.; De Lorenzi, E. CE can identify small molecules that selectively target soluble oligomers of amyloid β protein and display antifibrillogenic activity. Electrophoresis 2009, 30, 1418–1429. [Google Scholar] [CrossRef] [PubMed]
- Yang, L.; Peacey, E.; Dickson, J.; Donahue, C.P.; Zheng, S.; Varani, G.; Wolfe, M.S. Mitoxantrone analogues as ligands for a stem-loop structure of tau Pre-mRNA. J. Med. Chem. 2009, 52, 6523–6526. [Google Scholar] [CrossRef]
- Hong, C.; Luo, W.; Yao, D.; Su, Y. Bin; Zhang, X.; Tian, R.G.; Wang, C.J. Novel aromatic-polyamine conjugates as cholinesterase inhibitors with notable selectivity toward butyrylcholinesterase. Bioorg. Med. Chem. 2014, 22, 3213–3219. [Google Scholar] [CrossRef]
- Wang, J.; Li, W.; Qin, J.; Wang, L.; Wei, S.; Tang, H. Assessment of novel azaanthraquinone derivatives as potent multi-target inhibitors of inflammation and amyloid-β aggregation in Alzheimer’s disease. Bioorg. Chem. 2019, 83, 477–486. [Google Scholar] [CrossRef]
- Crowe, A.; Ballatore, C.; Hyde, E.; Trojanowski, J.Q.; Lee, V.M.Y. High throughput screening for small molecule inhibitors of heparin-induced tau fibril formation. Biochem. Biophys. Res. Commun. 2007, 358, 1–6. [Google Scholar] [CrossRef]
- Viayna, E.; Sola, I.; Bartolini, M.; De Simone, A.; Tapia-Rojas, C.; Serrano, F.G.; Sabaté, R.; Juárez-Jiménez, J.; Pérez, B.; Luque, F.J.; et al. Synthesis and multitarget biological profiling of a novel family of rhein derivatives as disease-modifying anti-Alzheimer agents. J. Med. Chem. 2014, 57, 2549–2567. [Google Scholar] [CrossRef]
- Camps, P.; Cusack, B.; Mallender, W.D.; El Achab, R.E.; Morral, J.; Muñoz-Torrero, D.; Rosenberry, T.L. Huprine X is a novel high-affinity inhibitor of acetylcholinesterase that is of interest for treatment of Alzheimer’s disease. Mol. Pharmacol. 2000, 57, 409–417. [Google Scholar] [PubMed]
- Pérez-Areales, F.J.; Betari, N.; Viayna, A.; Pont, C.; Espargaró, A.; Bartolini, M.; De Simone, A.; Rinaldi Alvarenga, J.F.; Pérez, B.; Sabate, R.; et al. Design, synthesis and multitarget biological profiling of second-generation anti-Alzheimer rhein–huprine hybrids. Future Med. Chem. 2017, 9, 965–981. [Google Scholar] [CrossRef] [PubMed]
- Li, S.Y.; Jiang, N.; Xie, S.S.; Wang, K.D.G.; Wang, X.B.; Kong, L.Y. Design, synthesis and evaluation of novel tacrine-rhein hybrids as multifunctional agents for the treatment of Alzheimer’s disease. Org. Biomol. Chem. 2014, 12, 801–814. [Google Scholar] [CrossRef] [PubMed]
- Carvajal, F.J.; Inestrosa, N.C. Interactions of AChE with Aβ Aggregates in Alzheimer’s Brain: Therapeutic Relevance of IDN. Front. Mol. Neurosci. 2011, 4, 19. [Google Scholar] [CrossRef]
- Turon-Estrada, A.; López-Pousa, S.; Gelada-Batlle, E.; Garre-Olmo, J.; Lozano-Gallego, M.; Hernández-Ferràndiz, M.; Fajardo-Tibau, C.; Morante-Muñoz, V.; Vilalta-Franch, J. Tolerance and adverse events of treatment with acetylcholinesterase inhibitors in a clinical sample of patients with very slight and mild Alzheimer’s disease over a six-month period. Rev. Neurol. 2003, 36, 421–424. [Google Scholar] [CrossRef]
- Delrieu, J.; Piau, A.; Caillaud, C.; Voisin, T.; Vellas, B. Managing Cognitive Dysfunction through the Continuum of Alzheimerʼs Disease. CNS Drugs 2011, 25, 213–226. [Google Scholar] [CrossRef]
- Scheltens, P.; Blennow, K.; Breteler, M.M.B.; de Strooper, B.; Frisoni, G.B.; Salloway, S.; Van der Flier, W.M. Alzheimer’s disease. Lancet 2016, 388, 505–517. [Google Scholar] [CrossRef]
- Prati, F.; Cavalli, A.; Bolognesi, M. Navigating the Chemical Space of Multitarget-Directed Ligands: From Hybrids to Fragments in Alzheimer’s Disease. Molecules 2016, 21, 466. [Google Scholar] [CrossRef]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Campora, M.; Francesconi, V.; Schenone, S.; Tasso, B.; Tonelli, M. Journey on Naphthoquinone and Anthraquinone Derivatives: New Insights in Alzheimer’s Disease. Pharmaceuticals 2021, 14, 33. https://doi.org/10.3390/ph14010033
Campora M, Francesconi V, Schenone S, Tasso B, Tonelli M. Journey on Naphthoquinone and Anthraquinone Derivatives: New Insights in Alzheimer’s Disease. Pharmaceuticals. 2021; 14(1):33. https://doi.org/10.3390/ph14010033
Chicago/Turabian StyleCampora, Marta, Valeria Francesconi, Silvia Schenone, Bruno Tasso, and Michele Tonelli. 2021. "Journey on Naphthoquinone and Anthraquinone Derivatives: New Insights in Alzheimer’s Disease" Pharmaceuticals 14, no. 1: 33. https://doi.org/10.3390/ph14010033
APA StyleCampora, M., Francesconi, V., Schenone, S., Tasso, B., & Tonelli, M. (2021). Journey on Naphthoquinone and Anthraquinone Derivatives: New Insights in Alzheimer’s Disease. Pharmaceuticals, 14(1), 33. https://doi.org/10.3390/ph14010033