51
|
Bortolami M, Rocco D, Messore A, Di Santo R, Costi R, Madia VN, Scipione L, Pandolfi F. Acetylcholinesterase inhibitors for the treatment of Alzheimer's disease - a patent review (2016-present). Expert Opin Ther Pat 2021; 31:399-420. [PMID: 33428491 DOI: 10.1080/13543776.2021.1874344] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Introduction - AD, the most common form of dementia, has a multifactorial etiology, and the current therapy (AChEIs and memantine) is unable to interrupt its progress and fatal outcome. This is reflected in the research programs that are oriented toward the development of new therapeutics able to operate on multiple targets involved in the disease progression.Areas covered - The patents from 2016 to present regarding the use of AChEIs in AD, concerns the development of new AChEIs, multitarget or multifunctional ligands, or the associations of currently used AChEIs with other compounds acting on different targets involved in the AD.Expert opinion - The development of new multitarget AChEIs promises to identify compounds with great therapeutic potential but requires more time and effort in order to obtain drugs with the optimal pharmacodynamic profile. Otherwise, the research on new combinations of existing drugs, with known pharmacodynamic and ADME profile, could shorten the time and reduce the costs to develop a new therapeutic treatment for AD. From the analyzed data, it seems more likely that a response to the urgent need to develop effective treatments for AD therapy could come more quickly from studies on drug combinations than from the development of new AChEIs.
Collapse
Affiliation(s)
- Martina Bortolami
- Department of Scienze Di Base E Applicate per l'Ingegneria, Sapienza University of Rome, Rome, Italy
| | - Daniele Rocco
- Department of Scienze Di Base E Applicate per l'Ingegneria, Sapienza University of Rome, Rome, Italy
| | - Antonella Messore
- Department of Chimica E Tecnologia Del Farmaco, Dipartimento Di Eccellenza 2018-2022, Istituto Pasteur-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy
| | - Roberto Di Santo
- Department of Chimica E Tecnologia Del Farmaco, Dipartimento Di Eccellenza 2018-2022, Istituto Pasteur-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy
| | - Roberta Costi
- Department of Chimica E Tecnologia Del Farmaco, Dipartimento Di Eccellenza 2018-2022, Istituto Pasteur-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy
| | - Valentina Noemi Madia
- Department of Chimica E Tecnologia Del Farmaco, Dipartimento Di Eccellenza 2018-2022, Istituto Pasteur-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy
| | - Luigi Scipione
- Department of Chimica E Tecnologia Del Farmaco, Dipartimento Di Eccellenza 2018-2022, Istituto Pasteur-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy
| | - Fabiana Pandolfi
- Department of Scienze Di Base E Applicate per l'Ingegneria, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
52
|
Panda SS, Jhanji N. Natural Products as Potential Anti-Alzheimer Agents. Curr Med Chem 2021; 27:5887-5917. [PMID: 31215372 DOI: 10.2174/0929867326666190618113613] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 03/20/2019] [Accepted: 05/28/2019] [Indexed: 01/18/2023]
Abstract
Medicinal plants have curative properties due to the presence of various complex chemical substances of different composition, which are found as secondary metabolites in one or more parts of the plant. The diverse secondary metabolites play an important role in the prevention and cure of various diseases including neurodegenerative diseases like Alzheimer's disease. Naturally occurring compounds such as flavonoids, polyphenols, alkaloids, and glycosides found in various parts of the plant and/or marine sources may potentially protect neurodegeneration as well as improve memory and cognitive function. Many natural compounds show anti-Alzheimer activity through specific pharmacological mechanisms like targeting β-amyloid, Beta-secretase 1 and Acetylcholinesterase. In this review, we have compiled more than 130 natural products with a broad diversity in the class of compounds, which were isolated from different sources showing anti- Alzheimer properties.
Collapse
Affiliation(s)
- Siva S Panda
- Department of Chemistry & Physics, Augusta University, Augusta, Georgia 30912, United States
| | - Nancy Jhanji
- Department of Chemistry & Physics, Augusta University, Augusta, Georgia 30912, United States
| |
Collapse
|
53
|
Zhang Z, Guo J, Cheng M, Zhou W, Wan Y, Wang R, Fang Y, Jin Y, Liu J, Xie SS. Design, synthesis, and biological evaluation of novel xanthone-alkylbenzylamine hybrids as multifunctional agents for the treatment of Alzheimer's disease. Eur J Med Chem 2021; 213:113154. [PMID: 33476932 DOI: 10.1016/j.ejmech.2021.113154] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 12/15/2020] [Accepted: 12/31/2020] [Indexed: 12/26/2022]
Abstract
In this study, a series of multifunctional hybrids against Alzheimer's disease were designed and obtained by conjugating the pharmacophores of xanthone and alkylbenzylamine through the alkyl linker. Biological activity results demonstrated that compound 4j was the most potent and balanced dual ChEs inhibitor with IC50 values 0.85 μM and 0.59 μM for eeAChE and eqBuChE, respectively. Kinetic analysis and docking study indicated that compound 4j was a mixed-type inhibitor for both AChE and BuChE. Additionally, it exhibited good abilities to penetrate BBB, scavenge free radicals (4.6 trolox equivalent) and selectively chelate with Cu2+ and Al3+ at a 1:1.4 ligand/metal molar ratio. Importantly, after assessments of cytotoxic and acute toxicity, we found compound 4j could improve memory function of scopolamine-induced amnesia mice. Hence, the compound 4j can be considered as a promising lead compound for further investigation in the treatment of AD.
Collapse
Affiliation(s)
- Zhipeng Zhang
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330006, PR China
| | - Jie Guo
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330006, PR China
| | - Maojun Cheng
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330006, PR China
| | - Weixin Zhou
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330006, PR China
| | - Yang Wan
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330006, PR China
| | - Rikang Wang
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330006, PR China
| | - Yuanying Fang
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330006, PR China
| | - Yi Jin
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330006, PR China
| | - Jing Liu
- School of Pharmacy, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330006, PR China.
| | - Sai-Sai Xie
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330006, PR China.
| |
Collapse
|
54
|
Campora M, Francesconi V, Schenone S, Tasso B, Tonelli M. Journey on Naphthoquinone and Anthraquinone Derivatives: New Insights in Alzheimer's Disease. Pharmaceuticals (Basel) 2021; 14:33. [PMID: 33466332 PMCID: PMC7824805 DOI: 10.3390/ph14010033] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 12/27/2020] [Accepted: 12/30/2020] [Indexed: 12/11/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease that is characterized by memory loss, cognitive impairment, and functional decline leading to dementia and death. AD imposes neuronal death by the intricate interplay of different neurochemical factors, which continue to inspire the medicinal chemist as molecular targets for the development of new agents for the treatment of AD with diverse mechanisms of action, but also depict a more complex AD scenario. Within the wide variety of reported molecules, this review summarizes and offers a global overview of recent advancements on naphthoquinone (NQ) and anthraquinone (AQ) derivatives whose more relevant chemical features and structure-activity relationship studies will be discussed with a view to providing the perspective for the design of viable drugs for the treatment of AD. In particular, cholinesterases (ChEs), β-amyloid (Aβ) and tau proteins have been identified as key targets of these classes of compounds, where the NQ or AQ scaffold may contribute to the biological effect against AD as main unit or significant substructure. The multitarget directed ligand (MTDL) strategy will be described, as a chance for these molecules to exhibit significant potential on the road to therapeutics for AD.
Collapse
Affiliation(s)
| | | | | | | | - Michele Tonelli
- Dipartimento di Farmacia, Università degli Studi di Genova, Viale Benedetto XV, 3, 16132 Genova, Italy; (M.C.); (V.F.); (S.S.); (B.T.)
| |
Collapse
|
55
|
Discovery of potent glycogen synthase kinase 3/cholinesterase inhibitors with neuroprotection as potential therapeutic agent for Alzheimer’s disease. Bioorg Med Chem 2021; 30:115940. [DOI: 10.1016/j.bmc.2020.115940] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 12/02/2020] [Accepted: 12/06/2020] [Indexed: 01/02/2023]
|
56
|
Li J, Du Q, Li N, Du S, Sun Z. Alpiniae oxyphyllae Fructus and Alzheimer's disease: An update and current perspective on this traditional Chinese medicine. Biomed Pharmacother 2020; 135:111167. [PMID: 33383373 DOI: 10.1016/j.biopha.2020.111167] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 12/05/2020] [Accepted: 12/14/2020] [Indexed: 02/01/2023] Open
Abstract
Alzheimer's disease (AD) is a common progressive neuro-degenerative disease, and the morbidity and mortality are still on the rise. In spite of recent advances in AD treatment, their clinical efficacy has been limited, non-curative and easy to drug resistance. Alpiniae oxyphyllae Fructus (AOF), derived from the dried and mature fruits of the Zingiberaceae plant Alpinia oxyphylla Miq, is a choice in traditional Chinese medicine to treat AD, which has a good effect and has been used for a long time. Recent studies have demonstrated its potent activities in modulating multiple signaling pathways associated with β-amyloid deposition, tau protein phosphorylation, chronic inflammation, oxidative stress. The neuropharmacological mechanism of AOF in AD have been fully illustrated in numerous studies. In this review, we first briefly described the active components of AOF and related mechanism for treating AD. And we also provide a systematic overview of recent progress on the pharmacokinetic characteristics of the active ingredients of AOF and analyzed their bioavailability differences in the development of AD. Thus, AOF hold a great therapeutic potential in the treatment of AD and is worthy of further research and promotion.
Collapse
Affiliation(s)
- Jia Li
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Qiuzheng Du
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Na Li
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Shuzhang Du
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| | - Zhi Sun
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
57
|
Pérez-Sánchez H, den Haan H, Pérez-Garrido A, Peña-García J, Chakraborty S, Erdogan Orhan I, Senol Deniz FS, Villalgordo JM. Combined Structure and Ligand-Based Design of Selective Acetylcholinesterase Inhibitors. J Chem Inf Model 2020; 61:467-480. [PMID: 33320652 DOI: 10.1021/acs.jcim.0c00463] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Acetylcholinesterase is a prime target for therapeutic intervention in Alzheimer's disease. Acetylcholinesterase inhibitors (AChEIs) are used to improve cognitive abilities, playing therefore an important role in disease management. Drug repurposing screening has been performed on a corporate chemical library containing 11 353 compounds using a target fishing approach comprising three-dimensional (3D) shape similarity and pharmacophore modeling against an approved drug database, Drugbank. This initial screening identified 108 hits. Among them, eight molecules showed structural similarity to the known AChEI drug, pyridostigmine. Further structure-based screening using a pharmacophore-guided rescoring method identifies one more potential hit. Experimental evaluations of the identified hits sieve out a highly selective AChEI scaffold. Further lead optimization using a substructure search approach identifies 24 new potential hits. Three of the 24 compounds (compounds 10b, 10h, and 10i) based on a 6-(2-(pyrrolidin-1-yl)pyrimidin-4-yl)-thiazolo[3,2-a]pyrimidine scaffold showed highly promising AChE inhibition ability with IC50 values of 13.10 ± 0.53, 16.02 ± 0.46, and 6.22 ± 0.54 μM, respectively. Moreover, these compounds are highly selective toward AChE. Compound 10i shows AChE inhibitory activity similar to a known Food and Drug Administration (FDA)-approved drug, galantamine, but with even better selectivity. Interaction analysis reveals that hydrophobic and hydrogen-bonding interactions are the primary driving forces responsible for the observed high affinity of the compound with AChE.
Collapse
Affiliation(s)
- Horacio Pérez-Sánchez
- Structural Bioinformatics and High Performance Computing Group (BIO-HPC), Universidad Católica San Antonio de Murcia (UCAM), Campus de los Jerónimos, Guadalupe 30107, Spain
| | - Helena den Haan
- Structural Bioinformatics and High Performance Computing Group (BIO-HPC), Universidad Católica San Antonio de Murcia (UCAM), Campus de los Jerónimos, Guadalupe 30107, Spain.,Parque Tecnológico de Fuente Álamo, Villapharma Research, Ctra. El Estrecho-Lobosillo, Km. 2,5- Av. Azul, 30320 Fuente Álamo de Murcia, Murcia, Spain
| | - Alfonso Pérez-Garrido
- Structural Bioinformatics and High Performance Computing Group (BIO-HPC), Universidad Católica San Antonio de Murcia (UCAM), Campus de los Jerónimos, Guadalupe 30107, Spain
| | - Jorge Peña-García
- Structural Bioinformatics and High Performance Computing Group (BIO-HPC), Universidad Católica San Antonio de Murcia (UCAM), Campus de los Jerónimos, Guadalupe 30107, Spain
| | | | - Ilkay Erdogan Orhan
- Department of Pharmacognosy, Faculty of Pharmacy, Gazi University, 06330 Ankara, Turkey
| | | | - José Manuel Villalgordo
- Parque Tecnológico de Fuente Álamo, Villapharma Research, Ctra. El Estrecho-Lobosillo, Km. 2,5- Av. Azul, 30320 Fuente Álamo de Murcia, Murcia, Spain
| |
Collapse
|
58
|
Makhaeva GF, Kovaleva NV, Rudakova EV, Boltneva NP, Lushchekina SV, Faingold II, Poletaeva DA, Soldatova YV, Kotelnikova RA, Serkov IV, Ustinov AK, Proshin AN, Radchenko EV, Palyulin VA, Richardson RJ. New Multifunctional Agents Based on Conjugates of 4-Amino-2,3-polymethylenequinoline and Butylated Hydroxytoluene for Alzheimer's Disease Treatment. Molecules 2020; 25:molecules25245891. [PMID: 33322783 PMCID: PMC7763995 DOI: 10.3390/molecules25245891] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 12/08/2020] [Accepted: 12/09/2020] [Indexed: 01/18/2023] Open
Abstract
New hybrids of 4-amino-2,3-polymethylenequinoline with different sizes of the aliphatic ring linked to butylated hydroxytoluene (BHT) by enaminoalkyl (7) or aminoalkyl (8) spacers were synthesized as potential multifunctional agents for Alzheimer's disease (AD) treatment. All compounds were potent inhibitors of acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) with selectivity toward BChE. Lead compound 8c, 2,6-di-tert-butyl-4-{[2-(7,8,9,10- tetrahydro-6H-cyclohepta[b]quinolin-11-ylamino)-ethylimino]-methyl}-phenol exhibited an IC50(AChE) = 1.90 ± 0.16 µM, IC50(BChE) = 0.084 ± 0.008 µM, and 13.6 ± 1.2% propidium displacement at 20 μM. Compounds possessed low activity against carboxylesterase, indicating likely absence of clinically unwanted drug-drug interactions. Kinetics were consistent with mixed-type reversible inhibition of both cholinesterases. Docking indicated binding to catalytic and peripheral AChE sites; peripheral site binding along with propidium displacement suggest the potential of the hybrids to block AChE-induced β-amyloid aggregation, a disease-modifying effect. Compounds demonstrated high antioxidant activity in ABTS and FRAP assays as well as inhibition of luminol chemiluminescence and lipid peroxidation in mouse brain homogenates. Conjugates 8 with amine-containing spacers were better antioxidants than those with enamine spacers 7. Computational ADMET profiles for all compounds predicted good blood-brain barrier distribution (permeability), good intestinal absorption, and medium cardiac toxicity risk. Overall, based on their favorable pharmacological and ADMET profiles, conjugates 8 appear promising as candidates for AD therapeutics.
Collapse
Affiliation(s)
- Galina F. Makhaeva
- Institute of Physiologically Active Compounds Russian Academy of Sciences, 142432 Chernogolovka, Russia; (G.F.M.); (N.V.K.); (E.V.R.); (N.P.B.); (S.V.L.); (I.V.S.); (A.K.U.); (A.N.P.); (E.V.R.); (V.A.P.)
| | - Nadezhda V. Kovaleva
- Institute of Physiologically Active Compounds Russian Academy of Sciences, 142432 Chernogolovka, Russia; (G.F.M.); (N.V.K.); (E.V.R.); (N.P.B.); (S.V.L.); (I.V.S.); (A.K.U.); (A.N.P.); (E.V.R.); (V.A.P.)
| | - Elena V. Rudakova
- Institute of Physiologically Active Compounds Russian Academy of Sciences, 142432 Chernogolovka, Russia; (G.F.M.); (N.V.K.); (E.V.R.); (N.P.B.); (S.V.L.); (I.V.S.); (A.K.U.); (A.N.P.); (E.V.R.); (V.A.P.)
| | - Natalia P. Boltneva
- Institute of Physiologically Active Compounds Russian Academy of Sciences, 142432 Chernogolovka, Russia; (G.F.M.); (N.V.K.); (E.V.R.); (N.P.B.); (S.V.L.); (I.V.S.); (A.K.U.); (A.N.P.); (E.V.R.); (V.A.P.)
| | - Sofya V. Lushchekina
- Institute of Physiologically Active Compounds Russian Academy of Sciences, 142432 Chernogolovka, Russia; (G.F.M.); (N.V.K.); (E.V.R.); (N.P.B.); (S.V.L.); (I.V.S.); (A.K.U.); (A.N.P.); (E.V.R.); (V.A.P.)
- Emanuel Institute of Biochemical Physics Russian Academy of Sciences, 119334 Moscow, Russia
| | - Irina I. Faingold
- Institute of Problems of Chemical Physics of Russian Academy of Sciences, 142432 Chernogolovka, Russia; (I.I.F.); (D.A.P.); (Y.V.S.); (R.A.K.)
| | - Darya A. Poletaeva
- Institute of Problems of Chemical Physics of Russian Academy of Sciences, 142432 Chernogolovka, Russia; (I.I.F.); (D.A.P.); (Y.V.S.); (R.A.K.)
| | - Yuliya V. Soldatova
- Institute of Problems of Chemical Physics of Russian Academy of Sciences, 142432 Chernogolovka, Russia; (I.I.F.); (D.A.P.); (Y.V.S.); (R.A.K.)
| | - Raisa A. Kotelnikova
- Institute of Problems of Chemical Physics of Russian Academy of Sciences, 142432 Chernogolovka, Russia; (I.I.F.); (D.A.P.); (Y.V.S.); (R.A.K.)
| | - Igor V. Serkov
- Institute of Physiologically Active Compounds Russian Academy of Sciences, 142432 Chernogolovka, Russia; (G.F.M.); (N.V.K.); (E.V.R.); (N.P.B.); (S.V.L.); (I.V.S.); (A.K.U.); (A.N.P.); (E.V.R.); (V.A.P.)
| | - Anatoly K. Ustinov
- Institute of Physiologically Active Compounds Russian Academy of Sciences, 142432 Chernogolovka, Russia; (G.F.M.); (N.V.K.); (E.V.R.); (N.P.B.); (S.V.L.); (I.V.S.); (A.K.U.); (A.N.P.); (E.V.R.); (V.A.P.)
| | - Alexey N. Proshin
- Institute of Physiologically Active Compounds Russian Academy of Sciences, 142432 Chernogolovka, Russia; (G.F.M.); (N.V.K.); (E.V.R.); (N.P.B.); (S.V.L.); (I.V.S.); (A.K.U.); (A.N.P.); (E.V.R.); (V.A.P.)
| | - Eugene V. Radchenko
- Institute of Physiologically Active Compounds Russian Academy of Sciences, 142432 Chernogolovka, Russia; (G.F.M.); (N.V.K.); (E.V.R.); (N.P.B.); (S.V.L.); (I.V.S.); (A.K.U.); (A.N.P.); (E.V.R.); (V.A.P.)
- Department of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Vladimir A. Palyulin
- Institute of Physiologically Active Compounds Russian Academy of Sciences, 142432 Chernogolovka, Russia; (G.F.M.); (N.V.K.); (E.V.R.); (N.P.B.); (S.V.L.); (I.V.S.); (A.K.U.); (A.N.P.); (E.V.R.); (V.A.P.)
- Department of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Rudy J. Richardson
- Department of Environmental Health Sciences, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
- Center of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
- Michigan Institute for Computational Discovery and Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- Correspondence: ; Tel.: +1-734-936-0769
| |
Collapse
|
59
|
Atali S, Dorandish S, Devos J, Williams A, Price D, Taylor J, Guthrie J, Heyl D, Evans HG. Interaction of amyloid beta with humanin and acetylcholinesterase is modulated by ATP. FEBS Open Bio 2020; 10:2805-2823. [PMID: 33145964 PMCID: PMC7714071 DOI: 10.1002/2211-5463.13023] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/24/2020] [Accepted: 11/02/2020] [Indexed: 12/31/2022] Open
Abstract
Humanin (HN) is known to bind amyloid beta (Aβ)‐inducing cytoprotective effects, while binding of acetylcholinesterase (AChE) to Aβ increases its aggregation and cytotoxicity. Previously, we showed that binding of HN to Aβ blocks aggregation induced by AChE and that HN decreases but does not abolish Aβ‐AChE interactions in A549 cell media. Here, we set out to shed light on factors that modulate the interactions of Aβ with HN and AChE. We found that binding of either HN or AChE to Aβ is not affected by heparan sulfate, while ATP, thought to reduce misfolding of Aβ, weakened interactions between AChE and Aβ but strengthened those between Aβ and HN. Using media from either A549 or H1299 lung cancer cells, we observed that more HN was bound to Aβ upon addition of ATP, while levels of AChE in a complex with Aβ were decreased by ATP addition to A549 cell media. Exogenous addition of ATP to either A549 or H1299 cell media increased interactions of endogenous HN with Aβ to a comparable extent despite differences in AChE expression in the two cell lines, and this was correlated with decreased binding of exogenously added HN to Aβ. Treatment with exogenous ATP had no effect on cell viability under all conditions examined. Exogenously added ATP did not affect viability of cells treated with AChE‐immunodepleted media, and there was no apparent protection against the cytotoxicity resulting from immunodepletion of HN. Moreover, exogenously added ATP had no effect on the relative abundance of oligomer versus total Aβ in either cell line.
Collapse
Affiliation(s)
- Sarah Atali
- Chemistry Department, Eastern Michigan University, Ypsilanti, MI, USA
| | - Sadaf Dorandish
- Chemistry Department, Eastern Michigan University, Ypsilanti, MI, USA
| | - Jonathan Devos
- Chemistry Department, Eastern Michigan University, Ypsilanti, MI, USA
| | - Asana Williams
- Chemistry Department, Eastern Michigan University, Ypsilanti, MI, USA
| | - Deanna Price
- Chemistry Department, Eastern Michigan University, Ypsilanti, MI, USA
| | - Jaylen Taylor
- Chemistry Department, Eastern Michigan University, Ypsilanti, MI, USA
| | - Jeffrey Guthrie
- Chemistry Department, Eastern Michigan University, Ypsilanti, MI, USA
| | - Deborah Heyl
- Chemistry Department, Eastern Michigan University, Ypsilanti, MI, USA
| | - Hedeel Guy Evans
- Chemistry Department, Eastern Michigan University, Ypsilanti, MI, USA
| |
Collapse
|
60
|
Comoletti D, Trobiani L, Chatonnet A, Bourne Y, Marchot P. Comparative mapping of selected structural determinants on the extracellular domains of cholinesterase-like cell-adhesion molecules. Neuropharmacology 2020; 184:108381. [PMID: 33166544 DOI: 10.1016/j.neuropharm.2020.108381] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 09/10/2020] [Accepted: 10/29/2020] [Indexed: 11/18/2022]
Abstract
Cell adhesion generally involves formation of homophilic or heterophilic protein complexes between two cells to form transcellular junctions. Neural cell-adhesion members of the α/β-hydrolase fold superfamily of proteins use their extracellular or soluble cholinesterase-like domain to bind cognate partners across cell membranes, as illustrated by the neuroligins. These cell-adhesion molecules currently comprise the synaptic organizers neuroligins found in all animal phyla, along with three proteins found only in invertebrates: the guidance molecule neurotactin, the glia-specific gliotactin, and the basement membrane protein glutactin. Although these proteins share a cholinesterase-like fold, they lack one or more residues composing the catalytic triad responsible for the enzymatic activity of the cholinesterases. Conversely, they are found in various subcellular localisations and display specific disulfide bonding and N-glycosylation patterns, along with individual surface determinants possibly associated with recognition and binding of protein partners. Formation of non-covalent dimers typical of the cholinesterases is documented for mammalian neuroligins, yet whether invertebrate neuroligins and their neurotactin, gliotactin and glutactin relatives also form dimers in physiological conditions is unknown. Here we provide a brief overview of the localization, function, evolution, and conserved versus individual structural determinants of these cholinesterase-like cell-adhesion proteins. This article is part of the special issue entitled 'Acetylcholinesterase Inhibitors: From Bench to Bedside to Battlefield'.
Collapse
Affiliation(s)
- Davide Comoletti
- School of Biological Sciences, Victoria University of Wellington, Wellington, 6012, New Zealand; Child Health Institute of New Jersey, New Brunswick, NJ 08901, USA; Department of Neuroscience and Cell Biology Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ 08901, USA.
| | - Laura Trobiani
- School of Biological Sciences, Victoria University of Wellington, Wellington, 6012, New Zealand
| | - Arnaud Chatonnet
- Lab 'Dynamique Musculaire et Métabolisme', Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement (INRAE) / Université Montpellier, Montpellier, France
| | - Yves Bourne
- Lab 'Architecture et Fonction des Macromolécules Biologiques (AFMB)', Centre National de la Recherche Scientifique (CNRS)/Aix-Marseille Univ, Faculté des Sciences - Campus Luminy, Marseille, France
| | - Pascale Marchot
- Lab 'Architecture et Fonction des Macromolécules Biologiques (AFMB)', Centre National de la Recherche Scientifique (CNRS)/Aix-Marseille Univ, Faculté des Sciences - Campus Luminy, Marseille, France.
| |
Collapse
|
61
|
Emon MA, Heinson A, Wu P, Domingo-Fernández D, Sood M, Vrooman H, Corvol JC, Scordis P, Hofmann-Apitius M, Fröhlich H. Clustering of Alzheimer's and Parkinson's disease based on genetic burden of shared molecular mechanisms. Sci Rep 2020; 10:19097. [PMID: 33154531 PMCID: PMC7645798 DOI: 10.1038/s41598-020-76200-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 10/23/2020] [Indexed: 02/07/2023] Open
Abstract
One of the visions of precision medicine has been to re-define disease taxonomies based on molecular characteristics rather than on phenotypic evidence. However, achieving this goal is highly challenging, specifically in neurology. Our contribution is a machine-learning based joint molecular subtyping of Alzheimer's (AD) and Parkinson's Disease (PD), based on the genetic burden of 15 molecular mechanisms comprising 27 proteins (e.g. APOE) that have been described in both diseases. We demonstrate that our joint AD/PD clustering using a combination of sparse autoencoders and sparse non-negative matrix factorization is reproducible and can be associated with significant differences of AD and PD patient subgroups on a clinical, pathophysiological and molecular level. Hence, clusters are disease-associated. To our knowledge this work is the first demonstration of a mechanism based stratification in the field of neurodegenerative diseases. Overall, we thus see this work as an important step towards a molecular mechanism-based taxonomy of neurological disorders, which could help in developing better targeted therapies in the future by going beyond classical phenotype based disease definitions.
Collapse
Affiliation(s)
- Mohammad Asif Emon
- Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), 53754, Sankt Augustin, Germany
- Bonn-Aachen International Center for IT, University of Bonn, Endenicher Allee 19c, 53115, Bonn, Germany
| | - Ashley Heinson
- UCB Pharma (UCB Celltech Ltd.), 208 Bath Road, Slough, SL1 3WE, Berkshire, UK
| | - Ping Wu
- UCB Pharma (UCB Celltech Ltd.), 208 Bath Road, Slough, SL1 3WE, Berkshire, UK
| | - Daniel Domingo-Fernández
- Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), 53754, Sankt Augustin, Germany
- Bonn-Aachen International Center for IT, University of Bonn, Endenicher Allee 19c, 53115, Bonn, Germany
| | - Meemansa Sood
- Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), 53754, Sankt Augustin, Germany
- Bonn-Aachen International Center for IT, University of Bonn, Endenicher Allee 19c, 53115, Bonn, Germany
| | - Henri Vrooman
- Department of Radiology and Nuclear Medicine, Department of Medical Informatics, Erasmus MC, University Medical Center Rotterdam, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
| | | | - Phil Scordis
- UCB Pharma (UCB Celltech Ltd.), 208 Bath Road, Slough, SL1 3WE, Berkshire, UK
| | - Martin Hofmann-Apitius
- Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), 53754, Sankt Augustin, Germany
- Bonn-Aachen International Center for IT, University of Bonn, Endenicher Allee 19c, 53115, Bonn, Germany
| | - Holger Fröhlich
- Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), 53754, Sankt Augustin, Germany.
- Bonn-Aachen International Center for IT, University of Bonn, Endenicher Allee 19c, 53115, Bonn, Germany.
- UCB Pharma (UCB Biosciences GmbH), Alfred-Nobel-Str. 10, 40789, Monheim, Germany.
| |
Collapse
|
62
|
Semenov VE, Zueva IV, Mukhamedyarov MA, Lushchekina SV, Petukhova EO, Gubaidullina LM, Krylova ES, Saifina LF, Lenina OA, Petrov KA. Novel Acetylcholinesterase Inhibitors Based on Uracil Moiety for Possible Treatment of Alzheimer Disease. Molecules 2020; 25:molecules25184191. [PMID: 32932702 PMCID: PMC7571187 DOI: 10.3390/molecules25184191] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 09/07/2020] [Accepted: 09/10/2020] [Indexed: 12/14/2022] Open
Abstract
In this study, novel derivatives based on 6-methyluracil and condensed uracil were synthesized, namely, 2,4-quinazoline-2,4-dione with ω-(ortho-nitrilebenzylethylamino) alkyl chains at the N atoms of the pyrimidine ring. In this series of synthesized compounds, the polymethylene chains were varied from having tetra- to hexamethylene chains, and secondary NH, tertiary ethylamino, and quaternary ammonium groups were introduced into the chains. The molecular modeling of the compounds indicated that they could function as dual binding site acetylcholinesterase inhibitors, binding to both the peripheral anionic site and active site. The data from in vitro experiments show that the most active compounds exhibit affinity toward acetylcholinesterase within a nanomolar range, with selectivity for acetylcholinesterase over butyrylcholinesterase reaching four orders of magnitude. In vivo biological assays demonstrated the potency of these compounds in the treatment of memory impairment using an animal model of Alzheimer disease.
Collapse
Affiliation(s)
- Vyacheslav E. Semenov
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, Arbuzov str. 8, 420088 Kazan, Russia; (I.V.Z.); (L.M.G.); (E.S.K.); (L.F.S.); (O.A.L.)
- Correspondence: (V.E.S.); (K.A.P.); Tel.: +7-843-279-47-09 (V.E.S.); +7-843-273-93-64 (K.A.P.)
| | - Irina V. Zueva
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, Arbuzov str. 8, 420088 Kazan, Russia; (I.V.Z.); (L.M.G.); (E.S.K.); (L.F.S.); (O.A.L.)
| | - Marat A. Mukhamedyarov
- Institute of Neuroscience, Kazan State Medical University, 420012 Kazan, Russia; (M.A.M.); (E.O.P.)
| | - Sofya V. Lushchekina
- Emanuel Institute of Biochemical Physics, Kosygina st. 4, 119334 Moscow, Russia;
| | - Elena O. Petukhova
- Institute of Neuroscience, Kazan State Medical University, 420012 Kazan, Russia; (M.A.M.); (E.O.P.)
| | - Lilya M. Gubaidullina
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, Arbuzov str. 8, 420088 Kazan, Russia; (I.V.Z.); (L.M.G.); (E.S.K.); (L.F.S.); (O.A.L.)
| | - Evgeniya S. Krylova
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, Arbuzov str. 8, 420088 Kazan, Russia; (I.V.Z.); (L.M.G.); (E.S.K.); (L.F.S.); (O.A.L.)
| | - Lilya F. Saifina
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, Arbuzov str. 8, 420088 Kazan, Russia; (I.V.Z.); (L.M.G.); (E.S.K.); (L.F.S.); (O.A.L.)
| | - Oksana A. Lenina
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, Arbuzov str. 8, 420088 Kazan, Russia; (I.V.Z.); (L.M.G.); (E.S.K.); (L.F.S.); (O.A.L.)
| | - Konstantin A. Petrov
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, Arbuzov str. 8, 420088 Kazan, Russia; (I.V.Z.); (L.M.G.); (E.S.K.); (L.F.S.); (O.A.L.)
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kremlyovskaya str., 18, 420008 Kazan, Russia
- Correspondence: (V.E.S.); (K.A.P.); Tel.: +7-843-279-47-09 (V.E.S.); +7-843-273-93-64 (K.A.P.)
| |
Collapse
|
63
|
New Hybrids of 4-Amino-2,3-polymethylene-quinoline and p-Tolylsulfonamide as Dual Inhibitors of Acetyl- and Butyrylcholinesterase and Potential Multifunctional Agents for Alzheimer's Disease Treatment. Molecules 2020; 25:molecules25173915. [PMID: 32867324 PMCID: PMC7504258 DOI: 10.3390/molecules25173915] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/24/2020] [Accepted: 08/24/2020] [Indexed: 01/31/2023] Open
Abstract
New hybrid compounds of 4-amino-2,3-polymethylene-quinoline containing different sizes of the aliphatic ring and linked to p-tolylsulfonamide with alkylene spacers of increasing length were synthesized as potential drugs for treatment of Alzheimer’s disease (AD). All compounds were potent inhibitors of acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) with selectivity toward BChE. The lead compound 4-methyl-N-(5-(1,2,3,4-tetrahydro-acridin-9-ylamino)-pentyl)-benzenesulfonamide (7h) exhibited an IC50 (AChE) = 0.131 ± 0.01 µM (five times more potent than tacrine), IC50(BChE) = 0.0680 ± 0.0014 µM, and 17.5 ± 1.5% propidium displacement at 20 µM. The compounds possessed low activity against carboxylesterase, indicating a likely absence of unwanted drug-drug interactions in clinical use. Kinetics studies were consistent with mixed-type reversible inhibition of both cholinesterases. Molecular docking demonstrated dual binding sites of the conjugates in AChE and clarified the differences in the structure-activity relationships for AChE and BChE inhibition. The conjugates could bind to the AChE peripheral anionic site and displace propidium, indicating their potential to block AChE-induced β-amyloid aggregation, thereby exerting a disease-modifying effect. All compounds demonstrated low antioxidant activity. Computational ADMET profiles predicted that all compounds would have good intestinal absorption, medium blood-brain barrier permeability, and medium cardiac toxicity risk. Overall, the results indicate that the novel conjugates show promise for further development and optimization as multitarget anti-AD agents.
Collapse
|
64
|
Phenethyl Esters and Amide of Ferulic Acid, Hydroferulic Acid, Homovanillic Acid, and Vanillic Acid: Synthesis, Free Radicals Scavenging Activity, and Molecular Modeling as Potential Cholinesterases Inhibitors. MOLBANK 2020. [DOI: 10.3390/m1151] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
As ferulic acid was reported to be involved in novel potential mechanisms associated with Alzheimer’s disease (AD) therapy, five closely related phenethyl esters and amide of this natural product were synthesized and screened for their free radicals scavenging activity. Ferulic acid and its analogue′s absorption, distribution, metabolism, and excretion (ADME) properties were predicted. All compounds obey Lipinski′s rules. Moreover, all evaluated compounds seem to present a high oral bioavailability and blood–brain barrier (BBB) permeation which is crucial for Alzheimer′s disease drug candidates. Molecular docking of analogues 4 and 8 with acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) showed interactions with the residues of the catalytic triad of AChE and BChE. In addition to their interactions with the anionic subsite, hydroferulic acid phenethyl ester 4 and homovanillic acid phenethyl ester 8 may have potential as inhibitors of AChE and BChE, respectively.
Collapse
|
65
|
Stavrakov G, Philipova I, Lukarski A, Atanasova M, Zheleva D, Zhivkova ZD, Ivanov S, Atanasova T, Konstantinov S, Doytchinova I. Galantamine-Curcumin Hybrids as Dual-Site Binding Acetylcholinesterase Inhibitors. Molecules 2020; 25:E3341. [PMID: 32717861 PMCID: PMC7435983 DOI: 10.3390/molecules25153341] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 07/16/2020] [Accepted: 07/21/2020] [Indexed: 02/07/2023] Open
Abstract
Galantamine (GAL) and curcumin (CU) are alkaloids used to improve symptomatically neurodegenerative conditions like Alzheimer's disease (AD). GAL acts mainly as an inhibitor of the enzyme acetylcholinesterase (AChE). CU binds to amyloid-beta (Aβ) oligomers and inhibits the formation of Aβ plaques. Here, we combine GAL core with CU fragments and design a combinatorial library of GAL-CU hybrids as dual-site binding AChE inhibitors. The designed hybrids are screened for optimal ADME properties and BBB permeability and docked on AChE. The 14 best performing compounds are synthesized and tested in vitro for neurotoxicity and anti-AChE activity. Five of them are less toxic than GAL and CU and show activities between 41 and 186 times higher than GAL.
Collapse
Affiliation(s)
- Georgi Stavrakov
- Faculty of Pharmacy, Medical University of Sofia, 1000 Sofia, Bulgaria; (A.L.); (M.A.); (D.Z.); (Z.D.Z.); (S.I.); (T.A.); (S.K.)
- Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria;
| | - Irena Philipova
- Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria;
| | - Atanas Lukarski
- Faculty of Pharmacy, Medical University of Sofia, 1000 Sofia, Bulgaria; (A.L.); (M.A.); (D.Z.); (Z.D.Z.); (S.I.); (T.A.); (S.K.)
| | - Mariyana Atanasova
- Faculty of Pharmacy, Medical University of Sofia, 1000 Sofia, Bulgaria; (A.L.); (M.A.); (D.Z.); (Z.D.Z.); (S.I.); (T.A.); (S.K.)
| | - Dimitrina Zheleva
- Faculty of Pharmacy, Medical University of Sofia, 1000 Sofia, Bulgaria; (A.L.); (M.A.); (D.Z.); (Z.D.Z.); (S.I.); (T.A.); (S.K.)
| | - Zvetanka D. Zhivkova
- Faculty of Pharmacy, Medical University of Sofia, 1000 Sofia, Bulgaria; (A.L.); (M.A.); (D.Z.); (Z.D.Z.); (S.I.); (T.A.); (S.K.)
| | - Stefan Ivanov
- Faculty of Pharmacy, Medical University of Sofia, 1000 Sofia, Bulgaria; (A.L.); (M.A.); (D.Z.); (Z.D.Z.); (S.I.); (T.A.); (S.K.)
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD 20850, USA
| | - Teodora Atanasova
- Faculty of Pharmacy, Medical University of Sofia, 1000 Sofia, Bulgaria; (A.L.); (M.A.); (D.Z.); (Z.D.Z.); (S.I.); (T.A.); (S.K.)
| | - Spiro Konstantinov
- Faculty of Pharmacy, Medical University of Sofia, 1000 Sofia, Bulgaria; (A.L.); (M.A.); (D.Z.); (Z.D.Z.); (S.I.); (T.A.); (S.K.)
| | - Irini Doytchinova
- Faculty of Pharmacy, Medical University of Sofia, 1000 Sofia, Bulgaria; (A.L.); (M.A.); (D.Z.); (Z.D.Z.); (S.I.); (T.A.); (S.K.)
| |
Collapse
|
66
|
Multitarget Therapeutic Strategies for Alzheimer's Disease: Review on Emerging Target Combinations. BIOMED RESEARCH INTERNATIONAL 2020; 2020:5120230. [PMID: 32714977 PMCID: PMC7354643 DOI: 10.1155/2020/5120230] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 02/12/2020] [Accepted: 06/02/2020] [Indexed: 12/12/2022]
Abstract
Neurodegenerative diseases represent nowadays one of the major health problems. Despite the efforts made to unveil the mechanism leading to neurodegeneration, it is still not entirely clear what triggers this phenomenon and what allows its progression. Nevertheless, it is accepted that neurodegeneration is a consequence of several detrimental processes, such as protein aggregation, oxidative stress, and neuroinflammation, finally resulting in the loss of neuronal functions. Starting from these evidences, there has been a wide search for novel agents able to address more than a single event at the same time, the so-called multitarget-directed ligands (MTDLs). These compounds originated from the combination of different pharmacophoric elements which endowed them with the ability to interfere with different enzymatic and/or receptor systems, or to exert neuroprotective effects by modulating proteins and metal homeostasis. MTDLs have been the focus of the latest strategies to discover a new treatment for Alzheimer's disease (AD), which is considered the most common form of dementia characterized by neurodegeneration and cognitive dysfunctions. This review is aimed at collecting the latest and most interesting target combinations for the treatment of AD, with a detailed discussion on new agents with favorable in vitro properties and on optimized structures that have already been assessed in vivo in animal models of dementia.
Collapse
|
67
|
Computational exploration and experimental validation to identify a dual inhibitor of cholinesterase and amyloid-beta for the treatment of Alzheimer’s disease. J Comput Aided Mol Des 2020; 34:983-1002. [DOI: 10.1007/s10822-020-00318-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 05/26/2020] [Indexed: 12/15/2022]
|
68
|
Price D, Dorandish S, Williams A, Iwaniec B, Stephens A, Marshall K, Guthrie J, Heyl D, Evans HG. Humanin Blocks the Aggregation of Amyloid-β Induced by Acetylcholinesterase, an Effect Abolished in the Presence of IGFBP-3. Biochemistry 2020; 59:1981-2002. [PMID: 32383868 PMCID: PMC8193794 DOI: 10.1021/acs.biochem.0c00274] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
It is known that the humanin (HN) peptide binding to amyloid-β (Aβ) protects against its cytotoxic effects, while acetylcholinesterase (AChE) binding to Aβ increases its aggregation and cytotoxicity. HN is also known to bind the insulin-like growth factor binding protein-3 (IGFBP-3). Here, we examined the regulation of Aβ conformations by HN, AChE, and IGFBP-3 both in vitro and in the conditioned media from A549 and H1299 lung cancer cells. Our in vitro results showed the following: IGFBP-3 binds HN and blocks it from binding Aβ in the absence or presence of AChE; HN and AChE can simultaneously bind Aβ but not when in the presence of IGFBP-3; HN is unable to reduce the aggregation of Aβ in the presence of IGFBP-3; and HN abolishes the aggregation of Aβ induced by the addition of AChE in the absence of IGFBP-3. In the media, AChE and HN can simultaneously bind Aβ. While both AChE and HN are detected when using 6E10 Aβ antibodies, only AChE is detected when using the Aβ 17-24 antibody 4G8, the anti-oligomer A11, and the anti-amyloid fibril LOC antibodies. No signal was observed for IGFBP-3 with any of the anti-amyloid antibodies used. Exogenously added IGFBP-3 reduced the amount of HN found in a complex when using 6E10 antibodies and correlated with a concomitant increase in the amyloid oligomers. Immunodepletion of HN from the media of the A549 and H1299 cells increased the relative abundance of the oligomer vs the total amount of Aβ, the A11-positive prefibrillar oligomers, and to a lesser extent the LOC-positive fibrillar oligomers, and was also correlated with diminished cell viability and increased apoptosis.
Collapse
Affiliation(s)
- Deanna Price
- Department of Chemistry, Eastern Michigan University, Ypsilanti, Michigan 48197, United States
| | - Sadaf Dorandish
- Department of Chemistry, Eastern Michigan University, Ypsilanti, Michigan 48197, United States
| | - Asana Williams
- Department of Chemistry, Eastern Michigan University, Ypsilanti, Michigan 48197, United States
| | - Brandon Iwaniec
- Department of Chemistry, Eastern Michigan University, Ypsilanti, Michigan 48197, United States
| | - Alexis Stephens
- Department of Chemistry, Eastern Michigan University, Ypsilanti, Michigan 48197, United States
| | - Keyan Marshall
- Department of Chemistry, Eastern Michigan University, Ypsilanti, Michigan 48197, United States
| | - Jeffrey Guthrie
- Department of Chemistry, Eastern Michigan University, Ypsilanti, Michigan 48197, United States
| | - Deborah Heyl
- Department of Chemistry, Eastern Michigan University, Ypsilanti, Michigan 48197, United States
| | - Hedeel Guy Evans
- Department of Chemistry, Eastern Michigan University, Ypsilanti, Michigan 48197, United States
| |
Collapse
|
69
|
Wilhelm EA, Torres MLCP, Pereira CF, Vogt AG, Cervo R, Dos Santos BGT, Cargnelutti R, Luchese C. Therapeutic potential of selanyl amide derivatives in the in vitro anticholinesterase activity and in in vivo antiamnesic action. Can J Physiol Pharmacol 2020; 98:304-313. [PMID: 31821013 DOI: 10.1139/cjpp-2019-0291] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The present study evaluated the in vitro acetylcholinesterase (AChE) inhibitor activity of two new selanyl amide derivatives in cerebral structures of mice. Our results demonstrated that N-(2-(3-(phenylselanyl)propoxy)phenyl)furan-2-carboxamide (1) and N-(2-(3-(phenylselanyl)propoxy)phenyl)thiophene-2-carboxamide (2) inhibited the in vitro AChE activity in mice. Another objective was to assess the effect of the best AChE inhibitor in an amnesic model induced by scopolamine (SCO) in male Swiss mice. The involvement of AChE activity and lipid peroxidation in the cerebral structures was investigated. Our results showed that compound 1 (10 mg/kg, intragastrically) attenuated the latency to find the escape box and the number of holes visited in the Barnes maze task, without altering the locomotor and exploratory activities in an open-field test. Compound 1 protected against increasing in lipid peroxidation levels and AChE activity caused by SCO in the cerebral cortex and hippocampus of mice. In conclusion, the present study evidenced the in vitro anticholinesterase effect of two new selanyl amide derivatives in the cerebral structures of mice. Moreover, compound 1, a selanyl amide derivative containing a furan ring, demonstrated antiamnesic action due to its antioxidant and anticholinesterase activities in cerebral structures.
Collapse
Affiliation(s)
- Ethel A Wilhelm
- Programa de Pós-graduação em Bioquímica e Bioprospecção, Laboratório de Pesquisa em Farmacologia Bioquímica (LaFarBio), Grupo de Pesquisa em Neurobiotecnologia (GPN), Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas (UFPel), CEP 96010-900 Pelotas, RS, Brazil
| | - Marina Laura C P Torres
- Programa de Pós-graduação em Bioquímica e Bioprospecção, Laboratório de Pesquisa em Farmacologia Bioquímica (LaFarBio), Grupo de Pesquisa em Neurobiotecnologia (GPN), Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas (UFPel), CEP 96010-900 Pelotas, RS, Brazil
| | - Caroline F Pereira
- Programa de Pós-graduação em Bioquímica e Bioprospecção, Laboratório de Pesquisa em Farmacologia Bioquímica (LaFarBio), Grupo de Pesquisa em Neurobiotecnologia (GPN), Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas (UFPel), CEP 96010-900 Pelotas, RS, Brazil
| | - Ane G Vogt
- Programa de Pós-graduação em Bioquímica e Bioprospecção, Laboratório de Pesquisa em Farmacologia Bioquímica (LaFarBio), Grupo de Pesquisa em Neurobiotecnologia (GPN), Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas (UFPel), CEP 96010-900 Pelotas, RS, Brazil
| | - Rodrigo Cervo
- Department of Chemistry, Universidade Federal de Santa Maria, LMI - Laboratório de Materiais Inorgânicos, 97105-900, Santa Maria, RS, Brazil
| | - Brenda G T Dos Santos
- Department of Chemistry, Universidade Federal de Santa Maria, LMI - Laboratório de Materiais Inorgânicos, 97105-900, Santa Maria, RS, Brazil
| | - Roberta Cargnelutti
- Department of Chemistry, Universidade Federal de Santa Maria, LMI - Laboratório de Materiais Inorgânicos, 97105-900, Santa Maria, RS, Brazil
| | - Cristiane Luchese
- Programa de Pós-graduação em Bioquímica e Bioprospecção, Laboratório de Pesquisa em Farmacologia Bioquímica (LaFarBio), Grupo de Pesquisa em Neurobiotecnologia (GPN), Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas (UFPel), CEP 96010-900 Pelotas, RS, Brazil
| |
Collapse
|
70
|
N-1,2,3-triazole-isatin derivatives for cholinesterase and β-amyloid aggregation inhibition: A comprehensive bioassay study. Bioorg Chem 2020; 98:103753. [DOI: 10.1016/j.bioorg.2020.103753] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 03/09/2020] [Accepted: 03/10/2020] [Indexed: 02/07/2023]
|
71
|
Liu Z, Qin G, Mana L, Dong Y, Huang S, Wang Y, Wu Y, Shi J, Tian J, Wang P. GAPT regulates cholinergic dysfunction and oxidative stress in the brains of learning and memory impairment mice induced by scopolamine. Brain Behav 2020; 10:e01602. [PMID: 32174034 PMCID: PMC7218254 DOI: 10.1002/brb3.1602] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 02/19/2020] [Accepted: 02/25/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Cholinergic dysfunction and oxidative stress are the crucial mechanisms of Alzheimer's disease (AD). GAPT, also called GEPT (a combination of several active components extracted from the Chinese herbs ginseng, epimedium, polygala and tuber curcumae) or Jinsiwei, is a patented Chinese herbal compound, has been clinically widely used to improve learning and memory impairment, but whether it can play a neuroprotective role by protecting cholinergic neurons and reducing oxidative stress injury remains unclear. METHODS Male ICR mice were intraperitoneally injected with scopolamine (3 mg/kg) to establish a learning and memory disordered model. An LC-MS method was established to study the chemical compounds and in vivo metabolites of GAPT. After scopolamine injection, a step-down passive-avoidance test (SDPA) and a Y maze test were used to estimate learning ability and cognitive function. In addition, ELISA detected the enzymatic activities of acetylcholinesterase (AChE), acetylcholine (ACh), choline acetyltransferase (ChAT), malondialdehyde (MDA), glutathione peroxidase (GPX), and total superoxide dismutase (T-SOD). The protein expressions of AChE, ChAT, SOD1, and GPX1 were observed by western blot, and the distribution of ChAT, SOD1, and GPX1 was observed by immunohistochemical staining. RESULTS After one-half or 1 month of intragastric administration, GAPT can ameliorate scopolamine-induced behavioral changes in learning and memory impaired mice. It can also decrease the activity of MDA and protein expression level of AChE, increase the activity of Ach, and increase activity and protein expression level of ChAT, SOD, and GPX in scopolamine-treated mice. After one and a half month of intragastric administration of GAPT, echinacoside, salvianolic acid A, ginsenoside Rb1, ginsenoside Rg2, pachymic acid, and beta asarone could be absorbed into mice blood and pass through BBB. CONCLUSIONS GAPT can improve the learning and memory ability of scopolamine-induced mice, and its mechanism may be related to protecting cholinergic neurons and reducing oxidative stress injury.
Collapse
Affiliation(s)
- Zhenhong Liu
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine (BUCM), Beijing, China
| | - Gaofeng Qin
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine (BUCM), Beijing, China
| | - Lulu Mana
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine (BUCM), Beijing, China.,Department of Integrative Medicine, School of TCM, Xinjiang Medical University, Urumqi, China
| | - Yunfang Dong
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine (BUCM), Beijing, China.,Zhongkang International Health Physical Examination Center-Qingdao Ruiyuan Hospital of Traditional Chinese Medicine, Qingdao, China
| | - Shuaiyang Huang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine (BUCM), Beijing, China
| | - Yahan Wang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine (BUCM), Beijing, China
| | - Yiqiong Wu
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine (BUCM), Beijing, China.,Jiangsu Province Hospital on Integrated Chinese and Western Medicines, Nanjing, China
| | - Jing Shi
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine (BUCM), Beijing, China.,BUCM Neurology Center, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Jinzhou Tian
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine (BUCM), Beijing, China.,BUCM Neurology Center, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Pengwen Wang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine (BUCM), Beijing, China
| |
Collapse
|
72
|
Mettupalayam Kaliyannan Sundaramoorthy P, Kilavan Packiam K. In vitro enzyme inhibitory and cytotoxic studies with Evolvulus alsinoides (Linn.) Linn. Leaf extract: a plant from Ayurveda recognized as Dasapushpam for the management of Alzheimer's disease and diabetes mellitus. BMC Complement Med Ther 2020; 20:129. [PMID: 32345272 PMCID: PMC7189705 DOI: 10.1186/s12906-020-02922-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Accepted: 04/07/2020] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Evolvulus alsinoides (Linn.) Linn. (Convolvulaceae) is a therapeutic herb alleviating brain patterns associated with three categories of regulatory principles of the body, mind, and behaviour. In the current research, enzyme inhibition and cytotoxic potentials of E. alsinoides (L.) L. leaf extract has been studied validating its potential application. METHODS The plant phenolics in the leaf extracts obtained via cold-maceration with solvents viz.: n-hexane, chloroform, ethyl acetate, methanol, and water were quantitatively analyzed. The antioxidant potency was evaluated using 2,2-diphenyl-1-picrylhydrazyl (DPPH) and Ferric Reducing Ability of Plasma (FRAP) assays at five concentrations (100-500 μg). The enzyme inhibition potential was performed with α-amylase, α-glucosidase, and acetylcholinesterase at seven concentrations (25-500 μg). The experiments were done in triplicates and statistically validated using Minitab-17 and SPSS 22. RESULTS Water extract contain 45.08 ± 0.02 mg GAE/g, 49.30 ± 0.07 mg GAE/g, 211.21 ± 0.02 mg QE/g tannins, phenolics, flavonoids respectively. Its antioxidant activity was supported by IC50 52.43 ± 0.2 μg/mL (DPPH assay) and 41.58 ± 0.03 (FRAP assay). Methanolic extract inhibits α-amylase with IC50 1.33 ± 0.05 μg/mL. Water extract inhibits α-glucosidase and acetylcholinesterase with IC50 3.58 ± 0.02 μg/mL and 4.46 ± 0.03 μg/mL. Cytotoxicity studies with SH-SY5Y cell-line substantiate the inhibition potential of water extract with IC50 103.0035 μg/mL. DISCUSSION AND CONCLUSIONS The extracts with potent antioxidant and enzyme-inhibiting activity were determined. The findings of the research are the first report about the inhibition effects of Evolvulus alsinoides (Linn.) Linn extracts against α-amylase, α-glucosidase and acetylcholinesterase. The extracts shall be examined in future studies to evaluate its pharmaceutical potential.
Collapse
Affiliation(s)
| | - Kannan Kilavan Packiam
- Department of Biotechnology, Bannari Amman Institute of Technology, Sathyamangalam, Tamilnadu, India
| |
Collapse
|
73
|
Biological and Computational Studies for Dual Cholinesterases Inhibitory Effect of Zerumbone. Nutrients 2020; 12:nu12051215. [PMID: 32344943 PMCID: PMC7281973 DOI: 10.3390/nu12051215] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 04/20/2020] [Accepted: 04/23/2020] [Indexed: 12/15/2022] Open
Abstract
Acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) mediate the degradation of acetylcholine (ACh), a primary neurotransmitter in the brain. Cholinergic deficiency occurs during the progression of Alzheimer’s disease (AD), resulting in widespread cognitive dysfunction and decline. We evaluated the potential effect of a natural cholinesterase inhibitor, zerumbone, using in vitro target enzyme assays, as well as in silico docking and ADMET (absorption, distribution, metabolism, excretion, and toxicity) simulation. Zerumbone showed a predominant cholinesterase inhibitory property with IC50 values of 2.74 ± 0.48 µM and 4.12 ± 0.42 µM for AChE and BChE, respectively; however, the modes of inhibition were different. Computational docking simulation indicated that Van der Waals interactions between zerumbone and both the cholinesterases were the main forces responsible for its inhibitory effects. Furthermore, zerumbone showed the best physicochemical properties for both bioavailability and blood–brain barrier (BBB) permeability. Together, in the present study, zerumbone was clearly identified as a unique dual AChE and BChE inhibitor with high permeability across the BBB, suggesting a strong potential for its physiological benefits and/or pharmacological efficacy in the prevention of AD.
Collapse
|
74
|
Abstract
Abstract
A new series of homobivalent Dimebon analogs, bis-γ-carbolines with alkylene, phenylenedialkylene, and triazole-containing spacers, was synthesized. Doubling the γ-carboline pharmacophore increased inhibitory potency against acetylcholinesterase (AChE) compared with Dimebon, while keeping Dimebon’s anti-butyrylcholinesterase activity; therefore, leading to inversion of selectivity. Molecular docking revealed the reasons for the increased anti-AChE activity and ability to block AChE-induced aggregation of β-amyloid for bis-γ-carbolines, which became double-site inhibitors of AChE. Conjugates with ditriazole-containing spacers were the most active antioxidants in both the ABTS-test and prevention of lipid peroxidation in brain homogenates without inhibiting the mitochondrial permeability transition (MPT). Conjugates with alkylene (4a–d), phenylenedialkylene (4e), and monotriazole (8) spacers were less active as antioxidants but prevented induction of the MPT and increased the calcium retention capacity of mitochondria. Lead compound 4e showed neuroprotective potential in a cellular calcium overload model of neurodegeneration. Computational studies showed that all the bis-γ-carbolines were expected to have high values for intestinal absorption and very good blood-brain barrier permeability along with good drug-likeness. Overall, the results showed that new homobivalent Dimebon analogs exhibit an expanded spectrum of biological activity and improved pharmacological properties, making them promising candidates for further research and optimization as multitarget agents for Alzheimer’s disease treatment.
Collapse
|
75
|
Makhaeva GF, Kovaleva NV, Boltneva NP, Lushchekina SV, Rudakova EV, Stupina TS, Terentiev AA, Serkov IV, Proshin AN, Radchenko EV, Palyulin VA, Bachurin SO, Richardson RJ. Conjugates of tacrine and 1,2,4-thiadiazole derivatives as new potential multifunctional agents for Alzheimer’s disease treatment: Synthesis, quantum-chemical characterization, molecular docking, and biological evaluation. Bioorg Chem 2020; 94:103387. [DOI: 10.1016/j.bioorg.2019.103387] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 10/12/2019] [Accepted: 10/21/2019] [Indexed: 12/13/2022]
|
76
|
Rullo M, Catto M, Carrieri A, de Candia M, Altomare CD, Pisani L. Chasing ChEs-MAO B Multi-Targeting 4-Aminomethyl-7-Benzyloxy-2 H-Chromen-2-ones. Molecules 2019; 24:E4507. [PMID: 31835376 PMCID: PMC6943664 DOI: 10.3390/molecules24244507] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 12/04/2019] [Accepted: 12/05/2019] [Indexed: 11/16/2022] Open
Abstract
A series of 4-aminomethyl-7-benzyloxy-2H-chromen-2-ones was investigated with the aim of identifying multiple inhibitors of cholinesterases (acetyl- and butyryl-, AChE and BChE) and monoamine oxidase B (MAO B) as potential anti-Alzheimer molecules. Starting from a previously reported potent MAO B inhibitor (3), we studied single-point modifications at the benzyloxy or at the basic moiety. The in vitro screening highlighted triple-acting compounds (6, 8, 9, 16, 20) showing nanomolar and selective MAO B inhibition along with IC50 against ChEs at the low micromolar level. Enzyme kinetics analysis toward AChE and docking simulations on the target enzymes were run in order to get insight into the mechanism of action and plausible binding modes.
Collapse
Affiliation(s)
| | | | | | | | | | - Leonardo Pisani
- Department of Pharmacy—Drug Sciences, University of Bari “Aldo Moro”, via E. Orabona 4, 70125 Bari, Italy
| |
Collapse
|
77
|
Han J, Ji Y, Youn K, Lim G, Lee J, Kim DH, Jun M. Baicalein as a Potential Inhibitor against BACE1 and AChE: Mechanistic Comprehension through In Vitro and Computational Approaches. Nutrients 2019; 11:E2694. [PMID: 31703329 PMCID: PMC6893645 DOI: 10.3390/nu11112694] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 10/28/2019] [Accepted: 11/04/2019] [Indexed: 12/17/2022] Open
Abstract
One of the major neurodegenerative features of Alzheimer's disease (AD) is the presence of neurotoxic amyloid plaques composed of amyloid beta peptide (Aβ). β-Secretase (BACE1) and acetylcholinesterase (AChE), which promote Aβ fibril formation, have become attractive therapeutic targets for AD. P-glycoprotein (P-gp), the major efflux pump of the blood-brain barrier (BBB), plays a critical role in limiting therapeutic molecules. In pursuit of discovering a natural anti-AD candidate, the bioactivity, physicochemical, drug-likeness, and molecular docking properties of baicalein, a major compound from Scutellaria baicalensis, was investigated. Baicalein exhibited strong BACE1 and AChE inhibitory properties (IC50 23.71 ± 1.91 µM and 45.95 ± 3.44 µM, respectively) and reacted in non-competitive and competitive manners with substrates, respectively. in Silico docking analysis was in full agreement with the in vitro results, demonstrating that the compound exhibited powerful binding interaction with target enzymes. Particularly, three continuous hydroxyl groups on the A ring demonstrated strong H-bond binding properties. It is also noteworthy that baicalein complied with all requirements of Lipinski's rule of five by its optimal physicochemical properties for both oral bioavailability and blood-brain barrier permeability. Overall, the present study strongly demonstrated the possibility of baicalein having in vivo pharmacological efficacy for specific targets in the prevention and/or treatment of AD.
Collapse
Affiliation(s)
- Jin Han
- Department of Food Science and Nutrition, College of Health Sciences, Dong-A University, Busan 49315, Korea; (J.H.); (Y.J.); (K.Y.)
- Center for Silver-Targeted Biomaterials, Brain Busan 21 Plus Program, Graduate School, Dong-A University, Busan 49315, Korea
| | - Yeongseon Ji
- Department of Food Science and Nutrition, College of Health Sciences, Dong-A University, Busan 49315, Korea; (J.H.); (Y.J.); (K.Y.)
- Center for Silver-Targeted Biomaterials, Brain Busan 21 Plus Program, Graduate School, Dong-A University, Busan 49315, Korea
| | - Kumju Youn
- Department of Food Science and Nutrition, College of Health Sciences, Dong-A University, Busan 49315, Korea; (J.H.); (Y.J.); (K.Y.)
| | - GyuTae Lim
- Korean Bioinformation Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea; (G.L.); (J.L.)
- Department of Bioinformatics, KRIBB School of Bioscience, Korea University of Sciences and Technology, Daejeon 34113, Korea
| | - Jinhyuk Lee
- Korean Bioinformation Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea; (G.L.); (J.L.)
- Department of Bioinformatics, KRIBB School of Bioscience, Korea University of Sciences and Technology, Daejeon 34113, Korea
| | - Dong Hyun Kim
- Department of Medicinal Biotechnology, College of Health Sciences, Dong-A University, Busan 49315, Korea;
- Institute of Convergence Bio-Health, Dong-A University, Busan 49315, Korea
| | - Mira Jun
- Department of Food Science and Nutrition, College of Health Sciences, Dong-A University, Busan 49315, Korea; (J.H.); (Y.J.); (K.Y.)
- Center for Silver-Targeted Biomaterials, Brain Busan 21 Plus Program, Graduate School, Dong-A University, Busan 49315, Korea
- Institute of Convergence Bio-Health, Dong-A University, Busan 49315, Korea
| |
Collapse
|
78
|
Korabecny J, Spilovska K, Mezeiova E, Benek O, Juza R, Kaping D, Soukup O. A Systematic Review on Donepezil-based Derivatives as Potential Cholinesterase Inhibitors for Alzheimer’s Disease. Curr Med Chem 2019; 26:5625-5648. [DOI: 10.2174/0929867325666180517094023] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 02/02/2018] [Accepted: 05/11/2018] [Indexed: 01/06/2023]
Abstract
:
Alzheimer’s Disease (AD) is a multifactorial progressive neurodegenerative disorder
characterized by memory loss, disorientation, and gradual deterioration of intellectual capacity.
Its etiology has not been elucidated yet. To date, only one therapeutic approach has
been approved for the treatment of AD. The pharmacotherapy of AD has relied on noncompetitive
N-methyl-D-aspartate (NMDA) receptor antagonist - memantine, and acetylcholinesterase
(AChE) inhibitors (AChEIs) - tacrine, donepezil, rivastigmine and galantamine.
Donepezil was able to ameliorate the symptoms related to AD mainly via AChE, but also
through reduction of β-amyloid burden. This review presents the overview of donepezilrelated
compounds as potential anti-AD drugs developed on the basis of cholinergic hypothesis
to act as solely AChE and butyrylcholinesterase (BChE) inhibitors.
Collapse
Affiliation(s)
- Jan Korabecny
- National Institute of Mental Health, Topolova 748, 250 67 Klecany, Czech Republic
| | - Katarina Spilovska
- National Institute of Mental Health, Topolova 748, 250 67 Klecany, Czech Republic
| | - Eva Mezeiova
- National Institute of Mental Health, Topolova 748, 250 67 Klecany, Czech Republic
| | - Ondrej Benek
- National Institute of Mental Health, Topolova 748, 250 67 Klecany, Czech Republic
| | - Radomir Juza
- National Institute of Mental Health, Topolova 748, 250 67 Klecany, Czech Republic
| | - Daniel Kaping
- National Institute of Mental Health, Topolova 748, 250 67 Klecany, Czech Republic
| | - Ondrej Soukup
- National Institute of Mental Health, Topolova 748, 250 67 Klecany, Czech Republic
| |
Collapse
|
79
|
Zueva I, Dias J, Lushchekina S, Semenov V, Mukhamedyarov M, Pashirova T, Babaev V, Nachon F, Petrova N, Nurullin L, Zakharova L, Ilyin V, Masson P, Petrov K. New evidence for dual binding site inhibitors of acetylcholinesterase as improved drugs for treatment of Alzheimer's disease. Neuropharmacology 2019; 155:131-141. [DOI: 10.1016/j.neuropharm.2019.05.025] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 05/06/2019] [Accepted: 05/19/2019] [Indexed: 01/22/2023]
|
80
|
Huang W, Liang M, Li Q, Zheng X, Zhang C, Wang Q, Tang L, Zhang Z, Wang B, Shen Z. Development of the “hidden” multifunctional agents for Alzheimer's disease. Eur J Med Chem 2019; 177:247-258. [DOI: 10.1016/j.ejmech.2019.05.051] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 04/26/2019] [Accepted: 05/18/2019] [Indexed: 01/13/2023]
|
81
|
Overview of novel multifunctional agents based on conjugates of γ-carbolines, carbazoles, tetrahydrocarbazoles, phenothiazines, and aminoadamantanes for treatment of Alzheimer's disease. Chem Biol Interact 2019; 308:224-234. [DOI: 10.1016/j.cbi.2019.05.020] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 04/25/2019] [Accepted: 05/13/2019] [Indexed: 01/10/2023]
|
82
|
Lana E, Gellerbring A, Jung S, Nordberg A, Unger Lithner C, Darreh-Shori T. Homomeric and Heteromeric Aβ Species Exist in Human Brain and CSF Regardless of Alzheimer's Disease Status and Risk Genotype. Front Mol Neurosci 2019; 12:176. [PMID: 31417354 PMCID: PMC6684963 DOI: 10.3389/fnmol.2019.00176] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 07/03/2019] [Indexed: 02/04/2023] Open
Abstract
Background: A fundamental question in Alzheimer’s disease (AD) is whether amyloid-β (Aβ) peptides and their deposition in the brain signify a direct pathological role or they are mere outcome of the disease pathophysiological events affecting neuronal function. It is therefore important to decipher their physiological role in the brain. So far, the overwhelming focus has been on the potential toxicity of Aβ, often studied outside the crucial AD characteristics, i.e.: (i) the slow, decades-long disease progression that precedes clinical symptoms; (ii) the link to apolipoprotein-E ε4 allele as major risk factor; (iii) the selective early degeneration of cholinergic neurons. Previous studies, in vitro and cerebrospinal fluid (CSF) only, indicated one possible native function of Aβ peptides is the allosteric modulation of acetylcholine homeostasis, via molecular interactions between Aβ, apolipoprotein-E, and the acetylcholine-degrading enzymes, cholinesterases, resulting in the formation of acetylcholine-hydrolyzing complexes (BAβACs). Methods: Here, by combining sucrose-density gradient fractionation of post-mortem brains and in-house developed sensitive ELISA assays on the obtained fractions, we investigated the presence, levels and molecular interactions between Aβ, apolipoprotein-E and cholinesterases for the first time in brain tissues. We examined three distinct brain regions of Alzheimer and non-demented subjects, plus a large number of Alzheimer CSF samples. Results: We report that both monomeric and oligomeric (homomeric and heteromeric) forms of Aβ peptides are present in the brain of Alzheimer and non-demented individuals. Heteromeric Aβ was found in stable complexes with apolipoprotein-E and/or cholinesterases, irrespective of APOE genotype or disease status, arguing in favor of a physiological dynamic formation and function for these complexes in the brain. The patterns and molecular sizes of the detected soluble Aβ forms were closely matched between CSF and brain samples. This evinces that the detected Aβ-apolipoprotein-E complexes and BAβACs in CSF most likely originate from the interstitial fluids of the brain. Conclusions: In conclusion, both light homomeric Aβ oligomers and heteromeric Aβ-ApoE and BAβACs are present and readily detectable in the brain, regardless of disease status and APOE4 genotype. Deeper knowledge of the physiological function of Aβ is crucial for better understanding the early pathological events that decades later lead to manifestation of AD.
Collapse
Affiliation(s)
- Erica Lana
- Department of Neurobiology, Care Sciences and Society, Division of Clinical Geriatrics, Center for Alzheimer Research, Karolinska Institutet (KI), Stockholm, Sweden
| | - Anna Gellerbring
- Department of Neurobiology, Care Sciences and Society, Division of Clinical Geriatrics, Center for Alzheimer Research, Karolinska Institutet (KI), Stockholm, Sweden
| | - Sabrina Jung
- Department of Neurobiology, Care Sciences and Society, Division of Clinical Geriatrics, Center for Alzheimer Research, Karolinska Institutet (KI), Stockholm, Sweden
| | - Agneta Nordberg
- Department of Neurobiology, Care Sciences and Society, Division of Clinical Geriatrics, Center for Alzheimer Research, Karolinska Institutet (KI), Stockholm, Sweden.,Theme Aging, The Aging Brain, Karolinska University Hospital, Stockholm, Sweden
| | - Christina Unger Lithner
- Department of Neurobiology, Care Sciences and Society, Division of Clinical Geriatrics, Center for Alzheimer Research, Karolinska Institutet (KI), Stockholm, Sweden
| | - Taher Darreh-Shori
- Department of Neurobiology, Care Sciences and Society, Division of Clinical Geriatrics, Center for Alzheimer Research, Karolinska Institutet (KI), Stockholm, Sweden
| |
Collapse
|
83
|
Panche AN, Chandra S, Diwan AD. Multi-Target β-Protease Inhibitors from Andrographis paniculata: In Silico and In Vitro Studies. PLANTS 2019; 8:plants8070231. [PMID: 31319560 PMCID: PMC6681301 DOI: 10.3390/plants8070231] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 06/15/2019] [Accepted: 06/20/2019] [Indexed: 12/29/2022]
Abstract
Natural products derived from plants play a vital role in the discovery of new drug candidates, and these are used for novel therapeutic drug development. Andrographis paniculata and Spilanthes paniculata are used extensively as medicinal herbs for the treatment of various ailments, and are reported to have neuroprotective properties. β-amyloid is a microscopic brain protein whose significant aggregation is detected in mild cognitive impairment and Alzheimer’s disease (AD) brains. The accumulation of β-amyloid disrupts cell communication and triggers inflammation by activating immune cells, leading to neuronal cell death and cognitive disabilities. The proteases acetylcholinesterase (AChE), butyrylcholinesterase (BChE), and beta secretase-1 (BACE-1) have been reported to be correlated with the synthesis and growth of β-amyloid plaques in the brains of AD patients. In the present study, the phenolic compounds from A. paniculata and S. paniculata that have been reported in the literature were selected for the current investigation. Furthermore, we employed molecular docking and molecular dynamics studies of the phenolic compounds with the proteins AChE, BChE, and BACE-1 in order to evaluate the binding characteristics and identify potent anti-amyloid agents against the neurodegenerative diseases such as AD. In this investigation, we predicted three compounds from A. paniculata with maximum binding affinities with cholinesterases and BACE-1. The computational investigations predicted that these compounds follow the rule of five. We further evaluated these molecules for in vitro inhibition activity against all the enzymes. In the in vitro investigations, 3,4-di-o-caffeoylquinic acid (5281780), apigenin (5280443), and 7-o-methylwogonin (188316) were found to be strong inhibitors of AChE, BChE, and BACE-1. These findings suggest that these compounds can be potent multi-target inhibitors of the proteases that might cumulatively work and inhibit the initiation and formation of β-amyloid plaques, which is a prime cause of neurotoxicity and dementia. According to our knowledge, these findings are the first report on natural compounds isolated from A. paniculata as multi-target potent inhibitors and anti-amyloid agents.
Collapse
Affiliation(s)
- Archana N Panche
- Department of Bio-Engineering, Birla Institute of Technology, Mesra, Ranchi 835215, India
- MGM's Institute of Biosciences & Technology, Mahatma Gandhi Mission, N-6, CIDCO, Aurangabad 431003, India
| | - Sheela Chandra
- Department of Bio-Engineering, Birla Institute of Technology, Mesra, Ranchi 835215, India.
| | - A D Diwan
- MGM's Institute of Biosciences & Technology, Mahatma Gandhi Mission, N-6, CIDCO, Aurangabad 431003, India
| |
Collapse
|
84
|
Makhaeva GF, Shevtsova EF, Boltneva NP, Kovaleva NV, Rudakova EV, Dubova LG, Shevtsov PN, Bachurin SO. Anticholinesterase and Antioxidant Activity of New Binary Conjugates of γ-Carbolines. DOKL BIOCHEM BIOPHYS 2019; 484:1-5. [DOI: 10.1134/s1607672919010010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Indexed: 11/23/2022]
|
85
|
Makhaeva GF, Rudakova EV, Kovaleva NV, Lushchekina SV, Boltneva NP, Proshin AN, Shchegolkov EV, Burgart YV, Saloutin VI. Cholinesterase and carboxylesterase inhibitors as pharmacological agents. Russ Chem Bull 2019. [DOI: 10.1007/s11172-019-2507-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
86
|
Guo H, Cao H, Cui X, Zheng W, Wang S, Yu J, Chen Z. Silymarin's Inhibition and Treatment Effects for Alzheimer's Disease. Molecules 2019; 24:E1748. [PMID: 31064071 PMCID: PMC6539875 DOI: 10.3390/molecules24091748] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 04/28/2019] [Accepted: 04/30/2019] [Indexed: 02/06/2023] Open
Abstract
As a longstanding problem, Alzheimer's disease (AD) has stymied researchers in the medical field with its increasing incidence and enormous treatment difficulty. Silymarin has always been valued by researchers for its good efficacy and safety in treating liver disease. Recent studies have shown that silymarin also has good pharmacological activity in the nervous system, especially for the treatment of AD. Silymarin can control the production of Aβ by inhibiting the precursor substance of Aβ (β-amyloid precursor protein), and it can inhibit the polymerization of Aβ. Silymarin can also increase the acetylcholine content in the nervous system by inhibiting cholinesterase activity. At the same time, it also has the effect of resisting oxidative stress and the inflammatory response of the nervous system. These pharmacological activities contribute to the inhibition of the onset of AD. The good efficacy of silymarin on AD and its high safety and availability give it huge potential for the treatment of AD.
Collapse
Affiliation(s)
- Hong Guo
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China.
| | - Hui Cao
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China.
| | - Xiaowei Cui
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China.
| | - Wenxiu Zheng
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China.
| | - Shanshan Wang
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China.
| | - Jiyang Yu
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China.
| | - Zhi Chen
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China.
| |
Collapse
|
87
|
Molochkina EM, Treshchenkova YA. The Effect of Alpha-Tocopherol on the Activity of Acetylcholinesterases from Different Sources. NEUROCHEM J+ 2019. [DOI: 10.1134/s1819712419010161] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
88
|
de Castro AA, Soares FV, Pereira AF, Polisel DA, Caetano MS, Leal DHS, da Cunha EFF, Nepovimova E, Kuca K, Ramalho TC. Non-conventional compounds with potential therapeutic effects against Alzheimer’s disease. Expert Rev Neurother 2019; 19:375-395. [DOI: 10.1080/14737175.2019.1608823] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Alexandre A. de Castro
- Laboratory of Molecular Modeling, Department of Chemistry, Federal University of Lavras, Lavras, Brazil
| | - Flávia V. Soares
- Laboratory of Molecular Modeling, Department of Chemistry, Federal University of Lavras, Lavras, Brazil
| | - Ander F. Pereira
- Laboratory of Molecular Modeling, Department of Chemistry, Federal University of Lavras, Lavras, Brazil
| | - Daniel A. Polisel
- Laboratory of Molecular Modeling, Department of Chemistry, Federal University of Lavras, Lavras, Brazil
| | - Melissa S. Caetano
- Institute of Exact and Biological Sciences, Federal University of Ouro Preto, Ouro Preto, Brazil
| | - Daniel H. S. Leal
- Laboratory of Molecular Modeling, Department of Chemistry, Federal University of Lavras, Lavras, Brazil
- Department of Health Sciences, Federal University of Espírito Santo, São Mateus, Brazil
| | - Elaine F. F. da Cunha
- Laboratory of Molecular Modeling, Department of Chemistry, Federal University of Lavras, Lavras, Brazil
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czech Republic
| | - Kamil Kuca
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czech Republic
| | - Teodorico C. Ramalho
- Laboratory of Molecular Modeling, Department of Chemistry, Federal University of Lavras, Lavras, Brazil
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czech Republic
| |
Collapse
|
89
|
Design, biological evaluation and X-ray crystallography of nanomolar multifunctional ligands targeting simultaneously acetylcholinesterase and glycogen synthase kinase-3. Eur J Med Chem 2019; 168:58-77. [DOI: 10.1016/j.ejmech.2018.12.063] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 12/21/2018] [Accepted: 12/24/2018] [Indexed: 11/20/2022]
|
90
|
Conjugates of methylene blue with γ-carboline derivatives as new multifunctional agents for the treatment of neurodegenerative diseases. Sci Rep 2019; 9:4873. [PMID: 30890752 PMCID: PMC6424957 DOI: 10.1038/s41598-019-41272-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 03/05/2019] [Indexed: 02/08/2023] Open
Abstract
We studied the inhibitory activity of methylene blue (MB) γ-carbolines (gC) conjugates (MB-gCs) against human erythrocyte acetylcholinesterase (AChE), equine serum butyrylcholinesterase (BChE), and a structurally related enzyme, porcine liver carboxylesterase (CaE). In addition, we determined the ability of MB-gCs to bind to the peripheral anionic site (PAS) of Electrophorus electricus AChE (EeAChE) and competitively displace propidium iodide from this site. Moreover, we examined the ability of MB-gCs to scavenge free radicals as well as their influence on mitochondrial potential and iron-induced lipid peroxidation. We found that MB-gCs effectively inhibited AChE and BChE with IC50 values in the range 1.73–10.5 μM and exhibited low potencies against CaE (9.8–26% inhibition at 20 μM). Kinetic studies showed that MB-gCs were mixed-type reversible inhibitors of both cholinesterases. Molecular docking results showed that the MB-gCs could bind both to the catalytic active site and to the PAS of human AChE and BChE. Accordingly, MB-gCs effectively displaced propidium from the peripheral anionic site of EeAChE. In addition, MB-gCs were extremely active in both radical scavenging tests. Quantum mechanical DFT calculations suggested that free radical scavenging was likely mediated by the sulfur atom in the MB fragment. Furthermore, the MB-gCs, in like manner to MB, can restore mitochondrial membrane potential after depolarization with rotenone. Moreover, MB-gCs possess strong antioxidant properties, preventing iron-induced lipid peroxidation in mitochondria. Overall, the results indicate that MB-gCs are promising candidates for further optimization as multitarget therapeutic agents for neurodegenerative diseases.
Collapse
|
91
|
Inhibition of β-site amyloid precursor protein cleaving enzyme 1 and cholinesterases by pterosins via a specific structure-activity relationship with a strong BBB permeability. Exp Mol Med 2019; 51:1-18. [PMID: 30755593 PMCID: PMC6372667 DOI: 10.1038/s12276-019-0205-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 11/05/2018] [Accepted: 11/08/2018] [Indexed: 01/18/2023] Open
Abstract
We extracted 15 pterosin derivatives from Pteridium aquilinum that inhibited β-site amyloid precursor protein cleaving enzyme 1 (BACE1) and cholinesterases involved in the pathogenesis of Alzheimer's disease (AD). (2R)-Pterosin B inhibited BACE1, acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) with an IC50 of 29.6, 16.2 and 48.1 µM, respectively. The Ki values and binding energies (kcal/mol) between pterosins and BACE1, AChE, and BChE corresponded to the respective IC50 values. (2R)-Pterosin B was a noncompetitive inhibitor against human BACE1 and BChE as well as a mixed-type inhibitor against AChE, binding to the active sites of the corresponding enzymes. Molecular docking simulation of mixed-type and noncompetitive inhibitors for BACE1, AChE, and BChE indicated novel binding site-directed inhibition of the enzymes by pterosins and the structure-activity relationship. (2R)-Pterosin B exhibited a strong BBB permeability with an effective permeability (Pe) of 60.3×10-6 cm/s on PAMPA-BBB. (2R)-Pterosin B and (2R,3 R)-pteroside C significantly decreased the secretion of Aβ peptides from neuroblastoma cells that overexpressed human β-amyloid precursor protein at 500 μM. Conclusively, our study suggested that several pterosins are potential scaffolds for multitarget-directed ligands (MTDLs) for AD therapeutics.
Collapse
|
92
|
Azure B affects amyloid precursor protein metabolism in PS70 cells. Chem Biol Interact 2019; 299:88-93. [PMID: 30500345 DOI: 10.1016/j.cbi.2018.11.023] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 10/22/2018] [Accepted: 11/27/2018] [Indexed: 01/01/2023]
Abstract
Alzheimer's disease (AD), the most common form of dementia, is characterized by abundant deposition of amyloid-β (Aβ) peptide that is the result of sequential cleavage of amyloid precursor protein (APP) by β-secretase and γ-secretase. Several studies have documented that inhibition of Aβ peptide synthesis or facilitating its degradation is one of the attractive therapeutic strategies in AD. Methylene blue (MethB), which has recently been investigated in Phase II clinical trials, is a prominent inhibitor in reducing Aβ oligomers. Herein, we wonder whether the mitigating effects of MethB on amyloid metabolism are related to the activity of its major metabolite, azure B. The goal of this study was to investigate the effects of azure B, which is also a cholinesterase inhibitor, on APP processing by using Chinese hamster ovary cells stably expressing human wild-type APP and presenilin 1 (PS70). Azure B significantly decreased the levels of secreted APPα (sAPPα) and Aβ40/42 in culture medium with a dose-dependent manner. A significant decrease was also observed in the levels of intracellular APP without affecting the cell viability. In parallel with the decrease of APP and APP metabolites, the activity of β-secretase 1 (BACE1) was significantly attenuated compared to control. Overall, our results show that azure B has a large contribution for the pharmacological profile of MethB in APP metabolism.
Collapse
|
93
|
Sun K, Bai Y, Zhao R, Guo Z, Su X, Li P, Yang P. Neuroprotective effects of matrine on scopolamine-induced amnesia via inhibition of AChE/BuChE and oxidative stress. Metab Brain Dis 2019; 34:173-181. [PMID: 30406376 DOI: 10.1007/s11011-018-0335-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 10/30/2018] [Indexed: 12/29/2022]
Abstract
The present study was designed to evaluate the effects of matrine (MAT) on scopolamine (SCOP)-induced learning and memory impairment. After successive oral administration of MAT to mice for three days at doses of 0.4, 2, and 10 mg/kg, we assessed improvements in learning and memory and investigated the mechanism of action of SCOP-induced amnesia. Donepezil at a dose of 3 mg/kg was used as a standard memory enhancer. MAT significantly improved SCOP-induced learning and memory impairment in novel object recognition and Y-maze tests at doses of 0.4, 2, and 10 mg/kg. Furthermore, MAT inhibited acetylcholinesterase (AChE) and butyrylcholinesterase (BuChE) activities and decreased oxidative stress in the brain, as evidenced by increased total antioxidant capacity, total superoxide dismutase levels, and catalase activities as well as decreased malondialdehyde levels. Additionally, there was a significant negative correlation between the percentage of spontaneous alternation in the Y maze and AChE activity in the cortex and hippocampus. MAT ameliorated SCOP-induced amnesia by the inhibition of both AChE/BuChE activities and oxidative stress. This study provides further evidence to encourage the development of MAT as a drug for the prevention or treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Kaiyue Sun
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, Shanxi, People's Republic of China.
| | - Yuting Bai
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, Shanxi, People's Republic of China
| | - Rong Zhao
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, Shanxi, People's Republic of China
| | - Zijiao Guo
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, Shanxi, People's Republic of China
| | - Xiang Su
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, Shanxi, People's Republic of China
| | - Peiqi Li
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, Shanxi, People's Republic of China
| | - Pengyu Yang
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, Shanxi, People's Republic of China
| |
Collapse
|
94
|
Lee J, Jun M. Dual BACE1 and Cholinesterase Inhibitory Effects of Phlorotannins from Ecklonia cava-An In Vitro and in Silico Study. Mar Drugs 2019; 17:E91. [PMID: 30717208 PMCID: PMC6410325 DOI: 10.3390/md17020091] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 01/24/2019] [Accepted: 01/28/2019] [Indexed: 01/09/2023] Open
Abstract
Alzheimer's disease (AD) is one of the most common neurodegenerative diseases with a multifactorial nature. β-Secretase (BACE1) and acetylcholinesterase (AChE), which are required for the production of neurotoxic β-amyloid (Aβ) and the promotion of Aβ fibril formation, respectively, are considered as prime therapeutic targets for AD. In our efforts towards the development of potent multi-target, directed agents for AD treatment, major phlorotannins such as eckol, dieckol, and 8,8'-bieckol from Ecklonia cava (E. cava) were evaluated. Based on the in vitro study, all tested compounds showed potent inhibitory effects on BACE1 and AChE. In particular, 8,8'-bieckol demonstrated the best inhibitory effect against BACE1 and AChE, with IC50 values of 1.62 ± 0.14 and 4.59 ± 0.32 µM, respectively. Overall, kinetic studies demonstrated that all the tested compounds acted as dual BACE1 and AChE inhibitors in a non-competitive or competitive fashion, respectively. In silico docking analysis exhibited that the lowest binding energies of all compounds were negative, and specifically different residues of each target enzyme interacted with hydroxyl groups of phlorotannins. The present study suggested that major phlorotannins derived from E. cava possess significant potential as drug candidates for therapeutic agents against AD.
Collapse
Affiliation(s)
- Jinhyuk Lee
- Korean Bioinformation Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125, Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea.
- Department of Bioinformatics, KIRBB School of Bioscience, Korea University of Sciences and Technology, 217 Gajung-ro, Yuseong-gu, Daejeon 34113, Korea.
| | - Mira Jun
- Department of Food Science and Nutrition, College of Health Sciences, Dong-A University, 37, Nakdong-daero 550 beon-gil, Saha-gu, Busan 49315, Korea.
- Center for Silver-Targeted Biomaterials, Brain Busan 21 Plus Program, Graduate School, Dong-A University, Nakdong-daero 550 beon-gil, Saha-gu, Busan 49315, Korea.
- Institute of Convergence Bio-Health, Dong-A University, Busan 49315, Korea.
| |
Collapse
|
95
|
Brewster JT, Dell’Acqua S, Thach DQ, Sessler JL. Classics in Chemical Neuroscience: Donepezil. ACS Chem Neurosci 2019; 10:155-167. [PMID: 30372021 DOI: 10.1021/acschemneuro.8b00517] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The discovery of acetylcholine and acetylcholinesterase provided the first insight into the intricacies of chemical signal transduction and neuronal communication. Further elucidation of the underlying mechanisms led to an attendant leveraging of this knowledge via the synthesis of new therapeutics designed to control aberrant biochemical processes. The central role of the cholinergic system within human memory and learning, as well as its implication in Alzheimer's disease, has made it a point of focus within the neuropharmacology and medicinal chemistry communities. This review is focused on donepezil and covers the background, synthetic routes, structure-activity relationships, binding interactions with acetylcholinesterase, pharmacokinetics and metabolism, efficacy, adverse effects, and historical importance of this leading therapeutic in the treatment of Alzheimer's disease and true Classic in Chemical Neuroscience.
Collapse
Affiliation(s)
- James T. Brewster
- Department of Chemistry, The University of Texas at Austin, Austin, Texas 78712-1224, United States
| | - Simone Dell’Acqua
- Department of Chemistry, University of Pavia, Via Taramelli 12, 27100 Pavia, Italy
| | - Danny Q. Thach
- Department of Chemistry, University of California—Berkeley, Berkeley, California 94720, United States
| | - Jonathan L. Sessler
- Department of Chemistry, The University of Texas at Austin, Austin, Texas 78712-1224, United States
| |
Collapse
|
96
|
Makhaeva GF, Kovaleva NV, Lushchekina SV, Rudakova EV, Boltneva NP, Proshin AN, Lednev BV, Serkov IV, Bachurin SO. Conjugates of Tacrine and Its Cyclic Homologues with p-Toluenesulfonamide as Novel Acetylcholinesterase and Butyrylcholinesterase Inhibitors. DOKL BIOCHEM BIOPHYS 2019; 483:369-373. [PMID: 30607741 DOI: 10.1134/s1607672918060200] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Indexed: 11/22/2022]
Abstract
Using the acylation reaction with tosyl chloride of N-aminopropyl analogues of tacrine and its cyclic homologues with different size of the aliphatic cycle (5-8), we synthesized a number of new derivatives of p-toluenesulfonamide. It is shown that the synthesized hybrid compounds of tacrine and p-toluenesulfonamide are effective inhibitors of acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) with the preferential inhibition of BChE. They also displace propidium from the peripheral anionic site of the electric eel AChE (Electrophorus electricus). The characteristics of the efficiency and selectivity of cholinesterase inhibition by the test compounds were confirmed by the results of molecular docking.
Collapse
Affiliation(s)
- G F Makhaeva
- Institute of Physiologically Active Compounds, Russian Academy of Sciences, Chernogolovka, Moscow oblast, 142432, Russia.
| | - N V Kovaleva
- Institute of Physiologically Active Compounds, Russian Academy of Sciences, Chernogolovka, Moscow oblast, 142432, Russia
| | - S V Lushchekina
- Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, Moscow, 119334, Russia
| | - E V Rudakova
- Institute of Physiologically Active Compounds, Russian Academy of Sciences, Chernogolovka, Moscow oblast, 142432, Russia
| | - N P Boltneva
- Institute of Physiologically Active Compounds, Russian Academy of Sciences, Chernogolovka, Moscow oblast, 142432, Russia
| | - A N Proshin
- Institute of Physiologically Active Compounds, Russian Academy of Sciences, Chernogolovka, Moscow oblast, 142432, Russia
| | - B V Lednev
- Institute of Physiologically Active Compounds, Russian Academy of Sciences, Chernogolovka, Moscow oblast, 142432, Russia
| | - I V Serkov
- Institute of Physiologically Active Compounds, Russian Academy of Sciences, Chernogolovka, Moscow oblast, 142432, Russia
| | - S O Bachurin
- Institute of Physiologically Active Compounds, Russian Academy of Sciences, Chernogolovka, Moscow oblast, 142432, Russia
| |
Collapse
|
97
|
He Q, Liu J, Lan JS, Ding J, Sun Y, Fang Y, Jiang N, Yang Z, Sun L, Jin Y, Xie SS. Coumarin-dithiocarbamate hybrids as novel multitarget AChE and MAO-B inhibitors against Alzheimer’s disease: Design, synthesis and biological evaluation. Bioorg Chem 2018; 81:512-528. [DOI: 10.1016/j.bioorg.2018.09.010] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 09/05/2018] [Accepted: 09/07/2018] [Indexed: 12/30/2022]
|
98
|
Lee SY, Chiu YJ, Yang SM, Chen CM, Huang CC, Lee-Chen GJ, Lin W, Chang KH. Novel synthetic chalcone-coumarin hybrid for Aβ aggregation reduction, antioxidation, and neuroprotection. CNS Neurosci Ther 2018; 24:1286-1298. [PMID: 30596401 DOI: 10.1111/cns.13058] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Revised: 08/11/2018] [Accepted: 08/11/2018] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Aggregation of misfolded amyloid β (Aβ) in senile plaques causes oxidative stress and neuronal death in Alzheimer's disease (AD). Compounds possessing antiaggregation and antioxidant properties are promising candidate compounds for AD treatment. METHODS We examined the potential of synthetic derivatives of licochalcone A and coumarin for inhibiting Aβ aggregation, scavenging reactive oxygen species (ROS), and providing neuroprotection by using biochemical assays and Tet-On Aβ-GFP 293/SH-SY5Y cell models for AD. RESULTS Among test compounds, LM-031, a novel chalcone-coumarin hybrid, inhibited Aβ aggregation and scavenged free oxygen radicals. LM-031 markedly reduced Aβ misfolding and ROS as well as promoted neurite outgrowth and inhibited acetylcholinesterase in Tet-On Aβ-GFP 293/SH-SY5Y cells. Mechanistic studies showed upregulation of the HSPB1 chaperone, NRF2/NQO1/GCLC pathway, and CREB/BDNF/BCL2 pathway. Decreased neurite outgrowth upon the induction of Aβ-GFP was rescued by LM-031, which was counteracted by knockdown of HSPB1, NRF2, or CREB. CONCLUSION Taken together, these findings demonstrate that LM-031 exhibited antiaggregation, antioxidant, and neuroprotective effects against Aβ toxicity by enhancing HSPB1 and the NRF2-related antioxidant pathway as well as by activating the CREB-dependent survival and antiapoptosis pathway. These results imply that LM-031 may be a new therapeutic compound for AD.
Collapse
Affiliation(s)
- Shin-Ying Lee
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Ya-Jen Chiu
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Shu-Mei Yang
- Department of Chemistry, National Taiwan Normal University, Taipei, Taiwan
| | - Chiung-Mei Chen
- Department of Neurology, Chang Gung Memorial Hospital-Linkou Medical Center, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Chin-Chang Huang
- Department of Neurology, Chang Gung Memorial Hospital-Linkou Medical Center, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Guey-Jen Lee-Chen
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Wenwei Lin
- Department of Chemistry, National Taiwan Normal University, Taipei, Taiwan
| | - Kuo-Hsuan Chang
- Department of Neurology, Chang Gung Memorial Hospital-Linkou Medical Center, Chang Gung University College of Medicine, Taoyuan, Taiwan
| |
Collapse
|
99
|
Feng LD, Tian Y, Wang X, Dai R, Cai S, Cao YJ, Si YC. Therapy of Dredging the Bowels Enhanced the Neuroprotective Effect of Nourishing Kidney Herbs on Hippocampal Cholinergic System in Alzheimer's Disease Model Rat Induced by A β 1-42. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2018; 2018:3282385. [PMID: 30298092 PMCID: PMC6157172 DOI: 10.1155/2018/3282385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 06/03/2018] [Accepted: 08/15/2018] [Indexed: 11/17/2022]
Abstract
BACKGROUND Therapy of nourishing kidney has been used for treating memory deficits of Alzheimer's disease (AD) for thousands of years based on traditional Chinese medicine. However, we found the therapy of dredging the bowels could alleviate both memory deficits and mental symptoms of AD in clinic. OBJECTIVE To determine whether the therapy of dredging the bowels could enhance the neuroprotective effect of nourishing kidney herbs for treating AD rats, and to explore the underlying mechanism of the combination of nourishing kidney and dredging the bowels (NKDB) herbs. METHODS 60 rats were randomly divided into sham-operated group (SOG), model group (MG), nourishing kidney group (NKG), dredging the bowels group (DBG), nourishing kidney and dredging the bowels group (NKDBG), and donepezil hydrochloride group (DHG). The model establishment was performed by injecting Aβ 1-42 into the hippocampal CA1 region. Animals received aqueous solution of Chinese herbal medicine or western medicine while SOG received only distilled water. Ability of learning and memory were assessed by Morris water maze. Acetylcholinesterase(AChE) and choline acetyltransferase (ChAT) activity and positive cells in the hippocampus were detected by the biochemical and immunofluorescent assay. RESULTS All rats were in the same baseline. While after model establishment, ability of learning and memory of MG, NKG, DBG, NKDBG, and DHG were significantly impaired compared with SOG. Whereas after treatment, ability of learning and memory of NKG, DBG, NKDBG, and DHG were significantly improved compared with MG. Additionally, AChE activity of NKG, DBG, and NKDBG was significantly decreased, meanwhile ChAT activity showed an increased tendency. The number of AChE-positive cells and ChAT-positive cells of both NKDBG and DHG were significantly decreased and increased respectively, superior to those when compared with NKG and DBG. What's more, there was no significant difference between NKDBG and DHG. CONCLUSION Therapy of dredging the bowels could enhance the neuroprotective effect of nourishing kidney herbs by reversing morphological damage of hippocampal cholinergic system. Furthermore, treatment with NKDB herbs could be effectively against AD, providing a practical therapeutic strategy in clinic.
Collapse
Affiliation(s)
- Lu-Da Feng
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Yang Tian
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing 100078, China
- Neuroscience Department, Tufts University, Boston MA, 02111, USA
| | - Xin Wang
- Research Center of TCM Information Engineering, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Run Dai
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Song Cai
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Yu-Jia Cao
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Yin-Chu Si
- Department of Anatomy, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| |
Collapse
|
100
|
Ţînţaş ML, Gembus V, Alix F, Barré A, Coadou G, Truong L, Sebban M, Papamicaël C, Oulyadi H, Levacher V. Rational design of carbamate-based dual binding site and central AChE inhibitors by a “biooxidisable” prodrug approach: Synthesis, in vitro evaluation and docking studies. Eur J Med Chem 2018; 155:171-182. [DOI: 10.1016/j.ejmech.2018.05.057] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 04/10/2018] [Accepted: 05/31/2018] [Indexed: 12/15/2022]
|