51
|
Heidarpoor Saremi L, Ebrahimi A, Lagzian M. Identification of new potential cyclooxygenase-2 inhibitors: insight from high throughput virtual screening of 18 million compounds combined with molecular dynamic simulation and quantum mechanics. J Biomol Struct Dyn 2020; 39:1717-1734. [PMID: 32122267 DOI: 10.1080/07391102.2020.1737574] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The cyclooxygenase isoenzymes (COX-1 and COX-2) have a critical role in inflammation, fever, and pain. In contrary to COX-1, COX-2 is specifically expressed in inflamed tissues. Because of the subtle difference between both enzyme active sites, targeting COX-2 represents an efficient strategy for the development of novel inhibitors against inflammation with fewer side effects. In order to identify potential inhibitors of COX-2, more than 18,000,000 small molecules were retrieved from the ZINC database and virtually screened against it with a gradual increase in the precision through combined multistep docking. The results were sorted according to the rank-by-rank, induced-fit docking, and MM-GBSA evaluation. Subsequently from the final hit list, two top hits along with an approved selective inhibitor (celecoxib) were further investigated by the molecular dynamics (MD) simulations. The results were indicated that ZINC16934653 and ZINC40484701 demonstrate the highest affinity for the COX-2 binding pocket. Both ligands were bound to the important active-site residues, which are necessary for the correct orientation of inhibitors inside the binding cavity. Their binding free energies were comparable to celecoxib. 100 ns MD simulation is revealed that ZINC40484701 is more preferred in comparison with ZINC16934653 and celecoxib. In addition, non-covalent interactions between the compounds and key residues located in 6 Å distance from the COX-2 binding site show similar patterns of bonding by the reduced density gradient and the independent gradient model. Therefore, ZINC40484701 can be a potential candidate for further in vitro and in vivo analysis after lead-optimization efforts.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Leily Heidarpoor Saremi
- Department of Chemistry, Computational Quantum Chemistry Laboratory, University of Sistan and Baluchestan, Zahedan, Iran
| | - Ali Ebrahimi
- Department of Chemistry, Computational Quantum Chemistry Laboratory, University of Sistan and Baluchestan, Zahedan, Iran
| | - Milad Lagzian
- Department of Biology, Faculty of Sciences, University of Sistan and Baluchestan, Zahedan, Iran
| |
Collapse
|
52
|
Fahmy NM, Al-Sayed E, Moghannem S, Azam F, El-Shazly M, Singab AN. Breaking Down the Barriers to a Natural Antiviral Agent: Antiviral Activity and Molecular Docking of Erythrina speciosa Extract, Fractions, and the Major Compound. Chem Biodivers 2020; 17:e1900511. [PMID: 31800173 DOI: 10.1002/cbdv.201900511] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 11/27/2019] [Indexed: 12/18/2022]
Abstract
The in vitro cytotoxic activity in Vero cells and the antiviral activity of Erythrina speciosa methanol extract, fractions, and isolated vitexin were studied. The results revealed that E. speciosa leaves ethyl acetate soluble fraction of the methanol extract (ESLE) was the most active against herpes simplex virus type 1 (HSV-1). Bioactivity-guided fractionation was performed on ESLE to isolate the bioactive compounds responsible for this activity. One sub-fraction from ESLE (ESLE IV) showed the highest activity against HSV-1 and Hepatitis A HAV-H10 viruses. Vitexin isolated from ESLE VI exhibited a significant antiviral activity (EC50 =35±2.7 and 18±3.3 μg/mL against HAV-H10 and HSV-1 virus, respectively), which was notably greater than the activity of the extract and the fractions. Molecular docking studies were carried out to explore the molecular interactions of vitexin with different macromolecular targets. Analysis of the in silico data together with the in vitro studies validated the antiviral activity associated with vitexin. These outcomes indicated that vitexin is a potential candidate to be utilized commendably in lead optimization for the development of antiviral agents.
Collapse
Affiliation(s)
- Nouran M Fahmy
- Department of Pharmacognosy, Faculty of Pharmacy, Ain-Shams University, Abassia, 11566, Cairo, Egypt
| | - Eman Al-Sayed
- Department of Pharmacognosy, Faculty of Pharmacy, Ain-Shams University, Abassia, 11566, Cairo, Egypt
| | - Saad Moghannem
- Department of Botany and Microbiology, Faculty of Science, Al-Azhar University, 11884, Cairo, Egypt
| | - Faizul Azam
- Department of Pharmaceutical Chemistry and Pharmacognosy, Unaizah College of Pharmacy, Qassim University, 51911, Unaizah, Saudi Arabia
| | - Mohamed El-Shazly
- Department of Pharmacognosy, Faculty of Pharmacy, Ain-Shams University, Abassia, 11566, Cairo, Egypt.,Department of Pharmaceutical Biology, Faculty of Pharmacy and Biotechnology, German University in Cairo, 11835, Cairo, Egypt
| | - Abdel Nasser Singab
- Department of Pharmacognosy, Faculty of Pharmacy, Ain-Shams University, Abassia, 11566, Cairo, Egypt.,Center for Drug Discovery and Development Research, Faculty of Pharmacy, Ain Shams University, 11566, Cairo, Egypt
| |
Collapse
|
53
|
Liu Y, Chu JMT, Yan T, Zhang Y, Chen Y, Chang RCC, Wong GTC. Short-term resistance exercise inhibits neuroinflammation and attenuates neuropathological changes in 3xTg Alzheimer's disease mice. J Neuroinflammation 2020; 17:4. [PMID: 31900170 PMCID: PMC6942350 DOI: 10.1186/s12974-019-1653-7] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 11/20/2019] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Both human and animal studies have shown beneficial effects of physical exercise on brain health but most tend to be based on aerobic rather than resistance type regimes. Resistance exercise has the advantage of improving both muscular and cardiovascular function, both of which can benefit the frail and the elderly. However, the neuroprotective effects of resistance training in cognitive impairment are not well characterized. METHODS We evaluated whether short-term resistant training could improve cognitive function and pathological changes in mice with pre-existing cognitive impairment. Nine-month-old 3xTg mouse underwent a resistance training protocol of climbing up a 1-m ladder with a progressively heavier weight loading. RESULTS Compared with sedentary counterparts, resistance training improved cognitive performance and reduced neuropathological and neuroinflammatory changes in the frontal cortex and hippocampus of mice. In line with these results, inhibition of pro-inflammatory intracellular pathways was also demonstrated. CONCLUSIONS Short-term resistance training improved cognitive function in 3xTg mice, and conferred beneficial effects on neuroinflammation, amyloid and tau pathology, as well as synaptic plasticity. Resistance training may represent an alternative exercise strategy for delaying disease progression in Alzheimer's disease.
Collapse
Affiliation(s)
- Yan Liu
- Department of Anaesthesiology, LKS Faculty of Medicine, The University of Hong Kong, Room K424, Queen Mary Hospital, Pokfulam, Hong Kong, SAR, China.,Laboratory of Neurodegenerative Diseases, LKS Faculty of MedicineSchool of Biomedical Sciences, The University of Hong Kong, Hong Kong, SAR, China
| | - John Man Tak Chu
- Department of Anaesthesiology, LKS Faculty of Medicine, The University of Hong Kong, Room K424, Queen Mary Hospital, Pokfulam, Hong Kong, SAR, China.,Laboratory of Neurodegenerative Diseases, LKS Faculty of MedicineSchool of Biomedical Sciences, The University of Hong Kong, Hong Kong, SAR, China
| | - Tim Yan
- Laboratory of Neurodegenerative Diseases, LKS Faculty of MedicineSchool of Biomedical Sciences, The University of Hong Kong, Hong Kong, SAR, China
| | - Yan Zhang
- Department of Anaesthesiology, LKS Faculty of Medicine, The University of Hong Kong, Room K424, Queen Mary Hospital, Pokfulam, Hong Kong, SAR, China.,Laboratory of Neurodegenerative Diseases, LKS Faculty of MedicineSchool of Biomedical Sciences, The University of Hong Kong, Hong Kong, SAR, China
| | - Ying Chen
- Department of Anaesthesiology, LKS Faculty of Medicine, The University of Hong Kong, Room K424, Queen Mary Hospital, Pokfulam, Hong Kong, SAR, China.,Laboratory of Neurodegenerative Diseases, LKS Faculty of MedicineSchool of Biomedical Sciences, The University of Hong Kong, Hong Kong, SAR, China
| | - Raymond Chuen Chung Chang
- Laboratory of Neurodegenerative Diseases, LKS Faculty of MedicineSchool of Biomedical Sciences, The University of Hong Kong, Hong Kong, SAR, China. .,State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, L4-49, Laboratory Block, Pokfulam, Hong Kong, SAR, China.
| | - Gordon Tin Chun Wong
- Department of Anaesthesiology, LKS Faculty of Medicine, The University of Hong Kong, Room K424, Queen Mary Hospital, Pokfulam, Hong Kong, SAR, China.
| |
Collapse
|
54
|
Gupta S, Dasmahapatra AK. Destabilization potential of phenolics on Aβ fibrils: mechanistic insights from molecular dynamics simulation. Phys Chem Chem Phys 2020; 22:19643-19658. [DOI: 10.1039/d0cp02459g] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Ellagic acid from pomegranate and walnuts is found to destabilize Aβ fibrils. It can be a potential drug to treat AD.
Collapse
Affiliation(s)
- Shivani Gupta
- Department of Chemical Engineering
- Indian Institute of Technology Guwahati
- Guwahati – 781039
- India
| | - Ashok Kumar Dasmahapatra
- Department of Chemical Engineering
- Indian Institute of Technology Guwahati
- Guwahati – 781039
- India
- Center for Nanotechnology
| |
Collapse
|
55
|
Azam F, Abodabos HS, Taban IM, Rfieda AR, Mahmood D, Anwar MJ, Khan S, Sizochenko N, Poli G, Tuccinardi T, Ali HI. Rutin as promising drug for the treatment of Parkinson’s disease: an assessment of MAO-B inhibitory potential by docking, molecular dynamics and DFT studies. MOLECULAR SIMULATION 2019. [DOI: 10.1080/08927022.2019.1662003] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Faizul Azam
- Department of Pharmaceutical Chemistry & Pharmacognosy, Unaizah College of Pharmacy, Qassim University, Unaizah, Saudi Arabia
| | - Honiwa Suliman Abodabos
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Misurata University, Misurata, Libya
| | - Ismail M. Taban
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Misurata University, Misurata, Libya
- School of Biosciences, Cardiff University, Cardiff, UK
| | - Abdalla R. Rfieda
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Misurata University, Misurata, Libya
| | - Danish Mahmood
- Department of Pharmacology & Toxicology, Unaizah College of Pharmacy, Qassim University, Unaizah, Saudi Arabia
| | - Md Jamir Anwar
- Department of Pharmacology & Toxicology, Unaizah College of Pharmacy, Qassim University, Unaizah, Saudi Arabia
| | - Shamshir Khan
- Department of Pharmacognosy & Medicinal Chemistry, Buraidah College of Dentistry & Pharmacy, Al-Qassim, Saudi Arabia
| | - Natalia Sizochenko
- Department of Chemistry, Physics and Atmospheric Sciences, Jackson State University, Jackson, MS, USA
- Department of Computer Science, Dartmouth College, Hanover, NH, USA
| | - Giulio Poli
- Department of Pharmacy, University of Pisa, Pisa, Italy
| | | | - Hamed I. Ali
- Rangel College of Pharmacy, Health Science Center, Texas A&M University, Kingsville, TX, USA
| |
Collapse
|
56
|
Aricov L, Angelescu DG, Băran A, Leontieş AR, Popa VT, Precupaş A, Sandu R, Stîngă G, Anghel DF. Interaction of piroxicam with bovine serum albumin investigated by spectroscopic, calorimetric and computational molecular methods. J Biomol Struct Dyn 2019; 38:2659-2671. [PMID: 31315508 DOI: 10.1080/07391102.2019.1645733] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The binding of drugs to serum proteins is governed by weak non-covalent forces. In this study, the nature and magnitude of the interactions between piroxicam (PRX) and bovine serum albumin (BSA) was assessed using spectroscopic, calorimetric and computational molecular methods. The fluorescence data revealed an atypical behavior during PRX and BSA interaction. The quenching process of tryptophan (Trp) by PRX is a dual one (approximately equal static and dynamic quenched components). The FRET results indicate that a non-radiative transfer of energy occurred. The association constant and the number of binding sites indicate moderate PRX and BSA binding. The competitive binding study indicates that PRX is bound to site I from the hydrophobic pocket of subdomain IIA of BSA. The synchronous spectra showed that the microenvironment around the BSA fluorophores and protein conformation do not change considerably. The Trp lifetimes revealed that PRX mainly quenches the fluorescence of Trp-213 situated in the hydrophobic domain. The CD and DSC investigation show that addition of PRX stabilizes the protein structure. ITC results revealed that BSA-PRX binding involves a combination of electrostatic, hydrophobic and hydrogen interactions. The analysis of the computational data is consistent with the experimental results. This thorough investigation of the PRX-BSA binding may provide support for other studies concerning moderate affinity drugs with serum protein.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Ludmila Aricov
- "Ilie Murgulescu" Institute of Physical Chemistry of the Romanian Academy, Bucharest, Romania
| | - Daniel George Angelescu
- "Ilie Murgulescu" Institute of Physical Chemistry of the Romanian Academy, Bucharest, Romania
| | - Adriana Băran
- "Ilie Murgulescu" Institute of Physical Chemistry of the Romanian Academy, Bucharest, Romania
| | - Anca Ruxandra Leontieş
- "Ilie Murgulescu" Institute of Physical Chemistry of the Romanian Academy, Bucharest, Romania
| | - Vlad Tudor Popa
- "Ilie Murgulescu" Institute of Physical Chemistry of the Romanian Academy, Bucharest, Romania
| | - Aurica Precupaş
- "Ilie Murgulescu" Institute of Physical Chemistry of the Romanian Academy, Bucharest, Romania
| | - Romică Sandu
- "Ilie Murgulescu" Institute of Physical Chemistry of the Romanian Academy, Bucharest, Romania
| | - Gabriela Stîngă
- "Ilie Murgulescu" Institute of Physical Chemistry of the Romanian Academy, Bucharest, Romania
| | - Dan-Florin Anghel
- "Ilie Murgulescu" Institute of Physical Chemistry of the Romanian Academy, Bucharest, Romania
| |
Collapse
|
57
|
E M Eid E, S Alanazi A, Koosha S, A Alrasheedy A, Azam F, M Taban I, Khalilullah H, Sadiq Al-Qubaisi M, A Alshawsh M. Zerumbone Induces Apoptosis in Breast Cancer Cells by Targeting αvβ3 Integrin upon Co-Administration with TP5-iRGD Peptide. Molecules 2019; 24:molecules24142554. [PMID: 31337024 PMCID: PMC6680663 DOI: 10.3390/molecules24142554] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 07/05/2019] [Accepted: 07/06/2019] [Indexed: 12/24/2022] Open
Abstract
Cell-penetrating peptides (CPPs) are highly promising tools to deliver therapeutic molecules into tumours. αVβ3 integrins are cell-matrix adhesion receptors, and are considered as an attractive target for anticancer therapies owing to their roles in the process of metastasis and angiogenesis. Therefore, this study aims to assess the effect of co-administration of zerumbone (ZER) and ZERencapsulated in hydroxypropyl-β-cyclodextrin with TP5-iRGD peptide towards cell cytotoxicity, apoptosis induction, and proliferation of normal and cancerous breast cells utilizing in vitro assays, as well as to study the molecular docking of ZER in complex with TP5-iRGD peptide. Cell viability assay findings indicated that ZER and ZERencapsulated in hydroxypropyl-β-cyclodextrin (ZER-HPβCD) inhibited the growth of estrogen receptor positivebreast cancer cells (ER+ MCF-7) at 72 h treatment with an inhibitory concentration (IC)50 of 7.51 ± 0.2 and 5.08 ± 0.2 µg/mL, respectively, and inhibited the growth of triple negative breast cancer cells (MDA-MB-231) with an IC50 of 14.96 ± 1.52 µg/mL and 12.18 ± 0.7 µg/mL, respectively. On the other hand, TP5-iRGD peptide showed no significant cytotoxicity on both cancer and normal cells. Interestingly, co-administration of TP5-iRGD peptide in MCF-7 cells reduced the IC50 of ZER from 7.51 ± 0.2 µg/mL to 3.13 ± 0.7 µg/mL and reduced the IC50 of ZER-HPβCD from 5.08 ± 0.2 µg/mL to 0.49 ± 0.004 µg/mL, indicating that the co-administration enhances the potency and increases the efficacy of ZER and ZER-HPβCD compounds. Acridine orange (AO)/propidium iodide (PI) staining under fluorescence microscopy showed evidence of early apoptosis after 72 h from the co-administration of ZER or ZER-HPβCD with TP5-iRGD peptide in MCF-7 breast cancer cells. The findings of the computational modelling experiment provide novel insights into the ZER interaction with integrin αvβ3 in the presence of TP5-iRGD, and this could explain why ZER has better antitumor activities when co-administered with TP5-iRGD peptide.
Collapse
Affiliation(s)
- Eltayeb E M Eid
- Department of Pharmaceutical Chemistry and Pharmacognosy, Unaizah College of Pharmacy, Qassim University, 51911 Unaizah, Saudi Arabia.
| | | | - Sanaz Koosha
- Department of Pharmacology, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Alian A Alrasheedy
- Department of Pharmaceutical Chemistry and Pharmacognosy, Unaizah College of Pharmacy, Qassim University, 51911 Unaizah, Saudi Arabia
| | - Faizul Azam
- Department of Pharmaceutical Chemistry and Pharmacognosy, Unaizah College of Pharmacy, Qassim University, 51911 Unaizah, Saudi Arabia
| | - Ismail M Taban
- School of Biosciences, Cardiff University, Cardiff CF10 3AT, UK
| | - Habibullah Khalilullah
- Department of Pharmaceutical Chemistry and Pharmacognosy, Unaizah College of Pharmacy, Qassim University, 51911 Unaizah, Saudi Arabia
| | | | - Mohammed A Alshawsh
- Department of Pharmacology, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia.
| |
Collapse
|
58
|
Uzzaman M, Uddin MN. Optimization of structures, biochemical properties of ketorolac and its degradation products based on computational studies. Daru 2019; 27:71-82. [PMID: 30784007 PMCID: PMC6593035 DOI: 10.1007/s40199-019-00243-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 01/10/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Ketorolac (KTR) is used as an analgesic drug with an efficacy close to that of the opioid family. It is mainly used for the short term treatment of post-operative pain. It can inhibit the prostaglandin synthesis by blocking cyclooxygenase (COX). METHODS In this investigation, the inherent stability and biochemical interaction of Ketorolac (KTR) and its degradation products have been studiedon the basis of quantum mechanical approaches. Density functional theory (DFT) with B3LYP/ 6-31G (d) has been employed to optimize the structures. Thermodynamic properties, frontier molecular orbital features, dipole moment, electrostatic potential, equilibrium geometry, vibrational frequencies and atomic partial charges of these optimized structureswere investigated. Molecular docking has been performed against prostaglandin H2 (PGH2) synthase protein 5F19 to search the binding affinity and mode(s). ADMET prediction has performed to evaluate the absorption, metabolism and carcinogenic properties. RESULTS The equilibrium geometry calculations support the optimized structures. Thermodynamic results disclosed the thermal stability of all structures. From molecular orbital data, all the degradents are chemically more reactive than parent drug (except K3). However, the substitution of carboxymethyl radicalin K4 improved the physicochemical properties and binding affinity. ADMET calculations predict the improved pharmacokinetic and non-carcinogenic properties of all degradents. CONCLUSION Based on physicochemical, molecular docking, and ADMET calculation, this study can be helpful to understand the biochemical activities of Ketorolac and its degradents and to design a potent analgesic drug.
Collapse
Affiliation(s)
- Monir Uzzaman
- Department of Chemistry, University of Chittagong, Chittagong, 4331, Bangladesh
- Department of Applied Chemistry and Biochemical Engineering, Shizuoka University, 3-5-1, Johoku, Hamamatsu, 432-8011, Japan
| | - Mohammad Nasir Uddin
- Department of Chemistry, University of Chittagong, Chittagong, 4331, Bangladesh.
| |
Collapse
|
59
|
Ali MM, Ghouri RG, Ans AH, Akbar A, Toheed A. Recommendations for Anti-inflammatory Treatments in Alzheimer's Disease: A Comprehensive Review of the Literature. Cureus 2019; 11:e4620. [PMID: 31312547 PMCID: PMC6615583 DOI: 10.7759/cureus.4620] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common cause of dementia in elderly patients, affecting individuals older than 60 years. It is a complex degenerative brain disease characterized by progressive cognitive impairment. AD constitutes a major global health concern. A central role for inflammation has been implicated in the pathogenesis of AD. Despite the understanding of multiple molecular pathways in the pathophysiology of AD, novel treatment agents with a possible role in modifying the disease activity are still lacking. Our article provides a comprehensive review of various observational studies and randomized trials encompassing the use of anti-inflammatory agents in the management of AD patients and utilizes the conclusions derived therefrom to give recommendations in this regard.
Collapse
Affiliation(s)
- Muhammad Mohsin Ali
- Internal Medicine, Mayo Hospital, King Edward Medical University, Lahore, PAK
| | - Raza G Ghouri
- Internal Medicine, Mayo Hospital, King Edward Medical University, Lahore, PAK
| | - Armghan H Ans
- Cardiology, University of Pennsylvania, Philadelphia, USA
| | - Arshia Akbar
- Internal Medicine, Rawalpindi Medical College, Rawalpindi, PAK
| | - Ahmed Toheed
- Internal Medicine, Mayo Hospital, King Edward Medical University, Lahore, PAK
| |
Collapse
|
60
|
Jethava DJ, Acharya PT, Vasava MS, Bhoi MN, Bhavsar ZA, Rathwa SK, Rajani DP, Patel HD. Design, synthesis, biological evaluation and computational study of novel triazolo [4,3-a]pyrazin analogues. J Mol Struct 2019. [DOI: 10.1016/j.molstruc.2019.01.091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
61
|
Saini RK, Shuaib S, Goyal D, Goyal B. Insights into the inhibitory mechanism of a resveratrol and clioquinol hybrid against Aβ42 aggregation and protofibril destabilization: A molecular dynamics simulation study. J Biomol Struct Dyn 2018; 37:3183-3197. [DOI: 10.1080/07391102.2018.1511475] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
- Rajneet Kaur Saini
- Department of Chemistry, Faculty of Basic and Applied Sciences, Sri Guru Granth Sahib World University, Fatehgarh Sahib, India
| | - Suniba Shuaib
- Department of Chemistry, Faculty of Basic and Applied Sciences, Sri Guru Granth Sahib World University, Fatehgarh Sahib, India
| | - Deepti Goyal
- Department of Chemistry, Faculty of Basic and Applied Sciences, Sri Guru Granth Sahib World University, Fatehgarh Sahib, India
| | - Bhupesh Goyal
- School of Chemistry and Biochemistry, Thapar Institute of Engineering and Technology, Patiala, India
| |
Collapse
|
62
|
Eid EEM, Azam F, Hassan M, Taban IM, Halim MA. Zerumbone binding to estrogen receptors: an in-silico investigation. J Recept Signal Transduct Res 2018; 38:342-351. [PMID: 30396310 DOI: 10.1080/10799893.2018.1531886] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Breast cancer is the most frequent malignancy among females worldwide. Estrogen receptor (ER) mediate important pathophysiological signaling pathways induced by estrogens, and is regarded as a promising target for the treatment of breast cancer. Zerumbone (2,6,9,9-tetramethylcycloundeca-2,6,10-trien-1-one; ZER), a chemical constituent present in the Zingiber zerumbet is known to exhibit anti-breast cancer activity by modulating several proteins to induce apoptosis. Medicinal chemists usually exploit lead compounds of natural origin to develop molecules with improved pharmacological properties. Current study is intended to utilize molecular modeling techniques to investigate the interaction of ZER with estrogen receptors. AutoDock was used to predict the binding modes of ZER and target receptors. Stability of the ZER-ER complex was verified by molecular dynamics simulation using Desmond software. Docked ZER was further optimized by density functional theory (DFT) using Gaussian09 program. Analysis of docked conformations in terms of binding energy disclosed estrogen receptor-β (ERβ) as more promising than estrogen receptor-α (ERα). Evaluation of MD trajectories of ZER bound to both ERα and ERβ showed appreciable stability with minimum Cα-atom root mean square deviation shifts. DFT based global reactivity descriptors such as electron affinity, hardness, chemical potential, electronegativity and electrophilicity index, calculated from the energies of highest occupied and lowest unoccupied molecular orbitals underscored the electronic features governing viability of the ZER for interaction with the target receptors. In conclusion, these findings can be exploited to design and develop novel anticancer agents based on the lead compound, ZER.
Collapse
Affiliation(s)
- Eltayeb E M Eid
- a Unaizah College of Pharmacy, Qassim University , Unaizah , Saudi Arabia
| | - Faizul Azam
- a Unaizah College of Pharmacy, Qassim University , Unaizah , Saudi Arabia
| | - Mahmoud Hassan
- b Swiss Tropical & Public Health Institute, University of Basel , Switzerland
| | - Ismail M Taban
- c School of Biosciences, Cardiff University , Cardiff , United Kingdom
| | - Mohammad A Halim
- d Division of Computer-aided Drug Design , The Red-Green Research Center , BICCB , Dhaka , Bangladesh.,e Institut Lumière Matière, Université Lyon 1-CNRS, Université de Lyon , Villeurbanne Cedex , France
| |
Collapse
|
63
|
Anti-Inflammatory, Anti-Diabetic, and Anti-Alzheimer's Effects of Prenylated Flavonoids from Okinawa Propolis: An Investigation by Experimental and Computational Studies. Molecules 2018; 23:molecules23102479. [PMID: 30262742 PMCID: PMC6222853 DOI: 10.3390/molecules23102479] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 09/24/2018] [Accepted: 09/26/2018] [Indexed: 12/15/2022] Open
Abstract
Okinawa propolis (OP) and its major ingredients were reported to have anti-cancer effects and lifespan-extending effects on Caenorhabditis elegans through inactivation of the oncogenic kinase, p21-activated kinase 1 (PAK1). Herein, five prenylated flavonoids from OP, nymphaeol-A (NA), nymphaeol-B (NB), nymphaeol-C (NC), isonymphaeol-B (INB), and 3'-geranyl-naringenin (GN), were evaluated for their anti-inflammatory, anti-diabetic, and anti-Alzheimer's effects using in vitro techniques. They showed significant anti-inflammatory effects through inhibition of albumin denaturation (half maximal inhibitory concentration (IC50) values of 0.26⁻1.02 µM), nitrite accumulation (IC50 values of 2.4⁻7.0 µM), and cyclooxygenase-2 (COX-2) activity (IC50 values of 11.74⁻24.03 µM). They also strongly suppressed in vitro α-glucosidase enzyme activity with IC50 values of 3.77⁻5.66 µM. However, only INB and NA inhibited acetylcholinesterase significantly compared to the standard drug donepezil, with IC50 values of 7.23 and 7.77 µM, respectively. Molecular docking results indicated that OP compounds have good binding affinity to the α-glucosidase and acetylcholinesterase proteins, making non-bonded interactions with their active residues and surrounding allosteric residues. In addition, none of the compounds violated Lipinski's rule of five and showed notable toxicity parameters. Density functional theory (DFT)-based global reactivity descriptors demonstrated their high reactive nature along with the kinetic stability. In conclusion, this combined study suggests that OP components might be beneficial in the treatment of inflammation, type 2 diabetes mellitus, and Alzheimer's disease.
Collapse
|
64
|
Shakour N, Bianconi V, Pirro M, Barreto GE, Hadizadeh F, Sahebkar A. In silico evidence of direct interaction between statins and β‐amyloid. J Cell Biochem 2018; 120:4710-4715. [DOI: 10.1002/jcb.27761] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 09/06/2018] [Indexed: 02/03/2023]
Affiliation(s)
- Neda Shakour
- Department of Medicinal Chemistry School of Pharmacy, Mashhad University of Medical Sciences Mashhad Iran
| | - Vanessa Bianconi
- Unit of Internal Medicine, Department of Medicine University of Perugia Perugia Italy
| | - Matteo Pirro
- Unit of Internal Medicine, Department of Medicine University of Perugia Perugia Italy
| | - George E. Barreto
- Departamento de Nutrición y Bioquímica Facultad de Ciencias, Pontificia Universidad Javeriana Bogotá Colombia
- Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile Santiago Chile
| | - Farzin Hadizadeh
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences Mashhad Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences Mashhad Iran
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences Mashhad Iran
- School of Pharmacy, Mashhad University of Medical Sciences Mashhad Iran
| |
Collapse
|
65
|
Recent Advances by In Silico and In Vitro Studies of Amyloid-β 1-42 Fibril Depicted a S-Shape Conformation. Int J Mol Sci 2018; 19:ijms19082415. [PMID: 30115846 PMCID: PMC6121414 DOI: 10.3390/ijms19082415] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Revised: 08/09/2018] [Accepted: 08/10/2018] [Indexed: 12/19/2022] Open
Abstract
The amyloid-β 1-42 (Aβ1-42) peptide is produced by proteolytic cleavage of the amyloid precursor protein (APP) by sequential reactions that are catalyzed by γ and β secretases. Aβ1-42, together with the Tau protein are two principal hallmarks of Alzheimer's disease (AD) that are related to disease genesis and progression. Aβ1-42 possesses a higher aggregation propensity, and it is able to form fibrils via nucleated fibril formation. To date, there are compounds available that prevent Aβ1-42 aggregation, but none have been successful in clinical trials, possibly because the Aβ1-42 structure and aggregation mechanisms are not thoroughly understood. New molecules have been designed, employing knowledge of the Aβ1-42 structure and are based on preventing or breaking the ionic interactions that have been proposed for formation of the Aβ1-42 fibril U-shaped structure. Recently, a new Aβ1-42 fibril S-shaped structure was reported that, together with its aggregation and catalytic properties, could be helpful in the design of new inhibitor molecules. Therefore, in silico and in vitro methods have been employed to analyze the Aβ1-42 fibril S-shaped structure and its aggregation to obtain more accurate Aβ1-42 oligomerization data for the design and evaluation of new molecules that can prevent the fibrillation process.
Collapse
|
66
|
Baruah P, Basumatary G, Yesylevskyy SO, Aguan K, Bez G, Mitra S. Novel coumarin derivatives as potent acetylcholinesterase inhibitors: insight into efficacy, mode and site of inhibition. J Biomol Struct Dyn 2018; 37:1750-1765. [DOI: 10.1080/07391102.2018.1465853] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Prayasee Baruah
- Centre for Advanced Studies in Chemistry and Department of Biotechnology & Bioinformatics, North-Eastern Hill University , Shillong, India
| | - Grace Basumatary
- Centre for Advanced Studies in Chemistry and Department of Biotechnology & Bioinformatics, North-Eastern Hill University , Shillong, India
| | - Semen O. Yesylevskyy
- Department of Physics of Biological Systems, Institute of Physics of the National Academy of Sciences of Ukraine , Kyiv, Ukraine
| | - Kripamoy Aguan
- Department of Physics of Biological Systems, Institute of Physics of the National Academy of Sciences of Ukraine , Kyiv, Ukraine
| | - Ghanashyam Bez
- Centre for Advanced Studies in Chemistry and Department of Biotechnology & Bioinformatics, North-Eastern Hill University , Shillong, India
| | - Sivaprasad Mitra
- Centre for Advanced Studies in Chemistry and Department of Biotechnology & Bioinformatics, North-Eastern Hill University , Shillong, India
| |
Collapse
|
67
|
Pascoini AL, Federico LB, Arêas ALF, Verde BA, Freitas PG, Camps I. In silico development of new acetylcholinesterase inhibitors. J Biomol Struct Dyn 2018; 37:1007-1021. [DOI: 10.1080/07391102.2018.1447513] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- A. L. Pascoini
- Laboratory of Computational Modeling-LaModel, Institute of Exact Sciences, Federal University of Alfenas, Alfenas, Brazil
| | - L. B. Federico
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | - A. L. F. Arêas
- Laboratory of Computational Modeling-LaModel, Institute of Exact Sciences, Federal University of Alfenas, Alfenas, Brazil
| | - B. A. Verde
- Laboratory of Computational Modeling-LaModel, Institute of Exact Sciences, Federal University of Alfenas, Alfenas, Brazil
| | - P. G. Freitas
- Laboratory of Molecular Modeling and Computer Simulations-MolMod-CS, Institute of Exact Sciences, Federal University of Alfenas, Alfenas, Brazil
| | - I. Camps
- Laboratory of Computational Modeling-LaModel, Institute of Exact Sciences, Federal University of Alfenas, Alfenas, Brazil
| |
Collapse
|