1
|
Abdallah AE. Review on anti-alzheimer drug development: approaches, challenges and perspectives. RSC Adv 2024; 14:11057-11088. [PMID: 38586442 PMCID: PMC10995770 DOI: 10.1039/d3ra08333k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 03/22/2024] [Indexed: 04/09/2024] Open
Abstract
Alzheimer is an irreversible progressive neurodegenerative disease that causes failure of cerebral neurons and disability of the affected person to practice normal daily life activities. There is no concrete evidence to identify the exact reason behind the disease, so several relevant hypotheses emerged, highlighting many possible therapeutic targets, such as acetylcholinesterase, cholinergic receptors, N-methyl d-aspartate receptors, phosphodiesterase, amyloid β protein, protein phosphatase 2A, glycogen synthase kinase-3 beta, β-secretase, γ-secretase, α-secretase, serotonergic receptors, glutaminyl cyclase, tumor necrosis factor-α, γ-aminobutyric acid receptors, and mitochondria. All of these targets have been involved in the design of new potential drugs. An extensive number of these drugs have been studied in clinical trials. However, only galantamine, donepezil, and rivastigmine (ChEIs), memantine (NMDA antagonist), and aducanumab and lecanemab (selective anti-Aβ monoclonal antibodies) have been approved for AD treatment. Many drugs failed in the clinical trials to such an extent that questions have been posed about the significance of some of the aforementioned targets. On the contrary, the data of other drugs were promising and shed light on the significance of their targets for the development of new potent anti-alzheimer drugs.
Collapse
Affiliation(s)
- Abdallah E Abdallah
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University 11884 Cairo Egypt
| |
Collapse
|
2
|
(2-Hydroxy-3-Methoxybenzylidene)thiazolo[3,2- a]pyrimidines: Synthesis, Self-Assembly in the Crystalline Phase and Cytotoxic Activity. Int J Mol Sci 2023; 24:ijms24032084. [PMID: 36768407 PMCID: PMC9917025 DOI: 10.3390/ijms24032084] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/16/2023] [Accepted: 01/18/2023] [Indexed: 01/21/2023] Open
Abstract
A series of new 2-hydroxy-3-methoxybenzylidenethiazolo[3,2-a]pyrimidines with different aryl substituents at the 5 position are synthesized and characterized by 1H/ 13C NMR and IR-spectroscopy and mass-spectrometry, as well as single crystal X-ray diffraction (SCXRD). It was demonstrated that the type of hydrogen bonding can play a key role in the chiral discrimination of these compounds in the crystalline phase. The hydrogen bond of the O-H...N type leads to 1D supramolecular heterochiral chains or conglomerate crystallization in the case of the formation of homochiral chains. The hydrogen bond of O-H...O type gave racemic dimers, which are packed into 2D supramolecular layers with a parallel or angular dimers arrangement. Halogen bonding of the N...Br or O...Br type brings a new motif into supramolecular self-assembly in the crystalline phase: the formation of 1D supramolecular homochiral chains instead 2D supramolecular layers. The study of cytotoxicity against various tumor cells in vitro was carried out. It was found that 2-hydroxy-3-methoxybenzylidenethiazolo[3,2-a]pyrimidines with 3-nitrophenyl substituent at C5 carbon atom demonstrated a high efficiency against M-HeLa (cervical adenocarcinoma) and low cytotoxicity against normal liver cells.
Collapse
|
3
|
Abdelwahab HE, Ibrahim HZ, Omar AZ. Design, Synthesis, DFT, Molecular Docking, and Biological Evalution of Pyrazole Derivatives as Potent Acetyl Cholinestrease Inhibitors. J Mol Struct 2023. [DOI: 10.1016/j.molstruc.2022.134137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|
4
|
Novel and Potent Acetylcholinesterase Inhibitors for the Treatment of Alzheimer's Disease from Natural (±)-7,8-Dihydroxy-3-methyl-isochroman-4-one. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27103090. [PMID: 35630563 PMCID: PMC9145193 DOI: 10.3390/molecules27103090] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/06/2022] [Accepted: 05/09/2022] [Indexed: 11/22/2022]
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disease that causes memory and cognitive decline as well as behavioral problems. It is a progressive and well recognized complex disease; therefore, it is very urgent to develop novel and effective anti-AD drugs. In this study, a series of novel isochroman-4-one derivatives from natural (±)-7,8-dihydroxy-3-methyl-isochroman-4-one [(±)-XJP] were designed and synthesized, and their anti-AD potential was evaluated. Among them, compound 10a [(Z)-3-acetyl-1-benzyl-4-((6,7-dimethoxy-4-oxoisochroman-3-ylidene)methyl)pyridin-1-ium bromide] possessed potent anti-acetylcholinesterase (AChE) activity as well as modest antioxidant activity. Further molecular modeling and kinetic investigations revealed that compound 10a was a dual-binding inhibitor that binds to both catalytic anionic site (CAS) and peripheral anionic site (PAS) of the enzyme AChE. In addition, compound 10a exhibited low cytotoxicity and moderate anti-Aβ aggregation efficacy. Moreover, the in silico screening suggested that these compounds could pass across the blood–brain barrier with high penetration. These findings show that compound 10a was a promising lead from a natural product with potent AChE inhibitory activity and deserves to be further developed for the prevention and treatment of AD.
Collapse
|
5
|
Dorababu A. Promising heterocycle-based scaffolds in recent (2019-2021) anti-Alzheimer's drug design and discovery. Eur J Pharmacol 2022; 920:174847. [PMID: 35218718 DOI: 10.1016/j.ejphar.2022.174847] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 02/06/2022] [Accepted: 02/18/2022] [Indexed: 12/28/2022]
Abstract
Alzheimer's disease (AD) is one of the neurodegenerative diseases that led to morbidity and mortality world-wide. It is a complex disease whose etiology is not completely known that leads to difficulty in prevent or cure of the AD. Also, there are only few approved drugs for AD treatment. Apart from deaths due to AD, expenditure of treatment and care of AD patients is higher than that of treatment of HIV and cancer diseases combined. Hence, it leads to an economic burden also. Although research is being carried out on designing drugs for AD, most of them have ended up in poor inhibitors with high toxicity. Hence, researchers should shoulder a great responsibility of discovery of efficient drugs for AD treatment. In the field of drug discovery, heterocycles played an important role. Also, most of the heterocyclic scaffolds have been used in design of potent anti-AD agents. In view of this, heterocyclic molecules reported recently are compiled and evaluated comprehensively. Especially, the molecules which exhibited pronounced activity are emphasized and described with respect to structure-activity relationship (SAR) in brief.
Collapse
Affiliation(s)
- Atukuri Dorababu
- SRMPP Government First Grade College, Huvinahadagali, 583219, India.
| |
Collapse
|
6
|
Adeowo FY, Elrashedy AA, Ejalonibu MA, Lawal IA, Lawal MM, Kumalo HM. Pharmacophore mapping of the crucial mediators of acetylcholinesterase and butyrylcholinesterase dual inhibition in Alzheimer's disease. Mol Divers 2022; 26:2761-2774. [PMID: 35067751 DOI: 10.1007/s11030-022-10377-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 01/03/2022] [Indexed: 11/25/2022]
Abstract
Optimization and re-optimization of bioactive molecules using in silico methods have found application in the design of more active ones. Herein, we applied a pharmacophore modeling approach to screen potent dual inhibitors of acetylcholinesterase (AChE) and butyrylcholinesterase (BuChE) aimed at Alzheimer's disease (AD) treatment. The investigation entails molecular dynamics simulation, docking, pharmacophore modeling, drug-like screening, and binding energy analysis. We prepared a pharmacophore model from approved inhibitors of AChE and BuChE to predict the crucial moieties required for optimum molecular interaction with these proteins. The obtained pharmacophore model, used for database screening via some critical criteria, showed 229 hit molecules. Further analyses showed 42 likely dual inhibitors of AChE/BuChE with drug-like and pharmacokinetics properties the same as the approved cholinesterase inhibitors. Finally, we identified 14 dual molecules with improved potentials over the existing inhibitors and simulated ZINC92385797 bound to human AChE and BuChE structure after noticing that these 14 molecules are similar. The selected compound maintained relative stability at the active sites of both proteins over 120 ns simulation. Our integrated protocols showed the pertinent recipes of anti-AD drug design through the in silico pipeline.
Collapse
Affiliation(s)
- Fatima Y Adeowo
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, University of KwaZulu-Natal, Durban, 4001, South Africa
| | - Ahmed A Elrashedy
- Natural and Microbial Product Department, National Research Centre, Dokki, Giza, 12622, Egypt
| | - Murtala A Ejalonibu
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, University of KwaZulu-Natal, Durban, 4001, South Africa
| | - Isiaka A Lawal
- Chemistry Department, Faculty of Applied and Computer Science, Vaal University of Technology, Vanderbijlpark Campus, Boulevard, Vanderbijlpark, 1900, South Africa
| | - Monsurat M Lawal
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, University of KwaZulu-Natal, Durban, 4001, South Africa.
| | - Hezekiel M Kumalo
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, University of KwaZulu-Natal, Durban, 4001, South Africa.
| |
Collapse
|
7
|
Girgis AS, D'Arcy P, Aboshouk DR, Bekheit MS. Synthesis and bio-properties of 4-piperidone containing compounds as curcumin mimics. RSC Adv 2022; 12:31102-31123. [DOI: 10.1039/d2ra05518j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 10/23/2022] [Indexed: 11/05/2022] Open
Abstract
3,5-Diyliden-4-piperidone scaffold are considered as curcumin mimic exhibiting diverse bio-properties.
Collapse
Affiliation(s)
- Adel S. Girgis
- Department of Pesticide Chemistry, National Research Centre, Dokki, Giza 12622, Egypt
| | - Padraig D'Arcy
- Department of Biomedical and Clinical Sciences, Linköping University, SE-581 83, Linköping, Sweden
| | - Dalia R. Aboshouk
- Department of Pesticide Chemistry, National Research Centre, Dokki, Giza 12622, Egypt
| | - Mohamed S. Bekheit
- Department of Pesticide Chemistry, National Research Centre, Dokki, Giza 12622, Egypt
| |
Collapse
|
8
|
Murtuja S, Shilkar D, Sarkar B, Sinha BN, Jayaprakash V. A short survey of dengue protease inhibitor development in the past 6 years (2015-2020) with an emphasis on similarities between DENV and SARS-CoV-2 proteases. Bioorg Med Chem 2021; 49:116415. [PMID: 34601454 PMCID: PMC8450225 DOI: 10.1016/j.bmc.2021.116415] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 08/06/2021] [Accepted: 09/11/2021] [Indexed: 11/26/2022]
Abstract
Dengue remains a disease of significant concern, responsible for nearly half of all arthropod-borne disease cases across the globe. Due to the lack of potent and targeted therapeutics, palliative treatment and the adoption of preventive measures remain the only available options. Compounding the problem further, the failure of the only dengue vaccine, Dengvaxia®, also delivered a significant blow to any hopes for the treatment of dengue fever. However, the success of Human Immuno-deficiency Virus (HIV) and Hepatitis C Virus (HCV) protease inhibitors in the past have continued to encourage researchers to investigate other viral protease targets. Dengue virus (DENV) NS2B-NS3 protease is an attractive target partly due to its role in polyprotein processing and also for being the most conserved domain in the viral genome. During the early days of the COVID-19 pandemic, a few cases of Dengue-COVID 19 co-infection were reported. In this review, we compared the substrate-peptide residue preferences and the residues lining the sub-pockets of the proteases of these two viruses and analyzed the significance of this similarity. Also, we attempted to abridge the developments in anti-dengue drug discovery in the last six years (2015-2020), focusing on critical discoveries that influenced the research.
Collapse
Affiliation(s)
- Sheikh Murtuja
- Department of Pharmaceutical Sciences & Technology, Birla Institute of Technology, Mesra, Ranchi 835215 (JH), India
| | - Deepak Shilkar
- Department of Pharmaceutical Sciences & Technology, Birla Institute of Technology, Mesra, Ranchi 835215 (JH), India
| | - Biswatrish Sarkar
- Department of Pharmaceutical Sciences & Technology, Birla Institute of Technology, Mesra, Ranchi 835215 (JH), India
| | - Barij Nayan Sinha
- Department of Pharmaceutical Sciences & Technology, Birla Institute of Technology, Mesra, Ranchi 835215 (JH), India
| | - Venkatesan Jayaprakash
- Department of Pharmaceutical Sciences & Technology, Birla Institute of Technology, Mesra, Ranchi 835215 (JH), India.
| |
Collapse
|
9
|
Adeowo FY, Oyetunji TP, Ejalonibu MA, Ndagi U, Kumalo HM, Lawal MM. Tailored Modeling of Rivastigmine Derivatives as Dual Acetylcholinesterase and Butyrylcholinesterase Inhibitors for Alzheimer's Disease Treatment. Chem Biodivers 2021; 18:e2100361. [PMID: 34547176 DOI: 10.1002/cbdv.202100361] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 09/21/2021] [Indexed: 12/13/2022]
Abstract
Rational modification of known drug candidates to design more potent ones using computational methods has found application in drug design, development, and discovery. Herein, we integrate computational and theoretical methodologies to unveil rivastigmine derivatives as dual inhibitors of acetylcholinesterase (AChE) and butyrylcholinesterase (BuChE) for Alzheimer's disease (AD) management. The investigation entails pharmacokinetics screening, density functional theory (DFT) mechanistic study, molecular docking, and molecular dynamics (MD) simulation. We designed over 20 rivastigmine substituents, subject them to some analyses, and identified RL2 with an appreciable blood-brain barrier score and no permeability glycoprotein binding. The compound shows higher acylation energy and a favored binding affinity to the cholinesterase enzymes. RL2 interacts with the AChE and BuChE active sites showing values of -41.1/-39.5 kcal mol-1 while rivastigmine binds with -32.7/-30.7 kcal mol-1 for these enzymes. The study revealed RL2 (4-fluorophenyl rivastigmine) as a potential dual inhibitor for AChE and BuChE towards Alzheimer's disorder management.
Collapse
Affiliation(s)
- Fatima Y Adeowo
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, University of KwaZulu-Natal, Durban, 4001, South Africa
| | | | - Murtala A Ejalonibu
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, University of KwaZulu-Natal, Durban, 4001, South Africa
| | - Umar Ndagi
- Center for Trans-Sahara Disease, Vaccine and Drug Research, IBB University Lapai, Niger State, Minna, Nigeria
| | - Hezekiel M Kumalo
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, University of KwaZulu-Natal, Durban, 4001, South Africa
| | - Monsurat M Lawal
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, University of KwaZulu-Natal, Durban, 4001, South Africa
| |
Collapse
|
10
|
Karasova JZ, Hrabinova M, Krejciova M, Jun D, Kuca K. Donepezil and Rivastigmine: Pharmacokinetic Profile and Brain-targeting After Intramuscular Administration in Rats. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2021; 19:95-102. [PMID: 33680013 PMCID: PMC7758017 DOI: 10.22037/ijpr.2019.1100723] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Current palliative pharmacotherapy of Alzheimer's disease based on the cholinergic hypothesis led to the development of four cholinesterase inhibitors. These compounds can bring prolongation of the symptom-free period in some patients. This is the first report directly comparing donepezil and rivastigmine plasma and brain levels in in-vivo study. Donepezil and rivastigmine were applied i.m. to rats; the dose was calculated from clinical recommendations. The samples were analysed on an Agilent 1260 Series LC with UV/VIS detector. An analytical column (Waters Spherisorb S5 W (250 mm × 4.6 i.d.; 5 μm particle size)) with guard column (Waters Spherisorb S5 W (30 mm × 4.6 mm i.d.)) was used. The mobile phase contained acetonitrile and 50 mM sodium dihydrogen phosphate (17:83; v/v); pH 3.1. The LLOQ in rat plasma was 0.5 ng/mL for donepezil and 0.8 ng/mL for rivastigmine, and the LLOQ in rat brain was 1.0 ng/mL for donepezil and 1.1 ng/mL for rivastigmine. Both compounds showed ability to target the central nervous system, with brain concentrations exceeding those in plasma. Maximum brain concentration after i.m. administration was reached in the 36 (8.34 ± 0.34 ng/mL) and 17 minute (6.18 ± 0.40 ng/mL), respectively for donepezil and rivastigmine. The differences in brain profile can be most easily expressed by plasma/brain AUCtotal ratios: donepezil ratio in the brain was nine-times higher than in plasma and rivastigmine ratio was less than two-times higher than in plasma.
Collapse
Affiliation(s)
- Jana Zdarova Karasova
- Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, University of Defence, Hradec Kralove, Czech Republic.,Biomedical Research Centre, University Hospital, Hradec Kralove, Czech Republic
| | - Martina Hrabinova
- Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, University of Defence, Hradec Kralove, Czech Republic
| | - Marketa Krejciova
- Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, University of Defence, Hradec Kralove, Czech Republic
| | - Daniel Jun
- Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, University of Defence, Hradec Kralove, Czech Republic.,Biomedical Research Centre, University Hospital, Hradec Kralove, Czech Republic
| | - Kamil Kuca
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czech Republic
| |
Collapse
|
11
|
Wang K, Shang F, Chen D, Cao T, Wang X, Jiao J, He S, Liang X. Protein liposomes-mediated targeted acetylcholinesterase gene delivery for effective liver cancer therapy. J Nanobiotechnology 2021; 19:31. [PMID: 33482834 PMCID: PMC7821407 DOI: 10.1186/s12951-021-00777-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Accepted: 01/13/2021] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Effective methods to deliver therapeutic genes to solid tumors and improve their bioavailability are the main challenges of current medical research on gene therapy. The development of efficient non-viral gene vector with tumor-targeting has very important application value in the field of cancer therapy. Proteolipid integrated with tumor-targeting potential of functional protein and excellent gene delivery performance has shown potential for targeted gene therapy. RESULTS Herein, we prepared transferrin-modified liposomes (Tf-PL) for the targeted delivery of acetylcholinesterase (AChE) therapeutic gene to liver cancer. We found that the derived Tf-PL/AChE liposomes exhibited much higher transfection efficiency than the commercial product Lipo 2000 and shown premium targeting efficacy to liver cancer SMMC-7721 cells in vitro. In vivo, the Tf-PL/AChE could effectively target liver cancer, and significantly inhibit the growth of liver cancer xenografts grafted in nude mice by subcutaneous administration. CONCLUSIONS This study proposed a transferrin-modified proteolipid-mediated gene delivery strategy for targeted liver cancer treatment, which has a promising potential for precise personalized cancer therapy.
Collapse
Affiliation(s)
- Kai Wang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine Affiliated Renji Hospital, Shanghai, 200032, People's Republic of China
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Public Health Clinical Center, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
| | - Fusheng Shang
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, People's Republic of China
| | - Dagui Chen
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, People's Republic of China
| | - Tieliu Cao
- Department of Hepatobiliary-pancreatic and Integrative Oncology, Minhang Branch, Fudan University Shanghai Cancer Center, Shanghai, 200240, People's Republic of China
| | - Xiaowei Wang
- Department of traditional Chinese medicine, Changzheng Hospital, Shanghai, 200001, People's Republic of China
| | - Jianpeng Jiao
- Department of traditional Chinese medicine, Changzheng Hospital, Shanghai, 200001, People's Republic of China
| | - Shengli He
- Department of Hepatobiliary-pancreatic and Integrative Oncology, Minhang Branch, Fudan University Shanghai Cancer Center, Shanghai, 200240, People's Republic of China.
| | - Xiaofei Liang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine Affiliated Renji Hospital, Shanghai, 200032, People's Republic of China.
| |
Collapse
|
12
|
Adeowo FY, Ejalonibu MA, Elrashedy AA, Lawal MM, Kumalo HM. Multi-target approach for Alzheimer's disease treatment: computational biomolecular modeling of cholinesterase enzymes with a novel 4- N-phenylaminoquinoline derivative reveal promising potentials. J Biomol Struct Dyn 2020; 39:3825-3841. [PMID: 33030113 DOI: 10.1080/07391102.2020.1826129] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
The identification of dual inhibitors targeting the active sites of the cholinesterase enzymes, acetylcholinesterase (AChE) and butyrylcholinesterase (BuChE), have lately surfaced as a multi-approach towards Alzheimer treatment. More recently, a novel series of 4-N-phenylaminoquinolines was synthesized and evaluated against AChE and BuChE in which one of the compounds displayed appreciable inhibition compared to the standard compound, galantamine. To provide a clearer picture of the inhibition mechanism of this potent compound at the molecular level, computational biomolecular modeling was carried out. The investigation was initiated with the exploration of the chemical properties of the identified compound 11 b and reference drug, galantamine. Density functional theory (DFT) calculations reveal some conceptual parameters that provide information on the stability and reactivity of the compounds as potential inhibitors. To unveil the binding mechanism, energetics and enzyme-ligand interactions, molecular dynamics (MD) simulations of six different systems were executed over a period. Calculated binding free energy values are in the same order with experimental IC50 data. Identification of the main residues driving optimum binding of the active compound 11 b to the binding region of both AChE and BuChE showed Trp81 and Trp110 as the most important, respectively. It was proposed that the studied compound could serve as a dual inhibitor for AChE and BuChE, therefore, would potentially be a promising moiety in a multi-target approach for the treatment of Alzheimer's disorder.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Fatima Y Adeowo
- Drug Research and Innovation Unit, Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, University of KwaZulu-Natal, Durban, South Africa
| | - Murtala A Ejalonibu
- Drug Research and Innovation Unit, Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, University of KwaZulu-Natal, Durban, South Africa
| | - Ahmed A Elrashedy
- Molecular Bio-computational and Drug Design Research Group, School of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Monsurat M Lawal
- Drug Research and Innovation Unit, Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, University of KwaZulu-Natal, Durban, South Africa
| | - Hezekiel M Kumalo
- Drug Research and Innovation Unit, Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
13
|
Design, synthesis and evaluation of new 4-arylthiazole-2-amine derivatives as acetylcholinesterase inhibitors. Bioorg Med Chem Lett 2020; 30:126985. [PMID: 32008906 DOI: 10.1016/j.bmcl.2020.126985] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 01/14/2020] [Accepted: 01/21/2020] [Indexed: 11/23/2022]
Abstract
A series of new 4-arylthiazole-2-amine derivatives as acetylcholinesterase inhibitors (AChEIs) were designed and synthesized, Furthermore, their inhibitory activities against acetylcholinesterase in vitro were tested by Ellman spectrophotometry, and the results of inhibitory activity test showed that most of them had a certain acetylcholinesterase inhibitory activity in vitro. Moreover, the IC50 value of compound 4f was to 0.66 μM, which was higher than that of Rivastigmine and Huperzine-A as reference compounds, and it had a weak inhibitory effect on butyrylcholinesterase. The potential binding mode of compound 4f with AChE was investigated by the molecular docking, and the results showed that 4f was strongly bound up with AChE with the optimal conformation, in addition, their binding energy reached -11.27 Kcal*mol-1. At last, in silico molecular property of the synthesized compounds were predicted by using Molinspiration online servers. It can be concluded that the lead AChEIs compound 4f presented satisfactory drug-like characteristics.
Collapse
|
14
|
Zhu J, Wang LN, Cai R, Geng SQ, Dong YF, Liu YM. Design, synthesis, evaluation and molecular modeling study of 4-N-phenylaminoquinolines for Alzheimer disease treatment. Bioorg Med Chem Lett 2019; 29:1325-1329. [DOI: 10.1016/j.bmcl.2019.03.050] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2019] [Revised: 03/22/2019] [Accepted: 03/30/2019] [Indexed: 12/12/2022]
|
15
|
Haouas B, Sbei N, Ayari H, Benkhoud ML, Batanero B. Efficient synthetic procedure to new 2-imino-1,3-thiazolines and thiazolidin-4-ones promoted by acetonitrile electrogenerated base. NEW J CHEM 2018. [DOI: 10.1039/c8nj01992d] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Regioselective sulfur–carbon bond formation from disubstituted thioureas and 1,2-dielectrophiles.
Collapse
Affiliation(s)
- Beya Haouas
- Laboratoire de Chimie Analytique et d’Electrochimie. Faculté des Sciences
- Université Tunis El Manar
- 2092 El-Manar (Tunis)
- Tunisia
| | - Najwa Sbei
- Laboratoire de Chimie Analytique et d’Electrochimie. Faculté des Sciences
- Université Tunis El Manar
- 2092 El-Manar (Tunis)
- Tunisia
| | - Hana Ayari
- Laboratoire de Chimie Analytique et d’Electrochimie. Faculté des Sciences
- Université Tunis El Manar
- 2092 El-Manar (Tunis)
- Tunisia
| | - M. Lamine Benkhoud
- Laboratoire de Chimie Analytique et d’Electrochimie. Faculté des Sciences
- Université Tunis El Manar
- 2092 El-Manar (Tunis)
- Tunisia
| | - Belen Batanero
- Department of Organic Chemistry
- University of Alcalá
- 28871 Alcalá de Henares (Madrid)
- Spain
- Instituto de Investigación Química “Andrés M. del Río” (IQAR)
| |
Collapse
|
16
|
Zdarova Karasova J, Mzik M, Hroch M, Korabecny J, Nepovimova E, Vorisek V, Palicka V, Kuca K. The New Acetylcholinesterase Inhibitors PC-37 and PC-48 (7-Methoxytacrine-Donepezil-Like Compounds): Characterization of Their Metabolites in Human Liver Microsomes, Pharmacokinetics and In Vivo Formation of the Major Metabolites in Rats. Basic Clin Pharmacol Toxicol 2017; 122:373-382. [PMID: 29067789 DOI: 10.1111/bcpt.12922] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 10/16/2017] [Indexed: 12/22/2022]
Abstract
The objective of this study was to elucidate the pharmacokinetics and metabolite formation of newly developed non-selective AChE/BChE 7-MEOTA-donepezil-like inhibitors for potential therapeutic use in Alzheimer's disease (AD) patients. The chemical structures of metabolites were defined during incubation with human liver microsomes, and subsequently, the metabolization was verified in in vivo study. In vitro metabolic profiling revealed the formation of nine major metabolites in the case of PC-37 and eight metabolites of PC-48. Hydroxylation and the enzymatic hydrolysis of bonds close to the piperazine ring appeared to be the principal metabolic pathways in vitro. Of these metabolites, M1-M7 of PC-37 and M1-M6 of PC-48 were confirmed under in vivo conditions. Pilot pharmacokinetic experiments in rats were focused on the absorption, distribution and elimination of these compounds. Absorption after i.m. application was relatively fast; the bioavailability expressed as AUCtotal was 28179 ± 4691 min.ng/mL for PC-37 and 23374 ± 4045 min.ng/mL for PC-48. Both compounds showed ability to target the central nervous system, with brain concentrations exceeding those in plasma. The maximal brain concentrations are approximately two times higher than the plasma concentrations. The relatively high brain concentrations persisted throughout the experiment until 24 hr after application. Elimination via the kidneys (urine) significantly exceeded elimination via the liver (bile). All these characteristics are crucial for new candidates intended for AD treatment. The principle metabolic pathways that were verified in the in vivo study do not show any evidence for formation of extremely toxic metabolites, but this needs to be confirmed by further studies.
Collapse
Affiliation(s)
- Jana Zdarova Karasova
- Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, University of Defense, Hradec Kralove, Brno, Czech Republic.,Biomedical Research Center, University Hospital, Hradec Kralove, Czech Republic
| | - Martin Mzik
- Institute of Clinical Biochemistry and Diagnostic, University Hospital, Hradec Kralove, Czech Republic
| | - Milos Hroch
- Department of Medical Biochemistry, Faculty of Medicine in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Jan Korabecny
- Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, University of Defense, Hradec Kralove, Brno, Czech Republic.,Biomedical Research Center, University Hospital, Hradec Kralove, Czech Republic
| | - Eugenie Nepovimova
- Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, University of Defense, Hradec Kralove, Brno, Czech Republic
| | - Viktor Vorisek
- Institute of Clinical Biochemistry and Diagnostic, University Hospital, Hradec Kralove, Czech Republic
| | - Vladimir Palicka
- Institute of Clinical Biochemistry and Diagnostic, University Hospital, Hradec Kralove, Czech Republic
| | - Kamil Kuca
- Biomedical Research Center, University Hospital, Hradec Kralove, Czech Republic.,Department of Cellular Biology and Pharmacology, Florida International University, Miami, FL, USA
| |
Collapse
|
17
|
Osman H, Idris NH, Kamarulzaman EE, Wahab HA, Hassan MZ. 3,5-Bis(arylidene)-4-piperidones as potential dengue protease inhibitors. Acta Pharm Sin B 2017; 7:479-484. [PMID: 28752033 PMCID: PMC5518655 DOI: 10.1016/j.apsb.2017.04.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 02/28/2017] [Accepted: 03/15/2017] [Indexed: 11/25/2022] Open
Abstract
Dengue is a severe mosquito-borne viral infection causing half a million deaths annually. Dengue virus NS2B/NS3 protease is a validated target for anti-dengue drug design. A series of hitherto unreported 3,5-bis(arylidene)-4-piperidones analogues 4a-4j were synthesized and screened in silico against DENV2 NS2B/NS3 protease to elucidate their binding mechanism and orientation around the active sites. Results were validated through an in vitro DENV2 NS2B/NS3 protease assay using a fluorogenic Boc-Gly-Arg-Arg-AMC substrate. Nitro derivatives of 3,5-bis(arylidene)-4-piperidones (4e and 4j) emerged as promising lead molecules for novel protease inhibitors with an IC50 of 15.22 and 16.23 µmol/L, respectively, compared to the standard, panduratin A, having IC50 of 57.28 µmol/L.
Collapse
|