1
|
Shahrivari-Baviloliaei S, Erdogan Orhan I, Abaci Kaplan N, Konopacka A, Waleron K, Plenis A, Viapiana A. Characterization of Phenolic Profile and Biological Properties of Astragalus membranaceus Fisch. ex Bunge Commercial Samples. Antioxidants (Basel) 2024; 13:993. [PMID: 39199238 PMCID: PMC11351125 DOI: 10.3390/antiox13080993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 08/01/2024] [Accepted: 08/12/2024] [Indexed: 09/01/2024] Open
Abstract
Astragalus membranaceus Fisch. ex Bunge (syn. Astragalus mongholicus Bunge) is one of the notable medicinal and food plants. Therefore, the aim of this study was to calculate the phenolic composition and antioxidant, antimicrobial, as well as enzyme inhibitory [acetylcholinesterase (AChE), butyrylcholinesterase (BChE), and tyrosinase (TYR)] activities with chemometric approaches of the hydromethanolic and water extracts of commercial A. membranaceus samples. Ten individual phenolic compounds were determined using high-performance liquid chromatography (HPLC), and only quercetin was found at a level of above 80 µg/g DW in both extracts. Moreover, the highest antioxidant activity in DPPH, FRAP, ABTS, and CUPRAC assays was found in the sample containing the roots in loose form from USA. A. membranaceus extracts displayed the inhibition zone diameters within the range from 10 to 22 mm antimicrobial activity against S. aureus, while there were no inhibition zones in any extracts in case of E. coli. The extracts of A. membranaceous showed an inhibition rate below 40% against TYR, and among tested extracts, only two samples were able to inhibit BChE with IC50 values of above 30 µg/mL. Correlation analysis showed a highly positive relationship between their phenolic composition and antioxidant activity. Concluding, the obtained results confirmed that A. membranaceus commercial samples could be an important dietary source of natural antioxidants.
Collapse
Affiliation(s)
- Saba Shahrivari-Baviloliaei
- Department of Analytical Chemistry, Medical University of Gdansk, Gen. J. Hallera 107, 80-416 Gdansk, Poland;
| | - Ilkay Erdogan Orhan
- Department of Pharmacognosy, Faculty of Pharmacy, Gazi University, 06330 Ankara, Türkiye; (N.A.K.); (I.E.O.)
- Department of Pharmacognosy, Faculty of Pharmacy, Lokman Hekim University, 06510 Ankara, Türkiye
| | - Nurten Abaci Kaplan
- Department of Pharmacognosy, Faculty of Pharmacy, Gazi University, 06330 Ankara, Türkiye; (N.A.K.); (I.E.O.)
| | - Agnieszka Konopacka
- Department of Pharmaceutical Microbiology, Medical University of Gdansk, Gen. J. Hallera 107, 80-416 Gdansk, Poland; (A.K.); (K.W.)
| | - Krzysztof Waleron
- Department of Pharmaceutical Microbiology, Medical University of Gdansk, Gen. J. Hallera 107, 80-416 Gdansk, Poland; (A.K.); (K.W.)
| | - Alina Plenis
- Department of Analytical Chemistry, Medical University of Gdansk, Gen. J. Hallera 107, 80-416 Gdansk, Poland;
| | - Agnieszka Viapiana
- Department of Analytical Chemistry, Medical University of Gdansk, Gen. J. Hallera 107, 80-416 Gdansk, Poland;
| |
Collapse
|
2
|
Abdul Manap AS, Almadodi R, Sultana S, Sebastian MG, Kavani KS, Lyenouq VE, Shankar A. Alzheimer's disease: a review on the current trends of the effective diagnosis and therapeutics. Front Aging Neurosci 2024; 16:1429211. [PMID: 39185459 PMCID: PMC11341404 DOI: 10.3389/fnagi.2024.1429211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 07/25/2024] [Indexed: 08/27/2024] Open
Abstract
The most prevalent cause of dementia is Alzheimer's disease. Cognitive decline and accelerating memory loss characterize it. Alzheimer's disease advances sequentially, starting with preclinical stages, followed by mild cognitive and/or behavioral impairment, and ultimately leading to Alzheimer's disease dementia. In recent years, healthcare providers have been advised to make an earlier diagnosis of Alzheimer's, prior to individuals developing Alzheimer's disease dementia. Regrettably, the identification of early-stage Alzheimer's disease in clinical settings can be arduous due to the tendency of patients and healthcare providers to disregard symptoms as typical signs of aging. Therefore, accurate and prompt diagnosis of Alzheimer's disease is essential in order to facilitate the development of disease-modifying and secondary preventive therapies prior to the onset of symptoms. There has been a notable shift in the goal of the diagnosis process, transitioning from merely confirming the presence of symptomatic AD to recognizing the illness in its early, asymptomatic phases. Understanding the evolution of disease-modifying therapies and putting effective diagnostic and therapeutic management into practice requires an understanding of this concept. The outcomes of this study will enhance in-depth knowledge of the current status of Alzheimer's disease's diagnosis and treatment, justifying the necessity for the quest for potential novel biomarkers that can contribute to determining the stage of the disease, particularly in its earliest stages. Interestingly, latest clinical trial status on pharmacological agents, the nonpharmacological treatments such as behavior modification, exercise, and cognitive training as well as alternative approach on phytochemicals as neuroprotective agents have been covered in detailed.
Collapse
Affiliation(s)
- Aimi Syamima Abdul Manap
- Department of Biomedical Science, College of Veterinary Medicine, King Faisal University, Al-Ahsa, Saudi Arabia
| | - Reema Almadodi
- Faculty of Pharmacy and Biomedical Sciences, MAHSA University, Selangor, Malaysia
| | - Shirin Sultana
- Faculty of Pharmacy and Biomedical Sciences, MAHSA University, Selangor, Malaysia
| | | | | | - Vanessa Elle Lyenouq
- Faculty of Pharmacy and Biomedical Sciences, MAHSA University, Selangor, Malaysia
| | - Aravind Shankar
- Faculty of Pharmacy and Biomedical Sciences, MAHSA University, Selangor, Malaysia
| |
Collapse
|
3
|
La Spada G, Miniero DV, Rullo M, Cipolloni M, Delre P, Colliva C, Colella M, Leonetti F, Liuzzi GM, Mangiatordi GF, Giacchè N, Pisani L. Structure-based design of multitargeting ChEs-MAO B inhibitors based on phenyl ring bioisosteres: AChE/BChE selectivity switch and drug-like characterization. Eur J Med Chem 2024; 274:116511. [PMID: 38820854 DOI: 10.1016/j.ejmech.2024.116511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 05/06/2024] [Accepted: 05/17/2024] [Indexed: 06/02/2024]
Abstract
A structure-based drug design approach was focused on incorporating phenyl ring heterocyclic bioisosteres into coumarin derivative 1, previously reported as potent dual AChE-MAO B inhibitor, with the aim of improving drug-like features. Structure-activity relationships highlighted that bioisosteric rings were tolerated by hMAO B enzymatic cleft more than hAChE. Interestingly, linker homologation at the basic nitrogen enabled selectivity to switch from hAChE to hBChE. In the present work, we identified thiophene-based isosteres 7 and 15 as dual AChE-MAO B (IC50 = 261 and 15 nM, respectively) and BChE-MAO B (IC50 = 375 and 20 nM, respectively) inhibitors, respectively. Both 7 and 15 were moderately water-soluble and membrane-permeant agents by passive diffusion (PAMPA-HDM). Moreover, they were able to counteract oxidative damage induced by both H2O2 and 6-OHDA in SH-SY5Y cells and predicted to penetrate into CNS in a cell-based model mimicking blood-brain barrier. Molecular dynamics (MD) simulations shed light on key differences in AChE and BChE recognition processes promoted by the basic chain homologation from 7 to 15.
Collapse
Affiliation(s)
- Gabriella La Spada
- Dept. of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, via E. Orabona 4, 70125, Bari, Italy
| | - Daniela Valeria Miniero
- Dept. of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, Via E. Orabona 4, 70125, Bari, Italy
| | - Mariagrazia Rullo
- Dept. of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, via E. Orabona 4, 70125, Bari, Italy
| | - Marco Cipolloni
- Tes Pharma s.r.l., via Palmiro Togliatti 20, 06073, Corciano, PG, Italy
| | - Pietro Delre
- CNR, Institute of Crystallography, 70126, Bari, Italy
| | - Carolina Colliva
- Tes Pharma s.r.l., via Palmiro Togliatti 20, 06073, Corciano, PG, Italy
| | - Marco Colella
- Dept. of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, via E. Orabona 4, 70125, Bari, Italy
| | - Francesco Leonetti
- Dept. of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, via E. Orabona 4, 70125, Bari, Italy
| | - Grazia Maria Liuzzi
- Dept. of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, Via E. Orabona 4, 70125, Bari, Italy
| | | | - Nicola Giacchè
- Tes Pharma s.r.l., via Palmiro Togliatti 20, 06073, Corciano, PG, Italy
| | - Leonardo Pisani
- Dept. of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, via E. Orabona 4, 70125, Bari, Italy.
| |
Collapse
|
4
|
Pont C, Sampietro A, Pérez-Areales FJ, Cristiano N, Albalat A, Pérez B, Bartolini M, De Simone A, Andrisano V, Barenys M, Teixidó E, Sabaté R, Loza MI, Brea J, Muñoz-Torrero D. Stepwise Structural Simplification of the Dihydroxyanthraquinone Moiety of a Multitarget Rhein-Based Anti-Alzheimer Lead to Improve Drug Metabolism and Pharmacokinetic Properties. Pharmaceutics 2024; 16:982. [PMID: 39204327 PMCID: PMC11359831 DOI: 10.3390/pharmaceutics16080982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 07/18/2024] [Accepted: 07/23/2024] [Indexed: 09/04/2024] Open
Abstract
Multitarget compounds have emerged as promising drug candidates to cope with complex multifactorial diseases, like Alzheimer's disease (AD). Most multitarget compounds are designed by linking two pharmacophores through a tether chain (linked hybrids), which results in rather large molecules that are particularly useful to hit targets with large binding cavities, but at the expense of suffering from suboptimal physicochemical/pharmacokinetic properties. Molecular size reduction by removal of superfluous structural elements while retaining the key pharmacophoric motifs may represent a compromise solution to achieve both multitargeting and favorable physicochemical/PK properties. Here, we report the stepwise structural simplification of the dihydroxyanthraquinone moiety of a rhein-huprine hybrid lead by hydroxy group removal-ring contraction-ring opening-ring removal, which has led to new analogs that retain or surpass the potency of the lead on its multiple AD targets while exhibiting more favorable drug metabolism and pharmacokinetic (DMPK) properties and safety profile. In particular, the most simplified acetophenone analog displays dual nanomolar inhibition of human acetylcholinesterase and butyrylcholinesterase (IC50 = 6 nM and 13 nM, respectively), moderately potent inhibition of human BACE-1 (48% inhibition at 15 µM) and Aβ42 and tau aggregation (73% and 68% inhibition, respectively, at 10 µM), favorable in vitro brain permeation, higher aqueous solubility (18 µM) and plasma stability (100/96/86% remaining in human/mouse/rat plasma after 6 h incubation), and lower acute toxicity in a model organism (zebrafish embryos; LC50 >> 100 µM) than the initial lead, thereby confirming the successful lead optimization by structural simplification.
Collapse
Affiliation(s)
- Caterina Pont
- Laboratory of Medicinal Chemistry (CSIC Associated Unit), Faculty of Pharmacy and Food Sciences, University of Barcelona, Av. Joan XXIII 27-31, E-08028 Barcelona, Spain
| | - Anna Sampietro
- Laboratory of Medicinal Chemistry (CSIC Associated Unit), Faculty of Pharmacy and Food Sciences, University of Barcelona, Av. Joan XXIII 27-31, E-08028 Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), E-08028 Barcelona, Spain
| | - F Javier Pérez-Areales
- Laboratory of Medicinal Chemistry (CSIC Associated Unit), Faculty of Pharmacy and Food Sciences, University of Barcelona, Av. Joan XXIII 27-31, E-08028 Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), E-08028 Barcelona, Spain
| | - Nunzia Cristiano
- Laboratory of Medicinal Chemistry (CSIC Associated Unit), Faculty of Pharmacy and Food Sciences, University of Barcelona, Av. Joan XXIII 27-31, E-08028 Barcelona, Spain
| | - Agustí Albalat
- Laboratory of Medicinal Chemistry (CSIC Associated Unit), Faculty of Pharmacy and Food Sciences, University of Barcelona, Av. Joan XXIII 27-31, E-08028 Barcelona, Spain
| | - Belén Pérez
- Department of Pharmacology, Therapeutics and Toxicology, Autonomous University of Barcelona, E-08193 Bellaterra, Spain
| | - Manuela Bartolini
- Department of Pharmacy and Biotechnology, University of Bologna, Via Belmeloro, 6, I-40126 Bologna, Italy
| | - Angela De Simone
- Department of Drug Science and Technology, University of Turin, I-10125 Torino, Italy
| | - Vincenza Andrisano
- Department for Life Quality Studies, Alma Mater Studiorum University of Bologna, Corso d'Augusto 237, I-47921 Rimini, Italy
| | - Marta Barenys
- Toxicology Unit, Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Av. Joan XXIII 27-31, E-08028 Barcelona, Spain
- German Centre for the Protection of Laboratory Animals (Bf3R), German Federal Institute for Risk Assessment (BfR), 10589 Berlin, Germany
| | - Elisabet Teixidó
- Toxicology Unit, Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Av. Joan XXIII 27-31, E-08028 Barcelona, Spain
- Institute of Nutrition and Food Safety of the University of Barcelona (INSA-UB), E-08921 Santa Coloma de Gramenet, Spain
| | - Raimon Sabaté
- Department of Pharmacy and Pharmaceutical Technology and Physical-Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Av. Joan XXIII 27-31, E-08028 Barcelona, Spain
| | - M Isabel Loza
- BioFarma Research Group, Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, Universidade de Santiago de Compostela, Av. de Barcelona s/n, E-15782 Santiago de Compostela, Spain
| | - José Brea
- BioFarma Research Group, Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, Universidade de Santiago de Compostela, Av. de Barcelona s/n, E-15782 Santiago de Compostela, Spain
| | - Diego Muñoz-Torrero
- Laboratory of Medicinal Chemistry (CSIC Associated Unit), Faculty of Pharmacy and Food Sciences, University of Barcelona, Av. Joan XXIII 27-31, E-08028 Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), E-08028 Barcelona, Spain
| |
Collapse
|
5
|
Sharma M, Pal P, Gupta SK. Advances in Alzheimer's disease: A multifaceted review of potential therapies and diagnostic techniques for early detection. Neurochem Int 2024; 177:105761. [PMID: 38723902 DOI: 10.1016/j.neuint.2024.105761] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 04/20/2024] [Accepted: 05/06/2024] [Indexed: 06/04/2024]
Abstract
Alzheimer's disease (AD) remains one of the most formidable neurological disorders, affecting millions globally. This review provides a holistic overview of the therapeutic strategies, both conventional and novel, aimed at mitigating the impact of AD. Initially, we delve into the conventional approach, emphasizing the role of Acetylcholinesterase (AChE) inhibition, which has been a cornerstone in AD management. As our understanding of AD evolves, several novel potential approaches emerge. We discuss the promising roles of Butyrylcholinesterase (BChE) inhibition, Tau Protein inhibitors, COX-2 inhibition, PPAR-γ agonism, and FAHH inhibition, among others. The potential of the endocannabinoids (eCB) system, cholesterol-lowering drugs, metal chelators, and MMPs inhibitors are also explored, culminating in the exploration of the pivotal role of microRNA in AD progression. Parallel to these therapeutic insights, we shed light on the novel tools and methodologies revolutionizing AD research. From the quantitative analysis of gene expression by qRTPCR to the evaluation of mitochondrial function using induced pluripotent stem cells (iPSCs), the advances in diagnostic and research tools offer renewed hope. Moreover, we explore the current landscape of clinical trials, highlighting the leading drug interventions and their respective stages of development. This comprehensive review concludes with a look into the future perspectives, capturing the potential breakthroughs and innovations on the horizon. Through a synthesis of current knowledge and emerging research, this article aims to provide a consolidated resource for clinicians, researchers, and academicians in the realm of Alzheimer's disease.
Collapse
Affiliation(s)
- Monika Sharma
- Faculty of Pharmacy, Department of Pharmacology, Swami Vivekanand Subharti University, Meerut, Uttar Pradesh, India
| | - Pankaj Pal
- Department of Pharmacy, Banasthali Vidyapith, Rajasthan, India.
| | - Sukesh Kumar Gupta
- KIET School of Pharmacy, KIET Group of Institutions, Ghaziabad, Uttar Pradesh, India; Department of Ophthalmology, Visual and Anatomical Sciences (OVAS), School of Medicine, Wayne State University, USA.
| |
Collapse
|
6
|
Basu S, Hendler-Neumark A, Bisker G. Rationally Designed Functionalization of Single-Walled Carbon Nanotubes for Real-Time Monitoring of Cholinesterase Activity and Inhibition in Plasma. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2309481. [PMID: 38358018 DOI: 10.1002/smll.202309481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 01/25/2024] [Indexed: 02/16/2024]
Abstract
Enzymes play a pivotal role in regulating numerous bodily functions. Thus, there is a growing need for developing sensors enabling real-time monitoring of enzymatic activity and inhibition. The activity and inhibition of cholinesterase (CHE) enzymes in blood plasma are fluorometrically monitored using near-infrared (NIR) fluorescent single-walled carbon nanotubes (SWCNTs) as probes, strategically functionalized with myristoylcholine (MC)- the substrate of CHE. A significant decrease in the fluorescence intensity of MC-suspended SWCNTs upon interaction with CHE is observed, attributed to the hydrolysis of the MC corona phase of the SWCNTs by CHE. Complementary measurements for quantifying choline, the product of MC hydrolysis, reveal a correlation between the fluorescence intensity decrease and the amount of released choline, rendering the SWCNTs optical sensors with real-time feedback in the NIR biologically transparent spectral range. Moreover, when synthetic and naturally abundant inhibitors inhibit the CHE enzymes present in blood plasma, no significant modulations of the MC-SWCNT fluorescence are observed, allowing effective detection of CHE inhibition. The rationally designed SWCNT sensors platform for monitoring of enzymatic activity and inhibition in clinically relevant samples is envisioned to not only advance the field of clinical diagnostics but also deepen further understanding of enzyme-related processes in complex biological fluids.
Collapse
Affiliation(s)
- Srestha Basu
- Department of Biomedical Engineering, Faculty of Engineering, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Adi Hendler-Neumark
- Department of Biomedical Engineering, Faculty of Engineering, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Gili Bisker
- Department of Biomedical Engineering, Faculty of Engineering, Tel Aviv University, Tel Aviv, 6997801, Israel
- Center for Physics and Chemistry of Living Systems, Tel Aviv University, Tel Aviv, 6997801, Israel
- Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, 6997801, Israel
- Center for Light-Matter Interaction, Tel Aviv University, Tel Aviv, 6997801, Israel
| |
Collapse
|
7
|
Sequeira L, Benfeito S, Fernandes C, Lima I, Peixoto J, Alves C, Machado CS, Gaspar A, Borges F, Chavarria D. Drug Development for Alzheimer's and Parkinson's Disease: Where Do We Go Now? Pharmaceutics 2024; 16:708. [PMID: 38931832 PMCID: PMC11206728 DOI: 10.3390/pharmaceutics16060708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 05/15/2024] [Accepted: 05/22/2024] [Indexed: 06/28/2024] Open
Abstract
Neurodegenerative diseases (NDs) are a set of progressive, chronic, and incurable diseases characterized by the gradual loss of neurons, culminating in the decline of cognitive and/or motor functions. Alzheimer's disease (AD) and Parkinson's disease (PD) are the most common NDs and represent an enormous burden both in terms of human suffering and economic cost. The available therapies for AD and PD only provide symptomatic and palliative relief for a limited period and are unable to modify the diseases' progression. Over the last decades, research efforts have been focused on developing new pharmacological treatments for these NDs. However, to date, no breakthrough treatment has been discovered. Hence, the development of disease-modifying drugs able to halt or reverse the progression of NDs remains an unmet clinical need. This review summarizes the major hallmarks of AD and PD and the drugs available for pharmacological treatment. It also sheds light on potential directions that can be pursued to develop new, disease-modifying drugs to treat AD and PD, describing as representative examples some advances in the development of drug candidates targeting oxidative stress and adenosine A2A receptors.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Fernanda Borges
- CIQUP-IMS—Centro de Investigação em Química da Universidade do Porto, Institute of Molecular Sciences, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, R. Campo Alegre s/n, 4169-007 Porto, Portugal
| | - Daniel Chavarria
- CIQUP-IMS—Centro de Investigação em Química da Universidade do Porto, Institute of Molecular Sciences, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, R. Campo Alegre s/n, 4169-007 Porto, Portugal
| |
Collapse
|
8
|
He J, Tam KY. Dual-target inhibitors of cholinesterase and GSK-3β to modulate Alzheimer's disease. Drug Discov Today 2024; 29:103914. [PMID: 38340951 DOI: 10.1016/j.drudis.2024.103914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 01/24/2024] [Accepted: 02/05/2024] [Indexed: 02/12/2024]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease that affects over 55 million patients worldwide. Most of the approved small-molecule drugs for AD have been designed to tackle a single pathological hallmark, such as cholinergic dysfunction or amyloid toxicity, and thus may not fully address the multifactorial nature of the disease. Inhibition of both cholinesterase and glycogen synthase kinase-3β (GSK-3β) has emerged as a promising strategy to modulate AD. However, the dual inhibition of these two targets posts challenges in molecular design: issues related to target engagements and biopharmaceutical properties in particular must be overcome. In this review, we discuss the physiopathological roles and structures of cholinesterase and GSK-3β as well as recently reported dual-target inhibitors. We critically evaluate the current status of the discovery of dual-target inhibitors of cholinesterase and GSK-3β, and highlight further perspectives.
Collapse
Affiliation(s)
- Junqiu He
- Faculty of Health Sciences, University of Macau SAR, Avenida de Universidade, Taipa, Macau SAR, China
| | - Kin Yip Tam
- Faculty of Health Sciences, University of Macau SAR, Avenida de Universidade, Taipa, Macau SAR, China.
| |
Collapse
|
9
|
Lu X, Li Y, Guan Q, Yang H, Liu Y, Du C, Wang L, Wang Q, Pei Y, Wu L, Sun H, Chen Y. Discovery, Structure-Based Modification, In Vitro, In Vivo, and In Silico Exploration of m-Sulfamoyl Benzoamide Derivatives as Selective Butyrylcholinesterase Inhibitors for Treating Alzheimer's Disease. ACS Chem Neurosci 2024; 15:1135-1156. [PMID: 38453668 DOI: 10.1021/acschemneuro.3c00737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2024] Open
Abstract
For the potential therapy of Alzheimer's disease (AD), butyrylcholinesterase (BChE) has gradually gained worldwide interest in the progression of AD. This study used a pharmacophore-based virtual screening (VS) approach to identify Z32439948 as a new BChE inhibitor. Aiding by molecular docking and molecular dynamics, essential binding information was disclosed. Specifically, a subpocket was found and structure-guided design of a series of novel compounds was conducted. Derivatives were evaluated in vitro for cholinesterase inhibition and physicochemical properties (BBB, log P, and solubility). The investigation involved docking, molecular dynamics, enzyme kinetics, and surface plasmon resonance as well. The study highlighted compounds 27a (hBChE IC50 = 0.078 ± 0.03 μM) and (R)-37a (hBChE IC50 = 0.005 ± 0.001 μM) as the top-ranked BChE inhibitors. These compounds showed anti-inflammatory activity and no apparent cytotoxicity against the human neuroblastoma (SH-SY5Y) and mouse microglia (BV2) cell lines. The most active compounds exhibited the ability to improve cognition in both scopolamine- and Aβ1-42 peptide-induced cognitive deficit models. They can be promising lead compounds with potential implications for treating the late stage of AD.
Collapse
Affiliation(s)
- Xin Lu
- Department of Pharmacy, College of Medicine, Institute of Translational Medicine, Yangzhou University, Yangzhou 225001, People's Republic of China
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou 225001, People's Republic of China
| | - Yueqing Li
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Qianwen Guan
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Huajing Yang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, People's Republic of China
| | - Yijun Liu
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Chenxi Du
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Lei Wang
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Qinghua Wang
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Yuqiong Pei
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, People's Republic of China
| | - Liang Wu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, People's Republic of China
| | - Haopeng Sun
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Yao Chen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, People's Republic of China
| |
Collapse
|
10
|
Ţînţaş ML, Peauger L, Barré A, Papamicaël C, Besson T, Sopkovà-de Oliveira Santos J, Gembus V, Levacher V. Design, synthesis and preliminary biological evaluation of rivastigmine-INDY hybrids as multitarget ligands against Alzheimer's disease by targeting butyrylcholinesterase and DYRK1A/CLK1 kinases. RSC Med Chem 2024; 15:963-980. [PMID: 38516603 PMCID: PMC10953492 DOI: 10.1039/d3md00708a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 02/16/2024] [Indexed: 03/23/2024] Open
Abstract
Based on a multitarget approach implementing rivastigmine-INDY hybrids 1, we identified a set of pseudo-irreversible carbamate-type inhibitors of eqBuChE that, after carbamate transfer at the active site serine residue, released the corresponding INDY analogues 2 endowed with hDYRK1A/hCLK1 kinases inhibitory properties. A SAR study and molecular docking investigation of both series of compounds 1 and 2 revealed that appropriate structural modifications at the carbamate moiety and at the N-appendage of the benzothiazole core led to potent and selective eqBuChE inhibitors with IC50 up to 27 nM and potent hDYRK1A and hCLK1 inhibitors with IC50 up to 106 nM and 17 nM respectively. Pleasingly, identification of the matched pair of compounds 1b/2b with a good balance between inhibition of eqBuChE and hDYRK1A/hCLK1 kinases (IC50 = 68 nM and IC50 = 529/54 nM, respectively) further validated our multitarget approach based on a sequential mechanism of action. In addition, target compound 1b exhibited a suitable ADMET profile, including good brain permeability and high stability in PBS, encouraging further biological investigation as a drug candidate.
Collapse
Affiliation(s)
- Mihaela-Liliana Ţînţaş
- INSA Rouen Normandie, Univ Rouen Normandie, CNRS, COBRA UMR 6014, Normandie Univ INC3M FR 3038 F-76000 Rouen France
| | | | - Anaïs Barré
- INSA Rouen Normandie, Univ Rouen Normandie, CNRS, COBRA UMR 6014, Normandie Univ INC3M FR 3038 F-76000 Rouen France
| | - Cyril Papamicaël
- INSA Rouen Normandie, Univ Rouen Normandie, CNRS, COBRA UMR 6014, Normandie Univ INC3M FR 3038 F-76000 Rouen France
| | - Thierry Besson
- INSA Rouen Normandie, Univ Rouen Normandie, CNRS, COBRA UMR 6014, Normandie Univ INC3M FR 3038 F-76000 Rouen France
| | - Jana Sopkovà-de Oliveira Santos
- UNICAEN, CERMN (Centre d'Etudes et de Recherche sur le Médicament de Normandie), Normandie Univ. Bd Becquerel F-14032 Caen France
| | - Vincent Gembus
- VFP Therapies 15 rue François Couperin 76000 Rouen France
| | - Vincent Levacher
- INSA Rouen Normandie, Univ Rouen Normandie, CNRS, COBRA UMR 6014, Normandie Univ INC3M FR 3038 F-76000 Rouen France
| |
Collapse
|
11
|
Bajad NG, Singh RB, T A G, Gutti G, Kumar A, Krishnamurthy S, Singh SK. Development of multi-targetable chalcone derivatives bearing N-aryl piperazine moiety for the treatment of Alzheimer's disease. Bioorg Chem 2024; 143:107082. [PMID: 38199142 DOI: 10.1016/j.bioorg.2023.107082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 12/26/2023] [Accepted: 12/28/2023] [Indexed: 01/12/2024]
Abstract
The multi-target directed ligand (MTDL) discovery has been gaining immense attention in the development of therapeutics for Alzheimer's disease (AD). The strategy has been evolved as an auspicious approach suitable to combat the heterogeneity and the multifactorial nature of AD. Therefore, multi-targetable chalcone derivatives bearing N-aryl piperazine moiety were designed, synthesized, and evaluated for the treatment of AD. All the synthesized compounds were screened for thein vitro activityagainst acetylcholinesterase (AChE), butylcholinesterase (BuChE), β-secretase-1 (BACE-1), and inhibition of amyloid β (Aβ) aggregation. Amongst all the tested derivatives, compound 41bearing unsubstituted benzylpiperazine fragment and para-bromo substitution at the chalcone scaffold exhibited balanced inhibitory profile against the selected targets. Compound 41 elicited favourable permeation across the blood-brain barrier in the PAMPA assay. The molecular docking and dynamics simulation studies revealed the binding mode analysis and protein-ligand stability ofthe compound with AChE and BACE-1. Furthermore,itameliorated cognitive dysfunctions and signified memory improvement in thein-vivobehavioural studies (scopolamine-induced amnesia model). Theex vivobiochemical analysis of mice brain homogenates established the reduced AChE and increased ACh levels. The antioxidant activity of compound 41 was accessed with the determination of catalase (CAT) and malondialdehyde (MDA) levels. The findings suggested thatcompound 41, containing a privileged chalcone scaffold, can act as a lead molecule for developing AD therapeutics.
Collapse
Affiliation(s)
- Nilesh Gajanan Bajad
- Pharmaceutical Chemistry Research Laboratory I, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi - 221005, India
| | | | - Gajendra T A
- Pharmaceutical Chemistry Research Laboratory I, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi - 221005, India
| | - Gopichand Gutti
- Pharmaceutical Chemistry Research Laboratory I, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi - 221005, India
| | - Ashok Kumar
- Pharmaceutical Chemistry Research Laboratory I, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi - 221005, India
| | - Sairam Krishnamurthy
- Pharmaceutical Chemistry Research Laboratory I, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi - 221005, India
| | - Sushil Kumar Singh
- Pharmaceutical Chemistry Research Laboratory I, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi - 221005, India.
| |
Collapse
|
12
|
Feyzi N, Ebadi A, Dastan D. Chimgin from Ferula haussknechtii as AChE inhibitor and confirmation of the absolute configuration. J Biomol Struct Dyn 2023:1-10. [PMID: 38109132 DOI: 10.1080/07391102.2023.2294176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 11/25/2023] [Indexed: 12/19/2023]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia worldwide and is classified as a neurodegenerative disorder. From a drug design perspective, natural products (NPs) are more drug-like and are highly compatible with biological systems compared to most synthetic libraries. NPs provide a more efficient and cost-effective approach to new drug discovery. However, the complexity of NPs makes their identification a challenging task. Chimgin, a bicyclic monoterpene with three chiral centers, exhibits a wide range of biological activity. Despite this, the exact structure of chimgin has remained unclear until now. In this study, we quantified the amount of chimgin in Ferula haussknechtii using analytical Reversed-phase high-pressure liquid chromatography equipped with photodiode array detector (RP-HPLC-PDA). Furthermore, we determined the absolute configuration of chimgin through electronic circular dichroism (ECD) spectroscopy and time-dependent density functional theory (TDDFT) calculations. Finally, we evaluated its inhibitory effect on AChE through in vitro and in silico studies. The extraction process yielded an output of 2.82 ± 0.10% with an exact amount of 0.62 ± 0.04 mg of chimgin per 100 g of plant. Based on the results of ECD and TDDFT calculation, the absolute configuration of chimgin was determined to be 1S, 2S, 4S. Chimgin exhibited an inhibitory effect on AChE with an IC50 of 37.43 µM and its mechanism of action was found to be competitive. HighlightsChimgin was isolated from the roots of Ferula haussknechtii.The amount of chimgin in the plant was determined by RP-HPLC-PDA.Its absolute configuration of chimgin was determined using ECD.In vitro acetylcholinesterase activity of the chimgin was evaluated.The docking and molecular dynamic simulation of chimgin was done.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Neda Feyzi
- Department of Pharmacognosy, School of Pharmacy, Medicinal Plants and Natural Products Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Ahmad Ebadi
- Department of Medicinal Chemistry, School of Pharmacy, Medicinal Plants and Natural Products Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Dara Dastan
- Department of Pharmacognosy, School of Pharmacy, Medicinal Plants and Natural Products Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
13
|
Sousa JLC, Albuquerque HMT, Silva AMS. Drug Discovery Based on Oxygen and Nitrogen (Non-)Heterocyclic Compounds Developed @LAQV-REQUI MTE/Aveiro. Pharmaceuticals (Basel) 2023; 16:1668. [PMID: 38139794 PMCID: PMC10747949 DOI: 10.3390/ph16121668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 11/18/2023] [Accepted: 11/27/2023] [Indexed: 12/24/2023] Open
Abstract
Artur Silva's research group has a long history in the field of medicinal chemistry. The development of new synthetic methods for oxygen (mostly polyphenols, e.g., 2- and 3-styrylchromones, xanthones, flavones) and nitrogen (e.g., pyrazoles, triazoles, acridones, 4-quinolones) heterocyclic compounds in order to be assessed as antioxidant, anti-inflammatory, antidiabetic, and anticancer agents has been the main core work of our research interests. Additionally, the synthesis of steroid-type compounds as anti-Alzheimer drugs as well as of several chromophores as important dyes for cellular imaging broadened our research scope. In this review article, we intend to provide an enlightened appraisal of all the bioactive compounds and their biological properties that were synthesized and studied by our research group in the last two decades.
Collapse
Affiliation(s)
| | | | - Artur M. S. Silva
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal; (J.L.C.S.); (H.M.T.A.)
| |
Collapse
|
14
|
Berrino E, Carradori S, Carta F, Melfi F, Gallorini M, Poli G, Tuccinardi T, Fernández-Bolaños JG, López Ó, Petzer JP, Petzer A, Guglielmi P, Secci D, Supuran CT. A Multitarget Approach against Neuroinflammation: Alkyl Substituted Coumarins as Inhibitors of Enzymes Involved in Neurodegeneration. Antioxidants (Basel) 2023; 12:2044. [PMID: 38136164 PMCID: PMC10740956 DOI: 10.3390/antiox12122044] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 11/14/2023] [Accepted: 11/21/2023] [Indexed: 12/24/2023] Open
Abstract
Neurodegenerative disorders (NDs) include a large range of diseases characterized by neural dysfunction with a multifactorial etiology. The most common NDs are Alzheimer's disease and Parkinson's disease, in which cholinergic and dopaminergic systems are impaired, respectively. Despite different brain regions being affected, oxidative stress and inflammation were found to be common triggers in the pathogenesis and progression of both diseases. By taking advantage of a multi-target approach, in this work we explored alkyl substituted coumarins as neuroprotective agents, capable to reduce oxidative stress and inflammation by inhibiting enzymes involved in neurodegeneration, among which are Carbonic Anhydrases (CAs), Monoamine Oxidases (MAOs), and Cholinesterases (ChEs). The compounds were synthesized and profiled against the three targeted enzymes. The binding mode of the most promising compounds (7 and 9) within MAO-A and -B was analyzed through molecular modeling studies, providing and explanation for the different selectivities observed for the MAO isoforms. In vitro biological studies using LPS-stimulated rat astrocytes showed that some compounds were able to counteract the oxidative stress-induced neuroinflammation and hamper interleukin-6 secretion, confirming the success of this multitarget approach.
Collapse
Affiliation(s)
- Emanuela Berrino
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy; (E.B.); (P.G.); (D.S.)
- NEUROFARBA Department, Sezione di Scienze Farmaceutiche e Nutraceutiche, Università degli Studi di Firenze, Via Ugo Schiff 6, 50019 Florence, Italy; (F.C.); (C.T.S.)
| | - Simone Carradori
- Department of Pharmacy, ‘‘G. d’Annunzio” University of Chieti-Pescara, via dei Vestini 31, 66100 Chieti, Italy; (F.M.); (M.G.)
| | - Fabrizio Carta
- NEUROFARBA Department, Sezione di Scienze Farmaceutiche e Nutraceutiche, Università degli Studi di Firenze, Via Ugo Schiff 6, 50019 Florence, Italy; (F.C.); (C.T.S.)
| | - Francesco Melfi
- Department of Pharmacy, ‘‘G. d’Annunzio” University of Chieti-Pescara, via dei Vestini 31, 66100 Chieti, Italy; (F.M.); (M.G.)
| | - Marialucia Gallorini
- Department of Pharmacy, ‘‘G. d’Annunzio” University of Chieti-Pescara, via dei Vestini 31, 66100 Chieti, Italy; (F.M.); (M.G.)
| | - Giulio Poli
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy; (G.P.); (T.T.)
| | - Tiziano Tuccinardi
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy; (G.P.); (T.T.)
| | - José G. Fernández-Bolaños
- Departamento de Química Orgánica, Facultad de Química, Universidad de Sevilla, Apartado 1203, 41012 Seville, Spain; (J.G.F.-B.); (Ó.L.)
| | - Óscar López
- Departamento de Química Orgánica, Facultad de Química, Universidad de Sevilla, Apartado 1203, 41012 Seville, Spain; (J.G.F.-B.); (Ó.L.)
| | - Jacobus P. Petzer
- Pharmaceutical Chemistry, School of Pharmacy and Centre of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom 2531, South Africa; (J.P.P.); (A.P.)
| | - Anél Petzer
- Pharmaceutical Chemistry, School of Pharmacy and Centre of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom 2531, South Africa; (J.P.P.); (A.P.)
| | - Paolo Guglielmi
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy; (E.B.); (P.G.); (D.S.)
| | - Daniela Secci
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy; (E.B.); (P.G.); (D.S.)
| | - Claudiu T. Supuran
- NEUROFARBA Department, Sezione di Scienze Farmaceutiche e Nutraceutiche, Università degli Studi di Firenze, Via Ugo Schiff 6, 50019 Florence, Italy; (F.C.); (C.T.S.)
| |
Collapse
|
15
|
Uliassi E, Bergamini C, Rizzardi N, Naldi M, Cores Á, Bartolini M, Carlos Menéndez J, Bolognesi ML. Quinolinetrione-tacrine hybrids as multi-target-directed ligands against Alzheimer's disease. Bioorg Med Chem 2023; 91:117419. [PMID: 37487339 DOI: 10.1016/j.bmc.2023.117419] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 07/04/2023] [Accepted: 07/18/2023] [Indexed: 07/26/2023]
Abstract
Multi-target drug discovery is one of the most active fields in the search for new drugs against Alzheimer's disease (AD). This is because the complexity of AD pathological network might be adequately tackled by multi-target-directed ligands (MTDLs) aimed at modulating simultaneously multiple targets of such a network. In a continuation of our efforts to develop MTDLs for AD, we have been focusing on the molecular hybridization of the acetylcholinesterase inhibitor tacrine with the aim of expanding its anti-AD profile. Herein, we manipulated the structure of a previously developed tacrine-quinone hybrid (1). We designed and synthesized a novel set of MTDLs (2-6) by replacing the naphthoquinone scaffold of 1 with that of 2,5,8-quinolinetrione. The most interesting hybrid 3 inhibited cholinesterase enzymes at nanomolar concentrations. In addition, 3 exerted antioxidant effects in menadione-induced oxidative stress of SH-SY5Y cells. Importantly, 3 also showed low hepatotoxicity and good anti-amyloid aggregation properties. Remarkably, we uncovered the potential of the quinolinetrione scaffold, as a novel anti-amyloid aggregation and antioxidant motif to be used in further anti-AD MTDL drug discovery endeavors.
Collapse
Affiliation(s)
- Elisa Uliassi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, 40126 Bologna, Italy
| | - Christian Bergamini
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, 40126 Bologna, Italy
| | - Nicola Rizzardi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, 40126 Bologna, Italy
| | - Marina Naldi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, 40126 Bologna, Italy
| | - Ángel Cores
- Department of Chemistry in Pharmaceutical Sciences, Organic and Medicinal Chemistry Unit, Faculty of Pharmacy, Universidad Complutense, 28040 Madrid, Spain
| | - Manuela Bartolini
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, 40126 Bologna, Italy
| | - J Carlos Menéndez
- Department of Chemistry in Pharmaceutical Sciences, Organic and Medicinal Chemistry Unit, Faculty of Pharmacy, Universidad Complutense, 28040 Madrid, Spain.
| | - Maria Laura Bolognesi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, 40126 Bologna, Italy.
| |
Collapse
|
16
|
Azmy EM, Nassar IF, Hagras M, Fawzy IM, Hegazy M, Mokhtar MM, Yehia AM, Ismail NS, Lashin WH. New indole derivatives as multitarget anti-Alzheimer's agents: synthesis, biological evaluation and molecular dynamics. Future Med Chem 2023; 15:473-495. [PMID: 37125532 DOI: 10.4155/fmc-2022-0228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2023] Open
Abstract
Background: Alzheimer's disease is a neurological disorder that causes brain cells to shrink and die. Aim: Thirteen novel 'oxathiolanyl', 'pyrazolyl' and 'pyrimidinyl' indole derivatives were designed and synthesized as anti-Alzheimer's disease treatment. Method: In vitro enzyme assay was performed against both AChE and BChE enzymes. In addition, antioxidant assay and cytotoxicity on a normal cell line were determined. Molecular docking and dynamic simulations were conducted to confirm the binding mode in both esterases' active sites. In silico absorption, distribution, metabolism, excretion and toxicity studies were also carried out. Results & conclusion: Compounds 5, 7 and 11 exhibited superior inhibitory activity against acetylcholinesterase and butyrylcholinesterase, with IC50 values of 0.042 and 3.003 μM, 2.54 and 0.207 μM and 0.052 and 2.529 μM, respectively, compared with donepezil.
Collapse
Affiliation(s)
- Eman M Azmy
- Department of Chemistry, Faculty of Women, Ain Shams University, Heliopolis, Cairo, 11457, Egypt
| | - Ibrahim F Nassar
- Faculty of Specific Education, Ain Shams University, 365 Ramsis Street, Abassia, Cairo, Egypt
| | - Mohamed Hagras
- Department of Pharmaceutical Organic Chemistry, College of Pharmacy (Boys), Al-Azhar University, Cairo, 11884, Egypt
| | - Iten M Fawzy
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Future University in Egypt, Cairo, 11835, Egypt
| | - Maghawry Hegazy
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
| | - Mahmoud Mohamed Mokhtar
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
| | - Amr Mohamed Yehia
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
| | - Nasser Sm Ismail
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Future University in Egypt, Cairo, 11835, Egypt
| | - Walaa H Lashin
- Department of Chemistry, Faculty of Women, Ain Shams University, Heliopolis, Cairo, 11457, Egypt
| |
Collapse
|
17
|
Wu D, Yu N, Gao Y, Xiong R, Liu L, Lei H, Jin S, Liu J, Liu Y, Xie J, Liu E, Zhou Q, Liu Y, Li S, Wei L, Lv J, Yu H, Zeng W, Zhou Q, Xu F, Luo MH, Zhang Y, Yang Y, Wang JZ. Targeting a vulnerable septum-hippocampus cholinergic circuit in a critical time window ameliorates tau-impaired memory consolidation. Mol Neurodegener 2023; 18:23. [PMID: 37060096 PMCID: PMC10103508 DOI: 10.1186/s13024-023-00614-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 03/12/2023] [Indexed: 04/16/2023] Open
Abstract
BACKGROUND Abnormal tau accumulation and cholinergic degeneration are hallmark pathologies in the brains of patients with Alzheimer's disease (AD). However, the sensitivity of cholinergic neurons to AD-like tau accumulation and strategies to ameliorate tau-disrupted spatial memory in terms of neural circuits still remain elusive. METHODS To investigate the effect and mechanism of the cholinergic circuit in Alzheimer's disease-related hippocampal memory, overexpression of human wild-type Tau (hTau) in medial septum (MS)-hippocampus (HP) cholinergic was achieved by specifically injecting pAAV-EF1α-DIO-hTau-eGFP virus into the MS of ChAT-Cre mice. Immunostaining, behavioral analysis and optogenetic activation experiments were used to detect the effect of hTau accumulation on cholinergic neurons and the MS-CA1 cholinergic circuit. Patch-clamp recordings and in vivo local field potential recordings were used to analyze the influence of hTau on the electrical signals of cholinergic neurons and the activity of cholinergic neural circuit networks. Optogenetic activation combined with cholinergic receptor blocker was used to detect the role of cholinergic receptors in spatial memory. RESULTS In the present study, we found that cholinergic neurons with an asymmetric discharge characteristic in the MS-hippocampal CA1 pathway are vulnerable to tau accumulation. In addition to an inhibitory effect on neuronal excitability, theta synchronization between the MS and CA1 subsets was significantly disrupted during memory consolidation after overexpressing hTau in the MS. Photoactivating MS-CA1 cholinergic inputs within a critical 3 h time window during memory consolidation efficiently improved tau-induced spatial memory deficits in a theta rhythm-dependent manner. CONCLUSIONS Our study not only reveals the vulnerability of a novel MS-CA1 cholinergic circuit to AD-like tau accumulation but also provides a rhythm- and time window-dependent strategy to target the MS-CA1 cholinergic circuit, thereby rescuing tau-induced spatial cognitive functions.
Collapse
Affiliation(s)
- Dongqin Wu
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Nana Yu
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yang Gao
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Rui Xiong
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Luping Liu
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, 999077, China
| | - Huiyang Lei
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Sen Jin
- The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 518055, China
| | - Jiale Liu
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yingzhou Liu
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jiazhao Xie
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Enjie Liu
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Qiuzhi Zhou
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yanchao Liu
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Shihong Li
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Linyu Wei
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jingru Lv
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Huilin Yu
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Wenbo Zeng
- State Key Laboratory of Virology, CAS Center for Excellence in Brain Science and Intelligence Technology (CEBSIT), Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Qiang Zhou
- State Key Laboratory of Chemical Oncogenomics, Guangdong Provincial Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Fuqiang Xu
- The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 518055, China
- Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Min-Hua Luo
- State Key Laboratory of Virology, CAS Center for Excellence in Brain Science and Intelligence Technology (CEBSIT), Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Yao Zhang
- Endocrine Department of Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430077, China.
| | - Ying Yang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226000, China.
| | - Jian-Zhi Wang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226000, China.
| |
Collapse
|
18
|
Martins MM, Branco PS, Ferreira LM. Enhancing the Therapeutic Effect in Alzheimer's Disease Drugs: The role of Polypharmacology and Cholinesterase inhibitors. ChemistrySelect 2023. [DOI: 10.1002/slct.202300461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2023]
Affiliation(s)
- M. Margarida Martins
- Department of Chemistry NOVA School of Science and Technology Campus da Caparica 2825-149 Caparica Portugal
| | - Paula S. Branco
- Department of Chemistry NOVA School of Science and Technology Campus da Caparica 2825-149 Caparica Portugal
| | - Luísa M. Ferreira
- Department of Chemistry NOVA School of Science and Technology Campus da Caparica 2825-149 Caparica Portugal
| |
Collapse
|
19
|
Cholinesterase Inhibitors from an Endophytic Fungus Aspergillus niveus Fv-er401: Metabolomics, Isolation and Molecular Docking. Molecules 2023; 28:molecules28062559. [PMID: 36985531 PMCID: PMC10052609 DOI: 10.3390/molecules28062559] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/04/2023] [Accepted: 03/08/2023] [Indexed: 03/14/2023] Open
Abstract
Alzheimer’s disease poses a global health concern with unmet demand requiring creative approaches to discover new medications. In this study, we investigated the chemical composition and the anticholinesterase activity of Aspergillus niveus Fv-er401 isolated from Foeniculum vulgare (Apiaceae) roots. Fifty-eight metabolites were identified using UHPLC-MS/MS analysis of the crude extract. The fungal extract showed acetylcholinesterase (AChE) and butyrylcholinesterase (BuChE) inhibitory effects with IC50 53.44 ± 1.57 and 48.46 ± 0.41 µg/mL, respectively. Two known metabolites were isolated, terrequinone A and citrinin, showing moderate AChE and BuChE inhibitory activity using the Ellman’s method (IC50 = 11.10 ± 0.38 µg/mL and 5.06 ± 0.15 µg/mL, respectively for AChE, and IC50 15.63 ± 1.27 µg/mL and 8.02 ± 0.08 µg/mL, respectively for BuChE). As evidenced by molecular docking, the isolated compounds and other structurally related metabolites identified by molecular networking had the required structural features for AChE and BuChE inhibition. Where varioxiranol G (−9.76 and −10.36 kcal/mol), penicitrinol B (−9.50 and −8.02 kcal/mol), dicitrinol A (−8.53 and −7.98 kcal/mol) and asterriquinone CT5 (−8.02 and −8.25 kcal/mol) showed better binding scores as AChE and BuChE inhibitors than the co-crystallized inhibitor (between −7.89 and 7.82 kcal/mol) making them promising candidates for the development of new drugs to treat Alzheimer’s.
Collapse
|
20
|
Jacob Y, Schneider B, Spies C, Heinrich M, von Haefen C, Kho W, Pohrt A, Müller A. In a secondary analysis from a randomised, double-blind placebo-controlled trial Dexmedetomidine blocks cholinergic dysregulation in delirium pathogenesis in patients with major surgery. Sci Rep 2023; 13:3971. [PMID: 36894596 PMCID: PMC9998872 DOI: 10.1038/s41598-023-30756-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 02/28/2023] [Indexed: 03/11/2023] Open
Abstract
Dexmedetomidine is an alpha-2 adrenoreceptor agonist with anti-inflammatory and anti-delirogenic properties. Pathogenesis of postoperative delirium (POD) includes cholinergic dysfunction and deregulated inflammatory response to surgical trauma. Acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) are discussed as biomarkers for both POD and severity in acute inflammation. To show whether there is a link between blood cholinesterase activities and dexmedetomidine, we performed a secondary analysis of a randomised, double-blind, placebo-controlled trial that recently showed a lower incidence of POD in the dexmedetomidine group. Abdominal or cardiac surgical patients aged ≥ 60 years were randomised to receive dexmedetomidine or placebo intra- and postoperatively in addition to standard general anaesthesia. We analysed the course of perioperative cholinesterase activities of 56 patients, measured preoperatively and twice postoperatively. Dexmedetomidine resulted in no change in AChE activity and caused a rapid recovery of BChE activity after an initial decrease, while placebo showed a significant decrease in both cholinesterase activities. There were no significant between-group differences at any point in time. From these data it can be assumed that dexmedetomidine could alleviate POD via altering the cholinergic anti-inflammatory pathway (CAIP). We advocate for further investigations to show the direct connection between dexmedetomidine and cholinesterase activity.
Collapse
Affiliation(s)
- Yanite Jacob
- Department of Anesthesiology and Operative Intensive Care Medicine (CCM, CVK), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, Charité Platz 1, 10117, Berlin, Germany
| | - Bill Schneider
- Department of Anesthesiology and Operative Intensive Care Medicine (CCM, CVK), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, Charité Platz 1, 10117, Berlin, Germany
| | - Claudia Spies
- Department of Anesthesiology and Operative Intensive Care Medicine (CCM, CVK), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, Charité Platz 1, 10117, Berlin, Germany
| | - Maria Heinrich
- Department of Anesthesiology and Operative Intensive Care Medicine (CCM, CVK), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, Charité Platz 1, 10117, Berlin, Germany.,Berlin Institute of Health (BIH), Anna-Louisa-Karsch 2, 10178, Berlin, Germany
| | - Clarissa von Haefen
- Department of Anesthesiology and Operative Intensive Care Medicine (CCM, CVK), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, Charité Platz 1, 10117, Berlin, Germany
| | - Widuri Kho
- Department of Anesthesiology and Operative Intensive Care Medicine (CCM, CVK), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, Charité Platz 1, 10117, Berlin, Germany
| | - Anne Pohrt
- Institute of Biometry and Clinical Epidemiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, Berlin, Germany
| | - Anika Müller
- Department of Anesthesiology and Operative Intensive Care Medicine (CCM, CVK), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, Charité Platz 1, 10117, Berlin, Germany.
| |
Collapse
|
21
|
Mota IFL, de Lima LS, Santana BDM, Gobbo GDAM, Bicca JVML, Azevedo JRM, Veras LG, Taveira RDAA, Pinheiro GB, Mortari MR. Alzheimer's Disease: Innovative Therapeutic Approaches Based on Peptides and Nanoparticles. Neuroscientist 2023; 29:78-96. [PMID: 34018874 DOI: 10.1177/10738584211016409] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Alzheimer's disease (AD) is the main cause of dementia in the world and its etiology is not yet fully understood. The pathology of AD is primarily characterized by intracellular neurofibrillary tangles and extracellular amyloid-β plaques. Unfortunately, few treatment options are available, and most treat symptoms, as is the case of acetylcholinesterase inhibitors (IAChE) and N-methyl-d-aspartate receptor antagonists. For more than 20 years pharmaceutical research has targeted the "amyloid cascade hypothesis," but this has not produced meaningful results, leading researchers to focus now on other characteristics of the disease and on multitarget approaches. This review aims to evaluate some new treatments that are being developed and studied. Among these are new treatments based on peptides, which have high selectivity and low toxicity; however, these compounds have a short half-life and encounter challenges when crossing the blood-brain barrier. The present review discusses up-and-coming peptides tested as treatments and explores some nanotechnological strategies to overcome the downsides. These compounds are promising, as they not only act on the symptoms but also aim to prevent progressive neuronal loss.
Collapse
Affiliation(s)
- Isabela F L Mota
- Laboratory of Neuropharmacology, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - Larissa S de Lima
- Laboratory of Neuropharmacology, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - Bruna de M Santana
- Laboratory of Neuropharmacology, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - Giovanna de A M Gobbo
- Laboratory of Neuropharmacology, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - João V M L Bicca
- Laboratory of Neuropharmacology, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - Juliana R M Azevedo
- Laboratory of Neuropharmacology, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - Letícia G Veras
- Laboratory of Neuropharmacology, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - Rodrigo de A A Taveira
- Laboratory of Neuropharmacology, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - Gabriela B Pinheiro
- Laboratory of Neuropharmacology, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - Márcia R Mortari
- Laboratory of Neuropharmacology, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| |
Collapse
|
22
|
Raghuvanshi R, Jamwal A, Nandi U, Bharate SB. Multitargeted C9-substituted ester and ether derivatives of berberrubine for Alzheimer's disease: Design, synthesis, biological evaluation, metabolic stability, and pharmacokinetics. Drug Dev Res 2023; 84:121-140. [PMID: 36461610 DOI: 10.1002/ddr.22017] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 10/20/2022] [Accepted: 11/08/2022] [Indexed: 12/04/2022]
Abstract
Berberrubine is a naturally occurring isoquinoline alkaloid and a bioactive metabolite of berberine. Berberine exhibits a wide range of pharmacological activities, including cholinesterase inhibition. The cholinesterase inhibitors provide symptomatic treatment for Alzheimer's disease; however, multitarget-directed ligands have the potential as disease-modifying therapeutics. Herein, we prepared a series of C9-substituted berberrubine derivatives intending to discover dual cholinesterase and beta-site amyloid-precursor protein cleaving enzyme 1 (BACE-1) inhibitors. Most synthesized derivatives possessed balanced dual inhibition (AChE and BChE) activity in the submicromolar range and a moderate inhibition against BACE-1. Two most active ester derivatives, 12a and 11d, display inhibition of AChE, BChE, and BACE-1. The 3-methoxybenzoyl ester derivative, 12a, inhibits electric eel acetylcholinesterase (EeAChE), equine serum butyrylcholinesterase (eqBChE), and human hBACE-1 with IC50 values of 0.5, 4.3, and 11.9 μM, respectively and excellent BBB permeability (Pe = 8 × 10-6 cm/s). The ester derivative 12a is metabolically unstable; however, its ether analog 13 is stable in HLM and exhibits inhibition of AChE, BChE, and BACE-1 with IC50 values of 0.44, 3.8, and 17.9 μM, respectively. The ether analog also inhibits self-aggregation of Aβ and crosses BBB (Pe = 7.3 × 10-6 cm/s). Administration of 13 at 5 mg/kg (iv) in Wistar rats showed excellent plasma exposure with AUC0-∞ of 28,834 ng min/ml. In conclusion, the multitargeted berberrubine ether derivative 13 is CNS permeable and has good ADME properties.
Collapse
Affiliation(s)
- Rinky Raghuvanshi
- Natural Products & Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India.,Academy of Scientific & Innovative Research, Ghaziabad, India
| | - Ashiya Jamwal
- Academy of Scientific & Innovative Research, Ghaziabad, India.,Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
| | - Utpal Nandi
- Academy of Scientific & Innovative Research, Ghaziabad, India.,Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
| | - Sandip B Bharate
- Natural Products & Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India.,Academy of Scientific & Innovative Research, Ghaziabad, India
| |
Collapse
|
23
|
Oliyaei N, Moosavi-Nasab M, Tanideh N, Iraji A. Multiple roles of fucoxanthin and astaxanthin against Alzheimer's disease: Their pharmacological potential and therapeutic insights. Brain Res Bull 2023; 193:11-21. [PMID: 36435362 DOI: 10.1016/j.brainresbull.2022.11.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 11/14/2022] [Accepted: 11/22/2022] [Indexed: 11/24/2022]
Abstract
Alzheimer's disease (AD) is the most devastating neurodegenerative disorder affecting the elderly. The exact pathology of AD is not yet fully understood and several hallmarks such as the deposition of amyloid-β, tau hyperphosphorylation, and neuroinflammation, as well as mitochondrial, metal ions, autophagy, and cholinergic dysfunctions are known as pathologic features of AD. Since no definitive treatment has been proposed to target AD to date, many natural products have shown promising preventive potentials and contributed to slowing down the disease progression. Algae is a promising source of novel bioactive substances known to prevent neurodegenerative disorders including AD. In this context, fucoxanthin and astaxanthin, natural carotenoids abundant in algae, has shown to possess neuroprotective properties through antioxidant, and anti-inflammatory characteristics in modulating the symptoms of AD. Fucoxanthin and astaxanthin exhibit anti-AD activities by inhibition of AChE, BuChE, BACE-1, and MAO, suppression of Aβ accumulation. Also, fucoxanthin and astaxanthin inhibit apoptosis induced by Aβ1-42 and H2O2-induced cytotoxicity, and modulate the antioxidant enzymes (SOD and CAT), through inhibition of the ERK pathway. Moreover, cellular and animal studies on the beneficial effects of fucoxanthin and astaxanthin against AD were also reviewed. The potential role of fucoxanthin and astaxanthin exhibits great efficacy for the management of AD by acting on multiple targets.
Collapse
Affiliation(s)
- Najmeh Oliyaei
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Seafood Processing Research Center, School of Agriculture, Shiraz University, Shiraz, Iran
| | - Marzieh Moosavi-Nasab
- Seafood Processing Research Center, School of Agriculture, Shiraz University, Shiraz, Iran; Department of Food Science and Technology, School of Agriculture, Shiraz University, Shiraz, Iran.
| | - Nader Tanideh
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Aida Iraji
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Central Research laboratory, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
24
|
Lista S, Vergallo A, Teipel SJ, Lemercier P, Giorgi FS, Gabelle A, Garaci F, Mercuri NB, Babiloni C, Gaire BP, Koronyo Y, Koronyo-Hamaoui M, Hampel H, Nisticò R. Determinants of approved acetylcholinesterase inhibitor response outcomes in Alzheimer's disease: relevance for precision medicine in neurodegenerative diseases. Ageing Res Rev 2023; 84:101819. [PMID: 36526257 DOI: 10.1016/j.arr.2022.101819] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 11/11/2022] [Accepted: 12/09/2022] [Indexed: 12/14/2022]
Abstract
Acetylcholinesterase inhibitors (ChEI) are the global standard of care for the symptomatic treatment of Alzheimer's disease (AD) and show significant positive effects in neurodegenerative diseases with cognitive and behavioral symptoms. Although experimental and large-scale clinical evidence indicates the potential long-term efficacy of ChEI, primary outcomes are generally heterogeneous across outpatient clinics and regional healthcare systems. Sub-optimal dosing or slow tapering, heterogeneous guidelines about the timing for therapy initiation (prodromal versus dementia stages), healthcare providers' ambivalence to treatment, lack of disease awareness, delayed medical consultation, prescription of ChEI in non-AD cognitive disorders, contribute to the negative outcomes. We present an evidence-based overview of determinants, spanning genetic, molecular, and large-scale networks, involved in the response to ChEI in patients with AD and other neurodegenerative diseases. A comprehensive understanding of cerebral and retinal cholinergic system dysfunctions along with ChEI response predictors in AD is crucial since disease-modifying therapies will frequently be prescribed in combination with ChEI. Therapeutic algorithms tailored to genetic, biological, clinical (endo)phenotypes, and disease stages will help leverage inter-drug synergy and attain optimal combined response outcomes, in line with the precision medicine model.
Collapse
Affiliation(s)
- Simone Lista
- Memory Resources and Research Center (CMRR), Neurology Department, Gui de Chauliac University Hospital, Montpellier, France; School of Pharmacy, University of Rome "Tor Vergata", Rome, Italy.
| | - Andrea Vergallo
- Sorbonne University, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France
| | - Stefan J Teipel
- German Center for Neurodegenerative Diseases (DZNE) Rostock/Greifswald, Rostock, Germany; Department of Psychosomatic Medicine and Psychotherapy, University Medicine Rostock, Rostock, Germany
| | - Pablo Lemercier
- Sorbonne University, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France
| | - Filippo Sean Giorgi
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Pisa, Italy
| | - Audrey Gabelle
- Memory Resources and Research Center (CMRR), Neurology Department, Gui de Chauliac University Hospital, Montpellier, France
| | - Francesco Garaci
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy; Casa di Cura "San Raffaele Cassino", Cassino, Italy
| | - Nicola B Mercuri
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy; IRCCS Santa Lucia Foundation, Rome, Italy
| | - Claudio Babiloni
- Department of Physiology and Pharmacology "Erspamer", Sapienza University of Rome, Rome, Italy; Hospital San Raffaele Cassino, Cassino, Italy
| | - Bhakta Prasad Gaire
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Yosef Koronyo
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Maya Koronyo-Hamaoui
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Department of Biomedical Sciences, Division of Applied Cell Biology and Physiology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Harald Hampel
- Sorbonne University, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France
| | - Robert Nisticò
- School of Pharmacy, University of Rome "Tor Vergata", Rome, Italy; Laboratory of Pharmacology of Synaptic Plasticity, EBRI Rita Levi-Montalcini Foundation, Rome, Italy.
| |
Collapse
|
25
|
Roy R, Khan J, Pradhan K, Nayak P, Sarkar A, Nandi S, Ghosh S, Ram H, Ghosh S. Short Peptoid Evolved from the Key Hydrophobic Stretch of Amyloid-β42 Peptide Serves as a Potent Therapeutic Lead of Alzheimer's Disease. ACS Chem Neurosci 2023; 14:246-260. [PMID: 36583718 DOI: 10.1021/acschemneuro.2c00549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Amyloid-β 42(Aβ42), an enzymatically cleaved (1-42 amino acid long) toxic peptide remnant, has long been reported to play the key role in Alzheimer's disease (AD). Aβ42 also plays the key role in the onset of other AD-related factors including hyperphosphorylation of tau protein that forms intracellular neurofibrillary tangles, imbalances in the function of the neurotransmitter acetylcholine, and even generation of reactive oxygen species (ROS), disrupting the cytoskeleton and homeostasis of the cell. To address these issues, researchers have tried to construct several strategies to target multiple aspects of the disease but failed to produce any clinically successful therapeutic molecules. In this article, we report a new peptoid called RA-1 that was designed and constructed from the hydrophobic stretch of the Aβ42 peptide, 16KLVFFA21. This hydrophobic stretch is primarily responsible for the Aβ42 peptide aggregation. Experimental study showed that the RA-1 peptoid is stable under proteolytic conditions, can stabilize the microtubule, and can inhibit the formation of toxic Aβ42 aggregates by attenuating hydrophobic interactions between Aβ42 monomers. Furthermore, results from various intracellular assays showed that RA-1 inhibits Aβ42 fibril formation caused by the imbalance in AchE activity, reduces the production of cytotoxic reactive oxygen species (ROS), and promotes neurite outgrowth even in the toxic environment. Remarkably, we have also demonstrated that our peptoid has significant ability to improve the cognitive ability and memory impairment in in vivo rats exposed to AlCl3 and d-galactose (d-gal) dementia model. These findings are also validated with histological studies. Overall, our newly developed peptoid emerges as a multimodal potent therapeutic lead molecule against AD.
Collapse
Affiliation(s)
- Rajsekhar Roy
- Department of Bioscience & Bioengineering, Indian Institute of Technology Jodhpur, NH 65, Surpura Bypass Road, Karwar, Jodhpur, Rajasthan 342037, India
| | - Juhee Khan
- Department of Bioscience & Bioengineering, Indian Institute of Technology Jodhpur, NH 65, Surpura Bypass Road, Karwar, Jodhpur, Rajasthan 342037, India.,Organic and Medicinal Chemistry and Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, West Bengal, India
| | - Krishnangsu Pradhan
- Organic and Medicinal Chemistry and Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, West Bengal, India
| | - Prasunpriya Nayak
- Department of Physiology, All India Institute of Medical Sciences, Basni, Phase II, Jodhpur, Rajasthan 342005, India
| | - Ankan Sarkar
- Department of Bioscience & Bioengineering, Indian Institute of Technology Jodhpur, NH 65, Surpura Bypass Road, Karwar, Jodhpur, Rajasthan 342037, India
| | - Subhadra Nandi
- Department of Bioscience & Bioengineering, Indian Institute of Technology Jodhpur, NH 65, Surpura Bypass Road, Karwar, Jodhpur, Rajasthan 342037, India
| | - Surojit Ghosh
- Interdisciplinary Research Platform, Smart Health Care, Indian Institute of Technology Jodhpur, NH65, Nagaur Road, Karwar, Jodhpur 342037, India
| | - Heera Ram
- Department of Zoology, Jai Narain Vyas University, Jodhpur, Rajasthan 342001, India
| | - Surajit Ghosh
- Department of Bioscience & Bioengineering, Indian Institute of Technology Jodhpur, NH 65, Surpura Bypass Road, Karwar, Jodhpur, Rajasthan 342037, India.,Organic and Medicinal Chemistry and Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, West Bengal, India.,Interdisciplinary Research Platform, Smart Health Care, Indian Institute of Technology Jodhpur, NH65, Nagaur Road, Karwar, Jodhpur 342037, India
| |
Collapse
|
26
|
Xia ZD, Ma RX, Wen JF, Zhai YF, Wang YQ, Wang FY, Liu D, Zhao XL, Sun B, Jia P, Zheng XH. Pathogenesis, Animal Models, and Drug Discovery of Alzheimer's Disease. J Alzheimers Dis 2023; 94:1265-1301. [PMID: 37424469 DOI: 10.3233/jad-230326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Alzheimer's disease (AD), the most common cause of dementia, is a chronic neurodegenerative disease induced by multiple factors. The high incidence and the aging of the global population make it a growing global health concern with huge implications for individuals and society. The clinical manifestations are progressive cognitive dysfunction and lack of behavioral ability, which not only seriously affect the health and quality of life of the elderly, but also bring a heavy burden to the family and society. Unfortunately, almost all the drugs targeting the classical pathogenesis have not achieved satisfactory clinical effects in the past two decades. Therefore, the present review provides more novel ideas on the complex pathophysiological mechanisms of AD, including classical pathogenesis and a variety of possible pathogenesis that have been proposed in recent years. It will be helpful to find out the key target and the effect pathway of potential drugs and mechanisms for the prevention and treatment of AD. In addition, the common animal models in AD research are outlined and we examine their prospect for the future. Finally, Phase I, II, III, and IV randomized clinical trials or on the market of drugs for AD treatment were searched in online databases (Drug Bank Online 5.0, the U.S. National Library of Medicine, and Alzforum). Therefore, this review may also provide useful information in the research and development of new AD-based drugs.
Collapse
Affiliation(s)
- Zhao-Di Xia
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Northwest University, Xi'an, PR China
| | - Ruo-Xin Ma
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Northwest University, Xi'an, PR China
| | - Jin-Feng Wen
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Northwest University, Xi'an, PR China
| | - Yu-Fei Zhai
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Northwest University, Xi'an, PR China
| | - Yu-Qi Wang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Northwest University, Xi'an, PR China
| | - Feng-Yun Wang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Northwest University, Xi'an, PR China
| | - Dan Liu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Northwest University, Xi'an, PR China
| | - Xiao-Long Zhao
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Northwest University, Xi'an, PR China
| | - Bao Sun
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Northwest University, Xi'an, PR China
- Department of Pharmacy, The Second Affiliated Hospital of Xi'an Medical University, Xi'an, PR China
| | - Pu Jia
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Northwest University, Xi'an, PR China
| | - Xiao-Hui Zheng
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Northwest University, Xi'an, PR China
| |
Collapse
|
27
|
8- Hydroxyquinolylnitrones as multifunctional ligands for the therapy of neurodegenerative diseases. Acta Pharm Sin B 2023; 13:2152-2175. [DOI: 10.1016/j.apsb.2023.01.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 12/28/2022] [Accepted: 01/05/2023] [Indexed: 01/20/2023] Open
|
28
|
Li Q, Ma Z, Qin S, Zhao WJ. Virtual Screening-Based Drug Development for the Treatment of Nervous System Diseases. Curr Neuropharmacol 2023; 21:2447-2464. [PMID: 36043797 PMCID: PMC10616913 DOI: 10.2174/1570159x20666220830105350] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 08/04/2022] [Accepted: 08/19/2022] [Indexed: 11/22/2022] Open
Abstract
The incidence rate of nervous system diseases has increased in recent years. Nerve injury or neurodegenerative diseases usually cause neuronal loss and neuronal circuit damage, which seriously affect motor nerve and autonomic nervous function. Therefore, safe and effective treatment is needed. As traditional drug research becomes slower and more expensive, it is vital to enlist the help of cutting- edge technology. Virtual screening (VS) is an attractive option for the identification and development of promising new compounds with high efficiency and low cost. With the assistance of computer- aided drug design (CADD), VS is becoming more and more popular in new drug development and research. In recent years, it has become a reality to transform non-neuronal cells into functional neurons through small molecular compounds, which provides a broader application prospect than transcription factor-mediated neuronal reprogramming. This review mainly summarizes related theory and technology of VS and the drug research and development using VS technology in nervous system diseases in recent years, and focuses more on the potential application of VS technology in neuronal reprogramming, thus facilitating new drug design for both prevention and treatment of nervous system diseases.
Collapse
Affiliation(s)
- Qian Li
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, Jiangsu, P.R. China
| | - Zhaobin Ma
- College of Life Science and Technology, Kunming University of Science and Technology, Kunming 650504, Yunnan, P.R. China
| | - Shuhua Qin
- College of Life Science and Technology, Kunming University of Science and Technology, Kunming 650504, Yunnan, P.R. China
| | - Wei-Jiang Zhao
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, Jiangsu, P.R. China
- Department of Cell Biology, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, Jiangsu, P.R. China
| |
Collapse
|
29
|
Alan E, Kerry Z, Sevin G. Molecular mechanisms of Alzheimer's disease: From therapeutic targets to promising drugs. Fundam Clin Pharmacol 2022; 37:397-427. [PMID: 36576325 DOI: 10.1111/fcp.12861] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 12/06/2022] [Accepted: 12/22/2022] [Indexed: 12/29/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease characterized by cognitive impairment so widespread that it interferes with a person's ability to complete daily activities. AD is becoming increasingly common, and it is estimated that the number of patients will reach 152 million by 2050. Current treatment options for AD are symptomatic and have modest benefits. Therefore, considering the human, social, and economic burden of the disease, the development of drugs with the potential to alter disease progression has become a global priority. In this review, the molecular mechanisms involved in the pathology of AD were evaluated as therapeutic targets. The main aim of the review is to focus on new knowledge about mitochondrial dysfunction, oxidative stress, and neuronal transmission in AD, as well as a range of cellular signaling mechanisms and associated treatments. Important molecular interactions leading to AD were described in amyloid cascade and in tau protein function, oxidative stress, mitochondrial dysfunction, cholinergic and glutamatergic neurotransmission, cAMP-regulatory element-binding protein (CREB), the silent mating type information regulation 2 homolog 1 (SIRT-1), neuroinflammation (glial cells), and synaptic alterations. This review summarizes recent experimental and clinical research in AD pathology and analyzes the potential of therapeutic applications based on molecular disease mechanisms.
Collapse
Affiliation(s)
- Elif Alan
- Department of Pharmacology, Faculty of Pharmacy, Ege University, Izmir, Turkey
| | - Zeliha Kerry
- Department of Pharmacology, Faculty of Pharmacy, Ege University, Izmir, Turkey
| | - Gulnur Sevin
- Department of Pharmacology, Faculty of Pharmacy, Ege University, Izmir, Turkey
| |
Collapse
|
30
|
Lamie PF, Abdel-Fattah MM, Philoppes JN. Design and synthesis of new indole drug candidates to treat Alzheimer's disease and targeting neuro-inflammation using a multi-target-directed ligand (MTDL) strategy. J Enzyme Inhib Med Chem 2022; 37:2660-2678. [PMID: 36146947 PMCID: PMC9518246 DOI: 10.1080/14756366.2022.2126464] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
A novel series of indole-based compounds was designed, synthesised, and evaluated as anti-Alzheimer’s and anti-neuroinflammatory agents. The designed compounds were in vitro evaluated for their AChE and BuChE inhibitory activities. The obtained results revealed that compound 3c had higher selectivity for AChE than BuChE, while, 4a, 4b, and 4d showed selectivity for BuChE over AChE. Compounds 5b, 6b, 7c, and 10b exerted dual AChE/BuChE inhibitory activities at nanomolar range. Compounds 5b and 6b had the ability to inhibit the self-induced Aβ amyloid aggregation. Different anti-inflammatory mediators (NO, COX-2, IL-1β, and TNF-α) were assessed for compounds 5b and 6b. Cytotoxic effect of 5b and 6b against human neuroblastoma (SH-SY5Y) and normal hepatic (THLE2) cell lines was screened in vitro. Molecular docking study inside rhAChE and hBuChE active sites, drug-likeness, and ADMET prediction were performed.
Collapse
Affiliation(s)
- Phoebe F Lamie
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Maha M Abdel-Fattah
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - John N Philoppes
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| |
Collapse
|
31
|
Saeedi M, Iraji A, Vahedi-Mazdabadi Y, Alizadeh A, Edraki N, Firuzi O, Eftekhari M, Akbarzadeh T. Cinnamomum verum J. Presl. Bark essential oil: in vitro investigation of anti-cholinesterase, anti-BACE1, and neuroprotective activity. BMC Complement Med Ther 2022; 22:303. [DOI: 10.1186/s12906-022-03767-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 10/25/2022] [Indexed: 11/19/2022] Open
Abstract
Abstract
Background
Cinnamomum verum J. Presl. (Lauraceae), Myrtus communis L. (Myrtaceae), Ruta graveolens L. (Rutaaceae), Anethum graveolens L. (Apiaceae), Myristica fragrans Houtt. (Myristicaceae), and Crocus sativus L. (Iridaceae) have been recommended for improvement of memory via inhalation, in Iranian Traditional Medicine (ITM). In this respect, the essential oils (EOs) from those plants were obtained and evaluated for cholinesterase (ChE) inhibitory activity as ChE inhibitors are the available drugs in the treatment of Alzheimer’s disease (AD).
Methods
EOs obtained from the plants under investigation, were evaluated for their potential to inhibit acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) in vitro based on the modified Ellman’s method. The most potent EO was candidate for the investigation of its beta-secretase 1 (BACE1) inhibitory activity and neuroprotectivity.
Results
Among all EOs, C. verum demonstrated the most potent activity toward AChE and BChE with IC50 values of 453.7 and 184.7 µg/mL, respectively. It also showed 62.64% and 41.79% inhibition against BACE1 at the concentration of 500 and 100 mg/mL, respectively. However, it depicted no neuroprotective potential against β-amyloid (Aβ)-induced neurotoxicity in PC12 cells. Also, identification of chemical composition of C. verum EO was achieved via gas chromatography-mass spectrometry (GC-MS) analysis and the major constituent; (E)-cinnamaldehyde, was detected as 68.23%.
Conclusion
Potent BChE inhibitory activity of C. verum EO can be considered in the development of cinnamon based dietary supplements for the management of patients with advanced AD.
Collapse
|
32
|
Keuler T, Lemke C, Elsinghorst PW, Iriepa I, Chioua M, Martínez-Grau MA, Beadle CD, Vetman T, López-Muñoz F, Wille T, Bartz U, Deuther-Conrad W, Marco-Contelles J, Gütschow M. The Chemotype of Chromanones as a Privileged Scaffold for Multineurotarget Anti-Alzheimer Agents. ACS Pharmacol Transl Sci 2022; 5:1097-1108. [PMID: 36407962 PMCID: PMC9667544 DOI: 10.1021/acsptsci.2c00097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Indexed: 11/28/2022]
Abstract
The multifactorial nature of Alzheimer's disease necessitates the development of agents able to interfere with different relevant targets. A series of 22 tailored chromanones was conceptualized, synthesized, and subjected to biological evaluation. We identified one representative bearing a linker-connected azepane moiety (compound 19) with balanced pharmacological properties. Compound 19 exhibited inhibitory activities against human acetyl-, butyrylcholinesterase and monoamine oxidase-B, as well as high affinity to both the σ1 and σ2 receptors. Our study provides a framework for the development of further chromanone-based multineurotarget agents.
Collapse
Affiliation(s)
- Tim Keuler
- Pharmaceutical
Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany
| | - Carina Lemke
- Pharmaceutical
Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany
| | - Paul W. Elsinghorst
- Pharmaceutical
Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany
- Central
Institute of the Bundeswehr Medical Service Munich, Ingolstädter Landstraße 102, 85748 Garching Germany
| | - Isabel Iriepa
- Universidad
de Alcalá, Departamento de Química
Orgánica y Química Inorgánica, Ctra. Madrid-Barcelona, 28871 Alcalá de Henares, Madrid España
| | - Mourad Chioua
- Laboratory
of Medicinal Chemistry, IQOG, CSIC, C/Juan de la Cierva 3, 28006 Madrid, Spain
| | | | - Christopher D. Beadle
- Lilly Research
Centre, Eli Lilly & Company, Erl Wood Manor, Windlesham, Surrey GU20
6PH, United Kingdom
| | - Tatiana Vetman
- Lilly
Research Laboratories, Eli Lilly & Company, Indianapolis, Indiana 46285, United States
| | - Francisco López-Muñoz
- Faculty
of Health, Camilo José Cela University of Madrid (UCJC), Neuropsychopharmacology Unit, “Hospital 12 de Octubre” Research
Institute, 28692 Madrid, Spain
| | - Timo Wille
- Bundeswehr
Institute of Pharmacology and Toxicology, Neuherbergstraße 11, 80937 München, Germany
| | - Ulrike Bartz
- Department
of Natural Sciences, University of Applied
Sciences Bonn-Rhein-Sieg, von-Liebig-Straße 20, 53359 Rheinbach, Germany
| | - Winnie Deuther-Conrad
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Department of Neuroradiopharmaceuticals, 04318 Leipzig, Germany
| | - José Marco-Contelles
- Laboratory
of Medicinal Chemistry, IQOG, CSIC, C/Juan de la Cierva 3, 28006 Madrid, Spain
| | - Michael Gütschow
- Pharmaceutical
Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany
| |
Collapse
|
33
|
Han N, Wen Y, Liu Z, Zhai J, Li S, Yin J. Advances in the roles and mechanisms of lignans against Alzheimer’s disease. Front Pharmacol 2022; 13:960112. [PMID: 36313287 PMCID: PMC9596774 DOI: 10.3389/fphar.2022.960112] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 08/24/2022] [Indexed: 11/30/2022] Open
Abstract
Background: Alzheimer’s disease (AD) is a serious neurodegenerative disease associated with the memory and cognitive impairment. The occurrence of AD is due to the accumulation of amyloid β-protein (Aβ) plaques and neurofibrillary tangles (NFTs) in the brain tissue as well as the hyperphosphorylation of Tau protein in neurons, doing harm to the human health and even leading people to death. The development of neuroprotective drugs with small side effects and good efficacy is focused by scientists all over the world. Natural drugs extracted from herbs or plants have become the preferred resources for new candidate drugs. Lignans were reported to effectively protect nerve cells and alleviate memory impairment, suggesting that they might be a prosperous class of compounds in treating AD. Objective: To explore the roles and mechanisms of lignans in the treatment of neurological diseases, providing proofs for the development of lignans as novel anti-AD drugs. Methods: Relevant literature was extracted and retrieved from the databases including China National Knowledge Infrastructure (CNKI), Elsevier, Science Direct, PubMed, SpringerLink, and Web of Science, taking lignan, anti-inflammatory, antioxidant, apoptosis, nerve regeneration, nerve protection as keywords. The functions and mechanisms of lignans against AD were summerized. Results: Lignans were found to have the effects of regulating vascular disorders, anti-infection, anti-inflammation, anti-oxidation, anti-apoptosis, antagonizing NMDA receptor, suppressing AChE activity, improving gut microbiota, so as to strengthening nerve protection. Among them, dibenzocyclooctene lignans were most widely reported and might be the most prosperous category in the develpment of anti-AD drugs. Conclusion: Lignans displayed versatile roles and mechanisms in preventing the progression of AD in in vitro and in vivo models, supplying potential candidates for the treatment of nerrodegenerative diseases.
Collapse
|
34
|
Guo Q, Cai Q, Huang F, Wei Z, Wang JZ, Zhang B, Liu R, Yang Y, Wang X, Li HL. The Therapeutic Effects of Seven Lycophyte Compounds on Cell Models of Alzheimer’s Disease. J Alzheimers Dis 2022; 90:795-809. [DOI: 10.3233/jad-220704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background: As an acetylcholinesterase inhibitor (AChEI), Huperzine-A (Hup-A) is marketed for treatment of mild to moderate Alzheimer’s disease (AD) for decades in China. However, Hup-A causes some side effects. To search for new analogs or derivatives of Hup-A, we produced five Lycophyte alkaloids and two analogues by chemical synthesis: Lyconadins A-E, H-R-NOB, and 2JY-OBZ4. Objective: To systematically evaluated the therapeutic effects of the seven compounds on AD cell models. Methods: We assessed the effects of the seven compounds on cell viability via CCK-8 kit and used HEK293-hTau cells and N2a-hAPP cells as AD cell models to evaluate their potential therapeutic effects. We examined their effects on cholinesterase activity by employing the mice primary neuron. Results: All compounds did not affect cell viability; in addition, Lyconadin A and 2JY-OBZ4 particularly increased cell viability. Lyconadin D and Lyconadin E restored tau phosphorylation at Thr231, and H-R-NOB and 2JY-OBZ4 restored tau phosphorylation at Thr231 and Ser396 in GSK-3β-transfected HEK293-hTau cells. 2JY-OBZ4 decreased the level of PP2Ac-pY307 and increased the level of PP2Ac-mL309, supporting that 2JY-OBZ4 may activate PP2A. Lyconadin B, Lyconadin D, Lyconadin E, H-R-NOB, and 2JY-OBZ4 increased sAβPPα level in N2a-hAPP cells. 2JY-OBZ4 decreased the levels of BACE1 and sAβPPβ, thereby reduced Aβ production. Seven compounds exhibited weaker AChE activity inhibition efficiency than Hup-A. Among them, 2JY-OBZ4 showed the strongest AChE inhibition activity with an inhibition rate of 17% at 10μM. Conclusion: Among the seven lycophyte compounds, 2JY-OBZ4 showed the most expected effects on promoting cell viability, downregulating tau hyperphosphorylation, and Aβ production and inhibiting AChE in AD.
Collapse
Affiliation(s)
- Qian Guo
- School of Basic Medicine, Key Laboratory of Education Ministry/Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Scienceand Technology, Wuhan, China
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong, JS, China
| | - Qinfeng Cai
- School of Mental Health and Psychological Science, Anhui Medical University, Hefei, Anhui, China
| | - Fang Huang
- School of Basic Medicine, Key Laboratory of Education Ministry/Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Scienceand Technology, Wuhan, China
| | - Zhen Wei
- School of Basic Medicine, Key Laboratory of Education Ministry/Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Scienceand Technology, Wuhan, China
| | - Jian-Zhi Wang
- School of Basic Medicine, Key Laboratory of Education Ministry/Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Scienceand Technology, Wuhan, China
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong, JS, China
| | - Bin Zhang
- School of Basic Medicine, Key Laboratory of Education Ministry/Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Scienceand Technology, Wuhan, China
| | - Rong Liu
- School of Basic Medicine, Key Laboratory of Education Ministry/Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Scienceand Technology, Wuhan, China
| | - Yang Yang
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiaochuan Wang
- School of Basic Medicine, Key Laboratory of Education Ministry/Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Scienceand Technology, Wuhan, China
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong, JS, China
- Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, China
| | - Hong-Lian Li
- School of Basic Medicine, Key Laboratory of Education Ministry/Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Scienceand Technology, Wuhan, China
| |
Collapse
|
35
|
Avetyan DL, Shatskiy A, Kärkäs MD, Stepanova EV. Scalable total synthesis of natural vanillin-derived glucoside ω-esters. Carbohydr Res 2022; 522:108683. [PMID: 36179617 DOI: 10.1016/j.carres.2022.108683] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 09/08/2022] [Accepted: 09/20/2022] [Indexed: 11/02/2022]
Abstract
The first total synthesis of vanilloloside, calleryanin, and a series of naturally occurring ω-esters of vanilloloside was realized through direct glycosylation of vanillin-based aglycones or late-stage derivatization of vanilloloside. All aglycones and their fragments were synthesized from vanillin as the sole aromatic precursor. Subsequently, these intermediates were used to construct various vanillin-derived glucoside ω-esters using a mild acidic deacetylation as the key synthetic step, providing the final products in the total yields of 10-50% and general purity of >95%. Additionally, the first operationally simple and sustainable synthesis of litseafoloside B was realized on large scale, avoiding the use of toxic solvents and reagents, providing an attractive alternative to isolation of this and other similar compounds from plant sources.
Collapse
Affiliation(s)
- David L Avetyan
- Tomsk Polytechnic University, Lenin Avenue 30, 634050, Tomsk, Russia
| | - Andrey Shatskiy
- KTH Royal Institute of Technology, SE-100 44, Stockholm, Sweden
| | - Markus D Kärkäs
- KTH Royal Institute of Technology, SE-100 44, Stockholm, Sweden
| | - Elena V Stepanova
- Tomsk Polytechnic University, Lenin Avenue 30, 634050, Tomsk, Russia.
| |
Collapse
|
36
|
Serotonin 5-HT 6 Receptor Ligands and Butyrylcholinesterase Inhibitors Displaying Antioxidant Activity-Design, Synthesis and Biological Evaluation of Multifunctional Agents against Alzheimer's Disease. Int J Mol Sci 2022; 23:ijms23169443. [PMID: 36012707 PMCID: PMC9409043 DOI: 10.3390/ijms23169443] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 08/18/2022] [Accepted: 08/20/2022] [Indexed: 11/17/2022] Open
Abstract
Neurodegeneration leading to Alzheimer’s disease results from a complex interplay of a variety of processes including misfolding and aggregation of amyloid beta and tau proteins, neuroinflammation or oxidative stress. Therefore, to address more than one of these, drug discovery programmes focus on the development of multifunctional ligands, preferably with disease-modifying and symptoms-reducing potential. Following this idea, herein we present the design and synthesis of multifunctional ligands and biological evaluation of their 5-HT6 receptor affinity (radioligand binding assay), cholinesterase inhibitory activity (spectroscopic Ellman’s assay), antioxidant activity (ABTS assay) and metal-chelating properties, as well as a preliminary ADMET properties evaluation. Based on the results we selected compound 14 as a well-balanced and potent 5-HT6 receptor ligand (Ki = 22 nM) and human BuChE inhibitor (IC50 = 16 nM) with antioxidant potential expressed as a reduction of ABTS radicals by 35% (150 μM). The study also revealed additional metal-chelating properties of compounds 15 and 18. The presented compounds modulating Alzheimer’s disease-related processes might be further developed as multifunctional ligands against the disease.
Collapse
|
37
|
Wu Y, Su X, Lu J, Wu M, Yang SY, Mai Y, Deng W, Xue Y. In Vitro and in Silico Analysis of Phytochemicals From Fallopia dentatoalata as Dual Functional Cholinesterase Inhibitors for the Treatment of Alzheimer’s Disease. Front Pharmacol 2022; 13:905708. [PMID: 35899116 PMCID: PMC9313597 DOI: 10.3389/fphar.2022.905708] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Accepted: 06/20/2022] [Indexed: 11/18/2022] Open
Abstract
Current studies have found that butyrylcholinesterase (BuChE) replaces the biological function of acetylcholinesterase (AChE) in the late stage of Alzheimer’s disease. Species in the genus of Fallopia, rich in polyphenols with diverse chemical structures and significant biological activities, are considered as an important resource for screening natural products to against AD. In this study, thirty-four compounds (1–34) were isolated from Fallopia dentatoalata (Fr. Schm.) Holub, and their inhibitory effects against AChE and BuChE were assessed. Compounds of the phenylpropanoid sucrose ester class emerged as the most promising members of the group, with 31–33 displaying moderate AChE inhibition (IC50 values ranging from 30.6 ± 4.7 to 56.0 ± 2.4 µM) and 30–34 showing potential inhibitory effects against BuChE (IC50 values ranging from 2.7 ± 1.7 to 17.1 ± 3.4 µM). Tacrine was used as a positive control (IC50: 126.7 ± 1.1 in AChE and 5.5 ± 1.7 nM in BuChE). Kinetic analysis highlighted compounds 31 and 32 as non-competitive inhibitors of AChE with Ki values of ∼30.0 and ∼34.4 µM, whilst 30–34 were revealed to competitively inhibit BuChE with Ki values ranging from ∼1.8 to ∼17.5 µM. Molecular binding studies demonstrated that 30–34 bound to the catalytic sites of BuChE with negative binding energies. The strong agreement between both in vitro and in silico studies highlights the phenylpropanoid sucrose esters 30–34 as promising candidates for use in future anti-cholinesterase therapeutics against Alzheimer’s disease.
Collapse
Affiliation(s)
- Yichuang Wu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, China
| | - Xiangdong Su
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, China
| | - Jielang Lu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, China
| | - Meifang Wu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, China
| | - Seo Young Yang
- Department of Pharmaceutical Engineering, Sangji University, Wonju, South Korea
| | - Yang Mai
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, China
| | - Wenbin Deng
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, China
- *Correspondence: Wenbin Deng, ; Yongbo Xue,
| | - Yongbo Xue
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, China
- *Correspondence: Wenbin Deng, ; Yongbo Xue,
| |
Collapse
|
38
|
The Aptamer Ob2, a novel AChE inhibitor, restores cognitive deficits and alleviates amyloidogenesis in 5×FAD transgenic mice. MOLECULAR THERAPY. NUCLEIC ACIDS 2022; 28:114-123. [PMID: 35402070 PMCID: PMC8938253 DOI: 10.1016/j.omtn.2022.02.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 02/25/2022] [Indexed: 02/07/2023]
Abstract
Loss of cerebral cholinergic neurons and decreased levels of acetylcholine (ACh) are considered to be major factors causing cognitive dysfunction in Alzheimer's disease (AD). Abnormally elevated levels of acetylcholinesterase (AChE) resulting in decreased levels of ACh are common in AD patients; thus, AChE inhibitors (AChEIs) are widely used for the treatment of AD. In our previous work, we acquired DNA aptamers Ob1, Ob2, and Ob3 against human brain AChE from systematic evolution of ligands by exponential enrichment (SELEX). In this study, we investigated the effect of these aptamers on learning and memory abilities, as well as the underlying mechanism in a 5×FAD transgenic AD mouse model. Here, we showed that only aptamer Ob2 exhibits a good inhibitory effect on both mouse and human AChE activity. In addition, chronic treatment with aptamer Ob2 significantly improved cognitive ability of 5×FAD mice in the Morris water maze. Moreover, the mechanism of aptamer Ob2 in 5×FAD mice may be associated with its inhibition of AChE activity, alleviation of the levels of Aβ by lowering the expression of β-secretase (BACE1), and activation of astrocytes in the brains of 5×FAD mice. These results indicate that aptamer Ob2 exhibits potential as an effective AChEI for the treatment of AD.
Collapse
|
39
|
Liu J, Liu L, Zheng L, Feng KW, Wang HT, Xu JP, Zhou ZZ. Discovery of novel 2,3-dihydro-1H-inden-1-ones as dual PDE4/AChE inhibitors with more potency against neuroinflammation for the treatment of Alzheimer's disease. Eur J Med Chem 2022; 238:114503. [DOI: 10.1016/j.ejmech.2022.114503] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 05/24/2022] [Accepted: 05/27/2022] [Indexed: 12/14/2022]
|
40
|
Pervaiz A, Jan MS, Hassan Shah SM, Khan A, Zafar R, Ansari B, Shahid M, Hussain F, Ijaz Khan M, Zeb A, Mukarram Shah SM. Comparative in-vitro anti-inflammatory, anticholinesterase and antidiabetic evaluation: computational and kinetic assessment of succinimides cyano-acetate derivatives. J Biomol Struct Dyn 2022:1-14. [PMID: 35507043 DOI: 10.1080/07391102.2022.2069862] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 04/19/2022] [Indexed: 12/12/2022]
Abstract
This research was planned to synthesize cyano-acetate derivatives of succinimide and evaluate its comparative biological efficacy as anti-inflammatory, anti-cholinesterase and anti-diabetic, which was further validated by molecular docking studies. The three cyano-acetate derivatives of succinimide including compound 23 Methyl 2-cyano-2-(2,5-dioxopyrrolidin-3-yl)acetate, compound 31 Methyl 2-cyano-2-(1-methyl-2,5-dioxopyrrolidin-3-yl)acetate and compound 44 Methyl 2-cyano-2-(1-ethyl-2,5-dioxopyrrolidin-3-yl) acetate were synthesized. The mentioned compounds were checked for in vitro anti-inflammatory, anti-cholinesterase and anti-diabetic (α-amylase inhibition) activity. To validate the in vitro results, computational studies were carried out using molecular operating environment to analyse the BE, i.e. binding energies of all synthesized compounds against the respective enzymes. The Compounds 23, 31, 44 exhibited anti-inflammatory via inhibiting COX-2 (IC50 value of 204.08, 68.60 and 50.93 µM, respectively), COX-1 (IC50 value of 287, 185, and 143 µM, respectively) and 5-LOX (IC50 value of 138, 50.76 and 20, 87 µM respectively). They exhibited choline-mimetic potential, such as compound 23, 31 and 44 inhibited AChE enzyme (IC50 value of 240, 174, and 134 µM, respectively) and BChE enzyme (IC50 value of 203, 134 and 97 µM, respectively). The Compounds 23, 31, 44 exhibited anti-diabetic effect via inhibiting α-amylase enzyme (IC50 values of 250, 106 and 60 µM, respectively). Molecular docking studies revealed that the synthesized compounds have good binding affinity in the binding pockets of AChE, BChE, COX-2, 5-LOX and α-amylase enzyme and showed high binding energies. The synthesized succinimide derivatives, i.e. compound 23, 31, 44 showed marked inhibitory activities against cyclooxygenase, lipoxygenase, α-amylase and cholinesterase enzymes. Among these three, compound 44 and 31 showed strong anti-inflammatory and anti-diabetic activity while they displayed moderate anti-cholinesterase activity supported by molecular docking results.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Aini Pervaiz
- Department of Pharmacy, University of Swabi, Swabi, KP, Pakistan
| | | | | | - Ali Khan
- Department of Pharmacy, University of Swabi, Swabi, KP, Pakistan
| | - Rehman Zafar
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Riphah International University, Islamabad, Pakistan
| | - Bushra Ansari
- Department of Pharmacy, Abdul Wali Khan University, Mardan, KP, Pakistan
| | - Muhammad Shahid
- Department of Pharmacy, Institute of Integrative Biosciences, CECOS University of IT and Emerging Sciences, Peshawar, KP, Pakistan
| | - Fida Hussain
- Department of Pharmacy, University of Swabi, Swabi, KP, Pakistan
| | | | - Anwar Zeb
- Department of Pharmacy, University of Swabi, Swabi, KP, Pakistan
| | | |
Collapse
|
41
|
O'Leary TP, Brown RE. Visuo-spatial learning and memory impairments in the 5xFAD mouse model of Alzheimer's disease: Effects of age, sex, albinism, and motor impairments. GENES, BRAIN, AND BEHAVIOR 2022; 21:e12794. [PMID: 35238473 PMCID: PMC9744519 DOI: 10.1111/gbb.12794] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 12/19/2021] [Indexed: 12/17/2022]
Abstract
The 5xFAD mouse model of Alzheimer's disease (AD) rapidly develops AD-related neuro-behavioral pathology. Learning and memory impairments in 5xFAD mice, however, are not always replicated and the size of impairments varies considerably across studies. To examine possible sources of this variability, we analyzed the effects of age, sex, albinism due to background genes (Tyrc , Oca2p ) and motor impairment on learning and memory performance of wild type and 5xFAD mice on the Morris water maze, from 3 to 15 months of age. The 5xFAD mice showed impaired learning at 6-9 months of age, but memory impairments were not detected with the test procedure used in this study. Performance of 5xFAD mice was profoundly impaired at 12-15 months of age, but was accompanied by slower swim speeds than wild-type mice and a frequent failure to locate the escape platform. Overall female mice performed worse than males, and reversal learning impairments in 5xFAD mice were more pronounced in females than males. Albino mice performed worse than pigmented mice, confirming that albinism can impair performance of 5xFAD mice independently of AD-related transgenes. Overall, these results show that 5xFAD mice have impaired learning performance at 6-9 months of age, but learning and memory performance at 12-15 months is confounded with motor impairments. Furthermore, sex and albinism should be controlled to provide an accurate assessment of AD-related transgenes on learning and memory. These results will help reduce variability across pre-clinical experiments with 5xFAD mice, and thus enhance the reliability of studies developing new therapeutics for AD.
Collapse
Affiliation(s)
- Timothy P. O'Leary
- Department of Psychology and NeuroscienceDalhousie UniversityHalifaxNova ScotiaCanada
| | - Richard E. Brown
- Department of Psychology and NeuroscienceDalhousie UniversityHalifaxNova ScotiaCanada
| |
Collapse
|
42
|
Role of Cholinergic Signaling in Alzheimer's Disease. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27061816. [PMID: 35335180 PMCID: PMC8949236 DOI: 10.3390/molecules27061816] [Citation(s) in RCA: 189] [Impact Index Per Article: 94.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 02/14/2022] [Accepted: 02/22/2022] [Indexed: 12/27/2022]
Abstract
Acetylcholine, a neurotransmitter secreted by cholinergic neurons, is involved in signal transduction related to memory and learning ability. Alzheimer’s disease (AD), a progressive and commonly diagnosed neurodegenerative disease, is characterized by memory and cognitive decline and behavioral disorders. The pathogenesis of AD is complex and remains unclear, being affected by various factors. The cholinergic hypothesis is the earliest theory about the pathogenesis of AD. Cholinergic atrophy and cognitive decline are accelerated in age-related neurodegenerative diseases such as AD. In addition, abnormal central cholinergic changes can also induce abnormal phosphorylation of ttau protein, nerve cell inflammation, cell apoptosis, and other pathological phenomena, but the exact mechanism of action is still unclear. Due to the complex and unclear pathogenesis, effective methods to prevent and treat AD are unavailable, and research to explore novel therapeutic drugs is various and active in the world. This review summaries the role of cholinergic signaling and the correlation between the cholinergic signaling pathway with other risk factors in AD and provides the latest research about the efficient therapeutic drugs and treatment of AD.
Collapse
|
43
|
Liu Y, Uras G, Onuwaje I, Li W, Yao H, Xu S, Li X, Li X, Phillips J, Allen S, Gong Q, Zhang H, Zhu Z, Liu J, Xu J. Novel inhibitors of AChE and Aβ aggregation with neuroprotective properties as lead compounds for the treatment of Alzheimer's disease. Eur J Med Chem 2022; 235:114305. [DOI: 10.1016/j.ejmech.2022.114305] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 02/26/2022] [Accepted: 03/16/2022] [Indexed: 01/27/2023]
|
44
|
Rullo M, Cipolloni M, Catto M, Colliva C, Miniero DV, Latronico T, de Candia M, Benicchi T, Linusson A, Giacchè N, Altomare CD, Pisani L. Probing Fluorinated Motifs onto Dual AChE-MAO B Inhibitors: Rational Design, Synthesis, Biological Evaluation, and Early-ADME Studies. J Med Chem 2022; 65:3962-3977. [PMID: 35195417 DOI: 10.1021/acs.jmedchem.1c01784] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Bioisosteric H/F or CH2OH/CF2H replacement was introduced in coumarin derivatives previously characterized as dual AChE-MAO B inhibitors to probe the effects on both inhibitory potency and drug-likeness. Along with in vitro screening, we investigated early-ADME parameters related to solubility and lipophilicity (Sol7.4, CHI7.4, log D7.4), oral bioavailability and central nervous system (CNS) penetration (PAMPA-HDM and PAMPA-blood-brain barrier (BBB) assays, Caco-2 bidirectional transport study), and metabolic liability (half-lives and clearance in microsomes, inhibition of CYP3A4). Both specific and nonspecific tissue toxicities were determined in SH-SY5Y and HepG2 lines, respectively. Compound 15 bearing a -CF2H motif emerged as a water-soluble, orally bioavailable CNS-permeant potent inhibitor of both human AChE (IC50 = 550 nM) and MAO B (IC50 = 8.2 nM, B/A selectivity > 1200). Moreover, 15 behaved as a safe and metabolically stable neuroprotective agent, devoid of cytochrome liability.
Collapse
Affiliation(s)
- Mariagrazia Rullo
- Department of Pharmacy─Pharmaceutical Sciences, University of Bari "Aldo Moro", via Orabona 4, 70125 Bari, Italy
| | | | - Marco Catto
- Department of Pharmacy─Pharmaceutical Sciences, University of Bari "Aldo Moro", via Orabona 4, 70125 Bari, Italy
| | | | - Daniela Valeria Miniero
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari "Aldo Moro", via Orabona, 4, 70125 Bari, Italy
| | - Tiziana Latronico
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari "Aldo Moro", via Orabona, 4, 70125 Bari, Italy
| | - Modesto de Candia
- Department of Pharmacy─Pharmaceutical Sciences, University of Bari "Aldo Moro", via Orabona 4, 70125 Bari, Italy
| | | | - Anna Linusson
- Department of Chemistry, Umeå University, 90187 Umeå, Sweden
| | - Nicola Giacchè
- TES Pharma s.r.l., Corso Vannucci 47, 06121 Perugia, Italy
| | - Cosimo Damiano Altomare
- Department of Pharmacy─Pharmaceutical Sciences, University of Bari "Aldo Moro", via Orabona 4, 70125 Bari, Italy
| | - Leonardo Pisani
- Department of Pharmacy─Pharmaceutical Sciences, University of Bari "Aldo Moro", via Orabona 4, 70125 Bari, Italy
| |
Collapse
|
45
|
Haghighijoo Z, Zamani L, Moosavi F, Emami S. Therapeutic potential of quinazoline derivatives for Alzheimer's disease: A comprehensive review. Eur J Med Chem 2022; 227:113949. [PMID: 34742016 DOI: 10.1016/j.ejmech.2021.113949] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 10/02/2021] [Accepted: 10/22/2021] [Indexed: 12/24/2022]
Abstract
Quinazolines are considered as a promising class of bioactive heterocyclic compounds with broad properties. Particularly, the quinazoline scaffold has an impressive role in the design and synthesis of new CNS-active drugs. The drug-like properties and pharmacological characteristics of quinazoline could lead to different drugs with various targets. Among CNS disorders, Alzheimer's disease (AD) is a progressive neurodegenerative disorder with memory loss, cognitive decline and language dysfunction. AD is a complex and multifactorial disease therefore, the need for finding multi-target drugs against this devastative disease is urgent. A literature survey revealed that quinazoline derivatives have diverse therapeutic potential for AD as modulators/inhibitors of β-amyloid, tau protein, cholinesterases, monoamine oxidases, and phosphodiesterases as well as other protective effects. Thus, we describe here the most relevant and recent studies about anti-AD agents with quinazoline structure which can further aid the development and discovery of new anti-AD agents.
Collapse
Affiliation(s)
- Zahra Haghighijoo
- Department of Chemistry, University of Louisiana at Lafayette, Lafayette, LA, 70504, USA
| | - Leila Zamani
- Worcester Polytechnic Institute, 100 Institute Road, Worcester, MA, 01609, USA
| | - Fatemeh Moosavi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Saeed Emami
- Department of Medicinal Chemistry and Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
46
|
Babu A, Joy MN, Sunil K, Sajith AM, Santra S, Zyryanov GV, Konovalova OA, Butorin II, Muniraju K. Towards novel tacrine analogues: Pd(dppf)Cl 2·CH 2Cl 2 catalyzed improved synthesis, in silico docking and hepatotoxicity studies. RSC Adv 2022; 12:22476-22491. [PMID: 36105950 PMCID: PMC9366599 DOI: 10.1039/d2ra03225b] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 08/03/2022] [Indexed: 01/23/2023] Open
Abstract
A plethora of 6-(hetero)aryl C–C and C–N bonded tacrine analogues has been made accessible by employing palladium mediated (Suzuki–Miyaura, Heck, Sonogashira, Stille and Buchwald) cross-coupling reactions, starting from either halogenated or borylated residues. The successful use of Pd(dppf)Cl2·CH2Cl2 as a common catalytic system in realizing all these otherwise challenging transformations is the highlight of our optimized protocols. The analogues thus synthesized allow the available chemical space around the C-6 of this biologically relevant tacrine core to be explored. The in silico docking studies of the synthesized compounds were carried out against the acetylcholinesterase (AChE) enzyme. The hepatotoxicity studies of these compounds were done against complexes of CYP1A2 and CYP3A4 proteins with known inhibitors like 7,8-benzoflavone and ketoconazole, respectively. 24 synthesized compounds by various cross-coupling reactions on 6-bromo tacrine. Molecular docking and toxicity prediction studies were also performed.![]()
Collapse
Affiliation(s)
- Aravinda Babu
- Department of Chemistry, SSIT, Sri Siddhartha Academy of Higher Education, Tumkur, Karnataka, India-572107
| | - Muthipeedika Nibin Joy
- Institute of Chemical Technology, Ural Federal University, 19 Mira Street, Yekaterinburg, Russia-620002
| | - K. Sunil
- Department of Chemistry, SSIT, Sri Siddhartha Academy of Higher Education, Tumkur, Karnataka, India-572107
| | | | - Sougata Santra
- Institute of Chemical Technology, Ural Federal University, 19 Mira Street, Yekaterinburg, Russia-620002
| | - Grigory V. Zyryanov
- Institute of Chemical Technology, Ural Federal University, 19 Mira Street, Yekaterinburg, Russia-620002
- I. Ya. Postovskiy Institute of Organic Synthesis, Ural Division of the Russian Academy of Sciences, 22 S. Kovalevskoy Street, Yekaterinburg, Russia-620219
| | - Olga A. Konovalova
- Institute of Chemical Technology, Ural Federal University, 19 Mira Street, Yekaterinburg, Russia-620002
| | - Ilya I. Butorin
- Institute of Chemical Technology, Ural Federal University, 19 Mira Street, Yekaterinburg, Russia-620002
| | - Keesaram Muniraju
- Government Degree College-Puttur (Affiliated to S. V. University, Tirupati), Narayanavanam Road, Puttur, Chittoor (Dt), Andhra Pradesh, India-517583
| |
Collapse
|
47
|
Ling W, Qin C, Tian C, Feng M, Wang C. Thermogravimetric infrared analysis for the pyrolysis mechanism and shelf life of lycoramine hydrobromide. INT J CHEM KINET 2021. [DOI: 10.1002/kin.21547] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Weihong Ling
- Key Laboratory of Hunan Forest Products and Chemical Industry Engineering Jishou University Zhangjiajie 427000 China
| | - Chong Qin
- School of Chemistry University of Chemistry Melbourne Parkville Victoria 3010 Australia
| | - Chunlian Tian
- Key Laboratory of Hunan Forest Products and Chemical Industry Engineering Jishou University Zhangjiajie 427000 China
| | - Miao Feng
- Key Laboratory of Hunan Forest Products and Chemical Industry Engineering Jishou University Zhangjiajie 427000 China
| | - Chaochun Wang
- Key Laboratory of Hunan Forest Products and Chemical Industry Engineering Jishou University Zhangjiajie 427000 China
| |
Collapse
|
48
|
Wichur T, Godyń J, Góral I, Latacz G, Bucki A, Siwek A, Głuch-Lutwin M, Mordyl B, Śniecikowska J, Walczak M, Knez D, Jukič M, Sałat K, Gobec S, Kołaczkowski M, Malawska B, Brazzolotto X, Więckowska A. Development and crystallography-aided SAR studies of multifunctional BuChE inhibitors and 5-HT 6R antagonists with β-amyloid anti-aggregation properties. Eur J Med Chem 2021; 225:113792. [PMID: 34530376 DOI: 10.1016/j.ejmech.2021.113792] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 07/26/2021] [Accepted: 08/17/2021] [Indexed: 12/16/2022]
Abstract
The lack of an effective treatment makes Alzheimer's disease a serious healthcare problem and a challenge for medicinal chemists. Herein we report interdisciplinary research on novel multifunctional ligands targeting proteins and processes involved in the development of the disease: BuChE, 5-HT6 receptors and β-amyloid aggregation. Structure-activity relationship analyses supported by crystallography and docking studies led to the identification of a fused-type multifunctional ligand 50, with remarkable and balanced potencies against BuChE (IC50 = 90 nM) and 5-HT6R (Ki = 4.8 nM), and inhibitory activity against Aβ aggregation (53% at 10 μM). In in vitro ADME-Tox and in vivo pharmacokinetic studies compound 50 showed good stability in the mouse liver microsomes, favourable safety profile and brain permeability with the brain to plasma ratio of 6.79 after p.o. administration in mice, thus being a promising candidate for in vivo pharmacology studies and a solid foundation for further research on effective anti-AD therapies.
Collapse
Affiliation(s)
- Tomasz Wichur
- Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland
| | - Justyna Godyń
- Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland
| | - Izabella Góral
- Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland
| | - Gniewomir Latacz
- Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland
| | - Adam Bucki
- Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland
| | - Agata Siwek
- Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland
| | - Monika Głuch-Lutwin
- Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland
| | - Barbara Mordyl
- Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland
| | - Joanna Śniecikowska
- Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland
| | - Maria Walczak
- Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland
| | - Damijan Knez
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva 7, 1000, Ljubljana, Slovenia
| | - Marko Jukič
- University of Maribor, Faculty of Chemistry and Chemical Engineering, Laboratory of Physical Chemistry and Chemical Thermodynamics, Smetanova ulica 17, SI-2000 Maribor, Slovenia
| | - Kinga Sałat
- Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland
| | - Stanislav Gobec
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva 7, 1000, Ljubljana, Slovenia
| | - Marcin Kołaczkowski
- Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland
| | - Barbara Malawska
- Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland
| | - Xavier Brazzolotto
- Département de Toxicologie et Risques Chimiques, Institut de Recherche Biomédicale des Armées, 91223, Brétigny sur Orge, France
| | - Anna Więckowska
- Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland.
| |
Collapse
|
49
|
Brandão P, López Ó, Leitzbach L, Stark H, Fernández-Bolaños JG, Burke AJ, Pineiro M. Ugi Reaction Synthesis of Oxindole-Lactam Hybrids as Selective Butyrylcholinesterase Inhibitors. ACS Med Chem Lett 2021; 12:1718-1725. [PMID: 34795859 PMCID: PMC8591717 DOI: 10.1021/acsmedchemlett.1c00344] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 07/20/2021] [Indexed: 12/17/2022] Open
Abstract
Molecular hybridization is a valuable approach in drug discovery. Combining it with multicomponent reactions is highly desirable, since structurally diverse libraries can be attained efficiently in an eco-friendly manner. In this work, isatin is used as the key building block for the Ugi 4-center 3-component reaction synthesis of oxindole-lactam hybrids, under catalyst-free conditions. The resulting oxindole-β-lactam and oxindole-γ-lactam hybrids were evaluated for their potential to inhibit relevant central nervous system targets, namely cholinesterases and monoamine oxidases. Druglikeness evaluation was also performed, and compounds 4eca and 5dab exhibited great potential as selective butyrylcholinesterase inhibitors, at the low micromolar range, with an interesting predictive pharmacokinetic profile. Our findings herein reported suggest oxindole-lactam hybrids as new potential agents for the treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Pedro Brandão
- University
of Coimbra, CQC and Department
of Chemistry, 3004-535 Coimbra, Portugal
- LAQV-REQUIMTE,
Rua Romão Ramalho, 59, University
of Évora, 7000 Évora, Portugal
| | - Óscar López
- Departamento
de Química Orgánica, Facultad de Química, Universidad de Sevilla, Apartado 1203, E-41071 Seville, Spain
| | - Luisa Leitzbach
- Institute
of Pharmaceutical and Medicinal Chemistry, Heinrich Heine University Düsseldorf, Universitaetsstrasse 1, 40225 Duesseldorf, NRW, Germany
| | - Holger Stark
- Institute
of Pharmaceutical and Medicinal Chemistry, Heinrich Heine University Düsseldorf, Universitaetsstrasse 1, 40225 Duesseldorf, NRW, Germany
| | - José G. Fernández-Bolaños
- Departamento
de Química Orgánica, Facultad de Química, Universidad de Sevilla, Apartado 1203, E-41071 Seville, Spain
| | - Anthony J. Burke
- LAQV-REQUIMTE,
Rua Romão Ramalho, 59, University
of Évora, 7000 Évora, Portugal
- University
of Evora, Department of Chemistry, Rua Romão Ramalho, 59, 7000 Évora, Portugal
| | - Marta Pineiro
- University
of Coimbra, CQC and Department
of Chemistry, 3004-535 Coimbra, Portugal
| |
Collapse
|
50
|
Bortolami M, Pandolfi F, Tudino V, Messore A, Madia VN, De Vita D, Di Santo R, Costi R, Romeo I, Alcaro S, Colone M, Stringaro A, Espargaró A, Sabatè R, Scipione L. New Pyrimidine and Pyridine Derivatives as Multitarget Cholinesterase Inhibitors: Design, Synthesis, and In Vitro and In Cellulo Evaluation. ACS Chem Neurosci 2021; 12:4090-4112. [PMID: 34652128 PMCID: PMC8569681 DOI: 10.1021/acschemneuro.1c00485] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
![]()
A new series of pyrimidine
and pyridine diamines was designed as
dual binding site inhibitors of cholinesterases (ChEs), characterized
by two small aromatic moieties separated by a diaminoalkyl flexible
linker. Many compounds are mixed or uncompetitive acetylcholinesterase
(AChE) and/or butyrylcholinesterase (BChE) nanomolar inhibitors, with
compound 9 being the most active on Electrophorus
electricus AChE (EeAChE) (Ki = 0.312 μM) and compound 22 on equine BChE (eqBChE) (Ki = 0.099 μM). Molecular docking and molecular dynamic
studies confirmed the interaction mode of our compounds with the enzymatic
active site. UV–vis spectroscopic studies showed that these
compounds can form complexes with Cu2+ and Fe3+ and that compounds 18, 20, and 30 have antioxidant properties. Interestingly, some compounds were
also able to reduce Aβ42 and tau aggregation, with
compound 28 being the most potent (22.3 and 17.0% inhibition
at 100 μM on Aβ42 and tau, respectively). Moreover,
the most active compounds showed low cytotoxicity on a human brain
cell line and they were predicted as BBB-permeable.
Collapse
Affiliation(s)
- Martina Bortolami
- Department of Scienze di Base e Applicate per l’Ingegneria, Sapienza University of Rome, via Castro Laurenziano 7, I-00161 Rome, Italy
| | - Fabiana Pandolfi
- Department of Scienze di Base e Applicate per l’Ingegneria, Sapienza University of Rome, via Castro Laurenziano 7, I-00161 Rome, Italy
| | - Valeria Tudino
- Department of Chimica e Tecnologia del Farmaco, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Antonella Messore
- Department of Chimica e Tecnologia del Farmaco, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Valentina Noemi Madia
- Department of Chimica e Tecnologia del Farmaco, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Daniela De Vita
- Department of Environmental Biology, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Roberto Di Santo
- Istituto Pasteur, Fondazione Cenci Bolognetti, Department of Chemistry and Technology of Drug, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Roberta Costi
- Istituto Pasteur, Fondazione Cenci Bolognetti, Department of Chemistry and Technology of Drug, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Isabella Romeo
- Net4Science s.r.l., Campus universitario ″S. Venuta″, Viale Europa, 88100 Catanzaro, Italy
- Dipartimento di Scienze della Salute, Università ″Magna Græcia″ di Catanzaro, Viale Europa, 88100 Catanzaro, Italy
| | - Stefano Alcaro
- Net4Science s.r.l., Campus universitario ″S. Venuta″, Viale Europa, 88100 Catanzaro, Italy
- Dipartimento di Scienze della Salute, Università ″Magna Græcia″ di Catanzaro, Viale Europa, 88100 Catanzaro, Italy
| | - Marisa Colone
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Viale Regina Elena, 00161 Rome, Italy
| | - Annarita Stringaro
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Viale Regina Elena, 00161 Rome, Italy
| | - Alba Espargaró
- Department of Pharmacy and Pharmaceutical Technology and Physical-Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Avda. Joan XXIII, 27-31 Barcelona, Catalonia, Spain
| | - Raimon Sabatè
- Department of Pharmacy and Pharmaceutical Technology and Physical-Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Avda. Joan XXIII, 27-31 Barcelona, Catalonia, Spain
| | - Luigi Scipione
- Department of Chimica e Tecnologia del Farmaco, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| |
Collapse
|