1
|
La Spada G, Miniero DV, Rullo M, Cipolloni M, Delre P, Colliva C, Colella M, Leonetti F, Liuzzi GM, Mangiatordi GF, Giacchè N, Pisani L. Structure-based design of multitargeting ChEs-MAO B inhibitors based on phenyl ring bioisosteres: AChE/BChE selectivity switch and drug-like characterization. Eur J Med Chem 2024; 274:116511. [PMID: 38820854 DOI: 10.1016/j.ejmech.2024.116511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 05/06/2024] [Accepted: 05/17/2024] [Indexed: 06/02/2024]
Abstract
A structure-based drug design approach was focused on incorporating phenyl ring heterocyclic bioisosteres into coumarin derivative 1, previously reported as potent dual AChE-MAO B inhibitor, with the aim of improving drug-like features. Structure-activity relationships highlighted that bioisosteric rings were tolerated by hMAO B enzymatic cleft more than hAChE. Interestingly, linker homologation at the basic nitrogen enabled selectivity to switch from hAChE to hBChE. In the present work, we identified thiophene-based isosteres 7 and 15 as dual AChE-MAO B (IC50 = 261 and 15 nM, respectively) and BChE-MAO B (IC50 = 375 and 20 nM, respectively) inhibitors, respectively. Both 7 and 15 were moderately water-soluble and membrane-permeant agents by passive diffusion (PAMPA-HDM). Moreover, they were able to counteract oxidative damage induced by both H2O2 and 6-OHDA in SH-SY5Y cells and predicted to penetrate into CNS in a cell-based model mimicking blood-brain barrier. Molecular dynamics (MD) simulations shed light on key differences in AChE and BChE recognition processes promoted by the basic chain homologation from 7 to 15.
Collapse
Affiliation(s)
- Gabriella La Spada
- Dept. of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, via E. Orabona 4, 70125, Bari, Italy
| | - Daniela Valeria Miniero
- Dept. of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, Via E. Orabona 4, 70125, Bari, Italy
| | - Mariagrazia Rullo
- Dept. of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, via E. Orabona 4, 70125, Bari, Italy
| | - Marco Cipolloni
- Tes Pharma s.r.l., via Palmiro Togliatti 20, 06073, Corciano, PG, Italy
| | - Pietro Delre
- CNR, Institute of Crystallography, 70126, Bari, Italy
| | - Carolina Colliva
- Tes Pharma s.r.l., via Palmiro Togliatti 20, 06073, Corciano, PG, Italy
| | - Marco Colella
- Dept. of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, via E. Orabona 4, 70125, Bari, Italy
| | - Francesco Leonetti
- Dept. of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, via E. Orabona 4, 70125, Bari, Italy
| | - Grazia Maria Liuzzi
- Dept. of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, Via E. Orabona 4, 70125, Bari, Italy
| | | | - Nicola Giacchè
- Tes Pharma s.r.l., via Palmiro Togliatti 20, 06073, Corciano, PG, Italy
| | - Leonardo Pisani
- Dept. of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, via E. Orabona 4, 70125, Bari, Italy.
| |
Collapse
|
2
|
Umar M, Rehman Y, Ambreen S, Mumtaz SM, Shaququzzaman M, Alam MM, Ali R. Innovative approaches to Alzheimer's therapy: Harnessing the power of heterocycles, oxidative stress management, and nanomaterial drug delivery system. Ageing Res Rev 2024; 97:102298. [PMID: 38604453 DOI: 10.1016/j.arr.2024.102298] [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: 02/01/2024] [Revised: 03/10/2024] [Accepted: 04/05/2024] [Indexed: 04/13/2024]
Abstract
Alzheimer's disease (AD) presents a complex pathology involving amyloidogenic proteolysis, neuroinflammation, mitochondrial dysfunction, and cholinergic deficits. Oxidative stress exacerbates AD progression through pathways like macromolecular peroxidation, mitochondrial dysfunction, and metal ion redox potential alteration linked to amyloid-beta (Aβ). Despite limited approved medications, heterocyclic compounds have emerged as promising candidates in AD drug discovery. This review highlights recent advancements in synthetic heterocyclic compounds targeting oxidative stress, mitochondrial dysfunction, and neuroinflammation in AD. Additionally, it explores the potential of nanomaterial-based drug delivery systems to overcome challenges in AD treatment. Nanoparticles with heterocyclic scaffolds, like polysorbate 80-coated PLGA and Resveratrol-loaded nano-selenium, show improved brain transport and efficacy. Micellar CAPE and Melatonin-loaded nano-capsules exhibit enhanced antioxidant properties, while a tetra hydroacridine derivative (CHDA) combined with nano-radiogold particles demonstrates promising acetylcholinesterase inhibition without toxicity. This comprehensive review underscores the potential of nanotechnology-driven drug delivery for optimizing the therapeutic outcomes of novel synthetic heterocyclic compounds in AD management. Furthermore, the inclusion of various promising heterocyclic compounds with detailed ADMET (Absorption, Distribution, Metabolism, Excretion, and Toxicity) data provides valuable insights for planning the development of novel drug delivery treatments for AD.
Collapse
Affiliation(s)
- Mohammad Umar
- Department of Pharmaceutical Chemistry, Delhi Pharmaceutical Sciences and Research University, Pushp Vihar, New Delhi 110017, India
| | - Yasir Rehman
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, Hamdard Nagar, New Delhi 110062, India
| | - Subiya Ambreen
- Department of Pharmaceutical Chemistry, Delhi Pharmaceutical Sciences and Research University, Pushp Vihar, New Delhi 110017, India
| | - Sayed Md Mumtaz
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, Hamdard Nagar, New Delhi 110062, India
| | - Mohd Shaququzzaman
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, Hamdard Nagar, New Delhi 110062, India
| | - Mohammad Mumtaz Alam
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, Hamdard Nagar, New Delhi 110062, India
| | - Ruhi Ali
- Department of Pharmaceutical Chemistry, Delhi Pharmaceutical Sciences and Research University, Pushp Vihar, New Delhi 110017, India.
| |
Collapse
|
3
|
Kumar S, Jaiswal S, Gupta SK, Ayyannan SR. Benzimidazole-derived carbohydrazones as dual monoamine oxidases and acetylcholinesterase inhibitors: design, synthesis, and evaluation. J Biomol Struct Dyn 2024; 42:4710-4729. [PMID: 37345530 DOI: 10.1080/07391102.2023.2224887] [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: 04/26/2023] [Accepted: 06/01/2023] [Indexed: 06/23/2023]
Abstract
A series of novel benzimidazole-derived carbohydrazones was designed, synthesized and evaluated for their dual inhibition potential against monoamine oxidases (MAOs) and acetylcholinesterase (AChE) using multitarget-directed ligand approach (MTDL). The investigated compounds have exhibited moderate to excellent in vitro MAOs/AChE inhibitory activity at micromolar to nanomolar concentrations. Compound 12, 2-(1H-Benzo[d]imidazol-1-yl)-N'-[1-(4-hydroxyphenyl) ethylidene]acetohydrazide has emerged as a lead dual MAO-AChE inhibitor by exhibiting superior multi-target activity profile against MAO-A (IC50 = 0.067 ± 0.018 µM), MAO-B (IC50 = 0.029 ± 0.005 µM) and AChE (IC50 = 1.37 ± 0.026 µM). SAR studies suggest that the site A (hydrophobic ring) and site C (semicarbazone linker) modifications attempted on the semicarbazone-based MTDL resulted in a significant enhancement in the MAO-A/B inhibitory potential and a drastic decrease in the AChE inhibitory activity. Further, molecular docking and dynamics simulation experiments disclosed the possible molecular interactions of inhibitors inside the active site of respective enzymes. Also, computational prediction of drug-likeness and ADME parameters of test compounds revealed their drug-like characteristics.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Sandeep Kumar
- Pharmaceutical Chemistry Research Laboratory II, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, India
| | - Shivani Jaiswal
- Pharmaceutical Chemistry Research Laboratory II, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, India
| | - Sukesh Kumar Gupta
- Neurotherapeutics Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi, India
| | - Senthil Raja Ayyannan
- Pharmaceutical Chemistry Research Laboratory II, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, India
| |
Collapse
|
4
|
de Sena Murteira Pinheiro P, Franco LS, Montagnoli TL, Fraga CAM. Molecular hybridization: a powerful tool for multitarget drug discovery. Expert Opin Drug Discov 2024; 19:451-470. [PMID: 38456452 DOI: 10.1080/17460441.2024.2322990] [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: 10/24/2023] [Accepted: 02/21/2024] [Indexed: 03/09/2024]
Abstract
INTRODUCTION The current drug discovery paradigm of 'one drug, multiple targets' has gained attention from both the academic medicinal chemistry community and the pharmaceutical industry. This is in response to the urgent need for effective agents to treat multifactorial chronic diseases. The molecular hybridization strategy is a useful tool that has been widely explored, particularly in the last two decades, for the design of multi-target drugs. AREAS COVERED This review examines the current state of molecular hybridization in guiding the discovery of multitarget small molecules. The article discusses the design strategies and target selection for a multitarget polypharmacology approach to treat various diseases, including cancer, Alzheimer's disease, cardiac arrhythmia, endometriosis, and inflammatory diseases. EXPERT OPINION Although the examples discussed highlight the importance of molecular hybridization for the discovery of multitarget bioactive compounds, it is notorious that the literature has focused on specific classes of targets. This may be due to a deep understanding of the pharmacophore features required for target binding, making targets such as histone deacetylases and cholinesterases frequent starting points. However, it is important to encourage the scientific community to explore diverse combinations of targets using the molecular hybridization strategy.
Collapse
Affiliation(s)
- Pedro de Sena Murteira Pinheiro
- Laboratório de Avaliação e Síntese de Substâncias Bioativas (LASSBio), Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Lucas Silva Franco
- Laboratório de Avaliação e Síntese de Substâncias Bioativas (LASSBio), Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Tadeu Lima Montagnoli
- Laboratório de Avaliação e Síntese de Substâncias Bioativas (LASSBio), Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Farmacologia e Química Medicinal, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Carlos Alberto Manssour Fraga
- Laboratório de Avaliação e Síntese de Substâncias Bioativas (LASSBio), Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Farmacologia e Química Medicinal, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
5
|
Manzoor S, Gabr MT, Nafie MS, Raza MK, Khan A, Nayeem SM, Arafa RK, Hoda N. Discovery of Quinolinone Hybrids as Dual Inhibitors of Acetylcholinesterase and Aβ Aggregation for Alzheimer's Disease Therapy. ACS Chem Neurosci 2024; 15:539-559. [PMID: 38149821 DOI: 10.1021/acschemneuro.3c00588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2023] Open
Abstract
The development of multitargeted therapeutics has evolved as a promising strategy to identify efficient therapeutics for neurological disorders. We report herein new quinolinone hybrids as dual inhibitors of acetylcholinesterase (AChE) and Aβ aggregation that function as multitargeted ligands for Alzheimer's disease. The quinoline hybrids (AM1-AM16) were screened for their ability to inhibit AChE, BACE1, amyloid fibrillation, α-syn aggregation, and tau aggregation. Among the tested compounds, AM5 and AM10 inhibited AChE activity by more than 80% at single-dose screening and possessed a remarkable ability to inhibit the fibrillation of Aβ42 oligomers at 10 μM. In addition, dose-dependent screening of AM5 and AM10 was performed, giving half-maximal AChE inhibitory concentration (IC50) values of 1.29 ± 0.13 and 1.72 ± 0.18 μM, respectively. In addition, AM5 and AM10 demonstrated concentration-dependent inhibitory profiles for the aggregation of Aβ42 oligomers with estimated IC50 values of 4.93 ± 0.8 and 1.42 ± 0.3 μM, respectively. Moreover, the neuroprotective properties of the lead compounds AM5 and AM10 were determined in SH-SY5Y cells incubated with Aβ oligomers. This work would enable future research efforts aiming at the structural optimization of AM5 and AM10 to develop potent dual inhibitors of AChE and amyloid aggregation. Furthermore, the in vivo assay confirmed the antioxidant activity of compounds AM5 and AM10 through increasing GSH, CAT, and SOD activities that are responsible for scavenging the ROS and restoring its normal level. Blood investigation illustrated the protective activity of the two compounds against lead-induced neurotoxicity through retaining hematological and liver enzymes near normal levels. Finally, immunohistochemistry investigation revealed the inhibitory activity of β-amyloid (Aβ) aggregation.
Collapse
Affiliation(s)
- Shoaib Manzoor
- Drug Design and Synthesis Laboratory, Department of Chemistry, Jamia Millia Islamia, New Delhi 110025, India
- Department of Pharmacy and Pharmacology, University of Bath, Bath BA2 7AY, U.K
| | - Moustafa T Gabr
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine, New York, New York10021, United States
| | - Mohamed S Nafie
- Department of Chemistry, College of Sciences, University of Sharjah, Sharjah (P.O. Box 27272), United Arab Emirates
- Chemistry Department, Faculty of Science, Suez Canal University, Ismailia 41522, Egypt
| | - Md Kausar Raza
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore 560012, India
| | - Ashma Khan
- Department of Chemistry, Aligarh Muslim University, Aligarh, UP 202002, India
| | - Shahid M Nayeem
- Department of Chemistry, Aligarh Muslim University, Aligarh, UP 202002, India
| | - Reem K Arafa
- Drug Design and Discovery Lab, Helmy Institute for Medical Sciences, Zewail City of Science, Technology and Innovation, Giza 12578, Egypt
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science, Technology and Innovation, Giza12578,Egypt
| | - Nasimul Hoda
- Drug Design and Synthesis Laboratory, Department of Chemistry, Jamia Millia Islamia, New Delhi 110025, India
| |
Collapse
|
6
|
Huang ST, Luo JC, Zhong GH, Teng LP, Yang CY, Tang CL, Jing L, Zhou ZB, Liu J, Jiang N. In vitro and in vivo Biological Evaluation of Newly Tacrine-Selegiline Hybrids as Multi-Target Inhibitors of Cholinesterases and Monoamine Oxidases for Alzheimer's Disease. Drug Des Devel Ther 2024; 18:133-159. [PMID: 38283137 PMCID: PMC10822116 DOI: 10.2147/dddt.s432170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 01/15/2024] [Indexed: 01/30/2024] Open
Abstract
Purpose Alzheimer's disease (AD) is the most common neurodegenerative disease, and its multifactorial nature increases the difficulty of medical research. To explore an effective treatment for AD, a series of novel tacrine-selegiline hybrids with ChEs and MAOs inhibitory activities were designed and synthesized as multifunctional drugs. Methods All designed compounds were evaluated in vitro for their inhibition of cholinesterases (AChE/BuChE) and monoamine oxidases (MAO-A/B) along with their blood-brain barrier permeability. Then, further biological activities of the optimizing compound 7d were determined, including molecular model analysis, in vitro cytotoxicity, acute toxicity studies in vivo, and pharmacokinetic and pharmacodynamic property studies in vivo. Results Most synthesized compounds demonstrated potent inhibitory activity against ChEs/MAOs. Particularly, compound 7d exhibited good and well-balanced activity against ChEs (hAChE: IC50 = 1.57 μM, hBuChE: IC50 = 0.43 μM) and MAOs (hMAO-A: IC50 = 2.30 μM, hMAO-B: IC50 = 4.75 μM). Molecular modeling analysis demonstrated that 7d could interact simultaneously with both the catalytic active site (CAS) and peripheral anionic site (PAS) of AChE in a mixed-type manner and also exhibits binding affinity towards BuChE and MAO-B. Additionally, 7d displayed excellent permeability of the blood-brain barrier, and under the experimental conditions, it elicited low or no toxicity toward PC12 and BV-2 cells. Furthermore, 7d was not acutely toxic in mice at doses up to 2500 mg/kg and could improve the cognitive function of mice with scopolamine-induced memory impairment. Lastly, 7d possessed well pharmacokinetic characteristics. Conclusion In light of these results, it is clear that 7d could potentially serve as a promising multi-functional drug for the treatment of AD.
Collapse
Affiliation(s)
- Shu-Tong Huang
- Department of Pharmacy, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, People’s Republic of China
- Pharmaceutical College, Guangxi Medical University, Nanning, Guangxi, People’s Republic of China
| | - Jin-Chong Luo
- Department of Pharmacy, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, People’s Republic of China
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, People’s Republic of China
| | - Guo-Hui Zhong
- Department of Pharmacy, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, People’s Republic of China
- Pharmaceutical College, Guangxi Medical University, Nanning, Guangxi, People’s Republic of China
| | - Li-Ping Teng
- School of Pharmacy, Youjiang Medical University for Nationalities, Baise, Guangxi, People’s Republic of China
| | - Cai-Yan Yang
- School of Pharmacy, Youjiang Medical University for Nationalities, Baise, Guangxi, People’s Republic of China
| | - Chun-Li Tang
- Department of Pharmacy, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, People’s Republic of China
| | - Lin Jing
- Department of Pharmacy, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, People’s Republic of China
| | - Zhong-Bo Zhou
- School of Pharmacy, Youjiang Medical University for Nationalities, Baise, Guangxi, People’s Republic of China
| | - Jing Liu
- Department of Pharmacy, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, People’s Republic of China
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, People’s Republic of China
| | - Neng Jiang
- Department of Pharmacy, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, People’s Republic of China
- Pharmaceutical College, Guangxi Medical University, Nanning, Guangxi, People’s Republic of China
| |
Collapse
|
7
|
Zou D, Liu R, Lv Y, Guo J, Zhang C, Xie Y. Latest advances in dual inhibitors of acetylcholinesterase and monoamine oxidase B against Alzheimer's disease. J Enzyme Inhib Med Chem 2023; 38:2270781. [PMID: 37955252 PMCID: PMC10653629 DOI: 10.1080/14756366.2023.2270781] [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: 08/07/2023] [Accepted: 09/27/2023] [Indexed: 11/14/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive brain disease characterised by progressive memory loss and cognition impairment, ultimately leading to death. There are three FDA-approved acetylcholinesterase inhibitors (donepezil, rivastigmine, and galantamine, AChEIs) for the symptomatic treatment of AD. Monoamine oxidase B (MAO-B) has been considered to contribute to pathologies of AD. Therefore, we reviewed the dual inhibitors of acetylcholinesterase (AChE) and MAO-B developed in the last five years. In this review, these dual-target inhibitors were classified into six groups according to the basic parent structure, including chalcone, coumarin, chromone, benzo-fused five-membered ring, imine and hydrazine, and other scaffolds. Their design strategies, structure-activity relationships (SARs), and molecular docking studies with AChE and MAO-B were analysed and discussed, giving valuable insights for the subsequent development of AChE and MAO-B dual inhibitors. Challenges in the development of balanced and potent AChE and MAO-B dual inhibitors were noted, and corresponding solutions were provided.
Collapse
Affiliation(s)
- Dajiang Zou
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| | - Renzheng Liu
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| | - Yangjing Lv
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| | - Jianan Guo
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| | - Changjun Zhang
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| | - Yuanyuan Xie
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceutical, Zhejiang University of Technology, Hangzhou, China
- Key Laboratory for Green Pharmaceutical Technologies and Related Equipment of Ministry of Education, Key Laboratory of Pharmaceutical Engineering of Zhejiang Province, Hangzhou, China
| |
Collapse
|
8
|
Zhong G, Guo J, Pang C, Su D, Tang C, Jing L, Zhang F, He P, Yan Y, Chen Z, Liu J, Jiang N. Novel AP2238-clorgiline hybrids as multi-target agents for the treatment of Alzheimer's disease: Design, synthesis, and biological evaluation. Bioorg Chem 2023; 130:106224. [DOI: 10.1016/j.bioorg.2022.106224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/19/2022] [Accepted: 10/20/2022] [Indexed: 11/02/2022]
|
9
|
Kpemissi M, Kantati YT, Veerapur VP, Eklu-Gadegbeku K, Hassan Z. Anti-cholinesterase, anti-inflammatory and antioxidant properties of Combretum micranthum G. Don: Potential implications in neurodegenerative disease. IBRO Neurosci Rep 2022; 14:21-27. [PMID: 36578633 PMCID: PMC9791815 DOI: 10.1016/j.ibneur.2022.12.001] [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] [Received: 08/04/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022] Open
Abstract
Background Brain damage is a severe and common pathology that leads to life-threatening diseases. Despite development in the research, the medical evidence of the effectiveness of potential neuroprotective medicines is insufficient. As a result, there is an immense and urgent demand for promising medication. For millennia, herbal remedies were a fundamental aspect of medical treatments. Combretum micranthum (CM), a plant of the family Combretaceae in sub-Saharan Africa, has been utilized in folklore medicine to cure diverse human ailments. In order to develop a neuroprotective phytomedicine, the current research was undertaken to explore the antioxidant, anti-inflammatory, anticholinesterase and neuroprotective potential of CM extract. Methods Colorimetric methods were used to determine CM antioxidant activity, in-vitro protein denaturation and membrane destabilization assays were used to evaluate its anti-inflammatory capacity, anticholinesterase activity was carried out using Ellman's method, and neuroprotective potential was assessed on brain homogenate stressed with ferric chloride and ascorbic acid (FeCl2-AA) by assessing the lipoperoxidation biomarker malondialdehyde (MDA). Results In Ferric Reducing Antioxidant Power (IC50 = 27.15 ± 0.06 µg/mL) and Total Antioxidant Capacity (IC50 = 31.13 ± 0.02 µg/mL), CM extract demonstrated strong antioxidant activity. Anti-inflammatory effect were improved in heat-induced Egg albumin and BSA denaturation (IC 50 = 46.35 ± 1.53 and 23.94 ± 1.10 µg/mL) as well as heat and hypotonia induced membrane destabilization (IC 50 = 20.96 ± 0.11 and 16.75 ± 0.94 µg/mL).CM extract showed strong anticholinesterase activity (IC 50 = 59.85 ± 0.91 µg/mL). In an ex-vivo neuroprotective model, CM extract showed substantial inhibition (p < 0.001) of oxidative damage caused by FeCl2-AA in brain tissue. Conclusion C. micranthum may be a good candidate for its probable neuroprotective potential. Its neuroprotective benefits might be attributed to its antioxidant, anti-inflammatory and anticholinesterase effects.
Collapse
Affiliation(s)
- Mabozou Kpemissi
- Centre for Drug Research, Universiti Sains Malaysia, 11800 Minden, Penang, Malaysia,University of Lomé, Togo,Sree Siddaganga College of Pharmacy, B.H. Road, Tumkur 572 102, Karnataka, India,Correspondence to: Major in Pharmacology and Physiology Faculty of Sciences, University of Lomé, Togo.
| | | | | | | | - Zurina Hassan
- Centre for Drug Research, Universiti Sains Malaysia, 11800 Minden, Penang, Malaysia,Corresponding author.
| |
Collapse
|
10
|
SAR studies of quinoline and derivatives as potential treatments for Alzheimer’s disease. ARAB J CHEM 2022. [DOI: 10.1016/j.arabjc.2022.104502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
11
|
Keuler T, Lemke C, Elsinghorst PW, Iriepa I, Chioua M, Martínez-Grau MA, Beadle CD, Vetman T, López-Muñoz F, Wille T, Bartz U, Deuther-Conrad W, Marco-Contelles J, Gütschow M. The Chemotype of Chromanones as a Privileged Scaffold for Multineurotarget Anti-Alzheimer Agents. ACS Pharmacol Transl Sci 2022; 5:1097-1108. [PMID: 36407962 PMCID: PMC9667544 DOI: 10.1021/acsptsci.2c00097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Indexed: 11/28/2022]
Abstract
The multifactorial nature of Alzheimer's disease necessitates the development of agents able to interfere with different relevant targets. A series of 22 tailored chromanones was conceptualized, synthesized, and subjected to biological evaluation. We identified one representative bearing a linker-connected azepane moiety (compound 19) with balanced pharmacological properties. Compound 19 exhibited inhibitory activities against human acetyl-, butyrylcholinesterase and monoamine oxidase-B, as well as high affinity to both the σ1 and σ2 receptors. Our study provides a framework for the development of further chromanone-based multineurotarget agents.
Collapse
Affiliation(s)
- Tim Keuler
- Pharmaceutical
Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany
| | - Carina Lemke
- Pharmaceutical
Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany
| | - Paul W. Elsinghorst
- Pharmaceutical
Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany
- Central
Institute of the Bundeswehr Medical Service Munich, Ingolstädter Landstraße 102, 85748 Garching Germany
| | - Isabel Iriepa
- Universidad
de Alcalá, Departamento de Química
Orgánica y Química Inorgánica, Ctra. Madrid-Barcelona, 28871 Alcalá de Henares, Madrid España
| | - Mourad Chioua
- Laboratory
of Medicinal Chemistry, IQOG, CSIC, C/Juan de la Cierva 3, 28006 Madrid, Spain
| | | | - Christopher D. Beadle
- Lilly Research
Centre, Eli Lilly & Company, Erl Wood Manor, Windlesham, Surrey GU20
6PH, United Kingdom
| | - Tatiana Vetman
- Lilly
Research Laboratories, Eli Lilly & Company, Indianapolis, Indiana 46285, United States
| | - Francisco López-Muñoz
- Faculty
of Health, Camilo José Cela University of Madrid (UCJC), Neuropsychopharmacology Unit, “Hospital 12 de Octubre” Research
Institute, 28692 Madrid, Spain
| | - Timo Wille
- Bundeswehr
Institute of Pharmacology and Toxicology, Neuherbergstraße 11, 80937 München, Germany
| | - Ulrike Bartz
- Department
of Natural Sciences, University of Applied
Sciences Bonn-Rhein-Sieg, von-Liebig-Straße 20, 53359 Rheinbach, Germany
| | - Winnie Deuther-Conrad
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Department of Neuroradiopharmaceuticals, 04318 Leipzig, Germany
| | - José Marco-Contelles
- Laboratory
of Medicinal Chemistry, IQOG, CSIC, C/Juan de la Cierva 3, 28006 Madrid, Spain
| | - Michael Gütschow
- Pharmaceutical
Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany
| |
Collapse
|
12
|
Pisani L, Catto M, Muncipinto G, Nicolotti O, Carrieri A, Rullo M, Stefanachi A, Leonetti F, Altomare C. A twenty-year journey exploring coumarin-based derivatives as bioactive molecules. Front Chem 2022; 10:1002547. [PMID: 36300022 PMCID: PMC9590106 DOI: 10.3389/fchem.2022.1002547] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 09/14/2022] [Indexed: 11/25/2022] Open
Abstract
The coumarin core (i.e., 1-benzopyran-2 (2H)-one) is a structural motif highly recurrent in both natural products and bioactive molecules. Indeed, depending on the substituents and branching positions around the byciclic core, coumarin-containing compounds have shown diverse pharmacological activities, ranging from anticoagulant activities to anti-inflammatory, antimicrobial, anti-HIV and antitumor effects. In this survey, we have reported the main scientific results of the 20-years investigation on the coumarin core, exploited by the research group headed by Prof. Angelo Carotti (Bari, Italy) either as a scaffold or a pharmacophore moiety in designing novel biologically active small molecules.
Collapse
Affiliation(s)
- Leonardo Pisani
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, Bari, Italy
| | - Marco Catto
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, Bari, Italy
| | | | - Orazio Nicolotti
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, Bari, Italy
| | - Antonio Carrieri
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, Bari, Italy
| | - Mariagrazia Rullo
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, Bari, Italy
| | - Angela Stefanachi
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, Bari, Italy
- *Correspondence: Angela Stefanachi, Francesco Leonetti,
| | - Francesco Leonetti
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, Bari, Italy
- *Correspondence: Angela Stefanachi, Francesco Leonetti,
| | - Cosimo Altomare
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, Bari, Italy
| |
Collapse
|
13
|
Design, Synthesis, and biological evaluation of pyrazolo-benzothiazole derivatives as a potential therapeutic agent for the treatment of Alzheimer’s disease. Med Chem Res 2022. [DOI: 10.1007/s00044-022-02953-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|
14
|
Ekström F, Gottinger A, Forsgren N, Catto M, Iacovino LG, Pisani L, Binda C. Dual Reversible Coumarin Inhibitors Mutually Bound to Monoamine Oxidase B and Acetylcholinesterase Crystal Structures. ACS Med Chem Lett 2022; 13:499-506. [PMID: 35300078 PMCID: PMC8919507 DOI: 10.1021/acsmedchemlett.2c00001] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 02/15/2022] [Indexed: 11/29/2022] Open
Abstract
![]()
Multitarget directed
ligands (MTDLs) represent a promising frontier
in tackling the complexity of multifactorial pathologies. The synergistic
inhibition of monoamine oxidase B (MAO B) and acetylcholinesterase
(AChE) is believed to provide a potentiated effect in the treatment
of Alzheimer’s disease. Among previously reported micromolar
or sub-micromolar coumarin-bearing dual inhibitors, compound 1 returned a tight-binding inhibition of MAO B (Ki = 4.5 μM) and a +5.5 °C
increase in the enzyme Tm value. Indeed,
the X-ray crystal structure revealed that binding of 1 produces unforeseen conformational changes at the MAO B entrance
cavity. Interestingly, 1 showed great shape complementarity
with the AChE enzymatic gorge, being deeply buried from the catalytic
anionic subsite (CAS) to the peripheral anionic subsite (PAS) and
causing significant structural changes in the active site. These findings
provide structural templates for further development of dual MAO B
and AChE inhibitors.
Collapse
Affiliation(s)
- Fredrik Ekström
- Swedish Defence Research Agency, CBRN Defence and Security, Umeå 901 82, Sweden
| | - Andrea Gottinger
- Department of Biology and Biotechnology, University of Pavia, 27100 Pavia, Italy
| | - Nina Forsgren
- Swedish Defence Research Agency, CBRN Defence and Security, Umeå 901 82, Sweden
| | - Marco Catto
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari “Aldo Moro”, via E. Orabona 4, 70125, Bari, Italy
| | - Luca G. Iacovino
- Department of Biology and Biotechnology, University of Pavia, 27100 Pavia, Italy
| | - Leonardo Pisani
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari “Aldo Moro”, via E. Orabona 4, 70125, Bari, Italy
| | - Claudia Binda
- Department of Biology and Biotechnology, University of Pavia, 27100 Pavia, Italy
| |
Collapse
|
15
|
Rullo M, Cipolloni M, Catto M, Colliva C, Miniero DV, Latronico T, de Candia M, Benicchi T, Linusson A, Giacchè N, Altomare CD, Pisani L. Probing Fluorinated Motifs onto Dual AChE-MAO B Inhibitors: Rational Design, Synthesis, Biological Evaluation, and Early-ADME Studies. J Med Chem 2022; 65:3962-3977. [PMID: 35195417 DOI: 10.1021/acs.jmedchem.1c01784] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Bioisosteric H/F or CH2OH/CF2H replacement was introduced in coumarin derivatives previously characterized as dual AChE-MAO B inhibitors to probe the effects on both inhibitory potency and drug-likeness. Along with in vitro screening, we investigated early-ADME parameters related to solubility and lipophilicity (Sol7.4, CHI7.4, log D7.4), oral bioavailability and central nervous system (CNS) penetration (PAMPA-HDM and PAMPA-blood-brain barrier (BBB) assays, Caco-2 bidirectional transport study), and metabolic liability (half-lives and clearance in microsomes, inhibition of CYP3A4). Both specific and nonspecific tissue toxicities were determined in SH-SY5Y and HepG2 lines, respectively. Compound 15 bearing a -CF2H motif emerged as a water-soluble, orally bioavailable CNS-permeant potent inhibitor of both human AChE (IC50 = 550 nM) and MAO B (IC50 = 8.2 nM, B/A selectivity > 1200). Moreover, 15 behaved as a safe and metabolically stable neuroprotective agent, devoid of cytochrome liability.
Collapse
Affiliation(s)
- Mariagrazia Rullo
- Department of Pharmacy─Pharmaceutical Sciences, University of Bari "Aldo Moro", via Orabona 4, 70125 Bari, Italy
| | | | - Marco Catto
- Department of Pharmacy─Pharmaceutical Sciences, University of Bari "Aldo Moro", via Orabona 4, 70125 Bari, Italy
| | | | - Daniela Valeria Miniero
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari "Aldo Moro", via Orabona, 4, 70125 Bari, Italy
| | - Tiziana Latronico
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari "Aldo Moro", via Orabona, 4, 70125 Bari, Italy
| | - Modesto de Candia
- Department of Pharmacy─Pharmaceutical Sciences, University of Bari "Aldo Moro", via Orabona 4, 70125 Bari, Italy
| | | | - Anna Linusson
- Department of Chemistry, Umeå University, 90187 Umeå, Sweden
| | - Nicola Giacchè
- TES Pharma s.r.l., Corso Vannucci 47, 06121 Perugia, Italy
| | - Cosimo Damiano Altomare
- Department of Pharmacy─Pharmaceutical Sciences, University of Bari "Aldo Moro", via Orabona 4, 70125 Bari, Italy
| | - Leonardo Pisani
- Department of Pharmacy─Pharmaceutical Sciences, University of Bari "Aldo Moro", via Orabona 4, 70125 Bari, Italy
| |
Collapse
|
16
|
Benazzouz-Touami A, Chouh A, Halit S, Terrachet-Bouaziz S, Makhloufi-Chebli M, Ighil-Ahriz K, Silva AM. New Coumarin-Pyrazole hybrids: Synthesis, Docking studies and Biological evaluation as potential cholinesterase inhibitors. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2021.131591] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
17
|
Mzezewa SC, Omoruyi SI, Zondagh LS, Malan SF, Ekpo OE, Joubert J. Design, synthesis, and evaluation of 3,7-substituted coumarin derivatives as multifunctional Alzheimer's disease agents. J Enzyme Inhib Med Chem 2021; 36:1607-1621. [PMID: 34281458 PMCID: PMC8291583 DOI: 10.1080/14756366.2021.1913137] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
Multitarget directed ligands (MTDLs) are emerging as promising treatment options for Alzheimer's disease (AD). Coumarin derivatives serve as a good starting point for designing MTDLs due to their inherent inhibition of monoamine oxidase (MAO) and cholinesterase enzymes, which are complicit in AD's complex pathophysiology. A preliminary series of 3,7-substituted coumarin derivatives were synthesised and evaluated for enzyme inhibitory activity, cytotoxicity as well as neuroprotective ability. The results indicated that the compounds are weak cholinesterase inhibitors with five compounds demonstrating relatively potent inhibition and selectivity towards MAO-B with IC50 values between 0.014 and 0.498 hx00B5;µM. Significant neuroprotective effects towards MPP+-compromised SH-SY5Y neuroblastoma cells were also observed, with no inherent cytotoxicity at 10 µM for all compounds. The overall results demonstrated that substitution of the phenylethyloxy moiety at the 7-position imparted superior general activity to the derivatives, with the propargylamine substitution at the 3-position, in particular, displaying the best MAO-B selectivity and neuroprotection.
Collapse
Affiliation(s)
- Sheunopa C Mzezewa
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of the Western Cape, Bellville, South Africa
| | - Sylvester I Omoruyi
- Department of Medical Biosciences, University of the Western Cape, Bellville, South Africa
| | - Luke S Zondagh
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of the Western Cape, Bellville, South Africa
| | - Sarel F Malan
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of the Western Cape, Bellville, South Africa
| | - Okobi E Ekpo
- Department of Medical Biosciences, University of the Western Cape, Bellville, South Africa
| | - Jacques Joubert
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of the Western Cape, Bellville, South Africa
| |
Collapse
|
18
|
Bouhaoui A, Eddahmi M, Dib M, Khouili M, Aires A, Catto M, Bouissane L. Synthesis and Biological Properties of Coumarin Derivatives. A Review. ChemistrySelect 2021. [DOI: 10.1002/slct.202101346] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Abderrazzak Bouhaoui
- Organic and Analytical Chemistry Laboratory Faculty of Sciences and Technologies Sultan Moulay Slimane University BP 523 23000 Beni-Mellal Morocco
| | - Mohammed Eddahmi
- Organic and Analytical Chemistry Laboratory Faculty of Sciences and Technologies Sultan Moulay Slimane University BP 523 23000 Beni-Mellal Morocco
| | - Mustapha Dib
- Organic and Analytical Chemistry Laboratory Faculty of Sciences and Technologies Sultan Moulay Slimane University BP 523 23000 Beni-Mellal Morocco
| | - Mostafa Khouili
- Organic and Analytical Chemistry Laboratory Faculty of Sciences and Technologies Sultan Moulay Slimane University BP 523 23000 Beni-Mellal Morocco
| | - Alfredo Aires
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences CITAB University of Trás-os-Montes e Alto Douro UTAD Vila Real Portugal
| | - Marco Catto
- Department of Pharmacy-Pharmaceutical Sciences University of Bari Aldo Moro via E. Orabona 4 70125 Bari Italy
| | - Latifa Bouissane
- Organic and Analytical Chemistry Laboratory Faculty of Sciences and Technologies Sultan Moulay Slimane University BP 523 23000 Beni-Mellal Morocco
| |
Collapse
|
19
|
Zhang C, Lv Y, Bai R, Xie Y. Structural exploration of multifunctional monoamine oxidase B inhibitors as potential drug candidates against Alzheimer's disease. Bioorg Chem 2021; 114:105070. [PMID: 34126574 DOI: 10.1016/j.bioorg.2021.105070] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 05/25/2021] [Accepted: 06/05/2021] [Indexed: 10/21/2022]
Abstract
AD is one of the most typical neurodegenerative disorders that suffer many seniors worldwide. Recently, MAO inhibitors have received increasing attention not only for their roles involved in monoamine neurotransmitters metabolism and oxidative stress but also for their additional neuroprotective and neurorescue effects against AD. The curiosity in MAO inhibitors is reviving, and novel MAO-B inhibitors recently developed with ancillary activities (e.g., Aβ aggregation and AChE inhibition, anti-ROS and chelating activities) have been proposed as multitarget drugs foreshadowing a positive outlook for the treatment of AD. The current review describes the recent development of the design, synthesis, and screening of multifunctional ligands based on MAO-B inhibition for AD therapy. Structure-activity relationships and rational design strategies of the synthetic or natural product derivatives (chalcones, coumarins, chromones, and homoisoflavonoids) are discussed.
Collapse
Affiliation(s)
- Changjun Zhang
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceutical, Zhejiang University of Technology, Hangzhou, PR China
| | - Yangjing Lv
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, PR China
| | - Renren Bai
- College of Pharmacy, School of Medicine, Hangzhou Normal University, Hangzhou, PR China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, PR China.
| | - Yuanyuan Xie
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceutical, Zhejiang University of Technology, Hangzhou, PR China; College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, PR China.
| |
Collapse
|
20
|
Titov AA, Kobzev MS, Catto M, Candia MD, Gambacorta N, Denora N, Pisani L, Nicolotti O, Borisova TN, Varlamov AV, Voskressensky LG, Altomare CD. Away from Flatness: Unprecedented Nitrogen-Bridged Cyclopenta[ a]indene Derivatives as Novel Anti-Alzheimer Multitarget Agents. ACS Chem Neurosci 2021; 12:340-353. [PMID: 33395258 DOI: 10.1021/acschemneuro.0c00706] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Nature-inspired, bridged polycyclic molecules share low similarity with currently available drugs, containing preferentially planar and/or achiral moieties. This "Escape from Flatland" scenario, aimed at exploring pharmacological properties of atypical molecular scaffolds, finds interest in synthetic routes leading to tridimensional-shaped molecules. Herein we report on the synthesis of N-bridged cyclopenta[a]indene derivatives, achieved through microwave-assisted thermal rearrangement of allene 3-benzazecines with high diastereoselectivity. The biological evaluation disclosed selective inhibition of human acetylcholinesterase or butyrylcholinesterase, depending on the substitution around the molecular core, which was rationalized by means of docking simulations. The most potent BChE inhibitor 31 was effective in neuroprotection from glutamatergic excitotoxicity and displayed low intrinsic cytotoxicity and good brain penetration. Overall, compound 31 and its close congeners 34 and 35 acted as multitarget agents addressing different biological events involved in neurodegeneration, particularly in the progression of Alzheimer's disease.
Collapse
Affiliation(s)
- Alexander A. Titov
- Organic Chemistry Department, Peoples’ Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya St, Moscow 117198, Russian Federation
| | - Maxim S. Kobzev
- Organic Chemistry Department, Peoples’ Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya St, Moscow 117198, Russian Federation
| | - Marco Catto
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, Via E. Orabona 4, 70125 Bari, Italy
| | - Modesto de Candia
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, Via E. Orabona 4, 70125 Bari, Italy
| | - Nicola Gambacorta
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, Via E. Orabona 4, 70125 Bari, Italy
| | - Nunzio Denora
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, Via E. Orabona 4, 70125 Bari, Italy
| | - Leonardo Pisani
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, Via E. Orabona 4, 70125 Bari, Italy
| | - Orazio Nicolotti
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, Via E. Orabona 4, 70125 Bari, Italy
| | - Tatiana N. Borisova
- Organic Chemistry Department, Peoples’ Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya St, Moscow 117198, Russian Federation
| | - Alexey V. Varlamov
- Organic Chemistry Department, Peoples’ Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya St, Moscow 117198, Russian Federation
| | - Leonid G. Voskressensky
- Organic Chemistry Department, Peoples’ Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya St, Moscow 117198, Russian Federation
| | - Cosimo D. Altomare
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, Via E. Orabona 4, 70125 Bari, Italy
| |
Collapse
|
21
|
Purgatorio R, Gambacorta N, Catto M, de Candia M, Pisani L, Espargaró A, Sabaté R, Cellamare S, Nicolotti O, Altomare CD. Pharmacophore Modeling and 3D-QSAR Study of Indole and Isatin Derivatives as Antiamyloidogenic Agents Targeting Alzheimer's Disease. Molecules 2020; 25:E5773. [PMID: 33297547 PMCID: PMC7731220 DOI: 10.3390/molecules25235773] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 12/02/2020] [Accepted: 12/04/2020] [Indexed: 11/23/2022] Open
Abstract
Thirty-six novel indole-containing compounds, mainly 3-(2-phenylhydrazono) isatins and structurally related 1H-indole-3-carbaldehyde derivatives, were synthesized and assayed as inhibitors of beta amyloid (Aβ) aggregation, a hallmark of pathophysiology of Alzheimer's disease. The newly synthesized molecules spanned their IC50 values from sub- to two-digit micromolar range, bearing further information into structure-activity relationships. Some of the new compounds showed interesting multitarget activity, by inhibiting monoamine oxidases A and B. A cell-based assay in tau overexpressing bacterial cells disclosed a promising additional activity of some derivatives against tau aggregation. The accumulated data of either about ninety published and thirty-six newly synthesized molecules were used to generate a pharmacophore hypothesis of antiamyloidogenic activity exerted in a wide range of potencies, satisfactorily discriminating the 'active' compounds from the 'inactive' (poorly active) ones. An atom-based 3D-QSAR model was also derived for about 80% of 'active' compounds, i.e., those achieving finite IC50 values lower than 100 μM. The 3D-QSAR model (encompassing 4 PLS factors), featuring acceptable predictive statistics either in the training set (n = 45, q2 = 0.596) and in the external test set (n = 14, r2ext = 0.695), usefully complemented the pharmacophore model by identifying the physicochemical features mainly correlated with the Aβ anti-aggregating potency of the indole and isatin derivatives studied herein.
Collapse
Affiliation(s)
- Rosa Purgatorio
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, Via E. Orabona 4, 70125 Bari, Italy; (R.P.); (N.G.); (M.d.C.); (L.P.); (S.C.); (O.N.); (C.D.A.)
| | - Nicola Gambacorta
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, Via E. Orabona 4, 70125 Bari, Italy; (R.P.); (N.G.); (M.d.C.); (L.P.); (S.C.); (O.N.); (C.D.A.)
| | - Marco Catto
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, Via E. Orabona 4, 70125 Bari, Italy; (R.P.); (N.G.); (M.d.C.); (L.P.); (S.C.); (O.N.); (C.D.A.)
| | - Modesto de Candia
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, Via E. Orabona 4, 70125 Bari, Italy; (R.P.); (N.G.); (M.d.C.); (L.P.); (S.C.); (O.N.); (C.D.A.)
| | - Leonardo Pisani
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, Via E. Orabona 4, 70125 Bari, Italy; (R.P.); (N.G.); (M.d.C.); (L.P.); (S.C.); (O.N.); (C.D.A.)
| | - Alba Espargaró
- Institute of Nanoscience and Nanotechnology (IN2UB), Department of Physical Chemistry, Faculty of Pharmacy, University of Barcelona, Joan XXIII 27-31, E-08028 Barcelona, Spain; (A.E.); (R.S.)
| | - Raimon Sabaté
- Institute of Nanoscience and Nanotechnology (IN2UB), Department of Physical Chemistry, Faculty of Pharmacy, University of Barcelona, Joan XXIII 27-31, E-08028 Barcelona, Spain; (A.E.); (R.S.)
| | - Saverio Cellamare
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, Via E. Orabona 4, 70125 Bari, Italy; (R.P.); (N.G.); (M.d.C.); (L.P.); (S.C.); (O.N.); (C.D.A.)
| | - Orazio Nicolotti
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, Via E. Orabona 4, 70125 Bari, Italy; (R.P.); (N.G.); (M.d.C.); (L.P.); (S.C.); (O.N.); (C.D.A.)
| | - Cosimo D. Altomare
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, Via E. Orabona 4, 70125 Bari, Italy; (R.P.); (N.G.); (M.d.C.); (L.P.); (S.C.); (O.N.); (C.D.A.)
| |
Collapse
|
22
|
Liu Z, Zhang B, Xia S, Fang L, Gou S. ROS-responsive and multifunctional anti-Alzheimer prodrugs: Tacrine-ibuprofen hybrids via a phenyl boronate linker. Eur J Med Chem 2020; 212:112997. [PMID: 33189440 DOI: 10.1016/j.ejmech.2020.112997] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 10/26/2020] [Accepted: 11/03/2020] [Indexed: 02/06/2023]
Abstract
Current drugs available in clinic for Alzheimer's disease (AD) treatment can only alleviate disease symptoms without clearly curing or delaying the process of AD. And some AD drugs failed in Phase III clinical trials are only focused on targeting amyloid-β (Aβ). Therefore, an alternative strategy in AD drug design is meaningful to be involved in the multiple pathogenic factors which can affect each other at multiple levels. Herein, we report a series of ROS-responsive prodrugs based on multi-target-directed ligands (MTDLs) approach, which can specifically release tacrine derivatives and ibuprofen under oxidation of ROS and show acetylcholinesterase (AChE)-inhibiting, neuron-protective and anti-inflammatory effects in extracellular or intracellular assays. Related biological study illustrated that compound 22 was able to permeate blood-brain-barrier (BBB) showing little hepatotoxicity in comparison to tacrine. Besides, 22 hinted a therapeutic clue in AD-treatment by regulating proinflammatory factors (IL-1β and TNF-α) and apoptosis related proteins (Bax, Bcl-2 and cleaved caspase-3). Further spatial memory assays in Aβ-induced AD model showed that 22 enhanced the ability of learning and memory. Our study proves that the strategy of ROS-responsive prodrugs has promise for AD treatments in future and offers a way for AD drug development.
Collapse
Affiliation(s)
- Zhikun Liu
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing, 211189, China; Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, China
| | - Bin Zhang
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing, 211189, China; Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, China
| | - Shengjin Xia
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing, 211189, China; Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, China
| | - Lei Fang
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing, 211189, China; Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, China.
| | - Shaohua Gou
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing, 211189, China; Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, China.
| |
Collapse
|
23
|
Reeta, Baek SC, Lee JP, Rangarajan TM, Ayushee, Singh RP, Singh M, Mangiatordi GF, Nicolotti O, Kim H, Mathew B. Ethyl Acetohydroxamate Incorporated Chalcones: Unveiling a Novel Class of Chalcones for Multitarget Monoamine Oxidase-B Inhibitors Against Alzheimer's Disease. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2020; 18:643-654. [PMID: 31550216 DOI: 10.2174/1871527318666190906101326] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 07/25/2019] [Accepted: 07/27/2019] [Indexed: 12/23/2022]
Abstract
BACKGROUND Chalcones are considered as the selective scaffold for the inhibition of MAO-B. OBJECTIVES A previously synthesized ethyl acetohydroxamate-chalcones (L1-L22) were studied for their inhibitory activities against human recombinant monoamine oxidase A and B (hMAO-A and hMAO-B, respectively) and acetylcholinesterase (AChE) as multi-target directed ligands for the treatment of Alzheimer's Disease (AD). METHODS Enzyme inhibition studies of MAO-A, MAO-B and AChE is carried out. Computational studies such as Molecular docking, Molecular Mechanics/Generalized Born Surface Area calculations, ADMET prediction, and protein target prediction are also performed. RESULTS Among the screened compounds, compound L3 has most potent hMAO-B inhibition with an IC50 value of 0.028 ± 0.0016 µM, and other compounds, L1, L2, L4, L8, L12, and L21 showed significant potent hMAO-B inhibition with IC50 values of 0.051 ± 0.0014, 0.086 ± 0.0035, 0.036 ± 0.0011, 0.096 ± 0.0061, 0.083 ± 0.0016, and 0.038 ± 0.0021 µM, respectively. On the other hand, among the tested compounds, compound L13 showed highest hMAO-A inhibition with an IC50 value of 0.51± 0.051 µM and L9 has a significant value of 1.85 ± 0.045 µM. However, the compounds L3 and L4 only showed high selectivities for hMAO-B with Selectivity Index (SI) values of 621.4 and 416.7, respectively. Among the substituents in ring A of ethyl acetohydroxamate-chalcones (L1-L9), F atom at p-position (L3) showed highest inhibitory effect against hMAO-B. This result supports the uniqness and bizarre behavior of fluorine. Moreover, chalcones L3, L4, L9, L11, and L12 showed potential AChE inhibitory effect with IC50 values of 0.67, 0.85, 0.39, 0.30, and 0.45 µM, respectively. Inhibitions of hMAO-B by L3 or L4 were recovered to the level of the reversible reference (lazabemide), and were competitive with Ki values of 0.0030 ± 0.0002 and 0.0046 ± 0.0005 µM, respectively. Inhibitions of AChE by L3 and L11 were of the competitive and mixed types with Ki values of 0.30 ± 0.044 and 0.14 ± 0.0054 µM, respectively. CONCLUSION The studies indicated that L3 and L4 are considered to be promising multitarget drug molecules with potent, selective, and reversible competitive inhibitors of hMAO-B and with highly potent AChE inhibitory effect.
Collapse
Affiliation(s)
- Reeta
- Centre for Fire, Explosive and Environment Saftey, DRDO, Delhi, India.,Department of Chemistry, University of Delhi, Delhi, India
| | - Seung Cheol Baek
- Department of Pharmacy, and Research Institute of Life Pharmaceutical Sciences, Sunchon National University, Suncheon 57922, Korea
| | - Jae Pil Lee
- Department of Pharmacy, and Research Institute of Life Pharmaceutical Sciences, Sunchon National University, Suncheon 57922, Korea
| | - T M Rangarajan
- Department of Chemistry, Sri Venketeswara College, University of Delhi, New Delhi, India
| | - Ayushee
- Department of Chemistry, University of Delhi, Delhi, India
| | - Rishi Pal Singh
- Department of Chemistry, Sri Venketeswara College, University of Delhi, New Delhi, India
| | - Manjula Singh
- Department of Chemistry, Shivaji College, University of Delhi, New Delhi, India
| | | | - Orazio Nicolotti
- Dipartimento di Farmacia- Scienze del Farmaco, Universitá degli Studi di Bari "Aldo Moro", via E. Orabona, 4, I-70125 Bari, Italy
| | - Hoon Kim
- Department of Pharmacy, and Research Institute of Life Pharmaceutical Sciences, Sunchon National University, Suncheon 57922, Korea
| | - Bijo Mathew
- Division of Drug Design and Medicinal Chemistry Research Lab, Department of Pharmaceutical Chemistry, Ahalia School of Pharmacy, Palakkad-678557, Kerala, India
| |
Collapse
|
24
|
Gouda MA, Abu‐Hashem AA, Salem MA, Helal MH, Al‐Ghorbani M, Hamama WS. Recent progress on coumarinscaffold‐basedanti‐microbial agents (PartIII). J Heterocycl Chem 2020. [DOI: 10.1002/jhet.4100] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Moustafa. A. Gouda
- Department of Chemistry Faculty of Science and Arts, Ulla, Taibah University Medina Saudi Arabia
- Department of Chemistry Faculty of Science, Mansoura University Mansoura Egypt
| | - Ameen A. Abu‐Hashem
- Photochemistry Department (Heterocyclic Unit) National Research Centre Giza Egypt
- Chemistry Department Faculty of Science, Jazan University Jazan Saudi Arabia
| | - Mohammed A. Salem
- Department of Chemistry Faculty of Science and Arts, King Khalid University Mohail Assir Saudi Arabia
- Department of Chemistry Faculty of Science, Al‐Azhar University Nasr City Cairo Egypt
| | - Mohamed H. Helal
- Department of Chemistry Faculty of Science, Al‐Azhar University Nasr City Cairo Egypt
- Department of Chemistry Faculty of Arts and Science, Northern Border University Rafha Saudi Arabia
| | - Mohammed Al‐Ghorbani
- Department of Chemistry Faculty of Science and Arts, Ulla, Taibah University Medina Saudi Arabia
- Department of Chemistry Faculty of Science, Thamar University Thamar Yemen
| | - Wafaa S. Hamama
- Department of Chemistry Faculty of Science, Mansoura University Mansoura Egypt
| |
Collapse
|
25
|
The structure-based optimization of δ-sultone-fused pyrazoles as selective BuChE inhibitors. Eur J Med Chem 2020; 201:112273. [DOI: 10.1016/j.ejmech.2020.112273] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 03/20/2020] [Accepted: 03/23/2020] [Indexed: 11/23/2022]
|
26
|
Xie SS, Liu J, Tang C, Pang C, Li Q, Qin Y, Nong X, Zhang Z, Guo J, Cheng M, Tang W, Liang N, Jiang N. Design, synthesis and biological evaluation of rasagiline-clorgyline hybrids as novel dual inhibitors of monoamine oxidase-B and amyloid-β aggregation against Alzheimer’s disease. Eur J Med Chem 2020; 202:112475. [DOI: 10.1016/j.ejmech.2020.112475] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 04/13/2020] [Accepted: 05/15/2020] [Indexed: 01/07/2023]
|
27
|
Canale V, Grychowska K, Kurczab R, Ryng M, Keeri AR, Satała G, Olejarz-Maciej A, Koczurkiewicz P, Drop M, Blicharz K, Piska K, Pękala E, Janiszewska P, Krawczyk M, Walczak M, Chaumont-Dubel S, Bojarski AJ, Marin P, Popik P, Zajdel P. A dual-acting 5-HT 6 receptor inverse agonist/MAO-B inhibitor displays glioprotective and pro-cognitive properties. Eur J Med Chem 2020; 208:112765. [PMID: 32949963 DOI: 10.1016/j.ejmech.2020.112765] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 08/03/2020] [Accepted: 08/15/2020] [Indexed: 01/06/2023]
Abstract
The complex etiology of Alzheimer's disease has initiated a quest for multi-target ligands to address the multifactorial causes of this neurodegenerative disorder. In this context, we designed dual-acting 5-HT6 receptor (5-HT6R) antagonists/MAO-B inhibitors using pharmacophore hybridization strategy. Our approach involved linking priviliged scaffolds of 5-HT6R with aryloxy fragments derived from reversible and irreversible MAO-B inhibitors. The study identified compound 48 that acts as an inverse agonist of 5-HT6R at Gs signaling and an irreversible MAO-B inhibitor. Compound 48 showed moderate metabolic stability in rat microsomal assay, artificial membrane permeability, no hepatotoxicity, and it was well distributed to the brain. Additionally, 48 showed glioprotective properties in a model of cultured astrocytes using 6-OHDA as the cytotoxic agent. Finally, compound 48 (MED = 1 mg/kg, p.o.) fully reversed memory deficits in the NOR task induced by scopolamine in rats. A better understanding of effects exerted by dual-acting 5-HT6R/MAO-B modulators may impact the future development of neurodegenerative-directed treatment strategies.
Collapse
Affiliation(s)
- Vittorio Canale
- Jagiellonian University Medical College, Department of Medicinal Chemistry, 9 Medyczna Str., 30-688, Kraków, Poland
| | - Katarzyna Grychowska
- Jagiellonian University Medical College, Department of Medicinal Chemistry, 9 Medyczna Str., 30-688, Kraków, Poland
| | - Rafał Kurczab
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Medicinal Chemistry, 12 Smętna Str., 31-324, Kraków, Poland
| | - Mateusz Ryng
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Medicinal Chemistry, 12 Smętna Str., 31-324, Kraków, Poland
| | - Abdul Raheem Keeri
- Jagiellonian University Medical College, Department of Medicinal Chemistry, 9 Medyczna Str., 30-688, Kraków, Poland
| | - Grzegorz Satała
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Medicinal Chemistry, 12 Smętna Str., 31-324, Kraków, Poland
| | - Agnieszka Olejarz-Maciej
- Jagiellonian University Medical College, Department of Technology and Biotechnology of Drugs, 9 Medyczna Str., 30-688, Kraków, Poland
| | - Paulina Koczurkiewicz
- Jagiellonian University Medical College, Department of Pharmaceutical Biochemistry, 9 Medyczna Str., 30-688, Kraków, Poland
| | - Marcin Drop
- Jagiellonian University Medical College, Department of Medicinal Chemistry, 9 Medyczna Str., 30-688, Kraków, Poland
| | - Klaudia Blicharz
- Jagiellonian University Medical College, Department of Medicinal Chemistry, 9 Medyczna Str., 30-688, Kraków, Poland
| | - Kamil Piska
- Jagiellonian University Medical College, Department of Pharmaceutical Biochemistry, 9 Medyczna Str., 30-688, Kraków, Poland
| | - Elżbieta Pękala
- Jagiellonian University Medical College, Department of Pharmaceutical Biochemistry, 9 Medyczna Str., 30-688, Kraków, Poland
| | - Paulina Janiszewska
- Jagiellonian University Medical College, Department of Toxicology, 9 Medyczna Str., 30-688, Kraków, Poland
| | - Martyna Krawczyk
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of New Drug Development, 12 Smętna Str., 31-324, Kraków, Poland
| | - Maria Walczak
- Jagiellonian University Medical College, Department of Toxicology, 9 Medyczna Str., 30-688, Kraków, Poland
| | - Severine Chaumont-Dubel
- Institut de Génomique Fonctionelle, Université de Montpellier, CNRS INSERM, 34094, Montpellier, France
| | - Andrzej J Bojarski
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Medicinal Chemistry, 12 Smętna Str., 31-324, Kraków, Poland
| | - Philippe Marin
- Institut de Génomique Fonctionelle, Université de Montpellier, CNRS INSERM, 34094, Montpellier, France
| | - Piotr Popik
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of New Drug Development, 12 Smętna Str., 31-324, Kraków, Poland
| | - Paweł Zajdel
- Jagiellonian University Medical College, Department of Medicinal Chemistry, 9 Medyczna Str., 30-688, Kraków, Poland.
| |
Collapse
|
28
|
Shi Z, Li N, Chen C, Wang Y, Lei Z, Chen L, Sun J. Novel NO-releasing scopoletin derivatives induce cell death via mitochondrial apoptosis pathway and cell cycle arrest. Eur J Med Chem 2020; 200:112386. [DOI: 10.1016/j.ejmech.2020.112386] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 04/23/2020] [Accepted: 04/23/2020] [Indexed: 01/15/2023]
|
29
|
Kavully FS, Oh JM, Dev S, Kaipakasseri S, Palakkathondi A, Vengamthodi A, Abdul Azeez RF, Tondo AR, Nicolotti O, Kim H, Bijo Mathew. Design of enamides as new selective monoamine oxidase-B inhibitors. J Pharm Pharmacol 2020; 72:916-926. [PMID: 32246471 DOI: 10.1111/jphp.13264] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 03/08/2020] [Indexed: 12/24/2022]
Abstract
OBJECTIVES To develop of new class of selective and reversible MAO-B inhibitors from enamides. METHODS Syntheses of the titled derivatives (AD1-AD11) were achieved by reacting cinnamoyl chloride and various primary and secondary amines in basic medium. All eleven compounds were investigated for in vitro inhibitory activities against recombinant human MAO-A and MAO-B. The reversibilities of lead compound inhibitions were analysed by dialysis. MTT assays of lead compounds were performed using normal VERO cell lines. KEY FINDINGS Compounds AD3 and AD9 exhibited the greatest inhibitory activity against MAO-B with IC50 values of 0.11 and 0.10 µm, respectively, and were followed by AD2 and AD1 (0.51 and 0.71 µm, respectively). Most of the compounds weakly inhibited MAO-A, with the exceptions AD9 and AD7, which had IC50 values of 4.21 and 5.95 µm, respectively. AD3 had the highest selectivity index (SI) value for MAO-B (>363.6) and was followed by AD9 (SI 42.1). AD3 and AD9 were found to be competitive inhibitors of MAO-B with Ki values of 0.044 ± 0.0036 and 0.039 ± 0.0047 µm, respectively. Reversibility experiments showed AD3 and AD9 were reversible inhibitors of MAO-B; dialysis restored the activity of MAO-B to the reference level. MTT assays revealed AD3 and AD9 were non-toxic to normal VERO cell lines with IC50 values of 153.96 and 194.04 µg/ml, respectively. Computational studies provided hypothetical binding modes for AD3 and AD9 in the binding cavities of MAO-A and MAO-B. CONCLUSIONS These results encourage further studies on the enamide scaffold as potential drug candidates for the treatment of Alzheimer's and Parkinson's diseases.
Collapse
Affiliation(s)
- Fathima Sahla Kavully
- Department of Pharmaceutical Chemistry, Al-Shifa College of Pharmacy, Perinthalmanna, India
| | - Jong Min Oh
- Department of Pharmacy, and Research Institute of Life Pharmaceutical Sciences, Sunchon National University, Suncheon, Korea
| | - Sanal Dev
- Department of Pharmaceutical Chemistry, Al-Shifa College of Pharmacy, Perinthalmanna, India
| | - Swafvan Kaipakasseri
- Department of Pharmaceutical Chemistry, Al-Shifa College of Pharmacy, Perinthalmanna, India
| | - Ashique Palakkathondi
- Department of Pharmaceutical Chemistry, Al-Shifa College of Pharmacy, Perinthalmanna, India
| | - Ajeesh Vengamthodi
- Department of Pharmaceutical Chemistry, Al-Shifa College of Pharmacy, Perinthalmanna, India
| | | | - Anna Rita Tondo
- Instituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Orazio Nicolotti
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari "Aldo Moro", Bari, Italy
| | - Hoon Kim
- Department of Pharmacy, and Research Institute of Life Pharmaceutical Sciences, Sunchon National University, Suncheon, Korea
| | - Bijo Mathew
- Division of Drug Design and Medicinal Chemistry Research Lab, Department of Pharmaceutical Chemistry, Ahalia School of Pharmacy, Palakkad, India
| |
Collapse
|
30
|
Rana M, Pareek A, Bhardwaj S, Arya G, Nimesh S, Arya H, Bhatt TK, Yaragorla S, Sharma AK. Aryldiazoquinoline based multifunctional small molecules for modulating Aβ42aggregation and cholinesterase activity related to Alzheimer's disease. RSC Adv 2020; 10:28827-28837. [PMID: 35520091 PMCID: PMC9055851 DOI: 10.1039/d0ra05172a] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 07/22/2020] [Indexed: 12/12/2022] Open
Abstract
Novel series of aryldiazoquinoline multifunctional molecules controls amyloid formation and neuro-protective role by inhibiting esterase enzymes.
Collapse
Affiliation(s)
- Monika Rana
- Department of Chemistry
- Central University of Rajasthan
- Ajmer
- India
| | - Abhishek Pareek
- School of Chemistry
- University of Hyderabad
- P.O. Central University
- Hyderabad
- India
| | - Shivani Bhardwaj
- Department of Chemistry
- Central University of Rajasthan
- Ajmer
- India
| | - Geeta Arya
- Department of Biotechnology
- Central University of Rajasthan
- Ajmer
- India
| | - Surendra Nimesh
- Department of Biotechnology
- Central University of Rajasthan
- Ajmer
- India
| | - Hemant Arya
- Department of Biotechnology
- Central University of Rajasthan
- Ajmer
- India
| | - Tarun K. Bhatt
- Department of Biotechnology
- Central University of Rajasthan
- Ajmer
- India
| | | | - Anuj K. Sharma
- Department of Chemistry
- Central University of Rajasthan
- Ajmer
- India
| |
Collapse
|
31
|
Lemke C, Christmann J, Yin J, Alonso JM, Serrano E, Chioua M, Ismaili L, Martínez-Grau MA, Beadle CD, Vetman T, Dato FM, Bartz U, Elsinghorst PW, Pietsch M, Müller CE, Iriepa I, Wille T, Marco-Contelles J, Gütschow M. Chromenones as Multineurotargeting Inhibitors of Human Enzymes. ACS OMEGA 2019; 4:22161-22168. [PMID: 31891098 PMCID: PMC6933783 DOI: 10.1021/acsomega.9b03409] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 11/21/2019] [Indexed: 06/01/2023]
Abstract
The complex nature of multifactorial diseases, such as Morbus Alzheimer, has produced a strong need to design multitarget-directed ligands to address the involved complementary pathways. We performed a purposive structural modification of a tetratarget small-molecule, that is contilisant, and generated a combinatorial library of 28 substituted chromen-4-ones. The compounds comprise a basic moiety which is linker-connected to the 6-position of the heterocyclic chromenone core. The syntheses were accomplished by Mitsunobu- or Williamson-type ether formations. The resulting library members were evaluated at a panel of seven human enzymes, all of which being involved in the pathophysiology of neurodegeneration. A concomitant inhibition of human acetylcholinesterase and human monoamine oxidase B, with IC50 values of 5.58 and 7.20 μM, respectively, was achieved with the dual-target 6-(4-(piperidin-1-yl)butoxy)-4H-chromen-4-one (7).
Collapse
Affiliation(s)
- Carina Lemke
- Pharmaceutical
Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany
| | - Joscha Christmann
- Bundeswehr
Institute of Pharmacology and Toxicology, Neuherbergstraße 11, 80937 München, Germany
| | - Jiafei Yin
- Pharmaceutical
Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany
| | - José M. Alonso
- Laboratory
of Medicinal Chemistry, IQOG, CSIC, C/Juan de la Cierva 3, 28006 Madrid, Spain
| | - Estefanía Serrano
- Laboratory
of Medicinal Chemistry, IQOG, CSIC, C/Juan de la Cierva 3, 28006 Madrid, Spain
| | - Mourad Chioua
- Laboratory
of Medicinal Chemistry, IQOG, CSIC, C/Juan de la Cierva 3, 28006 Madrid, Spain
| | - Lhassane Ismaili
- Neurosciences
intégratives et cliniques EA 481, Pôle de Chimie Organique
et Thérapeutique, Univ. Bourgogne
Franche-Comté, UFR Santé, 19, rue Ambroise Paré, 25000 Besançon, France
| | | | - Christopher D. Beadle
- Lilly Research
Centre, Eli Lilly & Company, Erl Wood Manor,
Windlesham, Surrey GU20 6PH, U.K.
| | - Tatiana Vetman
- Lilly
Research Laboratories, Eli Lilly & Company, Indianapolis 46285, Indiana, United States
| | - Florian M. Dato
- Institute
II of Pharmacology, Center of Pharmacology, Medical Faculty, University of Cologne, Gleueler Strasse 24, 50931 Cologne, Germany
| | - Ulrike Bartz
- Department
of Natural Sciences, University of Applied
Sciences Bonn-Rhein-Sieg, von-Liebig-Strasse 20, 53359 Rheinbach, Germany
| | - Paul W. Elsinghorst
- Pharmaceutical
Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany
- Central
Institute of the Bundeswehr Medical Service Munich, Ingolstädter Landstraße
102, 85748 Garching, Germany
| | - Markus Pietsch
- Institute
II of Pharmacology, Center of Pharmacology, Medical Faculty, University of Cologne, Gleueler Strasse 24, 50931 Cologne, Germany
| | - Christa E. Müller
- Pharmaceutical
Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany
| | - Isabel Iriepa
- Department of Organic and Inorganic Chemistry, University of Alcalá, Ctra. Madrid-Barcelona, Km. 33,6, 28871 Alcalá de Henares, Madrid, Spain
| | - Timo Wille
- Bundeswehr
Institute of Pharmacology and Toxicology, Neuherbergstraße 11, 80937 München, Germany
| | - José Marco-Contelles
- Laboratory
of Medicinal Chemistry, IQOG, CSIC, C/Juan de la Cierva 3, 28006 Madrid, Spain
| | - Michael Gütschow
- Pharmaceutical
Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany
| |
Collapse
|
32
|
Hasan AH, Amran SI, Saeed Hussain FH, Jaff BA, Jamalis J. Molecular Docking and Recent Advances in the Design and Development of Cholinesterase Inhibitor Scaffolds: Coumarin Hybrids. ChemistrySelect 2019. [DOI: 10.1002/slct.201903607] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Aso Hameed Hasan
- Department of ChemistryFaculty of ScienceUniversiti Teknologi Malaysia 81310 Johor Bahru, Johor Malaysia
- Department of ChemistryCollege of ScienceUniversity of Garmian- Kalar, Kurdistan Region-Iraq Iraq
| | - Syazwani Itri Amran
- Department of BiosciencesFaculty of ScienceUniversiti Teknologi Malaysia 81310 Johor Bahru, Johor Malaysia
| | | | - Baram Ahmed Jaff
- Charmo Research CenterChemistry DepartmentCharmo University 46023 Chamchamal, Kurdistan Region-Iraq Iraq
| | - Joazaizulfazli Jamalis
- Department of ChemistryFaculty of ScienceUniversiti Teknologi Malaysia 81310 Johor Bahru, Johor Malaysia
| |
Collapse
|
33
|
Pourshojaei Y, Eskandari K, Asadipour A. Highly Significant Scaffolds to Design and Synthesis Cholinesterase Inhibitors as Anti-Alzheimer Agents. Mini Rev Med Chem 2019; 19:1577-1598. [DOI: 10.2174/1389557519666190719143112] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Revised: 06/02/2019] [Accepted: 06/25/2019] [Indexed: 12/19/2022]
Abstract
:
Alzheimer, a progressive disease, is a common term for memory loss which interferes with
daily life through severe influence on cognitive abilities. Based on the cholinergic hypothesis, and Xray
crystallographic determination of the structure of acetylcholinesterase (AChE) enzyme, the level of
acetylcholine (ACh, an important neurotransmitter associated with memory) in the hippocampus and
cortex area of the brain has a direct effect on Alzheimer. This fact encourages scientists to design and
synthesize a wide range of acetylcholinesterase inhibitors (AChEIs) to control the level of ACh in the
brain, keeping in view the crystallographic structure of AChE enzyme and drugs approved by the Food
and Drug Administration (FDA).
:
AChEIs have slightly diverse pharmacological properties, but all of them work by inhibiting the segregation
of ACh by blocking AChE. We reviewed significant scaffolds introduced as AChEIs. In some
studies, the activity against butyrylcholinesterase (BuChE) has been evaluated as well because BuChE
is a similar enzyme to neuronal acetylcholinesterase and is capable of hydrolyzing ACh. In order to
study AChEIs effectively, we divided them structurally into 12 classes and briefly explained effective
AChEIs and compared their activities against AChE enzyme.
Collapse
Affiliation(s)
- Yaghoub Pourshojaei
- Department of Medicinal Chemistry, Faculty of Pharmacy & Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Khalil Eskandari
- Department of Medicinal Chemistry, Faculty of Pharmacy & Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Ali Asadipour
- Department of Medicinal Chemistry, Faculty of Pharmacy & Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
34
|
Fu J, Bao F, Gu M, Liu J, Zhang Z, Ding J, Xie SS, Ding J. Design, synthesis and evaluation of quinolinone derivatives containing dithiocarbamate moiety as multifunctional AChE inhibitors for the treatment of Alzheimer's disease. J Enzyme Inhib Med Chem 2019; 35:118-128. [PMID: 31694418 PMCID: PMC6844382 DOI: 10.1080/14756366.2019.1687460] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
A series of novel quinolinone derivatives bearing dithiocarbamate moiety were designed and synthesised as multifunctional AChE inhibitors for the treatment of AD. Most of these compounds exhibited strong and clearly selective inhibition to eeAChE. Among them, compound 4c was identified as the most potent inhibitor to both eeAChE and hAChE (IC50 = 0.22 μM for eeAChE; IC50 = 0.16 μM for hAChE), and it was also the best inhibitor to AChE-induced Aβ aggregation (29.02% at 100 μM) and an efficient inhibitor to self-induced Aβ aggregation (30.67% at 25 μM). Kinetic and molecular modelling studies indicated that compound 4c was a mixed-type inhibitor, which could interact simultaneously with the catalytic anionic site (CAS) and the peripheral anionic site (PAS) of AChE. In addition, 4c had good ability to cross the BBB, showed no toxicity on SH-SY5Y neuroblastoma cells and was well tolerated in mice at doses up to 2500 mg/kg (po).
Collapse
Affiliation(s)
- Jie Fu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China.,Jiangsu Zeyun Pharmaceutical Co., Ltd, Xibei Town Industrial Park, Wuxi, China
| | - Fengqi Bao
- Jiangsu Zeyun Pharmaceutical Co., Ltd, Xibei Town Industrial Park, Wuxi, China
| | - Min Gu
- Jiangsu Zeyun Pharmaceutical Co., Ltd, Xibei Town Industrial Park, Wuxi, China
| | - Jing Liu
- School of Pharmacy, Jiangxi University of Traditional Chinese Medicine, Nanchang, PR China
| | - Zhipeng Zhang
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Jiaoli Ding
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Sai-Sai Xie
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Jinsong Ding
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| |
Collapse
|
35
|
Paramonova MP, Ozerov AA, Chizhov AO, Snoeck R, Andrei G, Khandazhinskaya AL, Novikov MS. Synthesis of uracil–coumarin conjugates as potential inhibitors of virus replication. MENDELEEV COMMUNICATIONS 2019. [DOI: 10.1016/j.mencom.2019.11.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
36
|
Parambi DGT, Oh JM, Baek SC, Lee JP, Tondo AR, Nicolotti O, Kim H, Mathew B. Design, synthesis and biological evaluation of oxygenated chalcones as potent and selective MAO-B inhibitors. Bioorg Chem 2019; 93:103335. [PMID: 31606547 DOI: 10.1016/j.bioorg.2019.103335] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Revised: 09/30/2019] [Accepted: 10/02/2019] [Indexed: 01/10/2023]
Abstract
The present study documents the synthesis of oxygenated chalcone (O1-O26) derivatives and their abilities to inhibit monoamine oxidases. All 26 derivatives examined showed potent inhibitory activity against MAO-B. Compound O23 showed the greatest inhibitory activity against MAO-B with an IC50 value of 0.0021 µM, followed by compounds O10 and O17 (IC50 = 0.0030 and 0.0034 µM, respectively). In addition, most of the derivatives potently inhibited MAO-A and O6 was the most potent inhibitor with an IC50 value of 0.029 µM, followed by O3, O4, O9, and O2 (IC50 = 0.035, 0.053, 0.072, and 0.082 µM, respectively). O23 had a high selectivity index (SI) value for MAO-B of 138.1, and O20 (IC50 value for MAO-B = 0.010 µM) had an extremely high SI of >4000. In dialysis experiments, inhibitions of MAO-A and MAO-B by O6 and O23, respectively, were recovered to their respective reversible reference levels, demonstrating both are reversible inhibitors. Kinetic studies revealed that O6 and O23 competitively inhibited MAO-A and MAO-B, respectively, with respective Ki values of 0.016 ± 0.0007 and 0.00050 ± 0.00003 µM. Lead compound are also non-toxic at 200 µg/mL in normal rat spleen cells. Molecular docking simulations and subsequent Molecular Mechanics/Generalized Born Surface Area calculations provided a rationale that explained experimental data.
Collapse
Affiliation(s)
| | - Jong Min Oh
- Department of Pharmacy, and Research Institute of Life Pharmaceutical Sciences, Sunchon National University, Suncheon 57922, Republic of Korea
| | - Seung Cheol Baek
- Department of Pharmacy, and Research Institute of Life Pharmaceutical Sciences, Sunchon National University, Suncheon 57922, Republic of Korea
| | - Jae Pil Lee
- Department of Pharmacy, and Research Institute of Life Pharmaceutical Sciences, Sunchon National University, Suncheon 57922, Republic of Korea
| | - Anna Rita Tondo
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via la Masa 19, 20156 Milano, Italy
| | - Orazio Nicolotti
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari "Aldo Moro", via E. Orabona, 4, I-70125 Bari, Italy
| | - Hoon Kim
- Department of Pharmacy, and Research Institute of Life Pharmaceutical Sciences, Sunchon National University, Suncheon 57922, Republic of Korea.
| | - Bijo Mathew
- Division of Drug Design and Medicinal Chemistry Research Lab, Department of Pharmaceutical Chemistry, Ahalia School of Pharmacy, Palakkad 678557, Kerala, India.
| |
Collapse
|
37
|
Mathew B, Parambi DGT, Mathew GE, Uddin MS, Inasu ST, Kim H, Marathakam A, Unnikrishnan MK, Carradori S. Emerging therapeutic potentials of dual-acting MAO and AChE inhibitors in Alzheimer's and Parkinson's diseases. Arch Pharm (Weinheim) 2019; 352:e1900177. [PMID: 31478569 DOI: 10.1002/ardp.201900177] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 07/26/2019] [Accepted: 08/01/2019] [Indexed: 01/07/2023]
Abstract
No drug has been approved to prevent neuronal cell loss in patients suffering from Parkinson's disease (PD) or Alzheimer's disease (AD); despite increased comprehension of the underlying molecular causes, therapies target cognitive functional improvement and motor fluctuation control. Drug design strategies that adopt the "one protein, one target" philosophy fail to address the multifactorial aetiologies of neurodegenerative disorders such as AD and PD optimally. On the contrary, restoring neurotransmitter levels by combined combinatorial inhibition of cholinesterases, monoamine oxidases, and adenosine A2A A receptors, in conjunction with strategies to counter oxidative stress and beta-amyloid plaque accumulation, would constitute a therapeutically robust, multitarget approach. This extensive review delineates the therapeutic advantages of combining dual-acting molecules that inhibit monoamine oxidases and cholinesterases and/or adenosine A2A A receptors, and describes the structure-activity relationships of compound classes that include, but are not limited to, alkaloids, coumarins, chalcones, donepezil-propargylamine conjugates, homoisoflavonoids, resveratrol analogs, hydrazones, and pyrazolines. In the wake of recent advances in network biology, in silico approaches, and omics, this review emphasizes the need to consider conceptually informed research strategies for drug discovery, in the context of the mounting burden posed by chronic neurodegenerative diseases with complex aetiologies and pathophysiologies involving multiple signalling pathways and numerous drug targets.
Collapse
Affiliation(s)
- Bijo Mathew
- Department of Pharmaceutical Chemistry Research Lab, Division of Drug Design and Medicinal Chemistry, Ahalia School of Pharmacy, Palakkad, India
| | - Della G T Parambi
- Department of Pharmaceutical Chemistry, Jouf University, Sakaka, Saudi Arabia
| | - Githa E Mathew
- Department of Pharmacology, Grace College of Pharmacy, Palakkad, India
| | - Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh.,Pharmakon Neuroscience Research Network, Dhaka, Bangladesh
| | - Sini T Inasu
- Department of Pharmaceutical Chemistry Research Lab, Division of Drug Design and Medicinal Chemistry, Ahalia School of Pharmacy, Palakkad, India
| | - Hoon Kim
- Department of Pharmacy and Research, Institute of Life Pharmaceutical Sciences, Sunchon National University, Suncheon, Republic of Korea
| | - Akash Marathakam
- Department of Pharmaceutical Chemistry, National College of Pharmacy, Calicut, India
| | | | - Simone Carradori
- Department of Pharmacy, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| |
Collapse
|
38
|
Singh H, Singh JV, Bhagat K, Gulati HK, Sanduja M, Kumar N, Kinarivala N, Sharma S. Rational approaches, design strategies, structure activity relationship and mechanistic insights for therapeutic coumarin hybrids. Bioorg Med Chem 2019; 27:3477-3510. [PMID: 31255497 PMCID: PMC7970831 DOI: 10.1016/j.bmc.2019.06.033] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 06/12/2019] [Accepted: 06/19/2019] [Indexed: 01/01/2023]
Abstract
Hybrid molecules, furnished by combining two or more pharmacophores is an emerging concept in the field of medicinal chemistry and drug discovery that has attracted substantial traction in the past few years. Naturally occurring scaffolds such as coumarins display a wide spectrum of pharmacological activities including anticancer, antibiotic, antidiabetic and others, by acting on multiple targets. In this view, various coumarin-based hybrids possessing diverse medicinal attributes were synthesized in the last five years by conjugating coumarin moiety with other therapeutic pharmacophores. The current review summarizes the recent development (2014 and onwards) of these pharmacologically active coumarin hybrids and demonstrates rationale behind their design, structure-activity relationships (SAR) and mechanistic studies performed on these hybrid molecules. This review will be beneficial for medicinal chemist and chemical biologist, and in general to the drug discovery community and will facilitate the synthesis and development of novel, potent coumarin hybrid molecules serving as lead molecules for the treatment of complex disorders.
Collapse
Affiliation(s)
- Harbinder Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar 143005, Punjab, India
| | - Jatinder Vir Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar 143005, Punjab, India
| | - Kavita Bhagat
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar 143005, Punjab, India
| | - Harmandeep Kaur Gulati
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar 143005, Punjab, India
| | - Mohit Sanduja
- School of Pharmaceutical Sciences, MVN University, Palwal 121105, Haryana, India
| | - Nitish Kumar
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar 143005, Punjab, India
| | - Nihar Kinarivala
- Program in Chemical Biology, Sloan Kettering Institute, New York, NY 10065, USA.
| | - Sahil Sharma
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar 143005, Punjab, India; Program in Chemical Biology, Sloan Kettering Institute, New York, NY 10065, USA.
| |
Collapse
|
39
|
Guglielmi P, Carradori S, Ammazzalorso A, Secci D. Novel approaches to the discovery of selective human monoamine oxidase-B inhibitors: is there room for improvement? Expert Opin Drug Discov 2019; 14:995-1035. [PMID: 31268358 DOI: 10.1080/17460441.2019.1637415] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Introduction: Selective monoamine oxidase-B (MAO-B) inhibitors are currently used as coadjuvants for the treatment of early motor symptoms in Parkinson's disease. They can, based on their chemical structure and mechanism of inhibition, be categorized into reversible and irreversible agents. Areas covered: This review provides a comprehensive update on the development state of selective MAO-B inhibitors describing the results, structures, structure-activity relationships (SARs) and Medicinal chemistry strategies as well as the related shortcomings over the past five years. Expert opinion: Researchers have explored and implemented new and old chemical scaffolds achieving high inhibitory potencies and isoform selectivity. Most of them were characterized and proposed as multitarget agents able to act at different levels (including AChE inhibition, H3R or A2AR antagonism, antioxidant and chelating properties, Aβ1-42 aggregation reduction) in the network of aetiologies of neurodegenerative disorders. These results can also be used to avoid 'cheese-reaction' effects and the occurrence of serotonergic syndrome in patients.
Collapse
Affiliation(s)
- Paolo Guglielmi
- Dipartimento di Chimica e Tecnologie del Farmaco, Sapienza Università di Roma , Rome , Italy
| | - Simone Carradori
- Department of Pharmacy, University "G. d'Annunzio" of Chieti-Pescara , Chieti , Italy
| | | | - Daniela Secci
- Dipartimento di Chimica e Tecnologie del Farmaco, Sapienza Università di Roma , Rome , Italy
| |
Collapse
|
40
|
Lakshminarayanan B, Baek SC, Lee JP, Kannappan N, Mangiatordi GF, Nicolotti O, Subburaju T, Kim H, Mathew B. Ethoxylated Head of Chalcones as a New Class of Multi‐Targeted MAO Inhibitors. ChemistrySelect 2019. [DOI: 10.1002/slct.201901093] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Balasubramanian Lakshminarayanan
- Division of Drug Design and Medicinal Chemistry Research LabDepartment of Pharmaceutical ChemistryAhalia School of Pharmacy Palakkad- 678557, Kerala India
- Department of PharmacyAnnamalai University Chidambaram- 608002, Tamilnadu India
| | - Seung Cheol Baek
- Department of PharmacyResearch Institute of Life Pharmaceutical SciencesSunchon National University Suncheon 57922 Republic of Korea
| | - Jae Pil Lee
- Department of PharmacyResearch Institute of Life Pharmaceutical SciencesSunchon National University Suncheon 57922 Republic of Korea
| | - Nagappan Kannappan
- Department of PharmacyAnnamalai University Chidambaram- 608002, Tamilnadu India
| | | | - Orazio Nicolotti
- Dipartimento di Farmacia-Scienze del FarmacoUniversita degli Studi di Bari “Aldo Moro”, Via E. Orabona, 4 I-70125 Bari Italy
| | - Thillainayagam Subburaju
- Division of Drug Design and Medicinal Chemistry Research LabDepartment of Pharmaceutical ChemistryAhalia School of Pharmacy Palakkad- 678557, Kerala India
| | - Hoon Kim
- Department of PharmacyResearch Institute of Life Pharmaceutical SciencesSunchon National University Suncheon 57922 Republic of Korea
| | - Bijo Mathew
- Division of Drug Design and Medicinal Chemistry Research LabDepartment of Pharmaceutical ChemistryAhalia School of Pharmacy Palakkad- 678557, Kerala India
| |
Collapse
|
41
|
Highly potent and selective aryl-1,2,3-triazolyl benzylpiperidine inhibitors toward butyrylcholinesterase in Alzheimer's disease. Bioorg Med Chem 2019; 27:931-943. [DOI: 10.1016/j.bmc.2018.12.030] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 12/18/2018] [Accepted: 12/21/2018] [Indexed: 11/16/2022]
|
42
|
Tang YW, Shi CJ, Yang HL, Cai P, Liu QH, Yang XL, Kong LY, Wang XB. Synthesis and evaluation of isoprenylation-resveratrol dimer derivatives against Alzheimer's disease. Eur J Med Chem 2019; 163:307-319. [PMID: 30529634 DOI: 10.1016/j.ejmech.2018.11.040] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 11/16/2018] [Accepted: 11/16/2018] [Indexed: 01/09/2023]
Abstract
A series of resveratrol dimer derivatives against Alzheimer's disease (AD) was obtained by structural modification and transformation using resveratrol as substrate. Biological analysis revealed that these derivatives had moderate inhibitory activity against human monoamine oxidase B (hMAO-B). In particular, 3 and 7 showed the better inhibitory activity for hMAO-B (IC50 = 3.91 ± 0.23 μM, 0.90 ± 0.01 μM) respectively. Compound 3 (IC50 = 46.95 ± 0.21 μM for DPPH, 1.43 and 1.74 trolox equivalent by ABTS and FRAP method respectively), and 7 (IC50 = 35.33 ± 0.15 μM for DPPH, 1.70 and 1.97 trolox equivalent by ABTS method and FRAP method respectively) have excellent antioxidant effects. Cellular assay shown that 3 and 7 had lower toxicity and were resistant to neurotoxicity induced by oxidative toxins (H2O2, rotenone and oligomycin-A). More importantly, the selected compounds have neuroprotective effects against ROS generation, H2O2-induced apoptosis and a significant in vitro anti-inflammatory activity. The results of the parallel artificial membrane permeability assay for blood-brain barrier indicated that 3 and 7 would be predominant to cross the blood-brain barrier. In this study, mouse microglia BV2 cells were used to establish cell oxidative stress injury model with H2O2 and to explore the protective effect and mechanism of 3 and 7. In general, 3 and 7 can be considered candidates for potential treatment of AD.
Collapse
Affiliation(s)
- Yan-Wei Tang
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Cun-Jian Shi
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Hua-Li Yang
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Pei Cai
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Qiao-Hong Liu
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Xue-Lian Yang
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Ling-Yi Kong
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| | - Xiao-Bing Wang
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
43
|
Tripathi RKP, Ayyannan SR. Monoamine oxidase-B inhibitors as potential neurotherapeutic agents: An overview and update. Med Res Rev 2019; 39:1603-1706. [PMID: 30604512 DOI: 10.1002/med.21561] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 12/13/2018] [Accepted: 12/15/2018] [Indexed: 12/23/2022]
Abstract
Monoamine oxidase (MAO) inhibitors have made significant contributions and remain an indispensable approach of molecular and mechanistic diversity for the discovery of antineurodegenerative drugs. However, their usage has been hampered by nonselective and/or irreversible action which resulted in drawbacks like liver toxicity, cheese effect, and so forth. Hence, the search for selective MAO inhibitors (MAOIs) has become a substantial focus in current drug discovery. This review summarizes our current understanding on MAO-A/MAO-B including their structure, catalytic mechanism, and biological functions with emphases on the role of MAO-B as a potential therapeutic target for the development of medications treating neurodegenerative disorders. It also highlights the recent developments in the discovery of potential MAO-B inhibitors (MAO-BIs) belonging to diverse chemical scaffolds, arising from intensive chemical-mechanistic and computational studies documented during past 3 years (2015-2018), with emphases on their potency and selectivity. Importantly, readers will gain knowledge of various newly established MAO-BI scaffolds and their development potentials. The comprehensive information provided herein will hopefully accelerate ideas for designing novel selective MAO-BIs with superior activity profiles and critical discussions will inflict more caution in the decision-making process in the MAOIs discovery.
Collapse
Affiliation(s)
- Rati Kailash Prasad Tripathi
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi, India.,Department of Pharmaceutical Chemistry, Parul Institute of Pharmacy, Parul University, Vadodara, India
| | - Senthil Raja Ayyannan
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi, India
| |
Collapse
|
44
|
Investigating alkyl nitrates as nitric oxide releasing precursors of multitarget acetylcholinesterase-monoamine oxidase B inhibitors. Eur J Med Chem 2019; 161:292-309. [DOI: 10.1016/j.ejmech.2018.10.016] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 10/04/2018] [Accepted: 10/08/2018] [Indexed: 12/15/2022]
|
45
|
Hong R, Li X. Discovery of monoamine oxidase inhibitors by medicinal chemistry approaches. MEDCHEMCOMM 2019; 10:10-25. [PMID: 30774851 PMCID: PMC6350766 DOI: 10.1039/c8md00446c] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 11/22/2018] [Indexed: 12/15/2022]
Abstract
Neuropsychiatric disorders, such as Alzheimer's disease (AD), Parkinson's disease (PD) and depression, have seriously inconvenienced the lives of patients. Growing evidence indicates that these diseases are closely related to the monoamine oxidase (MAO) enzyme, making it an attractive target for the exploitation of potent MAO inhibitors (MAOIs) with high selectivity and low side effects. Although various MAOIs have been discovered, the discovery of an ideal MAOI is not an easy task. In this review, we discuss the currently available rational design strategies for obtaining ideal MAOIs, including ligand-based and receptor-based design strategies, and these strategies were further illustrated with the aid of specific examples from the recent literature. To better understanding the biological activity of MAO, we also highlight the binding modes of typical inhibitors against MAO. Besides, advanced strategies for finding upcoming potent MAOIs were prospected.
Collapse
Affiliation(s)
- Renyuan Hong
- Department of Medicinal Chemistry , Key Laboratory of Chemical Biology (Ministry of Education) , School of Pharmaceutical Sciences , Shandong University , 44 West Culture Road , 250012 , Jinan , Shandong , P. R. China . ; ; Tel: 86 531 88382005
| | - Xun Li
- Department of Medicinal Chemistry , Key Laboratory of Chemical Biology (Ministry of Education) , School of Pharmaceutical Sciences , Shandong University , 44 West Culture Road , 250012 , Jinan , Shandong , P. R. China . ; ; Tel: 86 531 88382005
| |
Collapse
|
46
|
He Q, Liu J, Lan JS, Ding J, Sun Y, Fang Y, Jiang N, Yang Z, Sun L, Jin Y, Xie SS. Coumarin-dithiocarbamate hybrids as novel multitarget AChE and MAO-B inhibitors against Alzheimer’s disease: Design, synthesis and biological evaluation. Bioorg Chem 2018; 81:512-528. [DOI: 10.1016/j.bioorg.2018.09.010] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 09/05/2018] [Accepted: 09/07/2018] [Indexed: 12/30/2022]
|
47
|
Hu J, An B, Pan T, Li Z, Huang L, Li X. Design, synthesis, and biological evaluation of histone deacetylase inhibitors possessing glutathione peroxidase-like and antioxidant activities against Alzheimer’s disease. Bioorg Med Chem 2018; 26:5718-5729. [DOI: 10.1016/j.bmc.2018.10.022] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 10/22/2018] [Accepted: 10/23/2018] [Indexed: 12/18/2022]
|
48
|
Qiu GL, He SS, Chen SC, Li B, Wu HH, Zhang J, Tang WJ. Design, synthesis and biological evaluation of tricyclic pyrazolo[1,5-c][1,3]benzoxazin-5(5H)-one scaffolds as selective BuChE inhibitors. J Enzyme Inhib Med Chem 2018; 33:1506-1515. [PMID: 30284486 PMCID: PMC6179045 DOI: 10.1080/14756366.2018.1488696] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Based on the structural analysis of tricyclic scaffolds as butyrylcholinesterase (BuChE) inhibitors, a series of pyrazolo[1,5-c][1,3]benzoxazin-5(5H)-one derivatives were designed, synthesized and evaluated for their acetylcholinesterase (AChE) and BuChE inhibitory activity. Compounds with 5-carbonyl and 7- or/and 9-halogen substitutions showed potential BuChE inhibitory activity, among which compounds 6a, 6c and 6g showed the best BuChE inhibition (IC50 = 1.06, 1.63 and 1.63 µM, respectively). The structure–activity relationship showed that the 5-carbonyl and halogen substituents significantly influenced BuChE activity. Compounds 6a and 6g were found nontoxic, lipophilic and exhibited remarkable neuroprotective activity and mixed-type inhibition against BuChE (Ki = 7.46 and 3.09 µM, respectively). Docking studies revealed that compound 6a can be accommodated into BuChE via five hydrogen bonds, one Pi–Sigma interaction and three Pi–Alkyl interactions.
Collapse
Affiliation(s)
- Guo-Liang Qiu
- a School of Pharmacy , Anhui Medical University , Hefei , PR China
| | - Shao-Sheng He
- a School of Pharmacy , Anhui Medical University , Hefei , PR China.,b Lujiang County People's Hospital , Lujiang , Anhui , PR China
| | - Shi-Chao Chen
- a School of Pharmacy , Anhui Medical University , Hefei , PR China
| | - Bo Li
- a School of Pharmacy , Anhui Medical University , Hefei , PR China
| | - Hui-Hui Wu
- c Anhui Prevention and Treatment Center for Occupational Disease , Hefei , PR China
| | - Jing Zhang
- c Anhui Prevention and Treatment Center for Occupational Disease , Hefei , PR China
| | - Wen-Jian Tang
- a School of Pharmacy , Anhui Medical University , Hefei , PR China
| |
Collapse
|
49
|
Rullo M, Niso M, Pisani L, Carrieri A, Colabufo NA, Cellamare S, Altomare CD. 1,2,3,4-Tetrahydroisoquinoline/2H-chromen-2-one conjugates as nanomolar P-glycoprotein inhibitors: Molecular determinants for affinity and selectivity over multidrug resistance associated protein 1. Eur J Med Chem 2018; 161:433-444. [PMID: 30384046 DOI: 10.1016/j.ejmech.2018.10.043] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Revised: 10/15/2018] [Accepted: 10/16/2018] [Indexed: 11/30/2022]
Abstract
A series of coniugates bearing a 1,2,3,4-tetrahydroisoquinoline motif linked to substituted 7-hydroxy-2H-chromen-2-ones was synthesized and assayed through calcein-AM test in Madin-Darby Canine Kidney (MDCK) cells overexpressing P-glycoprotein (P-gp) and closely related multidrug resistance associated protein 1 (MRP1) to probe the interference with efflux mechanisms mediated by P-gp and MRP1, respectively. A number of substituents at C3 and C4 of coumarin nucleus along with differently sized and shaped spacers was enrolled to investigate the effects of focused structural modifications over affinity and selectivity. Linker length and flexibility played a key role in enhancing P-gp affinity as proved by the most potent P-gp modulator (3h, IC50 = 70 nM). A phenyl ring within the spacer (3k, 3l, 3o) and bulkier groups (Br in 3r, Ph in 3u) at coumarin C3 led to derivatives showing nanomolar activity (160 nM < IC50 < 280 nM) along with outstanding selectivity over MRP1 (SI > 350). Molecular docking calculations carried out on a human MDR1 homology model structure contributed to gain insights into the ligands' binding modes. Some compounds (3d, 3h, 3l, 3r, 3t, 3u) reversed MDR thereby restoring doxorubicin cytotoxicity when co-administered with the drug into MDCK-MDR1 cells.
Collapse
Affiliation(s)
- Mariagrazia Rullo
- Dipartimento di Farmacia-Scienze Del Farmaco, Università Degli Studi di Bari "Aldo Moro", via E. Orabona 4, 70125, Bari, Italy
| | - Mauro Niso
- Dipartimento di Farmacia-Scienze Del Farmaco, Università Degli Studi di Bari "Aldo Moro", via E. Orabona 4, 70125, Bari, Italy
| | - Leonardo Pisani
- Dipartimento di Farmacia-Scienze Del Farmaco, Università Degli Studi di Bari "Aldo Moro", via E. Orabona 4, 70125, Bari, Italy.
| | - Antonio Carrieri
- Dipartimento di Farmacia-Scienze Del Farmaco, Università Degli Studi di Bari "Aldo Moro", via E. Orabona 4, 70125, Bari, Italy
| | - Nicola Antonio Colabufo
- Dipartimento di Farmacia-Scienze Del Farmaco, Università Degli Studi di Bari "Aldo Moro", via E. Orabona 4, 70125, Bari, Italy
| | - Saverio Cellamare
- Dipartimento di Farmacia-Scienze Del Farmaco, Università Degli Studi di Bari "Aldo Moro", via E. Orabona 4, 70125, Bari, Italy
| | - Cosimo Damiano Altomare
- Dipartimento di Farmacia-Scienze Del Farmaco, Università Degli Studi di Bari "Aldo Moro", via E. Orabona 4, 70125, Bari, Italy
| |
Collapse
|
50
|
Cai P, Fang SQ, Yang HL, Yang XL, Liu QH, Kong LY, Wang XB. Donepezil-butylated hydroxytoluene (BHT) hybrids as Anti-Alzheimer's disease agents with cholinergic, antioxidant, and neuroprotective properties. Eur J Med Chem 2018; 157:161-176. [PMID: 30096650 DOI: 10.1016/j.ejmech.2018.08.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 08/01/2018] [Accepted: 08/02/2018] [Indexed: 12/16/2022]
Abstract
The multifactorial nature of Alzheimer's disease (AD) calls for the development of multitarget agents addressing key pathogenic processes. A novel family of donepezil-butylated hydroxytoluene (BHT) hybrids were designed, synthesized and evaluated as multifunctional ligands against AD. The optimal compound 7d displayed a balanced multifunctional profile covering an intriguing acetylcholinesterase (AChE) inhibition (IC50, 0.075 μM for eeAChE and 0.75 μM for hAChE) and Monoamine oxidase B (MAO-B) inhibition (IC50, 7.4 μM for hMAO-B), excellent antioxidant activity (71.7 μM of IC50 by DPPH method, 0.82 and 1.62 trolox equivalent by ABTS method and ORAC method respectively), and inhibitory effects on self-induced, hAChE-induced Aβ aggregation. Moreover, 7d possessed neuroprotective potency against H2O2-induced oxidative damage on PC12 cells and Lipopolysaccharides (LPS)-stimulated inflammation on BV2 cells. Compound 7d was capable of penetrating BBB and presented good liver microsomal metabolic stability. Importantly, compound 7d could dose-dependently reverse scopolamine-induced memory deficit in mice without acute toxicity. Taken together, those outstanding results highlight the donepezil-BHT hybrid 7d as a promising prototype in the research of innovative compound for AD.
Collapse
Affiliation(s)
- Pei Cai
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Si-Qiang Fang
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Hua-Li Yang
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Xue-Lian Yang
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Qiao-Hong Liu
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Ling-Yi Kong
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, PR China.
| | - Xiao-Bing Wang
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, PR China.
| |
Collapse
|