1
|
Żołek T, Purgatorio R, Kłopotowski Ł, Catto M, Ostrowska K. Coumarin Derivative Hybrids: Novel Dual Inhibitors Targeting Acetylcholinesterase and Monoamine Oxidases for Alzheimer's Therapy. Int J Mol Sci 2024; 25:12803. [PMID: 39684512 DOI: 10.3390/ijms252312803] [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: 11/14/2024] [Revised: 11/23/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
Multi-target-directed ligands (MTDLs) represent a promising frontier in tackling the complexity of multifactorial pathologies like Alzheimer's disease (AD). The synergistic inhibition of MAO-B, MAO-A, and AChE is believed to enhance treatment efficacy. A novel coumarin-based molecule substituted with O-phenylpiperazine via three- and four-carbon linkers at the 5- and 7-positions, has been identified as an effective MTDL against AD. Employing a medicinal chemistry approach, combined with molecular docking, molecular dynamic simulation, and ΔGbind estimation, two series of derivatives emerged as potent MTDLs: 8-acetyl-7-hydroxy-4-methylcoumarin (IC50: 1.52-4.95 μM for hAChE, 6.97-7.65 μM for hMAO-A) and 4,7-dimethyl-5-hydroxycoumarin (IC50: 1.88-4.76 μM for hMAO-B). They displayed binding free energy (ΔGbind) of -76.32 kcal/mol (11) and -70.12 kcal/mol (12) against AChE and -66.27 kcal/mol (11) and -62.89 kcal/mol (12) against MAO-A. It is noteworthy that compounds 11 and 12 demonstrated efficient binding to both AChE and MAO-A, while compounds 3 and 10 significantly reduced MAO-B and AChE aggregation in vitro. These findings provide structural templates for the development of dual MAO and AChE inhibitors for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Teresa Żołek
- Department of Organic and Physical Chemistry, Faculty of Pharmacy, Medical University of Warsaw, Banacha 1, 02-097 Warsaw, Poland
| | - Rosa Purgatorio
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, Via E. Orabona 4, 70125 Bari, Italy
| | - Łukasz Kłopotowski
- Department of Organic and Physical Chemistry, Faculty of Pharmacy, Medical University of Warsaw, Banacha 1, 02-097 Warsaw, Poland
| | - Marco Catto
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, Via E. Orabona 4, 70125 Bari, Italy
| | - Kinga Ostrowska
- Department of Organic and Physical Chemistry, Faculty of Pharmacy, Medical University of Warsaw, Banacha 1, 02-097 Warsaw, Poland
| |
Collapse
|
2
|
Meletli F, Gündüz C, Alparslan MM, Attar A, Demir S, İskit E, Danış Ö. Design, synthesis and biological evaluation of novel benzocoumarin derivatives as potent inhibitors of MAO-B activity. Bioorg Med Chem Lett 2024; 113:129984. [PMID: 39384075 DOI: 10.1016/j.bmcl.2024.129984] [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: 02/25/2024] [Revised: 09/17/2024] [Accepted: 10/06/2024] [Indexed: 10/11/2024]
Abstract
The continued research of novel reversible inhibitors targeting monoamine oxidase (MAO) B remains crucial for effectively symptomatic treatment of Parkinson's disease. In this study we synthesized and evaluated a new series of 3-aryl benzo[g] and benzo[h] coumarin derivatives as MAO-B inhibitors. Compound A6 has been found to display the most potent inhibitory activity and selectivity against the MAO-B isoform (IC50 = 13 nM and SI = >7693.31 respectively). Inhibition mode of A6 on MAO-B was predicted as mixed reversible inhibition with a Ki value of 3.274 nM. Furthermore, in order to elaborate structure-activity relationships, the binding mode of A6 was investigated by molecular docking simulations.
Collapse
Affiliation(s)
- Furkan Meletli
- Department of Chemistry, Faculty of Science, Marmara University, Istanbul, Turkey.
| | - Cihan Gündüz
- Department of Chemistry and Biochemistry, Pace University, New York, USA
| | | | - Azade Attar
- Department of Bioengineering, Faculty of Chemical and Metallurgical Engineering, Yıldız Technical University, Istanbul, Turkey
| | - Serap Demir
- Department of Chemistry, Faculty of Science, Marmara University, Istanbul, Turkey
| | - Ece İskit
- Department of Chemistry, Faculty of Science, Marmara University, Istanbul, Turkey
| | - Özkan Danış
- Department of Chemistry, Faculty of Science, Marmara University, Istanbul, Turkey.
| |
Collapse
|
3
|
Olmo ED, Barboza B, Delgado-Esteban M, Escala N, Jiménez-Blasco D, Lopez-Pérez JL, Cillero de la Fuente L, Quezada E, Munín J, Viña D, Bolaños JP, Feliciano AS. Potent, selective and reversible hMAO-B inhibition by benzalphthalides: Synthesis, enzymatic and cellular evaluations and virtual docking and predictive studies. Bioorg Chem 2024; 146:107255. [PMID: 38457955 DOI: 10.1016/j.bioorg.2024.107255] [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: 11/09/2023] [Revised: 02/13/2024] [Accepted: 02/29/2024] [Indexed: 03/10/2024]
Abstract
Monoaminooxidases (MAOs) are important targets for drugs used in the treatment of neurological and psychiatric disorders and particularly on Parkinson's Disease (PD). Compounds containing a trans-stilbenoid skeleton have demonstrated good selective and reversible MAO-B inhibition. Here, twenty-two (Z)-3-benzylidenephthalides (benzalphthalides, BPHs) displaying a trans-stilbenoid skeleton have been synthesised and evaluated as inhibitors of the MAO-A and MAO-B isoforms. Some BPHs have selectively inhibited MAO-B, with IC50 values ranging from sub-nM to μM. The most potent compound with IC50 = 0.6 nM was the 3',4'-dichloro-BPH 16, which showed highly selective and reversible MAO-B inhibitory activity. Furthermore, the most selective BPHs displayed a significant protection against the apoptosis, and mitochondrial toxic effects induced by 6-hydroxydopamine (6OHDA) on SH-SY5Y cells, used as a cellular model of PD. The results of virtual binding studies on the most potent compounds docked in MAO-B and MAO-A were in agreement with the potencies and selectivity indexes found experimentally. Additionally, related to toxicity risks, drug-likeness and ADME properties, the predictions found for the most relevant BPHs in this research were within those ranges established for drug candidates.
Collapse
Affiliation(s)
- Esther Del Olmo
- Departamento de Ciencias Farmacéuticas: Química Farmacéutica. Facultad de Farmacia. Universidad de Salamanca, CIETUS, IBSAL. Campus Miguel de Unamuno s/n. 37007 Salamanca, Spain.
| | - Bianca Barboza
- Departamento de Ciencias Farmacéuticas: Química Farmacéutica. Facultad de Farmacia. Universidad de Salamanca, CIETUS, IBSAL. Campus Miguel de Unamuno s/n. 37007 Salamanca, Spain
| | - Maria Delgado-Esteban
- Institute of Functional Biology and Genomics (IBFG), Universidad de Salamanca, CSIC, Salamanca, Spain; Institute of Biomedical Research of Salamanca (IBSAL), Hospital Universitario de Salamanca, Salamanca, Spain; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain
| | - Nerea Escala
- Departamento de Ciencias Farmacéuticas: Química Farmacéutica. Facultad de Farmacia. Universidad de Salamanca, CIETUS, IBSAL. Campus Miguel de Unamuno s/n. 37007 Salamanca, Spain
| | - Daniel Jiménez-Blasco
- Institute of Functional Biology and Genomics (IBFG), Universidad de Salamanca, CSIC, Salamanca, Spain; Institute of Biomedical Research of Salamanca (IBSAL), Hospital Universitario de Salamanca, Salamanca, Spain; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain
| | - José L Lopez-Pérez
- Departamento de Ciencias Farmacéuticas: Química Farmacéutica. Facultad de Farmacia. Universidad de Salamanca, CIETUS, IBSAL. Campus Miguel de Unamuno s/n. 37007 Salamanca, Spain; Facultad de Medicina, Universidad de Panamá, Panamá, R. de Panamá
| | - Laura Cillero de la Fuente
- Institute of Functional Biology and Genomics (IBFG), Universidad de Salamanca, CSIC, Salamanca, Spain; Institute of Biomedical Research of Salamanca (IBSAL), Hospital Universitario de Salamanca, Salamanca, Spain; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain
| | - Elías Quezada
- Chronic Diseases Pharmacology Group, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela. Spain
| | - Javier Munín
- Chronic Diseases Pharmacology Group, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela. Spain
| | - Dolores Viña
- Chronic Diseases Pharmacology Group, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela. Spain.
| | - Juan P Bolaños
- Institute of Functional Biology and Genomics (IBFG), Universidad de Salamanca, CSIC, Salamanca, Spain; Institute of Biomedical Research of Salamanca (IBSAL), Hospital Universitario de Salamanca, Salamanca, Spain; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain.
| | - Arturo San Feliciano
- Departamento de Ciencias Farmacéuticas: Química Farmacéutica. Facultad de Farmacia. Universidad de Salamanca, CIETUS, IBSAL. Campus Miguel de Unamuno s/n. 37007 Salamanca, Spain; Programa de Pós-graduação em Ciências Farmacéuticas, Universidade do Vale do Itajaí, UNIVALI. Itajaí, SC, Brazil
| |
Collapse
|
4
|
Al-Saad OM, Gabr M, Darwish SS, Rullo M, Pisani L, Miniero DV, Liuzzi GM, Kany AM, Hirsch AKH, Abadi AH, Engel M, Catto M, Abdel-Halim M. Novel 6-hydroxybenzothiazol-2-carboxamides as potent and selective monoamine oxidase B inhibitors endowed with neuroprotective activity. Eur J Med Chem 2024; 269:116266. [PMID: 38490063 DOI: 10.1016/j.ejmech.2024.116266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 02/02/2024] [Accepted: 02/18/2024] [Indexed: 03/17/2024]
Abstract
In neurodegenerative diseases, using a single molecule that can exert multiple effects to modify the disease may have superior activity over the classical "one molecule-one target" approach. Herein, we describe the discovery of 6-hydroxybenzothiazol-2-carboxamides as highly potent and selective MAO-B inhibitors. Variation of the amide substituent led to several potent compounds having diverse side chains with cyclohexylamide 40 displaying the highest potency towards MAO-B (IC50 = 11 nM). To discover new compounds with extended efficacy against neurotoxic mechanisms in neurodegenerative diseases, MAO-B inhibitors were screened against PHF6, R3 tau, cellular tau and α-synuclein (α-syn) aggregation. We identified the phenethylamide 30 as a multipotent inhibitor of MAO-B (IC50 = 41 nM) and α-syn and tau aggregation. It showed no cytotoxic effects on SH-SY5Y neuroblastoma cells, while also providing neuroprotection against toxicities induced by α-syn and tau. The evaluation of key physicochemical and in vitro-ADME properties revealed a great potential as drug-like small molecules with multitarget neuroprotective activity.
Collapse
Affiliation(s)
- Omar M Al-Saad
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo, 11835, Egypt
| | - Moustafa Gabr
- Department of Radiology, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Sarah S Darwish
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo, 11835, Egypt; School of Life and Medical Sciences, University of Hertfordshire Hosted By Global Academic Foundation, New Administrative Capital, 11578, Cairo, Egypt
| | - Mariagrazia Rullo
- 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
| | - Daniela Valeria Miniero
- Department of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, Via E. Orabona 4, 70125, Bari, Italy
| | - Grazia Maria Liuzzi
- Department of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, Via E. Orabona 4, 70125, Bari, Italy
| | - Andreas M Kany
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), Saarland University Campus E8.1, 66123, Saarbrücken, Germany
| | - Anna K H Hirsch
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), Saarland University Campus E8.1, 66123, Saarbrücken, Germany; Department of Pharmacy, Saarland University, Campus E8.1, 66123, Saarbrücken, Germany
| | - Ashraf H Abadi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo, 11835, Egypt
| | - Matthias Engel
- Pharmaceutical and Medicinal Chemistry, Saarland University, Campus C2.3, D-66123, Saarbrücken, Germany
| | - Marco Catto
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, Via E. Orabona 4, 70125, Bari, Italy.
| | - Mohammad Abdel-Halim
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo, 11835, Egypt.
| |
Collapse
|
5
|
Yang C, Wang X, Gao C, Liu Y, Ma Z, Zang J, Wang H, Liu L, Liu Y, Sun H, Wang W. Molecular Mechanism and Structure-activity Relationship of the Inhibition Effect between Monoamine Oxidase and Selegiline Analogues. Curr Comput Aided Drug Des 2024; 20:474-485. [PMID: 37138424 DOI: 10.2174/1573409919666230503143055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 02/19/2023] [Accepted: 03/27/2023] [Indexed: 05/05/2023]
Abstract
INTRODUCTION To investigate the inhibition properties and structure-activity relationship between monoamine oxidase (MAO) and selected monoamine oxidase inhibitors (MAOIs, including selegiline, rasagiline and clorgiline). METHODS The inhibition effect and molecular mechanism between MAO and MAOIs were identified via the half maximal inhibitory concentration (IC50) and molecular docking technology. RESULTS It was indicated that selegiline and rasagiline were MAO B inhibitors, but clorgiline was MAO-A inhibitor based on the selectivity index (SI) of MAOIs (0.000264, 0.0197 and 14607.143 for selegiline, rasagiline and clorgiline, respectively). The high-frequency amino acid residues of the MAOIs and MAO were Ser24, Arg51, Tyr69 and Tyr407 for MAO-A and Arg42 and Tyr435 for MAO B. The MAOIs and MAO A/B pharmacophores included the aromatic core, hydrogen bond acceptor, hydrogen bond donor-acceptor and hydrophobic core. CONCLUSION This study shows the inhibition effect and molecular mechanism between MAO and MAOIs and provides valuable findings on the design and treatment of Alzheimer's and Parkinson's diseases.
Collapse
Affiliation(s)
- Chuanxi Yang
- School of Environmental and Municipal Engineering, Qingdao University of Technology, Qingdao, Shandong, 266520, China
| | - Xiaoning Wang
- School of Mechanical Engineering and Automation, Northeastern University, Shenyang, Liaoning, 110819, China
| | - Chang Gao
- Qingdao Jiaming Measurement and Control Technology Co., Ltd., Qingdao, Shandong, 266000, China
| | - Yunxiang Liu
- Environmental Monitoring Station of Yuncheng County Environmental Protection Bureau, Heze, Shandong, 274700, China
| | - Ziyi Ma
- School of Environmental and Municipal Engineering, Qingdao University of Technology, Qingdao, Shandong, 266520, China
| | - Jinqiu Zang
- School of Environmental and Municipal Engineering, Qingdao University of Technology, Qingdao, Shandong, 266520, China
| | - Haoce Wang
- School of Environmental and Municipal Engineering, Qingdao University of Technology, Qingdao, Shandong, 266520, China
| | - Lin Liu
- School of Environmental and Municipal Engineering, Qingdao University of Technology, Qingdao, Shandong, 266520, China
| | - Yonglin Liu
- School of Environmental and Municipal Engineering, Qingdao University of Technology, Qingdao, Shandong, 266520, China
| | - Haofen Sun
- School of Environmental and Municipal Engineering, Qingdao University of Technology, Qingdao, Shandong, 266520, China
| | - Weiliang Wang
- School of Environmental and Municipal Engineering, Qingdao University of Technology, Qingdao, Shandong, 266520, China
| |
Collapse
|
6
|
Lv Y, Zheng Z, Liu R, Guo J, Zhang C, Xie Y. Monoamine oxidase B inhibitors based on natural privileged scaffolds: A review of systematically structural modification. Int J Biol Macromol 2023; 251:126158. [PMID: 37549764 DOI: 10.1016/j.ijbiomac.2023.126158] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 07/28/2023] [Accepted: 08/04/2023] [Indexed: 08/09/2023]
Abstract
Monoamine oxidase is a flavin enzyme that catalyzes the oxidation of monoamine neurotransmitters in the brain. Various toxic by-products, aldehydes and hydrogen peroxide produced during the catalytic process, can cause oxidative stress and neuronal cell death. Overexpression of MAO-B and insufficient dopamine concentration are recognized as pathological factors in neurodegenerative diseases (NDs) including Parkinson's disease (PD) and Alzheimer's disease (AD). Therefore, the inhibition of MAO-B is an attractive target for the treatment of NDs. Despite significant efforts, few selective and reversible MAO-B inhibitors have been clinically approved. Natural products have emerged as valuable sources of lead compounds in drug discovery. Compounds such as chromone, coumarin, chalcone, caffeine, and aurone, present in natural structures, are considered as privileged scaffolds in the synthesis of MAO-B inhibitors. In this review, we summarized the structure-activity relationship (SAR) of MAO-B inhibitors based on the naturally privileged scaffolds over the past 20 years. Additionally, we proposed a balanced discussion on the advantages and limitations of natural scaffold-based MAO-B inhibitors with providing a future perspective in drug development.
Collapse
Affiliation(s)
- Yangjing Lv
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| | - Zhiyuan Zheng
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| | - Renzheng Liu
- 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, China; Key Laboratory of Pharmaceutical Engineering of Zhejiang Province, China.
| |
Collapse
|
7
|
Gambacorta N, Catto M, Pisani L, Carotti A, Nicolotti O. Informed Use of 3D-QSAR for the Rational Design of Coumarin Derivatives as Potent and Selective MAO B Inhibitors. Methods Mol Biol 2023; 2558:197-205. [PMID: 36169865 DOI: 10.1007/978-1-0716-2643-6_15] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
The quantitative structure-activity relationship method based on the three-dimensional structure of the target molecules (3D-QSAR) represents a valuable predictive tool for the design of new bioactive agents. Herewith, a detailed procedure is described which uses a pool comprising 67 derivatives substituted at position 4 and 7 of the common coumarin scaffold as a benchmark for deriving a predictive 3D-QSAR model employed for guiding the rational design of 10 new potent and selective MAO B inhibitors.
Collapse
Affiliation(s)
- Nicola Gambacorta
- 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
| | - Leonardo Pisani
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari "Aldo Moro", Bari, Italy
| | - Angelo Carotti
- 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.
| |
Collapse
|
8
|
Sudevan ST, Oh JM, Abdelgawad MA, Abourehab MAS, Rangarajan TM, Kumar S, Ahmad I, Patel H, Kim H, Mathew B. Introduction of benzyloxy pharmacophore into aryl/heteroaryl chalcone motifs as a new class of monoamine oxidase B inhibitors. Sci Rep 2022; 12:22404. [PMID: 36575270 PMCID: PMC9794710 DOI: 10.1038/s41598-022-26929-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 12/21/2022] [Indexed: 12/28/2022] Open
Abstract
The inhibitory action of fifteen benzyloxy ortho/para-substituted chalcones (B1-B15) was evaluated against human monoamine oxidases (hMAOs). All the molecules inhibited hMAO-B isoform more potently than hMAO-A. Furthermore, the majority of the molecules showed strong inhibitory actions against hMAO-B at 10 μM level with residual activities of less than 50%. Compound B10 has an IC50 value of 0.067 μM, making it the most potent inhibitor of hMAO-B, trailed by compound B15 (IC50 = 0.12 μM). The thiophene substituent (B10) in the A-ring exhibited the strongest hMAO-B inhibition structurally, however, increased residue synthesis did not result in a rise in hMAO-B inhibition. In contrast, the benzyl group at the para position of the B-ring displayed more hMAO-B inhibition than the other positions. Compounds B10 and B15 had relatively high selectivity index (SI) values for hMAO-B (504.791 and 287.600, respectively). Ki values of B10 and B15 were 0.030 ± 0.001 and 0.033 ± 0.001 μM, respectively. The reversibility study showed that B10 and B15 were reversible inhibitors of hMAO-B. PAMPA assay manifested that the benzyloxy chalcones (B10 and B15) had a significant permeability and CNS bioavailability with Pe value higher than 4.0 × 10-6 cm/s. Both compounds were stabilized in protein-ligand complexes by the π-π stacking, which enabled them to bind to the hMAO-B enzyme's active site incredibly effectively. The hMAO-B was stabilized by B10- and B15-hMAO-B complexes, with binding energies of - 74.57 and - 87.72 kcal/mol, respectively. Using a genetic algorithm and multiple linear regression, the QSAR model was created. Based on the best 2D and 3D descriptor-based QSAR model, the following statistics were displayed: R2 = 0.9125, Q2loo = 0.8347. These findings imply that B10 and B15 are effective, selective, and reversible hMAO-B inhibitors.
Collapse
Affiliation(s)
- Sachithra Thazhathuveedu Sudevan
- grid.411370.00000 0000 9081 2061Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi, 682 041 India
| | - Jong Min Oh
- grid.412871.90000 0000 8543 5345Department of Pharmacy, and Research Institute of Life Pharmaceutical Sciences, Sunchon National University, Suncheon, 57922 Republic of Korea
| | - Mohamed A. Abdelgawad
- grid.440748.b0000 0004 1756 6705Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University, Sakaka, 72341 Saudi Arabia ,grid.411662.60000 0004 0412 4932Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, 62514 Egypt
| | - Mohammed A. S. Abourehab
- grid.412832.e0000 0000 9137 6644Department of Pharmaceutics, College of Pharmacy, Umm Al-Qura University, Makkah, 21955 Saudi Arabia
| | - T. M. Rangarajan
- grid.8195.50000 0001 2109 4999Department of Chemistry, Sri Venketeswara College, University of Delhi, New Delhi, 110021 India
| | - Sunil Kumar
- grid.411370.00000 0000 9081 2061Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi, 682 041 India
| | - Iqrar Ahmad
- Department of Pharmaceutical Chemistry, Prof. Ravindra Nikam College of Pharmacy, Gondur, Dhule, 424002 Maharashtra India
| | - Harun Patel
- grid.412233.50000 0001 0641 8393Division of Computer Aided Drug Design, Department of Pharmaceutical Chemistry, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, 425405 Maharashtra India
| | - Hoon Kim
- grid.412871.90000 0000 8543 5345Department of Pharmacy, and Research Institute of Life Pharmaceutical Sciences, Sunchon National University, Suncheon, 57922 Republic of Korea
| | - Bijo Mathew
- grid.411370.00000 0000 9081 2061Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi, 682 041 India
| |
Collapse
|
9
|
In vitro and in silico investigation of inhibitory activities of 3-arylcoumarins and 3-phenylazo-4-hydroxycoumarin on MAO isoenzymes. Struct Chem 2022. [DOI: 10.1007/s11224-022-02092-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
10
|
Zheng L, Qin X, Wang J, Zhang M, An Q, Xu J, Qu X, Cao X, Niu B. Discovery of MAO-B Inhibitor with Machine Learning, Topomer CoMFA, Molecular Docking and Multi-Spectroscopy Approaches. Biomolecules 2022; 12:biom12101470. [PMID: 36291679 PMCID: PMC9599443 DOI: 10.3390/biom12101470] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/20/2022] [Accepted: 09/28/2022] [Indexed: 12/05/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common type of dementia and is a serious disruption to normal life. Monoamine oxidase-B (MAO-B) is an important target for the treatment of AD. In this study, machine learning approaches were applied to investigate the identification model of MAO-B inhibitors. The results showed that the identification model for MAO-B inhibitors with K-nearest neighbor(KNN) algorithm had a prediction accuracy of 94.1% and 88.0% for the 10-fold cross-validation test and the independent test set, respectively. Secondly, a quantitative activity prediction model for MAO-B was investigated with the Topomer CoMFA model. Two separate cutting mode approaches were used to predict the activity of MAO-B inhibitors. The results showed that the cut model with q2 = 0.612 (cross-validated correlation coefficient) and r2 = 0.824 (non-cross-validated correlation coefficient) were determined for the training and test sets, respectively. In addition, molecular docking was employed to analyze the interaction between MAO-B and inhibitors. Finally, based on our proposed prediction model, 1-(4-hydroxyphenyl)-3-(2,4,6-trimethoxyphenyl)propan-1-one (LB) was predicted as a potential MAO-B inhibitor and was validated by a multi-spectroscopic approach including fluorescence spectra and ultraviolet spectrophotometry.
Collapse
Affiliation(s)
- Linfeng Zheng
- School of Life Science, Shanghai University, Shanghai 200444, China
- Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Xiangyang Qin
- Department of Chemistry, School of Pharmacy, Air Force Medical University, Xi’an 710032, China
| | - Jiao Wang
- School of Life Science, Shanghai University, Shanghai 200444, China
| | - Mengying Zhang
- School of Life Science, Shanghai University, Shanghai 200444, China
| | - Quanlin An
- Institute of Clinical Science, Zhongshan Hospital, Shanghai Medical College, Fudan University, Shanghai 200444, China
| | - Jinzhi Xu
- Institute of Clinical Science, Zhongshan Hospital, Shanghai Medical College, Fudan University, Shanghai 200444, China
| | - Xiaosheng Qu
- National Engineering Laboratory of Southwest Endangered Medicinal Resources Development, Guangxi Botanical Garden of Medicinal Plants, Nanning 530023, China
| | - Xin Cao
- Institute of Clinical Science, Zhongshan Hospital, Shanghai Medical College, Fudan University, Shanghai 200444, China
- Correspondence: (X.C.); (B.N.)
| | - Bing Niu
- School of Life Science, Shanghai University, Shanghai 200444, China
- Correspondence: (X.C.); (B.N.)
| |
Collapse
|
11
|
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
|
12
|
Bhawna, Kumar A, Bhatia M, Kapoor A, Kumar P, Kumar S. Monoamine oxidase inhibitors: A concise review with special emphasis on structure activity relationship studies. Eur J Med Chem 2022; 242:114655. [PMID: 36037788 DOI: 10.1016/j.ejmech.2022.114655] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/30/2022] [Accepted: 08/01/2022] [Indexed: 12/29/2022]
Abstract
Monoamine oxidase enzyme is necessary for the management of brain functions. It oxidatively metabolizes monoamines and produces ammonia, aldehyde and hydrogen peroxide as by-products. Excessive production of by-products of monoamine metabolism generates free radicals which cause cellular apoptosis and several neurodegenerative disorders for example Alzheimer's disease, Parkinson's disease, depression and autism. The inhibition of MAOs is an attractive target for the treatment of neurological disorders. Clinically approved MAO inhibitors for example selegiline, rasagiline, clorgyline, pargyline etc. are irreversible in nature and cause some adverse effects while recently studied reversible MAO inhibitors are devoid of harmful effects of old monoamine oxidase inhibitors. In this review article we have listed various synthesized molecules containing different moieties like coumarin, chalcone, thiazole, thiourea, caffeine, pyrazole, chromone etc. along with their activity, mode of action, structure activity relationship and molecular docking studies.
Collapse
Affiliation(s)
- Bhawna
- Department of Pharmaceutical Sciences,Guru Jambheshwar University of Science and Technology, Hisar, 125001, Haryana, India
| | - Ashwani Kumar
- Department of Pharmaceutical Sciences,Guru Jambheshwar University of Science and Technology, Hisar, 125001, Haryana, India
| | - Meenakshi Bhatia
- Department of Pharmaceutical Sciences,Guru Jambheshwar University of Science and Technology, Hisar, 125001, Haryana, India
| | - Archana Kapoor
- Department of Pharmaceutical Sciences,Guru Jambheshwar University of Science and Technology, Hisar, 125001, Haryana, India
| | - Parvin Kumar
- Department of Chemistry, Kurukshetra University, Kurukshetra, 136119, Haryana, India
| | - Sunil Kumar
- Department of Pharmaceutical Sciences,Guru Jambheshwar University of Science and Technology, Hisar, 125001, Haryana, India.
| |
Collapse
|
13
|
Chen Z, Zeng P, Zhang S, Sun J. Recent Advances in Organic Synthesis of 3-Amino- or 4-Aminocoumarins. MINI-REV ORG CHEM 2022. [DOI: 10.2174/1570193x18666211001124004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
:
Coumarin is a privileged scaffold that contains the unique 2H-chromen-2-one motif, and its
derivatives are widely distributed in nature, especially in plants. In recent years, due to their diverse
pharmacological activities and remarkable photochemical properties, they have attracted significant
attention from scientists, which has also prompted the research on the synthesis approaches and the
availability of substrates for these compounds. This article is a brief description of the methods for the
synthesis of various coumarin derivatives via two- or multi-component reactions involving 3-amino
or 4-aminocoumarin reported during 2015-2021. This review may help expand the development of
various analogues with coumarin as the basic unit.
Collapse
Affiliation(s)
- Zhiwei Chen
- College of Pharmaceutical Sciences, Zhejiang University of Technology, and Institute of Drug Synthesis, Zhejiang University
of Technology, Hangzhou 310014, P.R. China
| | - Piaopiao Zeng
- College of Pharmaceutical Sciences, Zhejiang University of Technology, and Institute of Drug Synthesis, Zhejiang University
of Technology, Hangzhou 310014, P.R. China
| | - Shuo Zhang
- College of Pharmaceutical Sciences, Zhejiang University of Technology, and Institute of Drug Synthesis, Zhejiang University
of Technology, Hangzhou 310014, P.R. China
| | - Jie Sun
- College of Pharmaceutical Sciences, Zhejiang University of Technology, and Institute of Drug Synthesis, Zhejiang University
of Technology, Hangzhou 310014, P.R. China
| |
Collapse
|
14
|
Effects of Linkers and Substitutions on Multitarget Directed Ligands for Alzheimer’s Diseases: Emerging Paradigms and Strategies. Int J Mol Sci 2022; 23:ijms23116085. [PMID: 35682763 PMCID: PMC9181730 DOI: 10.3390/ijms23116085] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/24/2022] [Accepted: 05/25/2022] [Indexed: 02/06/2023] Open
Abstract
Alzheimer’s disease (AD) is multifactorial, progressive and the most predominant cause of cognitive impairment and dementia worldwide. The current “one-drug, one-target” approach provides only symptomatic relief to the condition but is unable to cure the disease completely. The conventional single-target therapeutic approach might not always induce the desired effect due to the multifactorial nature of AD. Hence, multitarget strategies have been proposed to simultaneously knock out multiple targets involved in the development of AD. Herein, we provide an overview of the various strategies, followed by the multitarget-directed ligand (MTDL) development, rationale designs and efficient examples. Furthermore, the effects of the linkers and substitutional functional groups on MTDLs against various targets of AD and their modes of action are also discussed.
Collapse
|
15
|
Sudevan ST, Rangarajan TM, Al-Sehemi AG, Nair AS, Koyiparambath VP, Mathew B. Revealing the role of the benzyloxy pharmacophore in the design of a new class of monoamine oxidase-B inhibitors. Arch Pharm (Weinheim) 2022; 355:e2200084. [PMID: 35567313 DOI: 10.1002/ardp.202200084] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 04/11/2022] [Accepted: 04/14/2022] [Indexed: 11/09/2022]
Abstract
The conceptual layout of monoamine oxidase (MAO) inhibitors has been modified to explore their potential biological application in the case of neurological disorders for the time being. The current review article is an effort to display the summation of innovative conceptual prospects of MAO inhibitors and their intriguing chemistry and bioactivity. Based on this scenario, we emphasize the pivotal role of the benzyloxy moiety attached to scaffolds like oxadiazolones, indolalkylamines, safinamide, caffeine, benzofurans, α-tetralones, β-nitrostyrene, benzoquinones, coumarins, indoles, chromones, and chromanone analogs, while acting as an MAO inhibitor.
Collapse
Affiliation(s)
- Sachithra T Sudevan
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi, India
| | - T M Rangarajan
- Department of Chemistry, Sri Venketeswara College, University of Delhi, New Delhi, India
| | - Abdullah G Al-Sehemi
- Research Center for Advanced Materials Science, King Khalid University, Abha, Saudi Arabia.,Department of Chemistry, KingKhalid University, 61413, Abha, Saudi Arabia
| | - Aathira S Nair
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi, India
| | - Vishal P Koyiparambath
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi, India
| | - Bijo Mathew
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi, India
| |
Collapse
|
16
|
Concu R, González-Durruthy M, Cordeiro MNDS. Developing a Multi-target Model to Predict the Activity of Monoamine Oxidase A and B Drugs. Curr Top Med Chem 2021; 20:1593-1600. [PMID: 32493193 DOI: 10.2174/1568026620666200603121224] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 11/19/2019] [Accepted: 11/28/2019] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Monoamine oxidase inhibitors (MAOIs) are compounds largely used in the treatment of Parkinson's disease (PD), Alzheimer's disease and other neuropsychiatric disorders since they are closely related to the MAO enzymes activity. The two isoforms of the MAO enzymes, MAO-A and MAO-B, are responsible for the degradation of monoamine neurotransmitters and due to this, relevant efforts have been devoted to finding new compounds with more selectivity and less side effects. One of the most used approaches is based on the use of computational approaches since they are time and money-saving and may allow us to find a more relevant structure-activity relationship. OBJECTIVE In this manuscript, we will review the most relevant computational approaches aimed at the prediction and development of new MAO inhibitors. Subsequently, we will also introduce a new multitask model aimed at predicting MAO-A and MAO-B inhibitors. METHODS The QSAR multi-task model herein developed was based on the use of the linear discriminant analysis. This model was developed gathering 5,759 compounds from the public dataset Chembl. The molecular descriptors used was calculated using the Dragon software. Classical statistical tests were performed to check the validity and robustness of the model. RESULTS The herein proposed model is able to correctly classify all the 5,759 compounds. All the statistical performed tests indicated that this model is robust and reproducible. CONCLUSION MAOIs are compounds of large interest since they are largely used in the treatment of very serious illness. These inhibitors may lose efficacy and produce severe side effects. Due to this, the development of selective MAO-A or MAO-B inhibitors is crucial for the treatment of these diseases and their effects. The herein proposed multi-target QSAR model may be a relevant tool in the development of new and more selective MAO inhibitors.
Collapse
Affiliation(s)
- Riccardo Concu
- LAQV@REQUIMTE/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, 4169-007 Porto, Portugal
| | - Michael González-Durruthy
- LAQV@REQUIMTE/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, 4169-007 Porto, Portugal
| | - Maria Natália D S Cordeiro
- LAQV@REQUIMTE/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, 4169-007 Porto, Portugal
| |
Collapse
|
17
|
Musa MA, Badisa VLD, Aghimien MO, Eyunni SVK, Latinwo LM. Identification of 7,8-dihydroxy-3-phenylcoumarin as a reversible monoamine oxidase enzyme inhibitor. J Biochem Mol Toxicol 2020; 35:e22651. [PMID: 33085988 DOI: 10.1002/jbt.22651] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 08/18/2020] [Accepted: 09/29/2020] [Indexed: 01/20/2023]
Abstract
We herein report the biological evaluation of 3-arylcoumarin derivatives (3a-l) as potential human monoamine oxidase-A and -B (hMAO-A and hMAO-B) inhibitors. The result indicated that 7,8-dihydroxy-3-(4-nitrophenyl)coumarin (3j) was most effective against MAO-A (inhibition concentration [IC50 ] = 6.46 ± 0.02 µM) and MAO-B (IC50 = 3.8 ± 0.3 µM) enzymes than other synthesized compounds and reference compounds (pargyline and moclobemide). Furthermore, compound (3j) showed (a) nonselectivity against hMAO enzymes, (b) reversible hMAO enzymes inhibition, and (c) neuroprotection against H2 O2 -treated human neuroblastoma (N2a) cells. Finally, a molecular modeling study revealed that the hMAO enzymes inhibitory activity of the compound (3j) may be due to the orientation where the nitro (NO2 ) group lies deep into the receptor and the phenyl ring directed toward flavin adenosine dinucleotide via hydrogen bond interaction, and possible π-π interaction with various important residues. Thus, the results of the present study demonstrate that compound (3j) can be considered as a promising scaffold for the development of hMAO-A and hMAO-B inhibitors.
Collapse
Affiliation(s)
- Musiliyu A Musa
- Department of Chemistry, Florida A&M University, Tallahassee, Florida
| | - Veera L D Badisa
- School of the Environment, Florida A&M University, Tallahassee, Florida
| | - Monica O Aghimien
- Department of Biological Sciences, Florida A&M University, Tallahassee, Florida
| | - Suresh V K Eyunni
- Department of Chemistry, Florida A&M University, Tallahassee, Florida.,College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, Florida
| | - Lekan M Latinwo
- Department of Biological Sciences, Florida A&M University, Tallahassee, Florida
| |
Collapse
|
18
|
Hydride Abstraction as the Rate-Limiting Step of the Irreversible Inhibition of Monoamine Oxidase B by Rasagiline and Selegiline: A Computational Empirical Valence Bond Study. Int J Mol Sci 2020; 21:ijms21176151. [PMID: 32858935 PMCID: PMC7503497 DOI: 10.3390/ijms21176151] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/11/2020] [Accepted: 08/21/2020] [Indexed: 12/23/2022] Open
Abstract
Monoamine oxidases (MAOs) catalyze the degradation of a very broad range of biogenic and dietary amines including many neurotransmitters in the brain, whose imbalance is extensively linked with the biochemical pathology of various neurological disorders, and are, accordingly, used as primary pharmacological targets to treat these debilitating cognitive diseases. Still, despite this practical significance, the precise molecular mechanism underlying the irreversible MAO inhibition with clinically used propargylamine inhibitors rasagiline and selegiline is still not unambiguously determined, which hinders the rational design of improved inhibitors devoid of side effects current drugs are experiencing. To address this challenge, we present empirical valence bond QM/MM simulations of the rate-limiting step of the MAO inhibition involving the hydride anion transfer from the inhibitor α-carbon onto the N5 atom of the flavin adenin dinucleotide (FAD) cofactor. The proposed mechanism is strongly supported by the obtained free energy profiles, which confirm a higher reactivity of selegiline over rasagiline, while the calculated difference in the activation Gibbs energies of ΔΔG‡ = 3.1 kcal mol-1 is found to be in very good agreement with that from the measured literature kinact values that predict a 1.7 kcal mol-1 higher selegiline reactivity. Given the similarity with the hydride transfer mechanism during the MAO catalytic activity, these results verify that both rasagiline and selegiline are mechanism-based irreversible inhibitors and offer guidelines in designing new and improved inhibitors, which are all clinically employed in treating a variety of neuropsychiatric and neurodegenerative conditions.
Collapse
|
19
|
Hu Y, Zhou G, Zhang C, Zhang M, Chen Q, Zheng L, Niu B. Identify Compounds' Target Against Alzheimer's Disease Based on In-Silico Approach. Curr Alzheimer Res 2020; 16:193-208. [PMID: 30605059 DOI: 10.2174/1567205016666190103154855] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 12/20/2018] [Accepted: 01/03/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Alzheimer's disease swept every corner of the globe and the number of patients worldwide has been rising. At present, there are as many as 30 million people with Alzheimer's disease in the world, and it is expected to exceed 80 million people by 2050. Consequently, the study of Alzheimer's drugs has become one of the most popular medical topics. METHODS In this study, in order to build a predicting model for Alzheimer's drugs and targets, the attribute discriminators CfsSubsetEval, ConsistencySubsetEval and FilteredSubsetEval are combined with search methods such as BestFirst, GeneticSearch and Greedystepwise to filter the molecular descriptors. Then the machine learning algorithms such as BayesNet, SVM, KNN and C4.5 are used to construct the 2D-Structure Activity Relationship(2D-SAR) model. Its modeling results are utilized for Receiver Operating Characteristic curve(ROC) analysis. RESULTS The prediction rates of correctness using Randomforest for AChE, BChE, MAO-B, BACE1, Tau protein and Non-inhibitor are 77.0%, 79.1%, 100.0%, 94.2%, 93.2% and 94.9%, respectively, which are overwhelming as compared to those of BayesNet, BP, SVM, KNN, AdaBoost and C4.5. CONCLUSION In this paper, we conclude that Random Forest is the best learner model for the prediction of Alzheimer's drugs and targets. Besides, we set up an online server to predict whether a small molecule is the inhibitor of Alzheimer's target at http://47.106.158.30:8080/AD/. Furthermore, it can distinguish the target protein of a small molecule.
Collapse
Affiliation(s)
- Yan Hu
- School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Guangya Zhou
- School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Chi Zhang
- Huaxia Eye Hospital of Foshan, Huaxia Eye Hospital Group, Foshan, Guangdong, China
| | - Mengying Zhang
- School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Qin Chen
- School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Linfeng Zheng
- Department of Radiology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai 200080, China.,Department of Radiology, Shanghai First People's Hospital, Baoshan Branch, Shanghai 200940, China
| | - Bing Niu
- School of Life Sciences, Shanghai University, Shanghai 200444, China
| |
Collapse
|
20
|
Ramesh M, Muthuraman A. Quantitative Structure-Activity Relationship (QSAR) Studies for the Inhibition of MAOs. Comb Chem High Throughput Screen 2020; 23:887-897. [PMID: 32208114 DOI: 10.2174/1386207323666200324173231] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 01/20/2020] [Accepted: 02/04/2020] [Indexed: 11/22/2022]
Abstract
Monoamine oxidases are the crucial drug targets for the treatment of neurodegenerative disorders like depression, Parkinson's disease, and Alzheimer's disease. The enzymes catalyze the oxidative deamination of several monoamine containing neurotransmitters, i.e. serotonin (5-HT), melatonin, epinephrine, norepinephrine, phenylethylamine, benzylamine, dopamine, tyramine, etc. The oxidative reaction of monoamine oxidases results in the production of hydrogen peroxide that leads to the neurodegeneration process. Therefore, the inhibition of monoamine oxidases has shown a profound effect against neurodegenerative diseases. At present, the design and development of newer lead molecules for the inhibition of monoamine oxidases are under intensive research in the field of medicinal chemistry. Recently, the advancement in QSAR methodologies has shown considerable interest in the development of monoamine oxidase inhibitors. The present review describes the development of QSAR methodologies, and their role in the design of newer monoamine oxidase inhibitors. It will assist the medicinal chemist in the identification of selective and potent monoamine oxidase inhibitors from various chemical scaffolds.
Collapse
Affiliation(s)
- Muthusamy Ramesh
- Department of Pharmaceutical Analysis, Omega College of Pharmacy, Hyderabad-501 301, India
| | - Arunachalam Muthuraman
- Pharmacology Unit, Faculty of Pharmacy, AIMST University, Semeling, 08100 Bedong, Kedah Darul Aman, Malaysia
| |
Collapse
|
21
|
Jafari B, Jalil S, Zaib S, Safarov S, Khalikova M, Khalikov D, Ospanov M, Yelibayeva N, Zhumagalieva S, Abilov ZA, Turmukhanova MZ, Kalugin SN, Salman GA, Ehlers P, Hameed A, Iqbal J, Langer P. Synthesis of 2‐Alkynyl‐ and2‐Amino‐12
H
‐benzothiazolo[2,3‐
b
]quinazolin‐12‐ones and Their Inhibitory Potential against Monoamine Oxidase A and B. ChemistrySelect 2019. [DOI: 10.1002/slct.201903300] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Behzad Jafari
- Institut für ChemieUniversität Rostock Albert-Einstein-Str. 3a 18059 Rostock Germany
| | - Saquib Jalil
- Centre for Advanced Drug ResearchCOMSATS University Islamabad, Abbottabad Campus Abbottabad- 22060 Pakistan
| | - Sumera Zaib
- Centre for Advanced Drug ResearchCOMSATS University Islamabad, Abbottabad Campus Abbottabad- 22060 Pakistan
| | - Sayfidin Safarov
- Institut für ChemieUniversität Rostock Albert-Einstein-Str. 3a 18059 Rostock Germany
- Institute of ChemistryTajikistan Academy of Sciences ul. Aini 299 Dushanbe 734063 Tajikistan
| | - Muattar Khalikova
- Institut für ChemieUniversität Rostock Albert-Einstein-Str. 3a 18059 Rostock Germany
- Institute of ChemistryTajikistan Academy of Sciences ul. Aini 299 Dushanbe 734063 Tajikistan
| | - Djurabay Khalikov
- Institut für ChemieUniversität Rostock Albert-Einstein-Str. 3a 18059 Rostock Germany
- Institute of ChemistryTajikistan Academy of Sciences ul. Aini 299 Dushanbe 734063 Tajikistan
| | - Meirambek Ospanov
- Institut für ChemieUniversität Rostock Albert-Einstein-Str. 3a 18059 Rostock Germany
- Al-Farabi Kazakh National University Al-Farabi ave. 71 050040 Almaty Kazakhstan
| | - Nazym Yelibayeva
- Institut für ChemieUniversität Rostock Albert-Einstein-Str. 3a 18059 Rostock Germany
- Al-Farabi Kazakh National University Al-Farabi ave. 71 050040 Almaty Kazakhstan
| | - Shynar Zhumagalieva
- Al-Farabi Kazakh National University Al-Farabi ave. 71 050040 Almaty Kazakhstan
| | | | | | - Sergey N. Kalugin
- Al-Farabi Kazakh National University Al-Farabi ave. 71 050040 Almaty Kazakhstan
| | - Ghazwan Ali Salman
- Institut für ChemieUniversität Rostock Albert-Einstein-Str. 3a 18059 Rostock Germany
- Department of ChemistryCollege of Science, University Al-Mustansiriyah Palestine St, Mustansiriya, Baghdad Iraq
| | - Peter Ehlers
- Institut für ChemieUniversität Rostock Albert-Einstein-Str. 3a 18059 Rostock Germany
| | - Abdul Hameed
- Centre for Advanced Drug ResearchCOMSATS University Islamabad, Abbottabad Campus Abbottabad- 22060 Pakistan
| | - Jamshed Iqbal
- Centre for Advanced Drug ResearchCOMSATS University Islamabad, Abbottabad Campus Abbottabad- 22060 Pakistan
| | - Peter Langer
- Institut für ChemieUniversität Rostock Albert-Einstein-Str. 3a 18059 Rostock Germany
- Leibniz Institut für Katalyse an der Universität Rostock e.V. (LIKAT) Albert-Einstein-Str. 29a 18059 Rostock Germany
| |
Collapse
|
22
|
Rullo M, Catto M, Carrieri A, de Candia M, Altomare CD, Pisani L. Chasing ChEs-MAO B Multi-Targeting 4-Aminomethyl-7-Benzyloxy-2 H-Chromen-2-ones. Molecules 2019; 24:E4507. [PMID: 31835376 PMCID: PMC6943664 DOI: 10.3390/molecules24244507] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 12/04/2019] [Accepted: 12/05/2019] [Indexed: 11/16/2022] Open
Abstract
A series of 4-aminomethyl-7-benzyloxy-2H-chromen-2-ones was investigated with the aim of identifying multiple inhibitors of cholinesterases (acetyl- and butyryl-, AChE and BChE) and monoamine oxidase B (MAO B) as potential anti-Alzheimer molecules. Starting from a previously reported potent MAO B inhibitor (3), we studied single-point modifications at the benzyloxy or at the basic moiety. The in vitro screening highlighted triple-acting compounds (6, 8, 9, 16, 20) showing nanomolar and selective MAO B inhibition along with IC50 against ChEs at the low micromolar level. Enzyme kinetics analysis toward AChE and docking simulations on the target enzymes were run in order to get insight into the mechanism of action and plausible binding modes.
Collapse
Affiliation(s)
| | | | | | | | | | - Leonardo Pisani
- Department of Pharmacy—Drug Sciences, University of Bari “Aldo Moro”, via E. Orabona 4, 70125 Bari, Italy
| |
Collapse
|
23
|
Wang D, Hong RY, Guo M, Liu Y, Chen N, Li X, Kong DX. Novel C7-Substituted Coumarins as Selective Monoamine Oxidase Inhibitors: Discovery, Synthesis and Theoretical Simulation. Molecules 2019; 24:molecules24214003. [PMID: 31694262 PMCID: PMC6864482 DOI: 10.3390/molecules24214003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 10/27/2019] [Accepted: 10/31/2019] [Indexed: 12/02/2022] Open
Abstract
There is a continued need to develop new selective human monoamine oxidase (hMAO) inhibitors that could be beneficial for the treatment of neurological diseases. However, hMAOs are closely related with high sequence identity and structural similarity, which hinders the development of selective MAO inhibitors. “Three-Dimensional Biologically Relevant Spectrum (BRS-3D)” method developed by our group has demonstrated its effectiveness in subtype selectivity studies of receptor and enzyme ligands. Here, we report a series of novel C7-substituted coumarins, either synthesized or commercially purchased, which were identified as selective hMAO inhibitors. Most of the compounds demonstrated strong activities with IC50 values (half-inhibitory concentration) ranging from sub-micromolar to nanomolar. Compounds, FR1 and SP1, were identified as the most selective hMAO-A inhibitors, with IC50 values of 1.5 nM (selectivity index (SI) < −2.82) and 19 nM (SI < −2.42), respectively. FR4 and FR5 showed the most potent hMAO-B inhibitory activity, with IC50 of 18 nM and 15 nM (SI > 2.74 and SI > 2.82). Docking calculations and molecular dynamic simulations were performed to elucidate the selectivity preference and SAR profiles.
Collapse
Affiliation(s)
- Dong Wang
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, China;
- Agricultural Bioinformatics Key Laboratory of Hubei Province, College of Informatics, Huazhong Agricultural University, Wuhan 430070, China; (M.G.); (Y.L.); (N.C.)
| | - Ren-Yuan Hong
- Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 West Culture Road, Ji’nan 250012, Shandong, China;
| | - Mengyao Guo
- Agricultural Bioinformatics Key Laboratory of Hubei Province, College of Informatics, Huazhong Agricultural University, Wuhan 430070, China; (M.G.); (Y.L.); (N.C.)
| | - Yi Liu
- Agricultural Bioinformatics Key Laboratory of Hubei Province, College of Informatics, Huazhong Agricultural University, Wuhan 430070, China; (M.G.); (Y.L.); (N.C.)
| | - Nianhang Chen
- Agricultural Bioinformatics Key Laboratory of Hubei Province, College of Informatics, Huazhong Agricultural University, Wuhan 430070, China; (M.G.); (Y.L.); (N.C.)
| | - Xun Li
- Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, No 18877, Jingshi Road, Ji’nan 250002, Shandong, China
- Correspondence: (X.L.); (D.-X.K.); Tel.: +86-531-88382005 (X.L.); +86-27-8728 0877 (D.-X.K.)
| | - De-Xin Kong
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, China;
- Agricultural Bioinformatics Key Laboratory of Hubei Province, College of Informatics, Huazhong Agricultural University, Wuhan 430070, China; (M.G.); (Y.L.); (N.C.)
- Correspondence: (X.L.); (D.-X.K.); Tel.: +86-531-88382005 (X.L.); +86-27-8728 0877 (D.-X.K.)
| |
Collapse
|
24
|
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
|
25
|
Dorababu A. Critical evaluation of current Alzheimer's drug discovery (2018-19) & futuristic Alzheimer drug model approach. Bioorg Chem 2019; 93:103299. [PMID: 31586701 DOI: 10.1016/j.bioorg.2019.103299] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 09/14/2019] [Accepted: 09/16/2019] [Indexed: 12/17/2022]
Abstract
Alzheimer's disease (AD), a neurodegenerative disease responsible for death of millions of people worldwide is a progressive clinical disorder which causes neurons to degenerate and ultimately die. It is one of the common causes of dementia wherein a person's incapability to independently think, behave and decline in social skills can be quoted as major symptoms. However the early signs include the simple non-clinical symptoms such as forgetting recent events and conversations. Onset of these symptoms leads to worsened conditions wherein the AD patient suffers severe memory impairment and eventually becomes unable to work out everyday tasks. Even though there is no complete cure for AD, rigorous research has been going on to reduce the progress of AD. Currently, a very few clinical drugs are prevailing for AD treatment. So this is the need of hour to design, develop and discovery of novel anti-AD drugs. The main factors for the cause of AD according to scientific research reveals structural changes in brain proteins such as beta amyloid, tau proteins into plaques and tangles respectively. The abnormal proteins distort the neurons. Despite the high potencies of the synthesized molecules; they could not get on the clinical tests up to human usage. In this review article, the recent research carried out with respect to inhibition of AChE, BuChE, NO, BACE1, MAOs, Aβ, H3R, DAPK, CSF1R, 5-HT4R, PDE, σ1R and GSK-3β is compiled and organized. The summary is focused mainly on cholinesterases, Aβ, BACE1 and MAOs classes of potential inhibitors. The review also covers structure activity relationship of most potent compounds of each class of inhibitors alongside redesign and remodeling of the most significant inhibitors in order to expect cutting edge inhibitory properties towards AD. Alongside the molecular docking studies of the some final compounds are discussed.
Collapse
Affiliation(s)
- Atukuri Dorababu
- Department of Studies in Chemistry, SRMPP Govt. First Grade College, Huvinahadagali 583219, Karnataka, India.
| |
Collapse
|
26
|
Coumarin analogue 3-methyl-7H-furo[3,2-g] chromen-7-one as a possible antiparkinsonian agent. ACTA ACUST UNITED AC 2019; 39:491-501. [PMID: 31584763 PMCID: PMC7357371 DOI: 10.7705/biomedica.4299] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Indexed: 11/30/2022]
Abstract
Introduction: Parkinson’s disease is the second most common neurodegenerative disease. Monoamine oxidase B inhibitors are used in the treatment of this disease concomitantly with levodopa or as monotherapy. Several substituted coumarins have shown activity as inhibitors of monoamine oxidase B. Objective: To evaluate the possible antiparkinsonian effects of the coumarin analogue FCS005 (3-methyl-7H-furo[3,2-g]chromen-7-one) in mouse models, as well as its inhibitory activity towards monoamine oxidases (MAO) and its antioxidant activity. Materials and methods: FCS005 was synthesized and the reversal of hypokinesia was evaluated in the reserpine and levodopa models. Moreover, in the haloperidol model, its anticataleptic effects were evaluated. Additionally, the monoamine oxidase inhibitory activity and antioxidant activity of FCS005 were evaluated using in vitro and ex vivo studies, respectively. Results: FCS005 (100 mg/kg) caused the reversal of hypokinesia in the reserpine and levodopa models. This furocoumarin also presented anti-cataleptic effects at the same dose. Besides, it showed selective inhibitory activity towards the MAO-B isoform and antioxidant activity. Conclusion: These results attribute interesting properties to the compound FCS005. It is important to continue research on this molecule considering that it could be a potential antiparkinsonian agent.
Collapse
|
27
|
Tandarić T, Vianello R. Computational Insight into the Mechanism of the Irreversible Inhibition of Monoamine Oxidase Enzymes by the Antiparkinsonian Propargylamine Inhibitors Rasagiline and Selegiline. ACS Chem Neurosci 2019; 10:3532-3542. [PMID: 31264403 DOI: 10.1021/acschemneuro.9b00147] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Monoamine oxidases (MAOs) are flavin adenine dinucleotide containing flavoenzymes that catalyze the degradation of a range of brain neurotransmitters, whose imbalance is extensively linked with the pathology of various neurological disorders. This is why MAOs have been the central pharmacological targets in treating neurodegeneration for more than 60 years. Still, despite this practical importance, the precise chemical mechanisms underlying the irreversible inhibition of the MAO B isoform with clinical drugs rasagiline (RAS) and selegiline (SEL) remained unknown. Here we employed a combination of MD simulations, MM-GBSA binding free energy evaluations, and QM cluster calculations to show the MAO inactivation proceeds in three steps, where, in the rate-limiting first step, FAD utilizes its N5 atom to abstracts a hydride anion from the inhibitor α-CH2 group to ultimately give the final inhibitor-FAD adduct matching crystallographic data. The obtained free energy profiles reveal a lower activation energy for SEL by 1.2 kcal mol-1 and a higher reaction exergonicity by 0.8 kcal mol-1, with the former being in excellent agreement with experimental ΔΔG‡EXP = 1.7 kcal mol-1, thus rationalizing its higher in vivo reactivity over RAS. The calculated ΔGBIND energies confirm SEL binds better due to its bigger size and flexibility allowing it to optimize hydrophobic C-H···π and π···π interactions with residues throughout both of enzyme's cavities, particularly with FAD, Gln206 and four active site tyrosines, thus overcoming a larger ability of RAS to form hydrogen bonds that only position it in less reactive orientations for the hydride abstraction. Offered results elucidate structural determinants affecting the affinity and rates of the inhibition reaction that should be considered to cooperate when designing more effective compounds devoid of untoward effects, which are of utmost significance and urgency with the growing prevalence of brain diseases.
Collapse
Affiliation(s)
- Tana Tandarić
- Computational Organic Chemistry and Biochemistry Group, Ruđer Bošković Institute, Bijenička cesta 54, HR-10000 Zagreb, Croatia
| | - Robert Vianello
- Computational Organic Chemistry and Biochemistry Group, Ruđer Bošković Institute, Bijenička cesta 54, HR-10000 Zagreb, Croatia
| |
Collapse
|
28
|
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
|
29
|
Li S, Lv X, Cheng K, Tian Y, Huang X, Kong H, Duan Y, Han J, Liao C, Xie Z. Discovery of novel 2,3-dihydro-1H-inden-1-amine derivatives as selective monoamine oxidase B inhibitors. Bioorg Med Chem Lett 2019; 29:1090-1093. [DOI: 10.1016/j.bmcl.2019.02.030] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 02/26/2019] [Accepted: 02/26/2019] [Indexed: 01/07/2023]
|
30
|
Cheng K, Li S, Lv X, Tian Y, Kong H, Huang X, Duan Y, Han J, Xie Z, Liao C. Design, synthesis and biological evaluation of novel human monoamine oxidase B inhibitors based on a fragment in an X-ray crystal structure. Bioorg Med Chem Lett 2019; 29:1012-1018. [DOI: 10.1016/j.bmcl.2019.02.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 01/17/2019] [Accepted: 02/07/2019] [Indexed: 12/30/2022]
|
31
|
Verma G, Khan MF, Akhtar W, Alam MM, Akhter M, Shaquiquzzaman M. A Review Exploring Therapeutic Worth of 1,3,4-Oxadiazole Tailored Compounds. Mini Rev Med Chem 2019; 19:477-509. [PMID: 30324877 DOI: 10.2174/1389557518666181015152433] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 12/27/2017] [Accepted: 09/30/2018] [Indexed: 02/01/2023]
Abstract
1,3,4-Oxadiazole, a five-membered aromatic ring can be seen in a number of synthetic molecules. The peculiar structural feature of 1,3,4-oxadiazole ring with pyridine type of nitrogen atom is beneficial for 1,3,4-oxadiazole derivatives to have effective binding with different enzymes and receptors in biological systems through numerous weak interactions, thereby eliciting an array of bioactivities. Research in the area of development of 1,3,4-oxadiazole-based derivatives has become an interesting topic for the scientists. A number of 1,3,4-oxadiazole based compounds with high therapeutic potency are being extensively used for the treatment of different ailments, contributing to enormous development value. This work provides a systematic and comprehensive review highlighting current developments of 1,3,4-oxadiazole based compounds in the entire range of medicinal chemistry such as anticancer, antifungal, antibacterial, antitubercular, anti-inflammatory, antineuropathic, antihypertensive, antihistaminic, antiparasitic, antiobesity, antiviral, and other medicinal agents. It is believed that this review will be of great help for new thoughts in the pursuit for rational designs for the development of more active and less toxic 1,3,4-oxadiazole based medicinal agents.
Collapse
Affiliation(s)
- Garima Verma
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi-110062, India
| | - Mohemmed F Khan
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi-110062, India
| | - Wasim Akhtar
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi-110062, India
| | - Mohammad Mumtaz Alam
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi-110062, India
| | - Mymoona Akhter
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi-110062, India
| | - Mohammad Shaquiquzzaman
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi-110062, India
| |
Collapse
|
32
|
Venko K, Novič M. An In Silico Approach for Assessment of the Membrane Transporter Activities of Phenols: A Case Study Based on Computational Models of Transport Activity for the Transporter Bilitranslocase. Molecules 2019; 24:E837. [PMID: 30818768 PMCID: PMC6429229 DOI: 10.3390/molecules24050837] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 02/19/2019] [Accepted: 02/26/2019] [Indexed: 12/03/2022] Open
Abstract
Phenols are the most abundant naturally accessible antioxidants present in a human normal diet. Since numerous beneficial applications of phenols as preventive agents in various diseases were revealed, the evaluation of phenols bioavailability is of high interest of researchers, consumers and drug manufacturers. The hydrophilic nature of phenols makes a cell membrane penetration difficult, which imply an alternative way of uptake via membrane transporters. However, the structural and functional data of membrane transporters are limited, thus the in silico modelling is really challenging and urgent tool in elucidation of transporter ligands. Focus of this research was a particular transporter bilitranslocase (BTL). BTL has a broad tissue expression (vascular endothelium, absorptive and excretory epithelia) and can transport wide variety of poly-aromatic compounds. With available BTL data (pKi [mmol/L] for 120 organic compounds) a robust and reliable QSAR models for BTL transport activity were developed and extrapolated on 300 phenolic compounds. For all compounds the transporter profiles were assessed and results show that dietary phenols and some drug candidates are likely to interact with BTL. Moreover, synopsis of predictions from BTL models and hits/predictions of 20 transporters from Metrabase and Chembench platforms were revealed. With such joint transporter analyses a new insights for elucidation of BTL functional role were acquired. Regarding limitation of models for virtual profiling of transporter interactions the computational approach reported in this study could be applied for further development of reliable in silico models for any transporter, if in vitro experimental data are available.
Collapse
Affiliation(s)
- Katja Venko
- Laboratory for Cheminformatics, Theory Department, National Institute of Chemistry, SI-1000 Ljubljana, Slovenia.
| | - Marjana Novič
- Laboratory for Cheminformatics, Theory Department, National Institute of Chemistry, SI-1000 Ljubljana, Slovenia.
| |
Collapse
|
33
|
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: 77] [Impact Index Per Article: 12.8] [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
|
34
|
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]
|
35
|
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: 34] [Impact Index Per Article: 5.7] [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
|
36
|
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]
|
37
|
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
|
38
|
Ishiki HM, Filho JMB, da Silva MS, Scotti MT, Scotti L. Computer-aided Drug Design Applied to Parkinson Targets. Curr Neuropharmacol 2018; 16:865-880. [PMID: 29189169 PMCID: PMC6080092 DOI: 10.2174/1570159x15666171128145423] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 11/24/2017] [Indexed: 12/01/2022] Open
Abstract
Background Parkinson’s disease (PD) is a progressive neurodegenerative disorder characterized by debilitating motor deficits, as well as autonomic problems, cognitive declines, changes in affect and sleep disturbances. Although the scientific community has performed great efforts in the study of PD, and from the most diverse points of view, the disease remains incurable. The exact mechanism underlying its progression is unclear, but oxidative stress, mitochondrial dysfunction and inflammation are thought to play major roles in the etiology. Objective Current pharmacological therapies for the treatment of Parkinson’s disease are mostly inadequate, and new therapeutic agents are much needed. Methods In this review, recent advances in computer-aided drug design for the rational design of new compounds against Parkinson disease; using methods such as Quantitative Structure-Activity Relationships (QSAR), molecular docking, molecular dynamics and pharmacophore modeling are discussed. Results In this review, four targets were selected: the enzyme monoamine oxidase, dopamine agonists, acetylcholine receptors, and adenosine receptors. Conclusion Computer aided-drug design enables the creation of theoretical models that can be used in a large database to virtually screen for and identify novel candidate molecules.
Collapse
Affiliation(s)
- Hamilton M Ishiki
- University of Western Sao Paulo (Unoeste), Presidente Prudente, SP, Brazil
| | | | | | - Marcus T Scotti
- Federal University of Paraiba, Campus I, Joao Pessoa-PB, Brazil
| | - Luciana Scotti
- Federal University of Paraiba, Campus I, Joao Pessoa-PB, Brazil
| |
Collapse
|
39
|
Design, synthesis and bioevalucation of novel 2,3-dihydro-1 H -inden-1-amine derivatives as potent and selective human monoamine oxidase B inhibitors based on rasagiline. Eur J Med Chem 2018; 145:588-593. [DOI: 10.1016/j.ejmech.2018.01.029] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 01/08/2018] [Accepted: 01/09/2018] [Indexed: 12/25/2022]
|
40
|
Tripathi AC, Upadhyay S, Paliwal S, Saraf SK. Privileged scaffolds as MAO inhibitors: Retrospect and prospects. Eur J Med Chem 2018; 145:445-497. [PMID: 29335210 DOI: 10.1016/j.ejmech.2018.01.003] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 12/01/2017] [Accepted: 01/01/2018] [Indexed: 12/24/2022]
Abstract
This review aims to be a comprehensive, authoritative, critical, and readable review of general interest to the medicinal chemistry community because it focuses on the pharmacological, chemical, structural and computational aspects of diverse chemical categories as monoamine oxidase inhibitors (MAOIs). Monoamine oxidases (MAOs), namely MAO-A and MAO-B represent an enormously valuable class of neuronal enzymes embodying neurobiological origin and functions, serving as potential therapeutic target in neuronal pharmacotherapy, and hence we have coined the term "Neurozymes" which is being introduced for the first time ever. Nowadays, therapeutic attention on MAOIs engrosses two imperative categories; MAO-A inhibitors, in certain mental disorders such as depression and anxiety, and MAO-B inhibitors, in neurodegenerative disorders like Alzheimer's disease (AD) and Parkinson's disease (PD). The use of MAOIs declined due to some potential side effects, food and drug interactions, and introduction of other classes of drugs. However, curiosity in MAOIs is reviving and the recent developments of new generation of highly selective and reversible MAOIs, have renewed the therapeutic prospective of these compounds. The initial section of the review emphasizes on the detailed classification, structural and binding characteristics, therapeutic potential, current status and future challenges of the privileged pharmacophores. However, the chemical prospective of privileged scaffolds such as; aliphatic and aromatic amines, amides, hydrazines, azoles, diazoles, tetrazoles, indoles, azines, diazines, xanthenes, tricyclics, benzopyrones, and more interestingly natural products, along with their conclusive SARs have been discussed in the later segment of review. The last segment of the article encompasses some patents granted in the field of MAOIs, in a simplistic way.
Collapse
Affiliation(s)
- Avinash C Tripathi
- Division of Pharmaceutical Chemistry, Faculty of Pharmacy, Babu Banarasi Das Northern India Institute of Technology, Lucknow 226028, UP, India
| | - Savita Upadhyay
- Division of Pharmaceutical Chemistry, Faculty of Pharmacy, Babu Banarasi Das Northern India Institute of Technology, Lucknow 226028, UP, India
| | - Sarvesh Paliwal
- Pharmacy Department, Banasthali Vidyapith, Banasthali, Tonk 304022, Rajasthan, India
| | - Shailendra K Saraf
- Division of Pharmaceutical Chemistry, Faculty of Pharmacy, Babu Banarasi Das Northern India Institute of Technology, Lucknow 226028, UP, India.
| |
Collapse
|
41
|
Costas-Lago MC, Besada P, Rodríguez-Enríquez F, Viña D, Vilar S, Uriarte E, Borges F, Terán C. Synthesis and structure-activity relationship study of novel 3-heteroarylcoumarins based on pyridazine scaffold as selective MAO-B inhibitors. Eur J Med Chem 2017; 139:1-11. [PMID: 28797881 DOI: 10.1016/j.ejmech.2017.07.045] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 07/19/2017] [Accepted: 07/21/2017] [Indexed: 10/19/2022]
Abstract
Compounds of hybrid structure pyridazine-coumarin were discovered as potent, selective and reversible inhibitors of monoamine oxidase B (MAO-B). These compounds were synthesized in good yield following a multistep approach based on Knoevenagel reaction and using as key intermediate pyridazinone 16, which was obtained from maleic anhydride and furan. Compounds 9b and 9d are the most active compounds of these series, with IC50 values in the sub-micromolar range, and lack of cytotoxic effects. Theoretical calculation of ADME properties also suggested a good pharmacokinetic profile for both compounds. Docking simulations provided insights into enzyme inhibitor interactions and allowed us to rationalize the observed structure-activity relationships (SARs).
Collapse
Affiliation(s)
- María Carmen Costas-Lago
- Departamento de Química Orgánica and Instituto de Investigación Sanitaria Galicia Sur (IISGS), Universidade de Vigo, 36310 Vigo, Spain
| | - Pedro Besada
- Departamento de Química Orgánica and Instituto de Investigación Sanitaria Galicia Sur (IISGS), Universidade de Vigo, 36310 Vigo, Spain
| | - Fernanda Rodríguez-Enríquez
- Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS) Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Dolores Viña
- Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS) Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Santiago Vilar
- Departamento de Química Orgánica, Facultad de Farmacia, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Eugenio Uriarte
- Departamento de Química Orgánica, Facultad de Farmacia, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; Instituto de Ciencias Químicas Aplicadas, Universidad Autónoma de Chile, 7500912 Santiago, Chile
| | - Fernanda Borges
- CIQUP/Departamento de Química e Bioquímica, Faculdade de Ciencias, Universidade do Porto, 4169-007 Porto, Portugal
| | - Carmen Terán
- Departamento de Química Orgánica and Instituto de Investigación Sanitaria Galicia Sur (IISGS), Universidade de Vigo, 36310 Vigo, Spain.
| |
Collapse
|
42
|
The evaluation of 1,4-benzoquinones as inhibitors of human monoamine oxidase. Eur J Med Chem 2017; 135:196-203. [DOI: 10.1016/j.ejmech.2017.04.055] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 04/03/2017] [Accepted: 04/20/2017] [Indexed: 02/06/2023]
|
43
|
Can ÖD, Osmaniye D, Demir Özkay Ü, Sağlık BN, Levent S, Ilgın S, Baysal M, Özkay Y, Kaplancıklı ZA. MAO enzymes inhibitory activity of new benzimidazole derivatives including hydrazone and propargyl side chains. Eur J Med Chem 2017; 131:92-106. [DOI: 10.1016/j.ejmech.2017.03.009] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 02/22/2017] [Accepted: 03/04/2017] [Indexed: 12/25/2022]
|
44
|
Mangiatordi GF, Alberga D, Pisani L, Gadaleta D, Trisciuzzi D, Farina R, Carotti A, Lattanzi G, Catto M, Nicolotti O. A rational approach to elucidate human monoamine oxidase molecular selectivity. Eur J Pharm Sci 2017; 101:90-99. [DOI: 10.1016/j.ejps.2017.02.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 01/27/2017] [Accepted: 02/05/2017] [Indexed: 02/05/2023]
|
45
|
Domínguez JL, Fernández-Nieto F, Brea JM, Catto M, Paleo MR, Porto S, Sardina FJ, Castro M, Pisani L, Carotti A, Soto-Otero R, Méndez-Alvarez E, Villaverde MC, Sussman F. 8-Aminomethyl-7-hydroxy-4-methylcoumarins as Multitarget Leads for Alzheimer's Disease. ChemistrySelect 2016. [DOI: 10.1002/slct.201600735] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- José L. Domínguez
- Departamento de Química Orgánica; Facultad de Química; Universidade de Santiago de Compostela; 15782 Santiago de Compostela Spain
| | - Fernando Fernández-Nieto
- Departamento de Química Orgánica and Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS); Universidade de Santiago de Compostela; 15782 Santiago de Compostela Spain
| | - José M. Brea
- Departamento de Farmacología; Instituto de Farmacia Industrial; Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS); Universidade de Santiago de Compostela; 15782 Santiago de Compostela Spain
| | - Marco Catto
- Dipartimento di Farmacia-Scienze del Farmaco; Università degli Studi di Bari “Aldo Moro”; 70125 Bari Italy
| | - M. Rita Paleo
- Departamento de Química Orgánica and Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS); Universidade de Santiago de Compostela; 15782 Santiago de Compostela Spain
| | - Silvia Porto
- Departamento de Química Orgánica and Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS); Universidade de Santiago de Compostela; 15782 Santiago de Compostela Spain
| | - F. Javier Sardina
- Departamento de Química Orgánica and Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS); Universidade de Santiago de Compostela; 15782 Santiago de Compostela Spain
| | - Marian Castro
- Departamento de Farmacología; Instituto de Farmacia Industrial; Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS); Universidade de Santiago de Compostela; 15782 Santiago de Compostela Spain
| | - Leonardo Pisani
- Dipartimento di Farmacia-Scienze del Farmaco; Università degli Studi di Bari “Aldo Moro”; 70125 Bari Italy
| | - Angelo Carotti
- Dipartimento di Farmacia-Scienze del Farmaco; Università degli Studi di Bari “Aldo Moro”; 70125 Bari Italy
| | - Ramón Soto-Otero
- Grupo de Neuroquímica; Departamento de Bioquímica y Biología Molecular; Facultad de Medicina; Universidade de Santiago de Compostela; 15782 Santiago de Compostela Spain
| | - Estefanía Méndez-Alvarez
- Grupo de Neuroquímica; Departamento de Bioquímica y Biología Molecular; Facultad de Medicina; Universidade de Santiago de Compostela; 15782 Santiago de Compostela Spain
| | - M. Carmen Villaverde
- Departamento de Química Orgánica; Facultad de Química; Universidade de Santiago de Compostela; 15782 Santiago de Compostela Spain
| | - Fredy Sussman
- Departamento de Química Orgánica; Facultad de Química; Universidade de Santiago de Compostela; 15782 Santiago de Compostela Spain
| |
Collapse
|
46
|
Pisani L, Farina R, Catto M, Iacobazzi RM, Nicolotti O, Cellamare S, Mangiatordi GF, Denora N, Soto-Otero R, Siragusa L, Altomare CD, Carotti A. Exploring Basic Tail Modifications of Coumarin-Based Dual Acetylcholinesterase-Monoamine Oxidase B Inhibitors: Identification of Water-Soluble, Brain-Permeant Neuroprotective Multitarget Agents. J Med Chem 2016; 59:6791-806. [DOI: 10.1021/acs.jmedchem.6b00562] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Leonardo Pisani
- Dipartimento
di Farmacia—Scienze del Farmaco, Università degli Studi di Bari “Aldo Moro”, via E. Orabona 4, I-70125 Bari, Italy
| | - Roberta Farina
- Dipartimento
di Farmacia—Scienze del Farmaco, Università degli Studi di Bari “Aldo Moro”, via E. Orabona 4, I-70125 Bari, Italy
| | - Marco Catto
- Dipartimento
di Farmacia—Scienze del Farmaco, Università degli Studi di Bari “Aldo Moro”, via E. Orabona 4, I-70125 Bari, Italy
| | - Rosa Maria Iacobazzi
- Dipartimento
di Farmacia—Scienze del Farmaco, Università degli Studi di Bari “Aldo Moro”, via E. Orabona 4, I-70125 Bari, Italy
| | - Orazio Nicolotti
- Dipartimento
di Farmacia—Scienze del Farmaco, Università degli Studi di Bari “Aldo Moro”, via E. Orabona 4, I-70125 Bari, Italy
| | - Saverio Cellamare
- Dipartimento
di Farmacia—Scienze del Farmaco, Università degli Studi di Bari “Aldo Moro”, via E. Orabona 4, I-70125 Bari, Italy
| | - Giuseppe Felice Mangiatordi
- Dipartimento
di Farmacia—Scienze del Farmaco, Università degli Studi di Bari “Aldo Moro”, via E. Orabona 4, I-70125 Bari, Italy
| | - Nunzio Denora
- Dipartimento
di Farmacia—Scienze del Farmaco, Università degli Studi di Bari “Aldo Moro”, via E. Orabona 4, I-70125 Bari, Italy
| | - Ramon Soto-Otero
- Departamento
de Bioquimica y Biologia Molecular, Facultad de Medicina, Universidad de Santiago de Compostela, San Francisco I, E-15782 Santiago de Compostela, Spain
| | - Lydia Siragusa
- Molecular Discovery Limited 215
Marsh Road, Pinner, Middlesex, London HA5 5NE, U.K
| | - Cosimo Damiano Altomare
- Dipartimento
di Farmacia—Scienze del Farmaco, Università degli Studi di Bari “Aldo Moro”, via E. Orabona 4, I-70125 Bari, Italy
| | - Angelo Carotti
- Dipartimento
di Farmacia—Scienze del Farmaco, Università degli Studi di Bari “Aldo Moro”, via E. Orabona 4, I-70125 Bari, Italy
| |
Collapse
|
47
|
Romero-Durán FJ, Alonso N, Yañez M, Caamaño O, García-Mera X, González-Díaz H. Brain-inspired cheminformatics of drug-target brain interactome, synthesis, and assay of TVP1022 derivatives. Neuropharmacology 2016; 103:270-8. [DOI: 10.1016/j.neuropharm.2015.12.019] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Revised: 11/22/2015] [Accepted: 12/18/2015] [Indexed: 01/22/2023]
|
48
|
Chen R, Xiao J, Ni Y, Xu HF, Zheng M, Tong X, Zhang TT, Liao C, Tang WJ. Novel tricyclic pyrazolo[1,5-d][1,4]benzoxazepin-5(6H)-one: Design, synthesis, model and use as hMAO-B inhibitors. Bioorg Med Chem 2016; 24:1741-8. [DOI: 10.1016/j.bmc.2016.02.045] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 02/26/2016] [Accepted: 02/29/2016] [Indexed: 01/24/2023]
|
49
|
Pisani L, Farina R, Soto-Otero R, Denora N, Mangiatordi GF, Nicolotti O, Mendez-Alvarez E, Altomare CD, Catto M, Carotti A. Searching for Multi-Targeting Neurotherapeutics against Alzheimer's: Discovery of Potent AChE-MAO B Inhibitors through the Decoration of the 2H-Chromen-2-one Structural Motif. Molecules 2016; 21:362. [PMID: 26999091 PMCID: PMC6273473 DOI: 10.3390/molecules21030362] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 03/05/2016] [Accepted: 03/10/2016] [Indexed: 11/16/2022] Open
Abstract
The need for developing real disease-modifying drugs against neurodegenerative syndromes, particularly Alzheimer’s disease (AD), shifted research towards reliable drug discovery strategies to unveil clinical candidates with higher therapeutic efficacy than single-targeting drugs. By following the multi-target approach, we designed and synthesized a novel class of dual acetylcholinesterase (AChE)-monoamine oxidase B (MAO-B) inhibitors through the decoration of the 2H-chromen-2-one skeleton. Compounds bearing a propargylamine moiety at position 3 displayed the highest in vitro inhibitory activities against MAO-B. Within this series, derivative 3h emerged as the most interesting hit compound, being a moderate AChE inhibitor (IC50 = 8.99 µM) and a potent and selective MAO-B inhibitor (IC50 = 2.8 nM). Preliminary studies in human neuroblastoma SH-SY5Y cell lines demonstrated its low cytotoxicity and disclosed a promising neuroprotective effect at low doses (0.1 µM) under oxidative stress conditions promoted by two mitochondrial toxins (oligomycin-A and rotenone). In a Madin-Darby canine kidney (MDCK)II-MDR1 cell-based transport study, Compound 3h was able to permeate the BBB-mimicking monolayer and did not result in a glycoprotein-p (P-gp) substrate, showing an efflux ratio = 0.96, close to that of diazepam.
Collapse
Affiliation(s)
- Leonardo Pisani
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari "Aldo Moro", via E. Orabona, 4, I-70125 Bari, Italy.
| | - Roberta Farina
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari "Aldo Moro", via E. Orabona, 4, I-70125 Bari, Italy.
| | - Ramon Soto-Otero
- Grupo de Neuroquimica, Departamento de Bioquimica y Biologia Molecular, Facultad de Medicina, Universidad de Santiago de Compostela, San Francisco I, E-15782 Santiago de Compostela, Spain.
| | - Nunzio Denora
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari "Aldo Moro", via E. Orabona, 4, I-70125 Bari, Italy.
| | - Giuseppe Felice Mangiatordi
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari "Aldo Moro", via E. Orabona, 4, I-70125 Bari, Italy.
| | - Orazio Nicolotti
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari "Aldo Moro", via E. Orabona, 4, I-70125 Bari, Italy.
| | - Estefania Mendez-Alvarez
- Grupo de Neuroquimica, Departamento de Bioquimica y Biologia Molecular, Facultad de Medicina, Universidad de Santiago de Compostela, San Francisco I, E-15782 Santiago de Compostela, Spain.
| | - Cosimo Damiano Altomare
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari "Aldo Moro", via E. Orabona, 4, I-70125 Bari, Italy.
| | - Marco Catto
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari "Aldo Moro", via E. Orabona, 4, I-70125 Bari, Italy.
| | - Angelo Carotti
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari "Aldo Moro", via E. Orabona, 4, I-70125 Bari, Italy.
| |
Collapse
|
50
|
Distinto S, Meleddu R, Yanez M, Cirilli R, Bianco G, Sanna ML, Arridu A, Cossu P, Cottiglia F, Faggi C, Ortuso F, Alcaro S, Maccioni E. Drug design, synthesis, in vitro and in silico evaluation of selective monoaminoxidase B inhibitors based on 3-acetyl-2-dichlorophenyl-5-aryl-2,3-dihydro-1,3,4-oxadiazole chemical scaffold. Eur J Med Chem 2016; 108:542-552. [DOI: 10.1016/j.ejmech.2015.12.026] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Revised: 12/10/2015] [Accepted: 12/12/2015] [Indexed: 12/18/2022]
|