1
|
Zhang YL, Yang HY, Gou J, Qi XM, Qiao YB, Li QS. Carvacrol/thymol derivatives as highly selective BuChE inhibitors with anti-inflammatory activities: Discovery and bio-evaluation. Bioorg Chem 2025; 160:108430. [PMID: 40209354 DOI: 10.1016/j.bioorg.2025.108430] [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: 01/08/2025] [Revised: 03/20/2025] [Accepted: 03/31/2025] [Indexed: 04/12/2025]
Abstract
In this study, nine novel carvacrol/thymol derivatives incorporating carbamate groups were designed, synthesized, and evaluated as multifunctional anti-AD agents. These derivatives displayed superior BuChE inhibitory and anti-inflammatory characteristics compared to the parent compounds. While the derivatives exhibited AChE IC50 values exceeding the detectable limit (>100 μM), they demonstrated high potency as BuChE inhibitors, with IC50 values ranging from 0.05 to 9.62 μM. In an inflammation model of BV2 microglial cells induced by lipopolysaccharide (LPS), the derivatives effectively reduced the levels of the pro-inflammatory cytokine interleukin-1β (IL1β), with inhibition rates of IL1β exceeding 50 % at 10 μM. Notably, compound SXF3 attained the highest BuChE inhibition efficacy (eqBuChE IC50 = 0.05 ± 0.003 μM, hBuChE IC50 = 0.04 ± 0.001 μM), the highest selectivity for BuChE (with a selectivity index, SI, exceeding 2000, calculated as the ratio of eeAChE IC50 to eqBuChE IC50) and high anti-inflammatory activity (inhibition of IL1β, IC50 = 8.33 ± 0.08 μM). In a scopolamine-induced AD mouse model, SXF3 (15 mg/kg) significantly reduced the latency to the platform and attenuated memory deficits. Biochemical analysis confirmed that SXF3 significantly increased acetylcholine (ACh) levels in the mice hippocampus, primarily due to the inhibition of BuChE rather than AChE, and that SXF3 significantly reduced IL1β levels to normal, further confirming its anti-inflammatory activities. Hence, the selective BuChE inhibitory properties and anti-inflammatory attributes of SXF3 render it a promising candidate for further investigation in the treatment of AD.
Collapse
Affiliation(s)
- Yuan-Lin Zhang
- Shanxi Key Laboratory of Innovative Drug for the Treatment of Serious Diseases Basing on the Chronic Inflammation, Shanxi University of Chinese Medicine, Taiyuan, Shanxi 030619, China; College of Traditional Chinese Medicine and Food Engineering, Shanxi University of Chinese Medicine, Taiyuan, Shanxi 030619, China
| | - Hao-Yan Yang
- Shanxi Key Laboratory of Innovative Drug for the Treatment of Serious Diseases Basing on the Chronic Inflammation, Shanxi University of Chinese Medicine, Taiyuan, Shanxi 030619, China; College of Traditional Chinese Medicine and Food Engineering, Shanxi University of Chinese Medicine, Taiyuan, Shanxi 030619, China
| | - Jie Gou
- Shanxi Key Laboratory of Innovative Drug for the Treatment of Serious Diseases Basing on the Chronic Inflammation, Shanxi University of Chinese Medicine, Taiyuan, Shanxi 030619, China; College of Traditional Chinese Medicine and Food Engineering, Shanxi University of Chinese Medicine, Taiyuan, Shanxi 030619, China
| | - Xiao-Ming Qi
- Shanxi Key Laboratory of Innovative Drug for the Treatment of Serious Diseases Basing on the Chronic Inflammation, Shanxi University of Chinese Medicine, Taiyuan, Shanxi 030619, China; College of Traditional Chinese Medicine and Food Engineering, Shanxi University of Chinese Medicine, Taiyuan, Shanxi 030619, China
| | - Yuan-Biao Qiao
- Shanxi Key Laboratory of Innovative Drug for the Treatment of Serious Diseases Basing on the Chronic Inflammation, Shanxi University of Chinese Medicine, Taiyuan, Shanxi 030619, China
| | - Qing-Shan Li
- Shanxi Key Laboratory of Innovative Drug for the Treatment of Serious Diseases Basing on the Chronic Inflammation, Shanxi University of Chinese Medicine, Taiyuan, Shanxi 030619, China; College of Traditional Chinese Medicine and Food Engineering, Shanxi University of Chinese Medicine, Taiyuan, Shanxi 030619, China.
| |
Collapse
|
2
|
Olivier N, Harvey BH, Gobec S, Shahid M, Košak U, Žakelj S, Brink CB. A novel butyrylcholinesterase inhibitor induces antidepressant, pro-cognitive, and anti-anhedonic effects in Flinders Sensitive Line rats: The role of the ghrelin-dopamine cascade. Biomed Pharmacother 2025; 187:118093. [PMID: 40318448 DOI: 10.1016/j.biopha.2025.118093] [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: 03/03/2025] [Revised: 04/04/2025] [Accepted: 04/24/2025] [Indexed: 05/07/2025] Open
Abstract
BACKGROUND AND PURPOSE Major depressive disorder (MDD) is often treatment resistant, particularly in addressing anhedonia and cognitive deficits. Novel pharmacological strategies are needed. While butyrylcholinesterase, ghrelin, and dopamine (DA) have been well studied in the context of stress and MDD, their interaction remains unclear. EXPERIMENTAL APPROACH The dose-dependent antidepressant effects of a novel butyrylcholinesterase inhibitor (BChEI) were evaluated in the Flinders Sensitive Line (FSL) rat model of MDD. Behavioural assessments included the forced swim test (despair), sucrose preference test (reward-related), and novel object recognition test (cognition). Brain-derived neurotrophic factor (BDNF), acetylcholine (ACh), and brain monoamines were analysed, as well as serum growth hormone and acyl- and desacyl-ghrelin. To confirm the role of ghrelin, pharmacological exploration was undertaken using the ghrelin receptor antagonist, D-Lys-3-GHRP-6. KEY RESULTS FSL rats had significantly lower ghrelin ratios, BDNF, ACh, DA and growth hormone levels. In FSL rats, both BChEI and escitalopram significantly reduced despair. BChEI significantly outperformed escitalopram in enhancing reward-related and cognitive behaviours. Biochemically, BChEI treatment significantly increased ghrelin ratios and brain DA levels without altering brain 5-HT, ACh or BDNF. D-Lys-3-GHRP-6 significantly reversed the antidepressant-like, rewarding, and pro-cognitive effects of BChEI, accompanied by significant reductions in BDNF and DA. CONCLUSIONS AND IMPLICATIONS FSL rats display impaired ghrelin, DA, serotonin, growth hormone, and BDNF signalling, akin to MDD. BChEI exerts antidepressant-like effects across despair, reward, and cognitive domains, most likely via the BChE-ghrelin-DA cascade. Reversal of these effects by ghrelin antagonism underscores the critical role of ghrelin, specifically via growth hormone secretagogue receptor-ghrelin interaction. These findings suggest a potentially novel multimodal neurobiological target for the treatment of MDD.
Collapse
Affiliation(s)
- Nadia Olivier
- Centre of Excellence for Pharmaceutical Sciences, School of Pharmacy (Pharmacology), Faculty of Health Sciences, North-West University, Potchefstroom 2520, South Africa
| | - Brian H Harvey
- Centre of Excellence for Pharmaceutical Sciences, School of Pharmacy (Pharmacology), Faculty of Health Sciences, North-West University, Potchefstroom 2520, South Africa; South African Medical Research Council Unit on Risk and Resilience in Mental Disorders, Department of Psychiatry and Neuroscience Institute, University of Cape Town, Cape Town 7505, South Africa; The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong 3220, Australia.
| | - Stanislav Gobec
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, Ljubljana 1000, Slovenia
| | | | - Urban Košak
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, Ljubljana 1000, Slovenia
| | - Simon Žakelj
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, Ljubljana 1000, Slovenia
| | - Christiaan B Brink
- Centre of Excellence for Pharmaceutical Sciences, School of Pharmacy (Pharmacology), Faculty of Health Sciences, North-West University, Potchefstroom 2520, South Africa
| |
Collapse
|
3
|
Colleoni A, Galli G, Dallanoce C, De Amici M, Gorostiza P, Matera C. Light-Activated Pharmacological Tools for Exploring the Cholinergic System. Med Res Rev 2025. [PMID: 40123150 DOI: 10.1002/med.22108] [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: 01/02/2025] [Revised: 02/27/2025] [Accepted: 03/06/2025] [Indexed: 03/25/2025]
Abstract
Cholinergic transmission plays a critical role in both the central and peripheral nervous systems, affecting processes such as learning, memory, and inflammation. Conventional cholinergic drugs generally suffer from poor selectivity and temporal precision, leading to undesired effects and limited therapeutic efficacy. Photopharmacology aims to overcome the limitations of traditional drugs using photocleavable or photoswitchable ligands and spatiotemporal patterns of illumination. Spanning from muscarinic and nicotinic modulators to cholinesterase inhibitors, this review explores the development and application of light-activated compounds as tools for unraveling the role of cholinergic signaling in both physiological and pathological contexts, while also paving the way for innovative phototherapeutic approaches.
Collapse
Grants
- This research was supported by the European Union-Next Generation EU, Mission 4, Component 1 (CUP J53C24002040004), EU Horizon 2020 Framework Programme for Research and Innovation, European Innovation Council Pathfinder (PHOTOTHERAPORT, 101130883), Human Brain Project (WaveScalES, SGA3, 945539), Information and Communication Technologies (Deeper, ICT-36-2020-101016787), and Piano di Sostegno alla Ricerca 2023 (Azione A, Linea 2, PSR2023_DIP_021_CMATE). It was also supported by the Government of Catalonia (CERCA Programme; AGAUR 2021-SGR-01410), Spanish Ministry of Science and Innovation (DEEP RED, grant PID2019-111493RB-I00; EPILLUM, grant AEI/10.13039/501100011033; and Research Network in Biomedicine eBrains-Spain, RED2022-134823-E).
Collapse
Affiliation(s)
- Alessio Colleoni
- Section of Medicinal Chemistry "Pietro Pratesi", Department of Pharmaceutical Sciences, University of Milan, Milan, Italy
- Department of Chemistry, Biology, and Biotechnology, University of Perugia, Perugia, Italy
| | - Giulia Galli
- Section of Medicinal Chemistry "Pietro Pratesi", Department of Pharmaceutical Sciences, University of Milan, Milan, Italy
| | - Clelia Dallanoce
- Section of Medicinal Chemistry "Pietro Pratesi", Department of Pharmaceutical Sciences, University of Milan, Milan, Italy
| | - Marco De Amici
- Section of Medicinal Chemistry "Pietro Pratesi", Department of Pharmaceutical Sciences, University of Milan, Milan, Italy
| | - Pau Gorostiza
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology, Barcelona, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - Carlo Matera
- Section of Medicinal Chemistry "Pietro Pratesi", Department of Pharmaceutical Sciences, University of Milan, Milan, Italy
| |
Collapse
|
4
|
Dingová D, Kučera M, Hodbod T, Fischmeister R, Krejci E, Hrabovská A. Cardiac acetylcholinesterase and butyrylcholinesterase have distinct localization and function. Am J Physiol Heart Circ Physiol 2025; 328:H526-H542. [PMID: 39836467 DOI: 10.1152/ajpheart.00672.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/16/2024] [Accepted: 12/29/2024] [Indexed: 01/22/2025]
Abstract
Cholinesterase (ChE) inhibitors are under consideration for use in the treatment of cardiovascular pathologies. A prerequisite to advancing ChE inhibitors into the clinic is their thorough characterization in the heart. The aim here was to provide a detailed analysis of cardiac ChE to understand their molecular composition, localization, and physiological functions. A battery of biochemical, microscopic, and physiological experiments was used to analyze two known ChE, acetylcholinesterase (AChE) and butyrylcholinesterase (BChE), in hearts of mutant mice lacking different ChE molecular forms. Overall, AChE activity was exceeded by BChE, while it was localized mainly in the atria and the ventricular epicardium of the heart base. AChE was anchored by collagen Q (ColQ) in the basal lamina or by PRiMA at the plasma membrane and co-localized with the neuronal marker TUJ1. In the absence of anchored AChE, the heart rate was unresponsive to a ChE inhibitor. BChE, the major ChE in the heart, was detected predominantly in ventricles, presumably as a precursor (soluble monomers/dimers). Mice lacking BChE were more sensitive to a ChE inhibitor. Nevertheless, the overall impact on heart physiology was subtle, showing mainly a role in cholinergic antagonism to the positive inotropic effect of β-adrenergic stimulation. Our results help to unravel the mechanisms of ChE in cardiovascular pathologies and provide a foundation to facilitate the design of novel, more effective pharmacotherapies, which may reduce morbidity and mortality of patients with various heart diseases.NEW & NOTEWORTHY Inhibition of cholinesterases has therapeutic potential in cardiovascular pathologies. Both acetylcholinesterase and butyrylcholinesterase are present in the heart. Each cholinesterase has distinct localization patterns in the heart and functions in cardiac physiology. Selective inhibition of acetylcholinesterase or butyrylcholinesterase may be used to alter specific cardiac functions. Butyrylcholinesterase polymorphism may have an impact on the outcome of the cholinesterase inhibitor treatment.
Collapse
Affiliation(s)
- Dominika Dingová
- Faculty of Pharmacy, Department of Pharmacology and Toxicology, Comenius University Bratislava, Bratislava, Slovakia
- Centre Borelli, CNRS, ENS Paris Saclay, Université Paris Cité, Paris, France
| | - Matej Kučera
- Faculty of Pharmacy, Department of Pharmacology and Toxicology, Comenius University Bratislava, Bratislava, Slovakia
- UMR-S 1180, Inserm, Université Paris-Saclay, Orsay, France
| | - Tibor Hodbod
- Faculty of Pharmacy, Department of Pharmacology and Toxicology, Comenius University Bratislava, Bratislava, Slovakia
| | | | - Eric Krejci
- Centre Borelli, CNRS, ENS Paris Saclay, Université Paris Cité, Paris, France
| | - Anna Hrabovská
- Faculty of Pharmacy, Department of Pharmacology and Toxicology, Comenius University Bratislava, Bratislava, Slovakia
- Toxicological and Antidoping Center, Faculty of Pharmacy, Comenius University Bratislava, Bratislava, Slovakia
| |
Collapse
|
5
|
Ferjančič Benetik S, Knez D, Obreza A, Košak U, Gobec S. Dual inhibition of butyrylcholinesterase and p38α mitogen-activated protein kinase: A new approach for the treatment of Alzheimer's disease. Pharmacol Ther 2024; 264:108748. [PMID: 39521443 DOI: 10.1016/j.pharmthera.2024.108748] [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: 07/02/2024] [Revised: 10/25/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
The simultaneous targeting of neuroinflammation and cholinergic hypofunction, the key pathological changes in Alzheimer's disease (AD), is not addressed by drugs currently in clinical trials, highlighting a critical therapeutic gap. We propose that dual-acting small molecules that inhibit butyrylcholinesterase (BChE) and mitogen-activated protein kinase p38α (p38α MAPK) represent a novel strategy to combat AD. This hypothesis is supported by cellular and animal studies as well as in silico modelling showing that it is possible to act simultaneously on both enzymes. Amyloid beta (Aβ) plaques trigger a pro-inflammatory microglial response that overactivates p38α MAPK, leading to increased Aβ synthesis, tau hyperphosphorylation, and altered synaptic plasticity. Overactivated microglia exacerbate neuroinflammation and cholinergic degeneration, ultimately leading to cognitive impairment. Structural similarities between the binding sites of BChE and p38α MAPK provide a promising basis for the development of dual inhibitors that could alleviate AD symptoms and address the underlying pathology.
Collapse
Affiliation(s)
| | - Damijan Knez
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva 7, 1000 Ljubljana, Slovenia
| | - Aleš Obreza
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva 7, 1000 Ljubljana, Slovenia
| | - Urban Košak
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva 7, 1000 Ljubljana, Slovenia
| | - Stanislav Gobec
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva 7, 1000 Ljubljana, Slovenia.
| |
Collapse
|
6
|
Hrabovska A. Potential mechanisms of acyl-ghrelin in heart failure: Insights into cholinergic modulation. ESC Heart Fail 2024; 11:3447-3448. [PMID: 39082104 PMCID: PMC11424330 DOI: 10.1002/ehf2.14990] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/01/2024] [Accepted: 07/08/2024] [Indexed: 09/27/2024] Open
Affiliation(s)
- Anna Hrabovska
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University Bratislava, Bratislava, Slovakia
- Department of Pharmacology, Faculty of Medicine, Slovak Medical University, Bratislava, Slovakia
| |
Collapse
|
7
|
Liu Chung Ming C, Wang X, Gentile C. Protective role of acetylcholine and the cholinergic system in the injured heart. iScience 2024; 27:110726. [PMID: 39280620 PMCID: PMC11402255 DOI: 10.1016/j.isci.2024.110726] [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] [Indexed: 09/18/2024] Open
Abstract
This review explores the roles of the cholinergic system in the heart, comprising the neuronal and non-neuronal cholinergic systems. Both systems are essential for maintaining cardiac homeostasis by regulating the release of acetylcholine (ACh). A reduction in ACh release is associated with the early onset of cardiovascular diseases (CVDs), and increasing evidence supports the protective roles of ACh against CVD. We address the challenges and limitations of current strategies to elevate ACh levels, including vagus nerve stimulation and pharmacological interventions such as cholinesterase inhibitors. Additionally, we introduce alternative strategies to increase ACh in the heart, such as stem cell therapy, gene therapy, microRNAs, and nanoparticle drug delivery methods. These findings offer new insights into advanced treatments for regenerating the injured human heart.
Collapse
Affiliation(s)
- Clara Liu Chung Ming
- School of Biomedical Engineering, Faculty of Engineering and Information Technology, University of Technology Sydney, Sydney, NSW, Australia
- Cardiovascular Regeneration Group, Heart Research Institute, Newtown, NSW 2042, Australia
| | - Xiaowei Wang
- Department of Medicine, Monash University, Melbourne, VIC 3800, Australia
- Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC 3010, Australia
- Molecular Imaging and Theranostics Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
| | - Carmine Gentile
- School of Biomedical Engineering, Faculty of Engineering and Information Technology, University of Technology Sydney, Sydney, NSW, Australia
- Cardiovascular Regeneration Group, Heart Research Institute, Newtown, NSW 2042, Australia
| |
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
|
Ganeshpurkar A, Akotkar L, Kumar D, Kumar D, Ganeshpurkar A. Machine learning-based virtual screening and molecular modelling studies for identification of butyrylcholinesterase inhibitors as anti-Alzheimer's agent. J Biomol Struct Dyn 2024:1-17. [PMID: 38466084 DOI: 10.1080/07391102.2024.2326664] [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: 04/17/2023] [Accepted: 02/28/2024] [Indexed: 03/12/2024]
Abstract
Butyrylcholinesterase (BChE) is a hydrolase involved in the metabolism and detoxification of specific esters in the blood. It is also implicated in the progression of Alzheimer's disease, a type of dementia. As the disease progresses, the level of BChE tends to increase, opting for a major role as an acetylcholine-degrading enzyme and surpassing the role of acetylcholinesterase. Hence, the development of BChE inhibitors could be beneficial for the latter stages of the disease. In the present study, machine learning (ML) models were developed and employed to identify new BChE inhibitors. Further, the identified molecules were subjected to molecular property filters. The filtered ligands were studied through molecular modelling techniques, viz. molecular docking and molecular dynamics (MD). Support vector machine-based ML models resulted in the identification of 3291 compounds that would have predicted IC50 values less than 200 nM. The docking study showed that compounds ART13069594, ART17350769 and LEG19710163 have mean binding energies of -9.62, -9.26 and -8.93 kcal/mol, respectively. The MD study displayed that all the selected ligands showed stable complexes with BChE. The trajectories of all the ligands were stable similar to the standard BChE inhibitors.
Collapse
Affiliation(s)
- Ankit Ganeshpurkar
- Department of Pharmaceutical Chemistry, Poona College of Pharmacy, Bharati Vidyapeeth (Deemed to be University), Pune, India
| | - Likhit Akotkar
- Department of Pharmacology, Poona College of Pharmacy, Bharati Vidyapeeth (Deemed to be University), Pune, India
| | - Devendra Kumar
- School of Pharmacy & Technology Management, SVKM's NMIMS University, Shirpur, India
| | - Dileep Kumar
- Department of Pharmaceutical Chemistry, Poona College of Pharmacy, Bharati Vidyapeeth (Deemed to be University), Pune, India
| | | |
Collapse
|
10
|
Chen T, Sang S, Wei Y, Ge Y, Jin J, Bian Y, Pei Y, Li N, Sun H, Chen Y. The structural modification and biological evaluation of tetrahydrothienopyridine derivatives as selective BChE inhibitors. Bioorg Med Chem Lett 2023; 93:129436. [PMID: 37549853 DOI: 10.1016/j.bmcl.2023.129436] [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: 05/16/2023] [Revised: 07/30/2023] [Accepted: 08/04/2023] [Indexed: 08/09/2023]
Abstract
A series of tetrahydrothienopyridine derivatives have been designed, synthesized, and evaluated as selective BChE inhibitors. Compounds were analyzed via HRMS, 1H NMR, and 13C NMR. The inhibitory effects were evaluated according to the method of Ellman et al. 6n was the most potent and selective inhibitor against BChE (eeAChE IC50 = 686.4 ± 478.6 μM, eqBChE IC50 = 10.5 ± 5.0 nM, SI = 6.5*104, hBChE IC50 = 32.5 ± 6.5 nM). Cell-based assays have confirmed the low neurotoxicity of 6a and 6n and their moderate neuroprotective effects. Compounds 6a and 6n provide novel chemical entities for the treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Tingkai Chen
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Shenghu Sang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yuqing Wei
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yujie Ge
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jisheng Jin
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yaoyao Bian
- Jiangsu Provincial Engineering Center of TCM External Medication Researching and Industrializing, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yuqiong Pei
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Nianguang Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Haopeng Sun
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China.
| | - Yao Chen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
11
|
Pasieka A, Panek D, Zaręba P, Sługocka E, Gucwa N, Espargaró A, Latacz G, Khan N, Bucki A, Sabaté R, Więckowska A, Malawska B. Novel drug-like fluorenyl derivatives as selective butyrylcholinesterase and β-amyloid inhibitors for the treatment of Alzheimer's disease. Bioorg Med Chem 2023; 88-89:117333. [PMID: 37236021 DOI: 10.1016/j.bmc.2023.117333] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 04/23/2023] [Accepted: 05/12/2023] [Indexed: 05/28/2023]
Abstract
Butyrylcholinesterase (BuChE) and amyloid β (Aβ) aggregation remain important biological target and mechanism in the search for effective treatment of Alzheimer's disease. Simultaneous inhibition thereof by the application of multifunctional agents may lead to improvement in terms of symptoms and causes of the disease. Here, we present the rational design, synthesis, biological evaluation and molecular modelling studies of novel series of fluorene-based BuChE and Aβ inhibitors with drug-like characteristics and advantageous Central Nervous System Multiparameter Optimization scores. Among 17 synthesized and tested compounds, we identified 22 as the most potent eqBuChE inhibitor with IC50 of 38 nM and 37.4% of Aβ aggregation inhibition at 10 μM. Based on molecular modelling studies, including molecular dynamics, we determined the binding mode of the compounds within BuChE and explained the differences in the activity of the two enantiomers of compound 22. A novel series of fluorenyl compounds meeting the drug-likeness criteria seems to be a promising starting point for further development as anti-Alzheimer agents.
Collapse
Affiliation(s)
- Anna Pasieka
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna St. 9, 30-688 Kraków, Poland
| | - Dawid Panek
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna St. 9, 30-688 Kraków, Poland.
| | - Paula Zaręba
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna St. 9, 30-688 Kraków, Poland
| | - Emilia Sługocka
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna St. 9, 30-688 Kraków, Poland; Doctoral School of Medical and Health Sciences, Jagiellonian University Medical College, 16 Łazarza St., 31-530 Kraków, Poland
| | - Natalia Gucwa
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna St. 9, 30-688 Kraków, Poland
| | - Alba Espargaró
- Department of Pharmacy and Pharmaceutical Technology and Physical-Chemistry, School of Pharmacy and Food Sciences, University of Barcelona, Av Joan XXIII 27-31, 08028 Barcelona, Spain; Institute of Nanoscience and Nanotechnology (IN2UB), Av Joan XXIII, S/N, 08028 Barcelona, Spain
| | - Gniewomir Latacz
- Department of Technology and Biotechnology of Drugs, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna St. 9, 30-688 Kraków, Poland
| | - Nadia Khan
- Department of Technology and Biotechnology of Drugs, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna St. 9, 30-688 Kraków, Poland; Departement of Pathophysiology, Jagiellonian University Medical College, 18 Czysta St., 31-121 Kraków, Poland
| | - Adam Bucki
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna St., 30-688 Kraków, Poland
| | - Raimon Sabaté
- Department of Pharmacy and Pharmaceutical Technology and Physical-Chemistry, School of Pharmacy and Food Sciences, University of Barcelona, Av Joan XXIII 27-31, 08028 Barcelona, Spain; Institute of Nanoscience and Nanotechnology (IN2UB), Av Joan XXIII, S/N, 08028 Barcelona, Spain
| | - Anna Więckowska
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna St. 9, 30-688 Kraków, Poland
| | - Barbara Malawska
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna St. 9, 30-688 Kraków, Poland
| |
Collapse
|
12
|
Panek D, Pasieka A, Latacz G, Zaręba P, Szczęch M, Godyń J, Chantegreil F, Nachon F, Brazzolotto X, Skrzypczak-Wiercioch A, Walczak M, Smolik M, Sałat K, Höfner G, Wanner K, Więckowska A, Malawska B. Discovery of new, highly potent and selective inhibitors of BuChE - design, synthesis, in vitro and in vivo evaluation and crystallography studies. Eur J Med Chem 2023; 249:115135. [PMID: 36696766 DOI: 10.1016/j.ejmech.2023.115135] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 01/09/2023] [Accepted: 01/16/2023] [Indexed: 01/19/2023]
Abstract
The symptomatic and disease-modifying effects of butyrylcholinesterase (BuChE) inhibitors provide an encouraging premise for researching effective treatments for Alzheimer's disease. Here, we examined a series of compounds with a new chemical scaffold based on 3-(cyclohexylmethyl)amino-2-hydroxypropyl, and we identified a highly selective hBuChE inhibitor (29). Based on extensive in vitro and in vivo evaluations of the compound and its enantiomers, (R)-29 was identified as a promising candidate for further development. Compound (R)-29 is a potent hBuChE inhibitor (IC50 = 40 nM) with selectivity over AChE and relevant off-targets, including H1, M1, α1A and β1 receptors. The compound displays high metabolic stability on human liver microsomes (90% of the parent compound after 2 h of incubation), and its safety was confirmed through examining the cytotoxicity on the HepG2 cell line (LC50 = 2.85 μM) and hERG inhibition (less than 50% at 10 μM). While (rac)-29 lacked an effect in vivo and showed limited penetration to the CNS in pharmacokinetics studies, compound (R)-29 exhibited a procognitive effect at 15 mg/kg in the passive avoidance task in scopolamine-treated mice.
Collapse
Affiliation(s)
- Dawid Panek
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna St. 9, 30-688, Kraków, Poland.
| | - Anna Pasieka
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna St. 9, 30-688, Kraków, Poland
| | - Gniewomir Latacz
- Department of Technology and Biotechnology of Drugs, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna St. 9, 30-688, Kraków, Poland
| | - Paula Zaręba
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna St. 9, 30-688, Kraków, Poland
| | - Michał Szczęch
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna St. 9, 30-688, Kraków, Poland
| | - Justyna Godyń
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna St. 9, 30-688, Kraków, Poland
| | - Fabien Chantegreil
- Département de Toxicologie et Risques Chimiques, Institut de Recherche Biomédicale des Armées, 91223, Brétigny sur Orge, France
| | - Florian Nachon
- Département de Toxicologie et Risques Chimiques, Institut de Recherche Biomédicale des Armées, 91223, Brétigny sur Orge, France
| | - Xavier Brazzolotto
- Département de Toxicologie et Risques Chimiques, Institut de Recherche Biomédicale des Armées, 91223, Brétigny sur Orge, France
| | - Anna Skrzypczak-Wiercioch
- Department of Animal Anatomy and Preclinical Sciences, University Centre of Veterinary Medicine JU-UA, University of Agriculture in Krakow, Mickiewicza 24/28, 30-059, Kraków, Poland
| | - Maria Walczak
- Chair and Department of Toxicology, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna St. 9, 30-688, Krakow, Poland
| | - Magdalena Smolik
- Chair and Department of Toxicology, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna St. 9, 30-688, Krakow, Poland
| | - Kinga Sałat
- Department of Pharmacodynamics, Chair of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna St. 9, 30-688, Krakow, Poland
| | - Georg Höfner
- Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians-Universität München, Butenandtstr, 5-13, 81377, Munich, Germany
| | - Klaus Wanner
- Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians-Universität München, Butenandtstr, 5-13, 81377, Munich, Germany
| | - Anna Więckowska
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna St. 9, 30-688, Kraków, Poland
| | - Barbara Malawska
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna St. 9, 30-688, Kraków, Poland
| |
Collapse
|
13
|
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
|
14
|
Patel KB, Patel DV, Patel NR, Kanhed AM, Teli DM, Gandhi B, Shah BS, Chaudhary BN, Prajapati NK, Patel KV, Yadav MR. Carbazole-based semicarbazones and hydrazones as multifunctional anti-Alzheimer agents. J Biomol Struct Dyn 2022; 40:10278-10299. [PMID: 34215173 DOI: 10.1080/07391102.2021.1942212] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
With the aim to combat a multi-faceted neurodegenerative Alzheimer's disease (AD), a series of carbazole-based semicarbazide and hydrazide derivatives were designed, synthesized and assessed for their cholinesterase (ChE) inhibitory, antioxidant and biometal chelating activity. Among them, (E)-2-((9-ethyl-9H-carbazol-3-yl)methylene)-N-(pyridin-2-yl)hydrazinecarbothioamide (62) and (E)-2-((9-ethyl-9H-carbazol-3-yl)methylene)-N-(5-chloropyridin-2-yl)hydrazinecarbothioamide (63) emerged as the premier candidates with good ChE inhibitory activities (IC50 values of 1.37 µM and 1.18 µM for hAChE, IC50 values of 2.69 µM and 3.31 µM for EqBuChE, respectively). All the test compounds displayed excellent antioxidant activity (reduction percentage of DPPH values for compounds (62) and (63) were 85.67% and 84.49%, respectively at 100 µM concentration). Compounds (62) and (63) conferred specific copper ion chelating property in metal chelation study. Molecular docking studies of compounds (62) and (63) indicate strong interactions within the active sites of both the ChE enzymes. Besides that, these compounds also exhibited significant in silico drug-like pharmacokinetic properties. Thus, taken together, they can serve as a starting point in the designing of multifunctional ligands in pursuit of potential anti-AD agents that might further prevent the progression of ADs.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Kishan B Patel
- Faculty of Pharmacy, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India
| | - Dushyant V Patel
- Faculty of Pharmacy, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India
| | - Nirav R Patel
- Faculty of Pharmacy, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India
| | - Ashish M Kanhed
- Shobhaben Pratapbhai Patel - School of Pharmacy & Technology Management, SVKM's NMIMS University, Mumbai, India
| | - Divya M Teli
- Department of Pharmaceutical Chemistry, L. M. College of Pharmacy, Navrangpura, Gujarat, India
| | - Bhumi Gandhi
- Faculty of Pharmacy, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India
| | - Bhavik S Shah
- Faculty of Pharmacy, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India
| | - Bharat N Chaudhary
- Faculty of Pharmacy, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India
| | - Navnit K Prajapati
- Faculty of Pharmacy, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India
| | - Kirti V Patel
- Faculty of Pharmacy, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India
| | - Mange Ram Yadav
- Faculty of Pharmacy, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India.,Centre of Research for Development, Parul University, Vadodara, Gujarat, India
| |
Collapse
|
15
|
Edem EE, Okhonmina UE, Nebo KE, Akinluyi ET, Ikuelogbon DA, Fafure AA, Olabiyi AA, Adedokun MA. Combined Exposure to Chronic Sleep Deprivation and Caffeine Potentiates Behavioural Deficits by Altering Neurochemical Profile and Synaptophysin Expression in Long-Evans Rats. Neurotox Res 2022; 40:2001-2015. [PMID: 36434357 DOI: 10.1007/s12640-022-00589-1] [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: 08/12/2022] [Revised: 09/22/2022] [Accepted: 10/08/2022] [Indexed: 11/27/2022]
Abstract
Using the Unpredictable Chronic Sleep Deprivation (UCSD) paradigm we developed, the combined effects of chronic sleep deprivation and high caffeine intake on prefrontal cortical synaptophysin expression, neurochemical profiles, and behavioural outcomes in Long-Evans rats were evaluated. The combination of chronic sleep deprivation and high-dose caffeine treatment produced varying degrees of behavioural impairments, depletion of antioxidants, serotonin, and an upregulation of acetylcholinesterase (AChE) activity in the prefrontal cortex. An immunohistochemical assessment revealed a reduction in synaptophysin protein expression in the prefrontal cortex following exposure to high-dose caffeine and chronic sleep deprivation. Overall, our findings support the advocacy for adequate sleep for optimal mental performance as a high intake of caffeine to attenuate the effects of sleep deprivation that may alter the neurochemical profile and synaptic plasticity in the prefrontal cortex, significantly increasing the risk of neuropsychiatric/degenerative disorders.
Collapse
Affiliation(s)
- Edem Ekpenyong Edem
- Neuroscience Unit, Department of Human Anatomy, College of Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, Nigeria. .,Department of Anatomy, College of Medicine, University of Lagos, Idi-Araba, Lagos State, Nigeria.
| | - Uyi Emmanuel Okhonmina
- Neuroscience Unit, Department of Human Anatomy, College of Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, Nigeria
| | - Kate Eberechukwu Nebo
- Neuroscience Unit, Department of Human Anatomy, College of Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, Nigeria
| | - Elizabeth Toyin Akinluyi
- Neuropharmacology Unit, Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, Nigeria
| | | | - Adedamola Adediran Fafure
- Neuroscience Unit, Department of Human Anatomy, College of Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, Nigeria
| | - Ayodeji Augustine Olabiyi
- Department of Medical Biochemistry, College of Medicine and Health Sciences, Afe Babalola University, Ekiti State, Ado-Ekiti, Nigeria
| | - Mujeeb Adekunle Adedokun
- Neuroscience Unit, Department of Human Anatomy, College of Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, Nigeria
| |
Collapse
|
16
|
Biological Characterization of Natural Peptide BcI-1003 from Boana cordobae (anura): Role in Alzheimer’s Disease and Microbial Infections. Int J Pept Res Ther 2022. [DOI: 10.1007/s10989-022-10472-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
17
|
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: 0.7] [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
|
18
|
Bestwick JS, Jones DJ, Jones HE, Kalomenopoulos PG, Szabla R, Lawrence AL. Total Synthesis and Prediction of Ulodione Natural Products Guided by DFT Calculations. Angew Chem Int Ed Engl 2022; 61:e202207004. [PMID: 35670364 PMCID: PMC9401604 DOI: 10.1002/anie.202207004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Indexed: 11/11/2022]
Abstract
A biomimetic synthetic strategy has resulted in a two-step total synthesis of (±)-ulodione A and the prediction of two potential natural products, (±)-ulodiones C and D. This work was guided by computational investigations into the selectivity of a proposed biosynthetic Diels-Alder dimerization, which was then utilized in the chemical synthesis. This work highlights how biosynthetic considerations can both guide the design of efficient synthetic strategies and lead to the anticipation of new natural products.
Collapse
Affiliation(s)
- Jacob S. Bestwick
- EaStCHEM School of ChemistryUniversity of Edinburgh Joseph Black BuildingDavid Brewster RoadEdinburghEH9 3FJUK
| | - David J. Jones
- EaStCHEM School of ChemistryUniversity of Edinburgh Joseph Black BuildingDavid Brewster RoadEdinburghEH9 3FJUK
| | - Helen E. Jones
- EaStCHEM School of ChemistryUniversity of Edinburgh Joseph Black BuildingDavid Brewster RoadEdinburghEH9 3FJUK
- Current address: Oncology R&DAstraZenecaCambridgeCB4 0WGUK
| | - Panagiotis G. Kalomenopoulos
- EaStCHEM School of ChemistryUniversity of Edinburgh Joseph Black BuildingDavid Brewster RoadEdinburghEH9 3FJUK
- Current address: Process ChemistryPharmaronHoddesdonEN11 9FHUK
| | - Rafal Szabla
- Department of Physical and Quantum ChemistryFaculty of ChemistryWrocław University of Science and TechnologyWrocławPoland
| | - Andrew L. Lawrence
- EaStCHEM School of ChemistryUniversity of Edinburgh Joseph Black BuildingDavid Brewster RoadEdinburghEH9 3FJUK
| |
Collapse
|
19
|
Ahmed ZB, Hefied F, Mahammed TH, Seidel V, Yousfi M. Identification of potential
anti‐Alzheimer
agents from
Pistacia atlantica
Desf. galls using
UPLC
fingerprinting, chemometrics, and molecular docking analyses. J FOOD PROCESS PRES 2022. [DOI: 10.1111/jfpp.16916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Ziyad Ben Ahmed
- Laboratoire des Sciences Fondamentale Université Amar Telidji Laghouat Algérie
- Department of Analytical Chemistry, Applied Chemometrics and Molecular Modelling, Vrije Universiteit Brussel (VUB), Brussels Belgium
| | - Fatiha Hefied
- Laboratoire des Sciences Fondamentale Université Amar Telidji Laghouat Algérie
| | | | - Veronique Seidel
- Natural Products Research Laboratory, Strathclyde Institute of Pharmacy and Biomedical Sciences University of Strathclyde Glasgow UK
| | - Mohamed Yousfi
- Laboratoire des Sciences Fondamentale Université Amar Telidji Laghouat Algérie
| |
Collapse
|
20
|
Bestwick JS, Jones DJ, Jones HE, Kalomenopoulos PG, Szabla R, Lawrence AL. Total Synthesis and Prediction of Ulodione Natural Products Guided by DFT Calculations. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202207004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Jacob S. Bestwick
- The University of Edinburgh EaStCHEM School of Chemistry UNITED KINGDOM
| | - David J. Jones
- The University of Edinburgh EaStCHEM School of Chemistry UNITED KINGDOM
| | - Helen E. Jones
- The University of Edinburgh EaStCHEM School of Chemistry UNITED KINGDOM
| | | | - Rafal Szabla
- Wroclaw University of Science and Technology: Politechnika Wroclawska Department of Physical and Quantum Chemistry POLAND
| | - Andrew Leslie Lawrence
- University of Edinburgh EaStCHEM School of Chemistry Joseph Black BuildingDavid Brewster Road EH9 3FJ Edinburgh UNITED KINGDOM
| |
Collapse
|
21
|
Cholinergic blockade of neuroinflammation – from tissue to RNA regulators. Neuronal Signal 2022; 6:NS20210035. [PMID: 35211331 PMCID: PMC8837817 DOI: 10.1042/ns20210035] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 01/06/2022] [Accepted: 01/06/2022] [Indexed: 11/17/2022] Open
Abstract
Inflammatory stimuli and consequent pro-inflammatory immune responses may facilitate neurodegeneration and threaten survival following pathogen infection or trauma, but potential controllers preventing these risks are incompletely understood. Here, we argue that small RNA regulators of acetylcholine (ACh) signaling, including microRNAs (miRs) and transfer RNA fragments (tRFs) may tilt the balance between innate and adaptive immunity, avoid chronic inflammation and prevent the neuroinflammation-mediated exacerbation of many neurological diseases. While the restrictive permeability of the blood–brain barrier (BBB) protects the brain from peripheral immune events, this barrier can be disrupted by inflammation and is weakened with age. The consequently dysregulated balance between pro- and anti-inflammatory processes may modify the immune activities of brain microglia, astrocytes, perivascular macrophages, oligodendrocytes and dendritic cells, leading to neuronal damage. Notably, the vagus nerve mediates the peripheral cholinergic anti-inflammatory reflex and underlines the consistent control of body–brain inflammation by pro-inflammatory cytokines, which affect cholinergic functions; therefore, the disruption of this reflex can exacerbate cognitive impairments such as attention deficits and delirium. RNA regulators can contribute to re-balancing the cholinergic network and avoiding its chronic deterioration, and their activities may differ between men and women and/or wear off with age. This can lead to hypersensitivity of aged patients to inflammation and higher risks of neuroinflammation-driven cholinergic impairments such as delirium and dementia following COVID-19 infection. The age- and sex-driven differences in post-transcriptional RNA regulators of cholinergic elements may hence indicate new personalized therapeutic options for neuroinflammatory diseases.
Collapse
|
22
|
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: 1.3] [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
|
23
|
Wichur T, Pasieka A, Godyń J, Panek D, Góral I, Latacz G, Honkisz-Orzechowska E, Bucki A, Siwek A, Głuch-Lutwin M, Knez D, Brazzolotto X, Gobec S, Kołaczkowski M, Sabate R, Malawska B, Więckowska A. Discovery of 1-(phenylsulfonyl)-1H-indole-based multifunctional ligands targeting cholinesterases and 5-HT 6 receptor with anti-aggregation properties against amyloid-beta and tau. Eur J Med Chem 2021; 225:113783. [PMID: 34461507 DOI: 10.1016/j.ejmech.2021.113783] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 08/12/2021] [Accepted: 08/13/2021] [Indexed: 12/31/2022]
Abstract
Multifunctional ligands as an essential variant of polypharmacology are promising candidates for the treatment of multi-factorial diseases like Alzheimer's disease. Based on clinical evidence and following the paradigm of multifunctional ligands we have rationally designed and synthesized a series of compounds targeting processes involved in the development of the disease. The biological evaluation led to the discovery of two compounds with favorable pharmacological characteristics and ADMET profile. Compounds 17 and 35 are 5-HT6R antagonists (Ki = 13 nM and Ki = 15 nM respectively) and cholinesterase inhibitors with distinct mechanisms of enzyme inhibition. Compound 17, a tacrine derivative is a reversible inhibitor of acetyl- and butyrylcholinesterase (IC50 = 8 nM and IC50 = 24 nM respectively), while compound 35 with rivastigmine-derived phenyl N-ethyl-N-methylcarbamate fragment is a selective, pseudo-irreversible inhibitor of butyrylcholinesterase (IC50 = 455 nM). Both compounds inhibit aggregation of amyloid β in vitro (75% for compound 17 and 68% for 35 at 10 μM) moreover, compound 35 is a potent tau aggregation inhibitor in cellulo (79%). In ADMET in vitro studies both compounds showed acceptable metabolic stability on mouse liver microsomes (28% and 60% for compound 17 and 35 respectively), no or little effect on CYP3A4 and 2D6 up to a concentration of 10 μM and lack of toxicity on HepG2 cell line (IC50 values of 80 and 21 μM, for 17 and 35 respectively). Based on the pharmacological characteristics and favorable pharmacokinetic properties, we propose compounds 17 and 35 as an excellent starting point for further optimization and in-depth biological studies.
Collapse
Affiliation(s)
- Tomasz Wichur
- Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| | - Anna Pasieka
- 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
| | - Dawid Panek
- 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
| | - Damijan Knez
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva 7, 1000 Ljubljana, Slovenia
| | - Xavier Brazzolotto
- Département de Toxicologie et Risques Chimiques, Institut de Recherche Biomédicale des Armées, 91223 Brétigny sur Orge, France
| | - 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
| | - Raimon Sabate
- Department of Pharmacy and Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy and Food Science, University of Barcelona, Barcelona, Spain; Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, Barcelona, Spain
| | - Barbara Malawska
- Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| | - Anna Więckowska
- Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland.
| |
Collapse
|
24
|
Wu C, Zhang G, Zhang ZW, Jiang X, Zhang Z, Li H, Qin HL, Tang W. Structure-activity relationship, in vitro and in vivo evaluation of novel dienyl sulphonyl fluorides as selective BuChE inhibitors for the treatment of Alzheimer's disease. J Enzyme Inhib Med Chem 2021; 36:1860-1873. [PMID: 34425715 PMCID: PMC8386747 DOI: 10.1080/14756366.2021.1959571] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/26/2021] [Accepted: 07/19/2021] [Indexed: 01/08/2023] Open
Abstract
To discover novel scaffolds as leads against dementia, a series of δ-aryl-1,3-dienesulfonyl fluorides with α-halo, α-aryl and α-alkynyl were assayed for ChE inhibitory activity, in which compound A10 was identified as a selective BuChE inhibitor (IC50 = 0.021 μM for eqBChE, 3.62 μM for hBuChE). SAR of BuChE inhibition showed: (i) o- > m- > p-; -OCH3 > -CH3 > -Cl (-Br) for δ-aryl; (ii) α-Br > α-Cl, α-I. Compound A10 exhibited neuroprotective, BBB penetration, mixed competitive inhibitory effect on BuChE (Ki = 29 nM), and benign neural and hepatic safety. Treatment with A10 could almost entirely recover the Aβ1-42-induced cognitive dysfunction to the normal level, and the assessment of total amount of Aβ1-42 confirmed its anti-amyloidogenic profile. Therefore, the potential BuChE inhibitor A10 is a promising effective lead for the treatment of AD.
Collapse
Affiliation(s)
- Chengyao Wu
- School of Pharmacy, Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, Hefei, China
| | - Guijuan Zhang
- Management Center of Anhui Continuing Education Network Park, Anhui Open University, Hefei, China
| | - Zai-Wei Zhang
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan, China
| | - Xia Jiang
- School of Pharmacy, Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, Hefei, China
| | - Ziwen Zhang
- School of Pharmacy, Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, Hefei, China
| | - Huanhuan Li
- School of Pharmacy, Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, Hefei, China
| | - Hua-Li Qin
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan, China
| | - Wenjian Tang
- School of Pharmacy, Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, Hefei, China
| |
Collapse
|
25
|
Magar P, Parravicini O, Štěpánková Š, Svrčková K, Garro AD, Jendrzejewska I, Pauk K, Hošek J, Jampílek J, Enriz RD, Imramovský A. Novel Sulfonamide-Based Carbamates as Selective Inhibitors of BChE. Int J Mol Sci 2021; 22:9447. [PMID: 34502357 PMCID: PMC8430704 DOI: 10.3390/ijms22179447] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 08/28/2021] [Indexed: 11/16/2022] Open
Abstract
A series of 14 target benzyl [2-(arylsulfamoyl)-1-substituted-ethyl]carbamates was prepared by multi-step synthesis and characterized. All the final compounds were tested for their ability to inhibit acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) in vitro, and the selectivity index (SI) was determined. Except for three compounds, all compounds showed strong preferential inhibition of BChE, and nine compounds were even more active than the clinically used rivastigmine. Benzyl {(2S)-1-[(2-methoxybenzyl)sulfamoyl]-4-methylpentan-2-yl}carbamate (5k), benzyl {(2S)-1-[(4-chlorobenzyl)sulfamoyl]-4-methylpentan-2-yl}carbamate (5j), and benzyl [(2S)-1-(benzylsulfamoyl)-4-methylpentan-2-yl]carbamate (5c) showed the highest BChE inhibition (IC50 = 4.33, 6.57, and 8.52 µM, respectively), indicating that derivatives 5c and 5j had approximately 5-fold higher inhibitory activity against BChE than rivastigmine, and 5k was even 9-fold more effective than rivastigmine. In addition, the selectivity index of 5c and 5j was approx. 10 and that of 5k was even 34. The process of carbamylation and reactivation of BChE was studied for the most active derivatives 5k, 5j. The detailed information about the mode of binding of these compounds to the active site of both BChE and AChE was obtained in a molecular modeling study. In this study, combined techniques (docking, molecular dynamic simulations, and QTAIM (quantum theory of atoms in molecules) calculations) were employed.
Collapse
Affiliation(s)
- Pratibha Magar
- Institute of Organic Chemistry and Technology, Faculty of Chemical Technology, University of Pardubice, Studentska 573, 532 10 Pardubice, Czech Republic; (P.M.); (K.P.)
| | - Oscar Parravicini
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Instituto Multidisciplinario de Investigaciones Biológicas (IMIBIO-SL), Chacabuco 915, 5700 San Luis, Argentina; (O.P.); (A.D.G.)
| | - Šárka Štěpánková
- Department of Biological and Biochemical Sciences, Faculty of Chemical Technology, University of Pardubice, Studentska 573, 532 10 Pardubice, Czech Republic; (Š.Š.); (K.S.)
| | - Katarina Svrčková
- Department of Biological and Biochemical Sciences, Faculty of Chemical Technology, University of Pardubice, Studentska 573, 532 10 Pardubice, Czech Republic; (Š.Š.); (K.S.)
| | - Adriana D. Garro
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Instituto Multidisciplinario de Investigaciones Biológicas (IMIBIO-SL), Chacabuco 915, 5700 San Luis, Argentina; (O.P.); (A.D.G.)
| | | | - Karel Pauk
- Institute of Organic Chemistry and Technology, Faculty of Chemical Technology, University of Pardubice, Studentska 573, 532 10 Pardubice, Czech Republic; (P.M.); (K.P.)
| | - Jan Hošek
- Department of Pharmacology and Toxicology, Veterinary Research Institute, Hudcova 296/70, 621 00 Brno, Czech Republic;
| | - Josef Jampílek
- Department of Analytical Chemistry, Faculty of Natural Sciences, Comenius University, Ilkovicova 6, 842 15 Bratislava, Slovakia;
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska Cesta 9, 845 10 Bratislava, Slovakia
| | - Ricardo D. Enriz
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Instituto Multidisciplinario de Investigaciones Biológicas (IMIBIO-SL), Chacabuco 915, 5700 San Luis, Argentina; (O.P.); (A.D.G.)
| | - Aleš Imramovský
- Institute of Organic Chemistry and Technology, Faculty of Chemical Technology, University of Pardubice, Studentska 573, 532 10 Pardubice, Czech Republic; (P.M.); (K.P.)
| |
Collapse
|
26
|
Winand L, Schneider P, Kruth S, Greven NJ, Hiller W, Kaiser M, Pietruszka J, Nett M. Mutasynthesis of Physostigmines in Myxococcus xanthus. Org Lett 2021; 23:6563-6567. [PMID: 34355569 DOI: 10.1021/acs.orglett.1c02374] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The alkaloid physostigmine is an approved anticholinergic drug and an important lead structure for the development of novel therapeutics. Using a complementary approach that merged chemical synthesis with pathway refactoring, we produced a series of physostigmine analogues with altered specificity and toxicity profiles in the heterologous host Myxococcus xanthus. The compounds that were generated by applying a simple feeding strategy include the promising drug candidate phenserine, which was previously accessible only by total synthesis.
Collapse
Affiliation(s)
- Lea Winand
- Department of Biochemical and Chemical Engineering, TU Dortmund University, Dortmund, 44227 Nordrhein-Westfalen, Germany
| | - Pascal Schneider
- Institute of Bioorganic Chemistry, Heinrich-Heine-University Düsseldorf at Forschungszentrum Jülich, Jülich, 44227 Nordrhein-Westfalen, Germany
| | - Sebastian Kruth
- Department of Biochemical and Chemical Engineering, TU Dortmund University, Dortmund, 44227 Nordrhein-Westfalen, Germany
| | - Nico-Joel Greven
- Department of Biochemical and Chemical Engineering, TU Dortmund University, Dortmund, 44227 Nordrhein-Westfalen, Germany
| | - Wolf Hiller
- Department of Chemistry and Chemical Biology, TU Dortmund University, Dortmund, 44227 Nordrhein-Westfalen, Germany
| | - Marcel Kaiser
- Parasite Chemotherapy Unit, Swiss Tropical and Public Health Institute, 4002 Basel, Switzerland.,University of Basel, 4001 Basel, Switzerland
| | - Jörg Pietruszka
- Institute of Bioorganic Chemistry, Heinrich-Heine-University Düsseldorf at Forschungszentrum Jülich, Jülich, 44227 Nordrhein-Westfalen, Germany.,Institut für Bio- und Geowissenschaften: Biotechnologie (IBG-1), Forschungszentrum Jülich, Jülich, 52428 Nordrhein-Westfalen, Germany
| | - Markus Nett
- Department of Biochemical and Chemical Engineering, TU Dortmund University, Dortmund, 44227 Nordrhein-Westfalen, Germany
| |
Collapse
|
27
|
Mamun AA, Pidaný F, Hulcová D, Maříková J, Kučera T, Schmidt M, Catapano MC, Hrabinová M, Jun D, Múčková L, Kuneš J, Janoušek J, Andrýs R, Nováková L, Peřinová R, Maafi N, Soukup O, Korábečný J, Cahlíková L. Amaryllidaceae Alkaloids of Norbelladine-Type as Inspiration for Development of Highly Selective Butyrylcholinesterase Inhibitors: Synthesis, Biological Activity Evaluation, and Docking Studies. Int J Mol Sci 2021; 22:8308. [PMID: 34361074 PMCID: PMC8348983 DOI: 10.3390/ijms22158308] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 07/26/2021] [Accepted: 07/28/2021] [Indexed: 01/01/2023] Open
Abstract
Alzheimer's disease (AD) is a multifactorial neurodegenerative condition of the central nervous system (CNS) that is currently treated by cholinesterase inhibitors and the N-methyl-d-aspartate receptor antagonist, memantine. Emerging evidence strongly supports the relevance of targeting butyrylcholinesterase (BuChE) in the more advanced stages of AD. Within this study, we have generated a pilot series of compounds (1-20) structurally inspired from belladine-type Amaryllidaceae alkaloids, namely carltonine A and B, and evaluated their acetylcholinesterase (AChE) and BuChE inhibition properties. Some of the compounds exhibited intriguing inhibition activity for human BuChE (hBuChE), with a preference for BuChE over AChE. Seven compounds were found to possess a hBuChE inhibition profile, with IC50 values below 1 µM. The most potent one, compound 6, showed nanomolar range activity with an IC50 value of 72 nM and an excellent selectivity pattern over AChE, reaching a selectivity index of almost 1400. Compound 6 was further studied by enzyme kinetics, along with in-silico techniques, to reveal the mode of inhibition. The prediction of CNS availability estimates that all the compounds in this survey can pass through the blood-brain barrier (BBB), as disclosed by the BBB score.
Collapse
Affiliation(s)
- Abdullah Al Mamun
- ADINACO Research Group, Department of Pharmaceutical Botany, Faculty of Pharmacy, Charles University, Heyrovskeho 1203, 500 05 Hradec Kralove, Czech Republic; (A.A.M.); (F.P.); (D.H.); (J.M.); (R.P.); (N.M.)
| | - Filip Pidaný
- ADINACO Research Group, Department of Pharmaceutical Botany, Faculty of Pharmacy, Charles University, Heyrovskeho 1203, 500 05 Hradec Kralove, Czech Republic; (A.A.M.); (F.P.); (D.H.); (J.M.); (R.P.); (N.M.)
| | - Daniela Hulcová
- ADINACO Research Group, Department of Pharmaceutical Botany, Faculty of Pharmacy, Charles University, Heyrovskeho 1203, 500 05 Hradec Kralove, Czech Republic; (A.A.M.); (F.P.); (D.H.); (J.M.); (R.P.); (N.M.)
- Department of Pharmacognosy, Faculty of Pharmacy, Charles University, Heyrovskeho 1203, 500 05 Hradec Kralove, Czech Republic;
| | - Jana Maříková
- ADINACO Research Group, Department of Pharmaceutical Botany, Faculty of Pharmacy, Charles University, Heyrovskeho 1203, 500 05 Hradec Kralove, Czech Republic; (A.A.M.); (F.P.); (D.H.); (J.M.); (R.P.); (N.M.)
- Department of Bioorganic and Organic Chemistry, Faculty of Pharmacy, Charles University, Heyrovskeho 1203, 500 05 Hradec Kralove, Czech Republic;
| | - Tomáš Kučera
- Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, University of Defence, Trebesska 1575, 500 01 Hradec Kralove, Czech Republic; (T.K.); (M.H.); (D.J.); (L.M.); (O.S.)
| | - Monika Schmidt
- Department of Chemistry, Faculty of Science, University of Hradec Králové, Rokitanského 62, 500 03 Hradec Králové, Czech Republic; (M.S.); (R.A.)
| | - Maria Carmen Catapano
- Department of Analytical Chemistry, Faculty of Pharmacy, Charles University, Heyrovskeho 1203, 500 05 Hradec Kralove, Czech Republic; (M.C.C.); (L.N.)
| | - Martina Hrabinová
- Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, University of Defence, Trebesska 1575, 500 01 Hradec Kralove, Czech Republic; (T.K.); (M.H.); (D.J.); (L.M.); (O.S.)
- Biomedical Research Centre, University Hospital Hradec Kralove, Sokolska 581, 500 05 Hradec Kralove, Czech Republic
| | - Daniel Jun
- Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, University of Defence, Trebesska 1575, 500 01 Hradec Kralove, Czech Republic; (T.K.); (M.H.); (D.J.); (L.M.); (O.S.)
| | - Lubica Múčková
- Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, University of Defence, Trebesska 1575, 500 01 Hradec Kralove, Czech Republic; (T.K.); (M.H.); (D.J.); (L.M.); (O.S.)
- Biomedical Research Centre, University Hospital Hradec Kralove, Sokolska 581, 500 05 Hradec Kralove, Czech Republic
| | - Jiří Kuneš
- Department of Bioorganic and Organic Chemistry, Faculty of Pharmacy, Charles University, Heyrovskeho 1203, 500 05 Hradec Kralove, Czech Republic;
| | - Jiří Janoušek
- Department of Pharmacognosy, Faculty of Pharmacy, Charles University, Heyrovskeho 1203, 500 05 Hradec Kralove, Czech Republic;
| | - Rudolf Andrýs
- Department of Chemistry, Faculty of Science, University of Hradec Králové, Rokitanského 62, 500 03 Hradec Králové, Czech Republic; (M.S.); (R.A.)
| | - Lucie Nováková
- Department of Analytical Chemistry, Faculty of Pharmacy, Charles University, Heyrovskeho 1203, 500 05 Hradec Kralove, Czech Republic; (M.C.C.); (L.N.)
| | - Rozálie Peřinová
- ADINACO Research Group, Department of Pharmaceutical Botany, Faculty of Pharmacy, Charles University, Heyrovskeho 1203, 500 05 Hradec Kralove, Czech Republic; (A.A.M.); (F.P.); (D.H.); (J.M.); (R.P.); (N.M.)
| | - Negar Maafi
- ADINACO Research Group, Department of Pharmaceutical Botany, Faculty of Pharmacy, Charles University, Heyrovskeho 1203, 500 05 Hradec Kralove, Czech Republic; (A.A.M.); (F.P.); (D.H.); (J.M.); (R.P.); (N.M.)
| | - Ondřej Soukup
- Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, University of Defence, Trebesska 1575, 500 01 Hradec Kralove, Czech Republic; (T.K.); (M.H.); (D.J.); (L.M.); (O.S.)
- Biomedical Research Centre, University Hospital Hradec Kralove, Sokolska 581, 500 05 Hradec Kralove, Czech Republic
| | - Jan Korábečný
- Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, University of Defence, Trebesska 1575, 500 01 Hradec Kralove, Czech Republic; (T.K.); (M.H.); (D.J.); (L.M.); (O.S.)
- Biomedical Research Centre, University Hospital Hradec Kralove, Sokolska 581, 500 05 Hradec Kralove, Czech Republic
| | - Lucie Cahlíková
- ADINACO Research Group, Department of Pharmaceutical Botany, Faculty of Pharmacy, Charles University, Heyrovskeho 1203, 500 05 Hradec Kralove, Czech Republic; (A.A.M.); (F.P.); (D.H.); (J.M.); (R.P.); (N.M.)
| |
Collapse
|
28
|
Gentzsch C, Hoffmann M, Ohshima Y, Nose N, Chen X, Higuchi T, Decker M. Synthesis and Initial Characterization of a Selective, Pseudo-irreversible Inhibitor of Human Butyrylcholinesterase as PET Tracer. ChemMedChem 2021; 16:1427-1437. [PMID: 33645891 PMCID: PMC8247983 DOI: 10.1002/cmdc.202000942] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/19/2021] [Indexed: 02/06/2023]
Abstract
The enzyme butyrylcholinesterase (BChE) represents a promising target for imaging probes to potentially enable early diagnosis of neurodegenerative diseases like Alzheimer's disease (AD) and to monitor disease progression in some forms of cancer. In this study, we present the design, facile synthesis, in vitro and preliminary ex vivo and in vivo evaluation of a morpholine-based, selective inhibitor of human BChE as a positron emission tomography (PET) tracer with a pseudo-irreversible binding mode. We demonstrate a novel protecting group strategy for 18 F radiolabeling of carbamate precursors and show that the inhibitory potency as well as kinetic properties of our unlabeled reference compound were retained in comparison to the parent compound. In particular, the prolonged duration of enzyme inhibition of such a morpholinocarbamate motivated us to design a PET tracer, possibly enabling a precise mapping of BChE distribution.
Collapse
Affiliation(s)
- Christian Gentzsch
- Pharmaceutical and Medicinal ChemistryInstitute of Pharmacy and Food ChemistryJulius-Maximilians-University of WürzburgAm Hubland97074WürzburgGermany
| | - Matthias Hoffmann
- Pharmaceutical and Medicinal ChemistryInstitute of Pharmacy and Food ChemistryJulius-Maximilians-University of WürzburgAm Hubland97074WürzburgGermany
| | - Yasuhiro Ohshima
- Comprehensive Heart Failure CenterUniversity Hospital of WürzburgAm Schwarzenberg 1597078WürzburgGermany
- Department of Nuclear MedicineUniversity Hospital of WürzburgOberdürrbacher Straße 697080WürzburgGermany
| | - Naoko Nose
- Graduate School of MedicineDentistry and Pharmaceutical SciencesOkayama University2-5-1 Shikata-cho, Kita-kuOkayamaJapan
| | - Xinyu Chen
- Department of Nuclear MedicineUniversity Hospital of AugsburgStenglinstraße 286156AugsburgGermany
- Comprehensive Heart Failure CenterUniversity Hospital of WürzburgAm Schwarzenberg 1597078WürzburgGermany
- Department of Nuclear MedicineUniversity Hospital of WürzburgOberdürrbacher Straße 697080WürzburgGermany
| | - Takahiro Higuchi
- Comprehensive Heart Failure CenterUniversity Hospital of WürzburgAm Schwarzenberg 1597078WürzburgGermany
- Department of Nuclear MedicineUniversity Hospital of WürzburgOberdürrbacher Straße 697080WürzburgGermany
- Graduate School of MedicineDentistry and Pharmaceutical SciencesOkayama University2-5-1 Shikata-cho, Kita-kuOkayamaJapan
| | - Michael Decker
- Pharmaceutical and Medicinal ChemistryInstitute of Pharmacy and Food ChemistryJulius-Maximilians-University of WürzburgAm Hubland97074WürzburgGermany
| |
Collapse
|
29
|
In Vitro Enzymatic and Kinetic Studies, and In Silico Drug-Receptor Interactions, and Drug-Like Profiling of the 5-Styrylbenzamide Derivatives as Potential Cholinesterase and β-Secretase Inhibitors with Antioxidant Properties. Antioxidants (Basel) 2021; 10:antiox10050647. [PMID: 33922328 PMCID: PMC8145986 DOI: 10.3390/antiox10050647] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 04/10/2021] [Accepted: 04/20/2021] [Indexed: 11/17/2022] Open
Abstract
The 5-(styryl)anthranilamides were transformed into the corresponding 5-styryl-2-(p-tolylsulfonamido)benzamide derivatives. These 5-styrylbenzamide derivatives were evaluated through enzymatic assays in vitro for their capability to inhibit acetylcholinesterase (AChE), butyrylcholinesterase (BChE), and β-secretase (BACE-1) activities as well as for antioxidant potential. An in vitro cell-based antioxidant activity assay involving lipopolysaccharides (LPS)-induced reactive oxygen species (ROS) production revealed that compounds 2a and 3b have the capability of scavenging free radicals. The potential of the most active compound, 5-styrylbenzamide (2a), to bind copper (II) or zinc (II) ions has also been evaluated spectrophotometrically. Kinetic studies of the most active derivatives from each series against the AChE, BChE, and β-secretase activities have been performed. The experimental results are complemented with molecular docking studies into the active sites of these enzymes to predict the hypothetical protein–ligand binding modes. Their drug likeness properties have also been predicted.
Collapse
|
30
|
Zhou Y, Lu X, Du C, Liu Y, Wang Y, Hong KH, Chen Y, Sun H. Novel BuChE-IDO1 inhibitors from sertaconazole: Virtual screening, chemical optimization and molecular modeling studies. Bioorg Med Chem Lett 2020; 34:127756. [PMID: 33359445 DOI: 10.1016/j.bmcl.2020.127756] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 11/14/2020] [Accepted: 12/16/2020] [Indexed: 12/17/2022]
Abstract
In our effort towards the identification of novel BuChE-IDO1 dual-targeted inhibitor for the treatment of Alzheimer's disease (AD), sertaconazole was identified through a combination of structure-based virtual screening followed by MM-GBSA rescoring. Preliminary chemical optimization was performed to develop more potent and selective sertaconazole analogues. In consideration of the selectivity and the inhibitory activity against target proteins, compounds 5c and 5d were selected for the next study. Further modification of compound 5c led to the generation of compound 10g with notably improved selectivity towards BuChE versus AChE. The present study provided us with a good starting point to further design potent and selective BuChE-IDO1 inhibitors, which may benefit the treatment of late stage AD.
Collapse
Affiliation(s)
- You Zhou
- College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing 400715, China.
| | - Xin Lu
- Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Chenxi Du
- Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Yijun Liu
- Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Yifan Wang
- College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing 400715, China
| | - Kwon Ho Hong
- Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota, Minneapolis, MN 55414, USA
| | - Yao Chen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Haopeng Sun
- Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
31
|
Cloete SJ, Petzer A, Petzer JP. Interactions of dye compounds that are structurally related to methylene blue with acetylcholinesterase and butyrylcholinesterase. Chem Biol Drug Des 2020. [DOI: 10.1111/cbdd.13814] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- Stephanus J. Cloete
- Centre of Excellence for Pharmaceutical Sciences North‐West University Potchefstroom South Africa
| | - Anél Petzer
- Centre of Excellence for Pharmaceutical Sciences North‐West University Potchefstroom South Africa
- Pharmaceutical Chemistry, School of Pharmacy North‐West University Potchefstroom South Africa
| | - Jacobus P. Petzer
- Centre of Excellence for Pharmaceutical Sciences North‐West University Potchefstroom South Africa
- Pharmaceutical Chemistry, School of Pharmacy North‐West University Potchefstroom South Africa
| |
Collapse
|
32
|
Patel DV, Patel NR, Kanhed AM, Teli DM, Patel KB, Gandhi PM, Patel SP, Chaudhary BN, Shah DB, Prajapati NK, Patel KV, Yadav MR. Further Studies on Triazinoindoles as Potential Novel Multitarget-Directed Anti-Alzheimer's Agents. ACS Chem Neurosci 2020; 11:3557-3574. [PMID: 33073564 DOI: 10.1021/acschemneuro.0c00448] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The inadequate clinical efficacy of the present anti-Alzheimer's disease (AD) drugs and their low impact on the progression of Alzheimer's disease in patients have revised the research focus from single targets to multitarget-directed ligands. A novel series of substituted triazinoindole derivatives were obtained by introducing various substituents on the indole ring for the development of multitarget-directed ligands as anti-AD agents. The experimental data indicated that some of these compounds exhibited significant anti-AD properties. Among them, 8-(piperidin-1-yl)-N-(6-(pyrrolidin-1-yl)hexyl)-5H-[1,2,4]triazino[5,6-b]indol-3-amine (60), the most potent cholinesterase inhibitor (AChE, IC50 value of 0.32 μM; BuChE, IC50 value of 0.21 μM), was also found to possess significant self-mediated Aβ1-42 aggregation inhibitory activity (54% at 25 μM concentration). Additionally, compound 60 showed strong antioxidant activity. In the PAMPA assay, compound 60 exhibited blood-brain barrier penetrating ability. An acute toxicity study in rats demonstrated no sign of toxicity at doses up to 2000 mg/kg. Furthermore, compound 60 significantly restored the cognitive deficits in the scopolamine-induced mice model and Aβ1-42-induced rat model. In the in silico ADMET prediction studies, the compound satisfied all the parameters of CNS acting drugs. These results highlighted the potential of compound 60 to be a promising multitarget-directed ligand for the development of potential anti-AD drugs.
Collapse
Affiliation(s)
- Dushyant V. Patel
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, 390001 Gujarat, India
| | - Nirav R. Patel
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, 390001 Gujarat, India
| | - Ashish M. Kanhed
- Shobhaben Pratapbhai Patel - School of Pharmacy & Technology Management, SVKM’s NMIMS University, Vile Parle, Mumbai 400056, India
| | - Divya M. Teli
- Department of Pharmaceutical Chemistry, L. M. College of Pharmacy, Navrangpura, Ahmedabad, 380009 Gujarat, India
| | - Kishan B. Patel
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, 390001 Gujarat, India
| | - Pallav M. Gandhi
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, 390001 Gujarat, India
| | - Sagar P. Patel
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, 390001 Gujarat, India
| | - Bharat N. Chaudhary
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, 390001 Gujarat, India
| | - Dharti B. Shah
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, 390001 Gujarat, India
| | - Navnit K. Prajapati
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, 390001 Gujarat, India
| | - Kirti V. Patel
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, 390001 Gujarat, India
| | - Mange Ram Yadav
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, 390001 Gujarat, India
- Director (R & D), Centre of Research for Development, Parul University, Limbda, Waghodia Road, Vadodara, 391760 Gujarat, India
| |
Collapse
|
33
|
Kanhed AM, Patel DV, Patel NR, Sinha A, Thakor PS, Patel KB, Prajapati NK, Patel KV, Yadav MR. Indoloquinoxaline derivatives as promising multi-functional anti-Alzheimer agents. J Biomol Struct Dyn 2020; 40:2498-2515. [DOI: 10.1080/07391102.2020.1840441] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Ashish M. Kanhed
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, India
- Shobhaben Pratapbhai Patel - School of Pharmacy & Technology Management, SVKMs NMIMS University, Mumbai, India
| | - Dushyant V. Patel
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, India
| | - Nirav R. Patel
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, India
| | - Anshuman Sinha
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, India
| | - Priyanka S. Thakor
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, India
| | - Kishan B. Patel
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, India
| | - Navnit K. Prajapati
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, India
| | - Kirti V. Patel
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, India
| | - Mange Ram Yadav
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, India
| |
Collapse
|
34
|
Miles JA, Kapure JS, Deora GS, Courageux C, Igert A, Dias J, McGeary RP, Brazzolotto X, Ross BP. Rapid discovery of a selective butyrylcholinesterase inhibitor using structure-based virtual screening. Bioorg Med Chem Lett 2020; 30:127609. [PMID: 33039562 DOI: 10.1016/j.bmcl.2020.127609] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 09/26/2020] [Accepted: 10/05/2020] [Indexed: 02/08/2023]
Abstract
Acetylcholinesterase inhibitors are the mainstay of Alzheimer's disease treatments, despite having only short-term symptomatic benefits and severe side effects. Selective butyrylcholinesterase inhibitors (BuChEIs) may be more effective treatments in late-stage Alzheimer's disease with fewer side effects. Virtual screening is a powerful tool for identifying potential inhibitors in large digital compound databases. This study used structure-based virtual screening combined with physicochemical filtering to screen the InterBioScreen and Maybridge databases for novel selective BuChEIs. The workflow rapidly identified 22 potential hits in silico, resulting in the discovery of a human BuChEI with low-micromolar potency in vitro (IC50 2.4 µM) and high selectivity for butyrylcholinesterase over acetylcholinesterase. The compound was a rapidly reversible BuChEI with mixed-model in vitro inhibition kinetics. The binding interactions were investigated using in silico molecular dynamics and by developing structure-activity relationships using nine analogues. The compound also displayed high permeability in an in vitro model of the blood-brain barrier.
Collapse
Affiliation(s)
- Jared A Miles
- The University of Queensland, School of Pharmacy, Brisbane, Queensland 4072, Australia
| | - Jeevak S Kapure
- The University of Queensland, School of Pharmacy, Brisbane, Queensland 4072, Australia
| | - Girdhar Singh Deora
- The University of Queensland, School of Pharmacy, Brisbane, Queensland 4072, Australia; Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Charlotte Courageux
- Département de Toxicologie et Risques Chimiques, Institut de Recherche Biomédicale des Armées, 91223 Brétigny sur Orge, France
| | - Alexandre Igert
- Département de Toxicologie et Risques Chimiques, Institut de Recherche Biomédicale des Armées, 91223 Brétigny sur Orge, France
| | - José Dias
- Département de Toxicologie et Risques Chimiques, Institut de Recherche Biomédicale des Armées, 91223 Brétigny sur Orge, France
| | - Ross P McGeary
- The University of Queensland, School of Chemistry and Molecular Biosciences, Brisbane, Queensland 4072, Australia
| | - Xavier Brazzolotto
- Département de Toxicologie et Risques Chimiques, Institut de Recherche Biomédicale des Armées, 91223 Brétigny sur Orge, France
| | - Benjamin P Ross
- The University of Queensland, School of Pharmacy, Brisbane, Queensland 4072, Australia.
| |
Collapse
|
35
|
Zhang Z, Fan F, Luo W, Zhao Y, Wang C. Molecular Dynamics Revealing a Detour-Forward Release Mechanism of Tacrine: Implication for the Specific Binding Characteristics in Butyrylcholinesterase. Front Chem 2020; 8:730. [PMID: 33195011 PMCID: PMC7477934 DOI: 10.3389/fchem.2020.00730] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 07/14/2020] [Indexed: 01/29/2023] Open
Abstract
Butyrylcholinesterase (BChE) is a non-specific enzyme with clinical pharmacological and toxicological significance, which was a renewed interest as therapeutic target in Alzheimer's disease (AD) nowadays. Here, all-atom molecular dynamics simulations of butyrylcholinesterase with tacrine complex were designed to characterize inhibitor binding modes, strengths, and the hydrogen-bond dependent non-covalent release mechanism. Four possible release channels were identified, and the most favorable channel was determined by random acceleration molecular dynamics molecular dynamics (RAMD MD) simulations. The thermodynamic and dynamic properties as well as the corresponding Detour-forward delivery mechanism were determined according to the classical molecular dynamics (MD) simulations accompanied with umbrella sampling. The free energy barrier of the tacrine release process for the most beneficial pathway is about 10.95 kcal/mol, which is related to the non-covalent interactions from the surrounding residues, revealing the specific binding characteristics in the active site. The residues including Asp70, Ser79, Trp82, Gly116, Thr120, Tyr332, and His438 were identified to play major roles in the stabilization of tacrine in the pocket of BChE, where hydrogen bonding and π-π interactions are significant factors. Tyr332 and Asp70, which act as gate keepers, play crucial roles in the substrate delivery. The present results provide a basic understanding for the ligand transport mechanism depending on the BChE enzymatic environment, which is useful for the design of BChE inhibitors in the future.
Collapse
Affiliation(s)
- Zhiyang Zhang
- The Key Laboratory of Natural Medicine and Immuno-Engineering, Henan University, Kaifeng, China
| | - Fangfang Fan
- School of Biological and Chemical Engineering, Zhejiang University of Science and Technology, Hangzhou, China
| | - Wen Luo
- The Key Laboratory of Natural Medicine and Immuno-Engineering, Henan University, Kaifeng, China
| | - Yuan Zhao
- The Key Laboratory of Natural Medicine and Immuno-Engineering, Henan University, Kaifeng, China
| | - Chaojie Wang
- The Key Laboratory of Natural Medicine and Immuno-Engineering, Henan University, Kaifeng, China
| |
Collapse
|
36
|
Meden A, Knez D, Malikowska-Racia N, Brazzolotto X, Nachon F, Svete J, Sałat K, Grošelj U, Gobec S. Structure-activity relationship study of tryptophan-based butyrylcholinesterase inhibitors. Eur J Med Chem 2020; 208:112766. [PMID: 32919297 DOI: 10.1016/j.ejmech.2020.112766] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/14/2020] [Accepted: 08/15/2020] [Indexed: 12/19/2022]
Abstract
A series of tryptophan-based selective nanomolar butyrylcholinesterase (BChE) inhibitors was designed and synthesized. Compounds were optimized in terms of potency, selectivity, and synthetic accessibility. The crystal structure of the inhibitor 18 in complex with BChE revealed the molecular basis for its low nanomolar inhibition (IC50 = 2.8 nM). The favourable in vitro results enabled a first-in-animal in vivo efficacy and safety trial, which demonstrated a positive impact on fear-motivated and spatial long-term memory retrieval without any concomitant adverse motor effects. Altogether, this research culminated in a handful of new lead compounds with promising potential for symptomatic treatment of patients with Alzheimer's disease.
Collapse
Affiliation(s)
- Anže Meden
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva 7, SI-1000, Ljubljana, Slovenia
| | - Damijan Knez
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva 7, SI-1000, Ljubljana, Slovenia
| | - Natalia Malikowska-Racia
- Department of Pharmacodynamics, Chair of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna St., 30-688, Krakow, Poland
| | - Xavier Brazzolotto
- Département de Toxicologie et Risques Chimiques, Institut de Recherche Biomédicale des Armées, 91223, Brétigny sur Orge, France
| | - Florian Nachon
- Département de Toxicologie et Risques Chimiques, Institut de Recherche Biomédicale des Armées, 91223, Brétigny sur Orge, France
| | - Jurij Svete
- University of Ljubljana, Faculty of Chemistry and Chemical Technology, Večna Pot 113, SI-1000, Ljubljana, Slovenia
| | - Kinga Sałat
- Department of Pharmacodynamics, Chair of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna St., 30-688, Krakow, Poland
| | - Uroš Grošelj
- University of Ljubljana, Faculty of Chemistry and Chemical Technology, Večna Pot 113, SI-1000, Ljubljana, Slovenia.
| | - Stanislav Gobec
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva 7, SI-1000, Ljubljana, Slovenia.
| |
Collapse
|
37
|
Monoclonal antibodies to fetal bovine serum acetylcholinesterase distinguish between acetylcholinesterases from ruminant and non-ruminant species. Chem Biol Interact 2020; 330:109225. [PMID: 32795450 DOI: 10.1016/j.cbi.2020.109225] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 08/06/2020] [Accepted: 08/11/2020] [Indexed: 11/24/2022]
Abstract
Two types of cholinesterases (ChEs) are present in mammalian blood and tissues: acetylcholinesterase (AChE) and butyrylcholinesterase (BChE). While AChE regulates neurotransmission by hydrolyzing acetylcholine at the postsynaptic membranes and neuromuscular junctions, BChE in plasma has been suggested to be involved in detoxifying toxic compounds. This study was undertaken to establish the identity of circulating ChE activity in plasmas from domestic animals (bovine, ovine, caprine, porcine and equine) by assessing sensitivity to AChE-specific inhibitors (BW284c51 and edrophonium) and BChE-specific inhibitors (dibucaine, ethopropazine and Iso-OMPA) as well as binding to anti-FBS AChE monoclonal antibodies (MAbs). Based on the inhibition of ChE activity by ChE-specific inhibitors, it was determined that bovine, ovine and caprine plasma predominantly contain AChE, while porcine and equine plasma contain BChE. Three of the anti-FBS AChE MAbs, 4E5, 5E8 and 6H9, inhibited 85-98% of enzyme activity in bovine, ovine and caprine plasma, confirming that the esterase in these plasmas was AChE. These MAbs did not bind to purified recombinant human or mouse AChE, demonstrating that these MAbs were specific for AChEs from ruminant species. These MAbs did not inhibit the activity of purified human BChE, or ChE activity in porcine and equine plasma, confirming that the ChE in these plasmas was BChE. Taken together, these results demonstrate that anti-FBS AChE MAbs can serve as useful tools for distinguishing between AChEs from ruminant and non-ruminant species and BChEs.
Collapse
|
38
|
Forebrain Cholinergic Signaling: Wired and Phasic, Not Tonic, and Causing Behavior. J Neurosci 2020; 40:712-719. [PMID: 31969489 DOI: 10.1523/jneurosci.1305-19.2019] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 10/29/2019] [Accepted: 11/04/2019] [Indexed: 01/21/2023] Open
Abstract
Conceptualizations of cholinergic signaling as primarily spatially diffuse and slow-acting are based largely on measures of extracellular brain ACh levels that require several minutes to generate a single data point. In addition, most such studies inhibited the highly potent catalytic enzyme for ACh, AChE, to facilitate measurement of ACh. Absent such inhibition, AChE limits the presence of ambient ACh and thus renders it unlikely that ACh influences target regions via slow changes in extracellular ACh concentrations. We describe an alternative view by which forebrain signaling in cortex driving cognition is largely phasic (milliseconds to perhaps seconds), and unlikely to be volume-transmitted. This alternative is supported by new evidence from real-time amperometric recordings of cholinergic signaling indicating a specific function of rapid, phasic, transient cholinergic signaling in attentional contexts. Previous neurochemical evidence may be reinterpreted in terms of integrated phasic cholinergic activity that mediates specific behavioral and cognitive operations; this reinterpretation fits well with recent computational models. Optogenetic studies support a causal relationship between cholinergic transients and behavior. This occurs in part via transient-evoked muscarinic receptor-mediated high-frequency oscillations in cortical regions. Such oscillations outlast cholinergic transients and thus link transient ACh signaling with more sustained postsynaptic activity patterns to support relatively persistent attentional biases. Reconceptualizing cholinergic function as spatially specific, phasic, and modulating specific cognitive operations is theoretically powerful and may lead to pharmacologic treatments more effective than those based on traditional views.Dual Perspectives Companion Paper: Diverse Spatiotemporal Scales of Cholinergic Signaling in the Neocortex, by Anita A. Disney and Michael J. Higley.
Collapse
|
39
|
Computational exploration and experimental validation to identify a dual inhibitor of cholinesterase and amyloid-beta for the treatment of Alzheimer’s disease. J Comput Aided Mol Des 2020; 34:983-1002. [DOI: 10.1007/s10822-020-00318-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 05/26/2020] [Indexed: 12/15/2022]
|
40
|
Patel DV, Patel NR, Kanhed AM, Teli DM, Patel KB, Joshi PD, Patel SP, Gandhi PM, Chaudhary BN, Prajapati NK, Patel KV, Yadav MR. Novel carbazole-stilbene hybrids as multifunctional anti-Alzheimer agents. Bioorg Chem 2020; 101:103977. [PMID: 32485470 DOI: 10.1016/j.bioorg.2020.103977] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 05/20/2020] [Accepted: 05/24/2020] [Indexed: 01/03/2023]
Abstract
Molecules capable of engaging with multiple targets associated with pathological condition of Alzheimer's disease have proved to be potential anti-Alzheimer's agents. In our goal to develop multitarget-directed ligands for the treatment of Alzheimer's disease, a novel series of carbazole-based stilbene derivatives were designed by the fusion of carbazole ring with stilbene scaffold. The designed compounds were synthesized and evaluated for their anti-AD activities including cholinesterase inhibition, Aβ aggregation inhibition, antioxidant and metal chelation properties. Amongst them, (E)-1-(4-(2-(9-ethyl-9H-carbazol-3-yl)vinyl)phenyl)-3-(2-(pyrrolidin-1-yl)ethyl)thiourea (50) appeared to be the best candidate with good inhibitory activities against AChE (IC50 value of 2.64 μM) and BuChE (IC50 value of 1.29 μM), and significant inhibition of self-mediated Aβ1-42 aggregation (51.29% at 25 μM concentration). The metal chelation study showed that compound (50) possessed specific copper ion chelating property. Additionally, compound (50) exhibited moderate antioxidant activity. To understand the binding mode of 50, molecular docking studies were performed, and the results indicated strong non-covalent interactions of 50 with the enzymes in the active sites of AChE, BuChE as well as of the Aβ1-42 peptide. Additionally, it showed promising in silico ADMET properties. Putting together, these findings evidently showed compound (50) as a potential multitarget-directed ligand in the course of developing novel anti-AD drugs.
Collapse
Affiliation(s)
- Dushyant V Patel
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara 390001, Gujarat, India
| | - Nirav R Patel
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara 390001, Gujarat, India
| | - Ashish M Kanhed
- Shobhaben Pratapbhai Patel - School of Pharmacy & Technology Management, SVKM's NMIMS University, Vile Parle, Mumbai 400056, India
| | - Divya M Teli
- Department of Pharmaceutical Chemistry, L. M. College of Pharmacy, Navrangpura, Ahmedabad 380009, Gujarat, India
| | - Kishan B Patel
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara 390001, Gujarat, India
| | - Prashant D Joshi
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara 390001, Gujarat, India
| | - Sagar P Patel
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara 390001, Gujarat, India
| | - Pallav M Gandhi
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara 390001, Gujarat, India
| | - Bharat N Chaudhary
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara 390001, Gujarat, India
| | - Navnit K Prajapati
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara 390001, Gujarat, India
| | - Kirti V Patel
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara 390001, Gujarat, India
| | - Mange Ram Yadav
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara 390001, Gujarat, India.
| |
Collapse
|
41
|
Enderlin J, Igert A, Auvin S, Nachon F, Dal Bo G, Dupuis N. Characterization of organophosphate-induced brain injuries in a convulsive mouse model of diisopropylfluorophosphate exposure. Epilepsia 2020; 61:e54-e59. [PMID: 32359085 DOI: 10.1111/epi.16516] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 04/05/2020] [Accepted: 04/06/2020] [Indexed: 11/30/2022]
Abstract
Organophosphate (OP) compounds constitute a class of highly toxic molecules, characterized by irreversible cholinesterase (ChE) inhibition. Being either pesticides or chemical warfare agents, they present a major health issue in some countries, as well as a terrorist or military threat. Prompted by the need for suitable animal models to test novel medical countermeasures, we developed a new convulsive mouse model of OP poisoning using diisopropylfluorophosphate (DFP). Using electrocorticography (ECoG), we analyzed seizure and status epilepticus (SE) occurrences, as well as relative power of ECoG frequency band modifications after DFP injection in male Swiss mice. Next, we investigated DFP effect on ChE inhibition. Histological changes on neuronal activity and neuronal damage were examined by c-Fos immunolabeling and Fluoro-Jade C staining. We showed that mice exposed to DFP presented electrocorticographic seizures that rapidly progressed to SE within 20 minutes. Lasting >8 hours, DFP-induced SE was associated with major power spectrum modifications in seizing DFP animals compared to control animals. Seizures and SE development were concomitant with profound ChE inhibition and induced massive neuronal degeneration. Presenting all hallmarks of convulsive OP poisoning, we showed that our mouse model is valuable for studying pathophysiological mechanisms and preclinical testing of newly available therapeutic molecules.
Collapse
Affiliation(s)
- Julie Enderlin
- NeuroDiderot, Inserm U1141, Sorbonne Paris Cité, Paris Diderot University, Paris, France.,Pediatric Neurology Department, Robert Debré Hospital, Public Hospital Network of Paris, Paris, France
| | - Alexandre Igert
- Department of Toxicology and Chemical Risks, Institute for Biomedical Research of the Armed Forces, Brétigny sur Orge, France
| | - Stéphane Auvin
- NeuroDiderot, Inserm U1141, Sorbonne Paris Cité, Paris Diderot University, Paris, France.,Pediatric Neurology Department, Robert Debré Hospital, Public Hospital Network of Paris, Paris, France
| | - Florian Nachon
- Department of Toxicology and Chemical Risks, Institute for Biomedical Research of the Armed Forces, Brétigny sur Orge, France
| | - Grégory Dal Bo
- Department of Toxicology and Chemical Risks, Institute for Biomedical Research of the Armed Forces, Brétigny sur Orge, France
| | - Nina Dupuis
- Department of Toxicology and Chemical Risks, Institute for Biomedical Research of the Armed Forces, Brétigny sur Orge, France
| |
Collapse
|
42
|
Ha ZY, Mathew S, Yeong KY. Butyrylcholinesterase: A Multifaceted Pharmacological Target and Tool. Curr Protein Pept Sci 2020; 21:99-109. [DOI: 10.2174/1389203720666191107094949] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Revised: 09/10/2019] [Accepted: 09/24/2019] [Indexed: 12/22/2022]
Abstract
Butyrylcholinesterase is a serine hydrolase that catalyzes the hydrolysis of esters in the body. Unlike its sister enzyme acetylcholinesterase, butyrylcholinesterase has a broad substrate scope and lower acetylcholine catalytic efficiency. The difference in tissue distribution and inhibitor sensitivity also points to its involvement external to cholinergic neurotransmission. Initial studies on butyrylcholinesterase showed that the inhibition of the enzyme led to the increment of brain acetylcholine levels. Further gene knockout studies suggested its involvement in the regulation of amyloid-beta, a brain pathogenic protein. Thus, it is an interesting target for neurological disorders such as Alzheimer’s disease. The substrate scope of butyrylcholinesterase was recently found to include cocaine, as well as ghrelin, the “hunger hormone”. These findings led to the development of recombinant butyrylcholinesterase mutants and viral gene therapy to combat cocaine addiction, along with in-depth studies on the significance of butyrylcholinesterase in obesity. It is observed that the pharmacological impact of butyrylcholinesterase increased in tandem with each reported finding. Not only is the enzyme now considered an important pharmacological target, it is also becoming an important tool to study the biological pathways in various diseases. Here, we review and summarize the biochemical properties of butyrylcholinesterase and its roles, as a cholinergic neurotransmitter, in various diseases, particularly neurodegenerative disorders.
Collapse
Affiliation(s)
- Zhe Ying Ha
- School of Science, Monash University Malaysia Campus, Jalan Lagoon Selatan, Bandar Sunway, 47500, Selangor, Malaysia
| | - Shintu Mathew
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), ITI Compound, Raebareli, 229010, India
| | - Keng Yoon Yeong
- School of Science, Monash University Malaysia Campus, Jalan Lagoon Selatan, Bandar Sunway, 47500, Selangor, Malaysia
| |
Collapse
|
43
|
Exploring Biological Activity of 4-Oxo-4 H-furo[2,3- h]chromene Derivatives as Potential Multi-Target-Directed Ligands Inhibiting Cholinesterases, β-Secretase, Cyclooxygenase-2, and Lipoxygenase-5/15. Biomolecules 2019; 9:biom9110736. [PMID: 31766252 PMCID: PMC6920776 DOI: 10.3390/biom9110736] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 11/06/2019] [Accepted: 11/07/2019] [Indexed: 12/12/2022] Open
Abstract
A series of 5-oxo-5H-furo[3,2-g]chromene-6-carbaldehydes and their hydrazone derivatives were evaluated as potential multi-target-directed ligands in vitro against cholinesterases, β-secretase, cyclooxygenase-2, and lipoxygenase-15 (LOX-15), as well as for free radical-scavenging activities. The most active compounds against LOX-15 were also evaluated for activity against the human lipoxygenase-5 (LOX-5). Kinetic studies against AChE, BChE, and β-secretase (BACE-1) were performed on 2-(3-fluorophenyl)- (3b) and 2-(4-chlorophenyl)-6-[(4-trifluoromethylphenyl)hydrazonomethyl]furo[3,2-h]chromen-5-one (3e) complemented with molecular docking (in silico) to determine plausible protein-ligand interactions on a molecular level. The docking studies revealed hydrogen and/or halogen bonding interactions between the strong electron-withdrawing fluorine atoms of the trifluoromethyl group with several residues of the enzyme targets, which are probably responsible for the observed increased biological activity of these hydrazone derivatives. The two compounds were found to moderately inhibit COX-2 and lipoxygenases (LOX-5 and LOX-15). Compounds 3b and 3e were also evaluated for cytotoxicity against the breast cancer MCF-7 cell line and Hek293-T cells.
Collapse
|
44
|
Scheiner M, Dolles D, Gunesch S, Hoffmann M, Nabissi M, Marinelli O, Naldi M, Bartolini M, Petralla S, Poeta E, Monti B, Falkeis C, Vieth M, Hübner H, Gmeiner P, Maitra R, Maurice T, Decker M. Dual-Acting Cholinesterase-Human Cannabinoid Receptor 2 Ligands Show Pronounced Neuroprotection in Vitro and Overadditive and Disease-Modifying Neuroprotective Effects in Vivo. J Med Chem 2019; 62:9078-9102. [PMID: 31609608 PMCID: PMC7640639 DOI: 10.1021/acs.jmedchem.9b00623] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
We have designed and synthesized a series of 14 hybrid molecules out of the cholinesterase (ChE) inhibitor tacrine and a benzimidazole-based human cannabinoid receptor subtype 2 (hCB2R) agonist and investigated them in vitro and in vivo. The compounds are potent ChE inhibitors, and for the most promising hybrids, the mechanism of human acetylcholinesterase (hAChE) inhibition as well as their ability to interfere with AChE-induced aggregation of β-amyloid (Aβ), and Aβ self-aggregation was assessed. All hybrids were evaluated for affinity and selectivity for hCB1R and hCB2R. To ensure that the hybrids retained their agonist character, the expression of cAMP-regulated genes was quantified, and potency and efficacy were determined. Additionally, the effects of the hybrids on microglia activation and neuroprotection on HT-22 cells were investigated. The most promising in vitro hybrids showed pronounced neuroprotection in an Alzheimer's mouse model at low dosage (0.1 mg/kg, i.p.), lacking hepatotoxicity even at high dose (3 mg/kg, i.p.).
Collapse
Affiliation(s)
- Matthias Scheiner
- Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy and Food Chemistry, Julius Maximilian University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Dominik Dolles
- Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy and Food Chemistry, Julius Maximilian University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Sandra Gunesch
- Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy and Food Chemistry, Julius Maximilian University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Matthias Hoffmann
- Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy and Food Chemistry, Julius Maximilian University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Massimo Nabissi
- School of Pharmacy, University of Camerino, Via Madonna delle Carceri 9, 62032 Camerino, Italy
| | - Oliviero Marinelli
- School of Pharmacy, University of Camerino, Via Madonna delle Carceri 9, 62032 Camerino, Italy
| | - Marina Naldi
- Department of Pharmacy and Biotechnology, University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Manuela Bartolini
- Department of Pharmacy and Biotechnology, University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Sabrina Petralla
- Department of Pharmacy and Biotechnology, University of Bologna, Via Selmi 3, 40126 Bologna, Italy
| | - Eleonora Poeta
- Department of Pharmacy and Biotechnology, University of Bologna, Via Selmi 3, 40126 Bologna, Italy
| | - Barbara Monti
- Department of Pharmacy and Biotechnology, University of Bologna, Via Selmi 3, 40126 Bologna, Italy
| | - Christina Falkeis
- Pathology, Clinical Center Bayreuth, Preuschwitzer Straße 101, 95445 Bayreuth, Germany
| | - Michael Vieth
- Pathology, Clinical Center Bayreuth, Preuschwitzer Straße 101, 95445 Bayreuth, Germany
| | - Harald Hübner
- Medicinal Chemistry, Department of Chemistry and Pharmacy, Friedrich-Alexander University Erlangen-Nürnberg, Schuhstraße 19, 91052 Erlangen, Germany
| | - Peter Gmeiner
- Medicinal Chemistry, Department of Chemistry and Pharmacy, Friedrich-Alexander University Erlangen-Nürnberg, Schuhstraße 19, 91052 Erlangen, Germany
| | - Rangan Maitra
- Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, North Carolina 27709, United States
| | - Tangui Maurice
- MMDN, University of Montpellier, INSERM, EPHE, UMR-S1198, 34095 Montpellier, France
| | - Michael Decker
- Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy and Food Chemistry, Julius Maximilian University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| |
Collapse
|
45
|
Jasiecki J, Wasąg B. Butyrylcholinesterase Protein Ends in the Pathogenesis of Alzheimer's Disease-Could BCHE Genotyping Be Helpful in Alzheimer's Therapy? Biomolecules 2019; 9:biom9100592. [PMID: 31601022 PMCID: PMC6843418 DOI: 10.3390/biom9100592] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 10/04/2019] [Accepted: 10/08/2019] [Indexed: 01/19/2023] Open
Abstract
Late-onset Alzheimer’s disease (AD) is clinically characterized by a progressive decline of memory and other cognitive functions leading to the loss of the ability to perform everyday activities. Only a few drugs have been approved to treat AD dementia over the past century since the first AD patient was diagnosed. Drugs increasing the availability of neurotransmitters at synapses in the brain are used clinically in the treatment of AD dementia, and cholinesterase inhibitors (ChEIs) are the mainstay of the therapy. A detrimental effect on cognitive function has been reported in patients with pharmacological inhibition of acetylcholinesterase (AChE) by ChEIs and reduced butyrylcholinesterase (BChE) activity due to the single nucleotide polymorphisms. The BChE K-variant (rs1803274), the most common genetic variant of the BCHE gene, was thought to reduce enzyme activity reflecting the lower clinical response to rivastigmine in AD patients. During ChEIs therapy, patients carrying reduced-activity BChE do not present such improved attention like patients with the wild-type enzyme. On the other hand, alterations in the BCHE gene causing enzyme activity reduction may delay AD onset in patients at risk by preserving the level of cortical acetylcholine (ACh). Based on our previous results, we conclude that SNPs localized outside of the coding sequence, in 5’UTR (rs1126680) and/or intron 2 (rs55781031) of the BCHE gene, but not solely K-variant alteration (p.A539T) itself, are responsible for reduced enzyme activity. Therefore, we suspect that not BChE-K itself, but these coexisting SNPs (rs1126680 and rs55781031), could be associated with deleterious changes in cognitive decline in patients treated with ChEIs. Based on the results, we suggest that SNPs (rs1126680) and/or (rs55781031) genotyping should be performed to identify subjects at risk for lowered efficacy ChEIs therapy, and such patients should be treated with a lower rivastigmine dosage. Finally, our sequence analysis of the N-terminal end of N-BChE revealed evolutionarily conserved amino acid residues that can be involved in disulfide bond formation and anchoring of N-BChE in the cell membrane.
Collapse
Affiliation(s)
- Jacek Jasiecki
- Faculty of Pharmacy with Subfaculty of Laboratory Medicine, Medical University of Gdańsk, 80-416 Gdańsk, Poland
- Correspondence: ; Tel.: +48-58-3491972
| | - Bartosz Wasąg
- Department of Biology and Medical Genetics, Medical University of Gdańsk, 80-211 Gdańsk, Poland;
- Laboratory of Clinical Genetics, University Clinical Centre, 80-952 Gdańsk, Poland
| |
Collapse
|
46
|
Sinclair LI, Kumar A, Darreh-Shori T, Love S. Visual hallucinations in Alzheimer's disease do not seem to be associated with chronic hypoperfusion of to visual processing areas V2 and V3 but may be associated with reduced cholinergic input to these areas. ALZHEIMERS RESEARCH & THERAPY 2019; 11:80. [PMID: 31511061 PMCID: PMC6740037 DOI: 10.1186/s13195-019-0519-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 07/08/2019] [Indexed: 12/31/2022]
Abstract
Background Up to 20% of patients with AD experience hallucinations. The pathological substrate is not known. Visual hallucinations (VH) are more common in dementia with Lewy bodies (DLB). In autopsy studies, up to 60% of patients with AD have concomitant Lewy body pathology. Decreased perfusion of the occipital lobe has been implicated in DLB patients with VH, and post-mortem studies point to both decreased cholinergic activity and reduced oxygenation of the occipital cortex in DLB. Methods We used biochemical methods to assess microvessel density (level of von Willebrand factor, a marker of endothelial cell content), ante-mortem oxygenation (vascular endothelial growth factor, a marker of tissue hypoxia; myelin-associated glycoprotein to proteolipid protein-1 ratio, a measure of tissue oxygenation relative to metabolic demand), cholinergic innervation (acetylcholinesterase and choline acetyltransferase), butyrylcholinesterase and insoluble α-synuclein content in the BA18 and BA19 occipital cortex obtained post-mortem from 23 AD patients who had experienced visual hallucinations, 19 AD patients without hallucinations, 19 DLB patients, and 36 controls. The cohorts were matched for age, gender and post-mortem interval. Results There was no evidence of reduced microvessel density, hypoperfusion or reduction in ChAT activity in AD with visual hallucinations. Acetylcholinesterase activity was reduced in both BA18 and BA19, in all 3 dementia groups, and the concentration was also reduced in BA19 in the DLB and AD without visual hallucinations groups. Insoluble α-synuclein was raised in the DLB group in both areas but not in AD either with or without visual hallucinations. Conclusions Our results suggest that visual hallucinations in AD are associated with cholinergic denervation rather than chronic hypoperfusion or α-synuclein accumulation in visual processing areas of the occipital cortex. Electronic supplementary material The online version of this article (10.1186/s13195-019-0519-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Lindsey Isla Sinclair
- Population Health Sciences, Oakfield House, University of Bristol, Clifton, Bristol, BS8 2BN, UK. .,Translational Health Sciences, Level 1 Learning & Research Building, Southmead Hospital, University of Bristol, Bristol, BS10 5NB, UK.
| | - Amit Kumar
- Division of Clinical Geriatrics, NEO Plan 7, Department of Neurobiology, Care Sciences and Society (NVS), H1, 141 52, Huddinge, Sweden
| | - Taher Darreh-Shori
- Division of Clinical Geriatrics, NEO Plan 7, Department of Neurobiology, Care Sciences and Society (NVS), H1, 141 52, Huddinge, Sweden
| | - Seth Love
- Translational Health Sciences, Level 1 Learning & Research Building, Southmead Hospital, University of Bristol, Bristol, BS10 5NB, UK
| |
Collapse
|
47
|
Novel 12-hydroxydehydroabietylamine derivatives act as potent and selective butyrylcholinesterase inhibitors. Bioorg Chem 2019; 90:103092. [DOI: 10.1016/j.bioorg.2019.103092] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Revised: 06/23/2019] [Accepted: 06/26/2019] [Indexed: 01/08/2023]
|
48
|
Patel DV, Patel NR, Kanhed AM, Patel SP, Sinha A, Kansara DD, Mecwan AR, Patel SB, Upadhyay PN, Patel KB, Shah DB, Prajapati NK, Murumkar PR, Patel KV, Yadav MR. Novel Multitarget Directed Triazinoindole Derivatives as Anti-Alzheimer Agents. ACS Chem Neurosci 2019; 10:3635-3661. [PMID: 31310717 DOI: 10.1021/acschemneuro.9b00226] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The multifaceted nature of Alzheimer's disease (AD) demands treatment with multitarget-directed ligands (MTDLs) to confront the key pathological aberrations. A novel series of triazinoindole derivatives were designed and synthesized. In vitro studies revealed that all the compounds showed moderate to good anticholinesterase activity; the most active compound 23e showed an IC50 value of 0.56 ± 0.02 μM for AChE and an IC50 value of 1.17 ± 0.09 μM for BuChE. These derivatives are also endowed with potent antioxidant activity. To understand the plausible binding mode of the compound 23e, molecular docking studies and molecular dynamics simulation studies were performed, and the results indicated significant interactions of 23e within the active sites of AChE as well as BuChE. Compound 23e successfully diminished H2O2-induced oxidative stress in SH-SY5Y cells and displayed excellent neuroprotective activity against H2O2 as well as Aβ-induced toxicity in SH-SY5Y cells in a concentration dependent manner. Furthermore, it did not show any significant toxicity in neuronal SH-SY5Y cells in the cytotoxicity assay. Compound 23e did not show any acute toxicity in rats at doses up to 2000 mg/kg, and it significantly reversed scopolamine-induced memory deficit in mice model. Additionally, compound 23e showed notable in silico ADMET properties. Taken collectively, these findings project compound 23e as a potential balanced MTDL in the evolution process of novel anti-AD drugs.
Collapse
Affiliation(s)
- Dushyant V. Patel
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara-390001 Gujarat, India
| | - Nirav R. Patel
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara-390001 Gujarat, India
| | - Ashish M. Kanhed
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara-390001 Gujarat, India
| | - Sagar P. Patel
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara-390001 Gujarat, India
| | - Anshuman Sinha
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara-390001 Gujarat, India
| | - Deep D. Kansara
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara-390001 Gujarat, India
| | - Annie R. Mecwan
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara-390001 Gujarat, India
| | - Sarvangee B. Patel
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara-390001 Gujarat, India
| | - Pragnesh N. Upadhyay
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara-390001 Gujarat, India
| | - Kishan B. Patel
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara-390001 Gujarat, India
| | - Dharti B. Shah
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara-390001 Gujarat, India
| | - Navnit K. Prajapati
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara-390001 Gujarat, India
| | - Prashant R. Murumkar
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara-390001 Gujarat, India
| | - Kirti V. Patel
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara-390001 Gujarat, India
| | - Mange Ram Yadav
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara-390001 Gujarat, India
| |
Collapse
|
49
|
Overview of novel multifunctional agents based on conjugates of γ-carbolines, carbazoles, tetrahydrocarbazoles, phenothiazines, and aminoadamantanes for treatment of Alzheimer's disease. Chem Biol Interact 2019; 308:224-234. [DOI: 10.1016/j.cbi.2019.05.020] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 04/25/2019] [Accepted: 05/13/2019] [Indexed: 01/10/2023]
|
50
|
Zhou S, Yuan Y, Zheng F, Zhan CG. Structure-based virtual screening leading to discovery of highly selective butyrylcholinesterase inhibitors with solanaceous alkaloid scaffolds. Chem Biol Interact 2019; 308:372-376. [PMID: 31152736 PMCID: PMC6613991 DOI: 10.1016/j.cbi.2019.05.051] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 05/08/2019] [Accepted: 05/29/2019] [Indexed: 11/29/2022]
Abstract
According to recent research advance, it is interesting to identify new, potent and selective inhibitors of human butyrylcholinesterase (BChE) for therapeutic treatment of both the Alzheimer's disease (AD) and heroin abuse. In this study, we carried out a structure-based virtual screening followed by in vitro activity assays, with the goal to identify new inhibitors that are selective for BChE over acetylcholinesterase (AChE). As a result, a set of new, selective inhibitors of human BChE were identified from natural products with solanaceous alkaloid scaffolds. The most active one of the natural products (compound 1) identified has an IC50 of 16.8 nM against BChE. It has been demonstrated that the desirable selectivity of these inhibitors for BChE over AChE is mainly controlled by three key residues in the active site cavity, i.e. residues Q119, A277, and A328 in BChE versus the respective residues Y124, W286, and Y337 in AChE. Based on this structural insight, future rational design of new, potent and selective BChE inhibitors may focus on these key structural differences in the active site cavity.
Collapse
Affiliation(s)
- Shuo Zhou
- Molecular Modeling and Biopharmaceutical Center, 789 South Limestone Street, Lexington, KY, 40536, USA; Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY, 40536, USA
| | - Yaxia Yuan
- Molecular Modeling and Biopharmaceutical Center, 789 South Limestone Street, Lexington, KY, 40536, USA; Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY, 40536, USA
| | - Fang Zheng
- Molecular Modeling and Biopharmaceutical Center, 789 South Limestone Street, Lexington, KY, 40536, USA; Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY, 40536, USA.
| | - Chang-Guo Zhan
- Molecular Modeling and Biopharmaceutical Center, 789 South Limestone Street, Lexington, KY, 40536, USA; Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY, 40536, USA.
| |
Collapse
|