1
|
Thaikkad A, Henna F, Thomas SD, John L, Raju R, Jayanandan A. Cangrelor and AVN-944 as repurposable candidate drugs for hMPV: analysis entailed by AI-driven in silico approach. Mol Divers 2025:10.1007/s11030-025-11206-6. [PMID: 40316857 DOI: 10.1007/s11030-025-11206-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2025] [Accepted: 04/22/2025] [Indexed: 05/04/2025]
Abstract
Human metapneumovirus (hMPV) primarily causes respiratory tract infections in young children and older adults. According to the 2024 Human Pneumonia Etiology Research for Child Health (PERCH) study, hMPV is the second leading common cause of pneumonia in children under five in Asia and Africa. The virus encodes nine proteins, including the essential Fusion (F) and G glycoproteins, which facilitate entry to the host cells. Currently, there are no approved vaccines or antiviral treatments for hMPV; supportive care is the primary way it is managed. Hence, this study focuses on the F protein as a therapeutic target to find a repurposable drug to fight hMPV. Refolding of the F protein and its binding to heparan sulfate enable hMPV infection. Heparin sulfate is important for hMPV binding, and we have found that cangrelor and AVN 944 can prevent the fusion of membranes. We developed a deep learning-based pharmacophore to identify potential drugs targeting hMPV, from which we could narrowed a list of 2400 FDA-approved drugs and 255 antiviral drugs to 792 and 72 drugs, respectively. We then conducted quantitative validation using the ROC curve. Further virtual screening of the drugs was performed, leading us to select the one with the highest docking score. The validation of the deep learning prediction in virtual screening Pearson correlation was done. Further, the MD simulation of these drugs confirmed that the protein-drug complex stability remained in dynamic condition. Further, the stability of protein-drug complexes than unbound protein was confirmed by Free Energy Landscape and Dynamic Cross Correlation Matrices. Further in vitro and in vivo experiments need to determine the efficacy of the identified candidates.
Collapse
Affiliation(s)
- Amritha Thaikkad
- Centre for Integrative Omics Data Science, Yenepoya (Deemed to Be University), Mangalore, Karnataka, 575018, India
| | - Fathimath Henna
- Centre for Integrative Omics Data Science, Yenepoya (Deemed to Be University), Mangalore, Karnataka, 575018, India
| | - Sonet Daniel Thomas
- Centre for Integrative Omics Data Science, Yenepoya (Deemed to Be University), Mangalore, Karnataka, 575018, India
| | - Levin John
- Institute of Regeneration and Repair (IRR), The University of Edinburgh, 4-5 Little France Dr, Edinburgh EH16 4UU, United Kingdom, Edinburgh, Scotland
| | - Rajesh Raju
- Centre for Integrative Omics Data Science, Yenepoya (Deemed to Be University), Mangalore, Karnataka, 575018, India.
| | - Abhithaj Jayanandan
- Centre for Integrative Omics Data Science, Yenepoya (Deemed to Be University), Mangalore, Karnataka, 575018, India.
| |
Collapse
|
2
|
Savita MK, Dwivedi V, Srivastava P. Deployment of in-silico analysis to reveal the antibacterial profiles of Allium sativum against Aeromonas hydrophila. J Biomol Struct Dyn 2025; 43:2320-2334. [PMID: 38116953 DOI: 10.1080/07391102.2023.2294832] [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: 12/30/2022] [Accepted: 10/02/2023] [Indexed: 12/21/2023]
Abstract
The key challenges in aquaculture are the emergence of antimicrobial resistance in fish cultivation due to the frequent use of antibiotics. Over the past three decades, this led to a major threat in the persistence of multidrug-resistant bacteria. Aeromonas hydrophila is a Gram-negative bacterium, a common causative agent of motile bacterial septicemia in fisheries. Combining these two key factors of the presented narrative, the essential type II topoisomerase enzyme 'DNA gyrase' (encoded by the gyrA and gyrB genes) as a potential drug target in Aeromonas hydrophila was taken, retrieve its sequence from UniProtKB (Id-A0KKQ2), constructs the 3-D structure using SWISS-MODEL (in absence of the experimental structure), and performs an in-silico screening of selected drug-like compounds (25 antibacterial phytochemicals) most of which are bioactive compounds of A. sativum through molecular docking. Quercetin a derivative of A. sativum was observed as a more potent drug molecule than other studied molecules based on ligand binding energy as docking score -7.812, showed highly encouraging results, supported by a study using structural dynamics of the receptor-ligand complex for a duration of 100 ns by Molecular Dynamic Simulations and confirm binding stability with MM-GBSA calculations. This study also provides theoretical grounds for drug discovery against other pathogenic bacteria posing threats to the ecosystem. Switching to herbal products is the best way to combat the plurality of problems to avoid seen or unseen post-treatment side effects.
Collapse
Affiliation(s)
| | - Vinay Dwivedi
- Department of Biotechnology, Naraina Vidyapeeth Engineering and Management Institute, Kanpur, Uttar Pradesh, India
| | - Prachi Srivastava
- Amity Institute of Biotechnology, Amity University, Lucknow, Uttar Pradesh, India
| |
Collapse
|
3
|
Lee S, Wong AR, Mehmet H, Yang AWH, Hung A. Elucidating the mechanisms of a herbal compound fumarine and its modulation on the estrogen receptor 1. J Biomol Struct Dyn 2024:1-14. [PMID: 39663629 DOI: 10.1080/07391102.2024.2438357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 05/15/2024] [Indexed: 12/13/2024]
Abstract
Stroke-related numbness and weakness (SRNW) are resultant disabilities following a stroke episode and may present with muscle weakness, numbness, tightness, spasticity, and pain in up to 85% of patients. Huangqi Guizhi Wuwu Decoction (HGWD) has been widely investigated to manage the sensorimotor deficiencies at the herb and formula level. However, detailed molecular mechanisms of its constituents are presently lacking. This project employed computational molecular modelling and docking methods to identify candidate compounds of HGWD which may serve as effective modulators of target proteins involved in SRNW. Estrogen Receptor 1 was identified as a promising target for HGWD compounds, while the herbal compound fumarine, a constituent of Jujubae Fructus, was predicted to exhibit high binding affinity and favourable ligand-receptor interactions with ESR1. There is currently a lack of scientific evidence for specific atomic-level interactions between ESR1 and this compound. Therefore, molecular docking and molecular dynamics simulations were used to elucidate the interaction mechanisms of fumarine with ESR1; and the molecular-level structural and functional consequences of ligand binding. Ligand-receptor contact analysis and free energy decomposition calculations identified Glu419 and Leu38 as stable hydrogen bond partners, while favourable contributions to the binding free energy include in Met421 (-10.74 kJ/mol) and Leu525 (-10.02 kJ/mol). This work provides the basis for further studies on discovering lead compounds which modulate the activity of ESR1.
Collapse
Affiliation(s)
- Sanghyun Lee
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia
| | - Ann Rann Wong
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia
| | - Hanife Mehmet
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia
| | - Angela Wei Hong Yang
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia
| | - Andrew Hung
- School of Science, RMIT University, Melbourne, Victoria, Australia
| |
Collapse
|
4
|
Yu X, Xu L, Su C, Wang C, Wang Z, Wang Y, Lu X, Sun H. Luteolin Protects against Vascular Calcification by Modulating SIRT1/CXCR4 Signaling Pathway and Promoting Autophagy. AAPS J 2024; 26:111. [PMID: 39438407 DOI: 10.1208/s12248-024-00982-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 10/08/2024] [Indexed: 10/25/2024] Open
Abstract
Vascular calcification (VC) is a common pathological manifestation of atherosclerosis, hypertension, diabetes vascular disease, vascular injury, chronic kidney disease and aging, which is mainly manifested as increased stiffness of the vascular wall. Oxidative stress and autophagy dysfunction are key factors in the pathogenesis of vascular calcification, but the specific mechanisms and the therapeutic strategy of vascular calcification have not been clarified. In the present study, Sirtuin 1 (SIRT1) was screened as the therapeutic targets for vascular calcification by the bioinformatics. SIRT1 is a nicotinamide adenine dinucleotide, which plays an important role in inhibiting oxidative stress and promoting autophagy. Luteolin (LUT), a kind of natural tetrahydroxyl flavonoid, exists in many plants and has many pharmacological effects such as anti-oxidation and anti-apoptosis. We have reported that luteolin has certain anti-osteoporosis effects in the previous study, and it is accepted that the development of vascular calcification is similar to bone formation, indicating that luteolin may also resist vascular calcification. And luteolin is known to activate SIRT1 to some extent. Moreover, the molecular docking analysis predicted that SIRT1 could bind directly to luteolin. Therefore, the purpose of this study was to investigate the potential role of luteolin in inhibiting oxidative stress and promoting autophagy during vascular calcification via modulating SIRT1 expression. The results showed that luteolin significantly improved vascular calcification induced by a high-fat diet (HFD) and vitamin D3 in rats in vivo. In addition, luteolin significantly repressed the formation of mineralized nodules and ALP activity in H2O2-treated A7r5 cells. Luteolin reduced the level of MDA, LDH and ROS generation, inhibited the protein expression of cleaved caspase-3, cleaved caspase-9, β-catenin and BMP-2 in the aortic tissue of the rat and rat smooth muscle cells (A7r5) treated with hydrogen peroxide. At the same time, luteolin could promote the expression of autophagy related proteins. Moreover, luteolin also produced effects to increase the protein expression levels of SIRT1 more than 2 times both in vivo and in vitro. In terms of mechanism, luteolin attenuated vascular calcification by inhibiting oxidative stress and improving autophagy level, via modulating SIRT1 / CXCR4 signaling pathway. In conclusion, this experiment for the first time revealed that LUT protected against VC via modulating SIRT1 / CXCR4 signaling pathway to promote autophagy and inhibit vascular calcification and may be developed as a new therapeutic agent for vascular calcification and atherosclerosis.
Collapse
Affiliation(s)
- Xiaoyu Yu
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, 9 West Section, Lvshun South Road, Lvshunkou District, Dalian, 116044, China
| | - Lei Xu
- Office of Ethics Committee, the First Affiliated Hospital, Dalian Medical University, No. 222, Zhongshan Road, Xigang District, Dalian, 116011, China
| | - Ce Su
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, 9 West Section, Lvshun South Road, Lvshunkou District, Dalian, 116044, China
| | - Changyuan Wang
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, 9 West Section, Lvshun South Road, Lvshunkou District, Dalian, 116044, China
| | - Zimeng Wang
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, 9 West Section, Lvshun South Road, Lvshunkou District, Dalian, 116044, China
| | - Yanna Wang
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, 9 West Section, Lvshun South Road, Lvshunkou District, Dalian, 116044, China
| | - Xiaolong Lu
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, 9 West Section, Lvshun South Road, Lvshunkou District, Dalian, 116044, China.
| | - Huijun Sun
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, 9 West Section, Lvshun South Road, Lvshunkou District, Dalian, 116044, China.
| |
Collapse
|
5
|
Soto-Sánchez J, Garza-Treviño G. Combination Therapy and Phytochemical-Loaded Nanosytems for the Treatment of Neglected Tropical Diseases. Pharmaceutics 2024; 16:1239. [PMID: 39458571 PMCID: PMC11510106 DOI: 10.3390/pharmaceutics16101239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 09/04/2024] [Accepted: 09/17/2024] [Indexed: 10/28/2024] Open
Abstract
Background: Neglected tropical diseases (NTDs), including leishmaniasis, trypanosomiasis, and schistosomiasis, impose a significant public health burden, especially in developing countries. Despite control efforts, treatment remains challenging due to drug resistance and lack of effective therapies. Objective: This study aimed to synthesize the current research on the combination therapy and phytochemical-loaded nanosystems, which have emerged as promising strategies to enhance treatment efficacy and safety. Methods/Results: In the present review, we conducted a systematic search of the literature and identified several phytochemicals that have been employed in this way, with the notable efficacy of reducing the parasite load in the liver and spleen in cases of visceral leishmaniasis, as well as lesion size in cutaneous leishmaniasis. Furthermore, they have a synergistic effect against Trypanosoma brucei rhodesiense rhodesain; reduce inflammation, parasitic load in the myocardium, cardiac hypertrophy, and IL-15 production in Chagas disease; and affect both mature and immature stages of Schistosoma mansoni, resulting in improved outcomes compared to the administration of phytochemicals alone or with conventional drugs. Moreover, the majority of the combinations studied demonstrated enhanced solubility, efficacy, and selectivity, as well as increased immune response and reduced cytotoxicity. Conclusions: These formulations appear to offer significant therapeutic benefits, although further research is required to validate their clinical efficacy in humans and their potential to improve treatment outcomes in affected populations.
Collapse
Affiliation(s)
- Jacqueline Soto-Sánchez
- Section for Postgraduate Studies and Research, Escuela Nacional de Medicina y Homeopatía, Instituto Politécnico Nacional, Guillermo Massieu Helguera #239, Fracc. La Escalera, Ticomán, Ciudad de México 07320, Mexico
| | - Gilberto Garza-Treviño
- Section for Postgraduate Studies and Research, Escuela Nacional de Medicina y Homeopatía, Instituto Politécnico Nacional, Guillermo Massieu Helguera #239, Fracc. La Escalera, Ticomán, Ciudad de México 07320, Mexico
| |
Collapse
|
6
|
Mia MM, Allaie IM, Zhang X, Li K, Khan SM, Kadotani S, Witola WH. Characterization of a unique catechol-O-methyltransferase as a molecular drug target in parasitic filarial nematodes. PLoS Negl Trop Dis 2024; 18:e0012473. [PMID: 39213433 PMCID: PMC11392244 DOI: 10.1371/journal.pntd.0012473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 09/12/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Filarial nematodes cause severe illnesses in humans and canines including limb deformities and disfigurement, heart failure, blindness, and death, among others. There are no vaccines, and current drugs against filarial nematodes infections have only modest effects and are prone to complications. METHODOLOGY/PRINCIPAL FINDINGS We identified a gene (herein called DiMT) encoding an S-adenosyl-L-methionine (SAM)-dependent methyltransferase with orthologs in parasite filarial worms but not in mammals. By in silico analysis, DiMT possesses catalytic sites for binding SAM and catecholamines with high affinity. We expressed and purified recombinant DiMT protein and used it as an enzyme in a series of SAM-dependent methylation assays. DiMT acted specifically as a catechol-O-methyltransferase (COMT), catalyzing catabolic methylation of dopamine, and depicted Michaelis Menten kinetics on substrate and co-substrate. Among a set of SAM-dependent methyltransferase inhibitors, we identified compounds that bound with high affinity to DiMT's catalytic sites and inhibited its enzymatic activity. By testing the efficacy of DiMT inhibitors against microfilariae of Dirofilaria immitis in culture, we identified three inhibitors with concentration- and time-dependent effect of killing D. immitis microfilariae. Importantly, RNAi silencing of a DiMT ortholog in Caenorhabditis elegans has been shown to be lethal, likely as a result of excessive accumulation of active catecholamines that inhibit worm locomotion, pharyngeal pumping and fecundity. CONCLUSIONS/SIGNIFICANCE Together, we have unveiled DiMT as an essential COMT that is conserved in parasitic filarial nematodes, but is significantly different from mammalian COMTs and, therefore, is a viable target for development of novel drugs against filarial nematode infections.
Collapse
Affiliation(s)
- Md Mukthar Mia
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois Urbana-Champaign, Urbana, Illinois, United States of America
| | - Idrees Mehraj Allaie
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois Urbana-Champaign, Urbana, Illinois, United States of America
| | - Xuejin Zhang
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois Urbana-Champaign, Urbana, Illinois, United States of America
| | - Kun Li
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois Urbana-Champaign, Urbana, Illinois, United States of America
- Institute of Traditional Chinese Veterinary Medicine, MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Shahbaz M Khan
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois Urbana-Champaign, Urbana, Illinois, United States of America
| | - Saki Kadotani
- Department of Veterinary Clinical Medicine, College of Veterinary Medicine, University of Illinois Urbana-Champaign, Urbana, Illinois, United States of America
| | - William H Witola
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois Urbana-Champaign, Urbana, Illinois, United States of America
| |
Collapse
|
7
|
Hossen MS, Islam MN, Pramanik MEA, Rahman MH, Amin MA, Antora ST, Sraboni FS, Chowdhury RN, Farha N, Sathi AA, Sadaf S, Banna F, Karim MR, Akter N, Gofur MR, Islam MS, Miah MMZ, Akhter M, Islam MS, Hasan MS, Fahmin F, Rahman MM, Basak PM, Sonnyashi AK, Das HS, Al Mahtab M, Akbar SMF. Molecular Characterization and Potential Inhibitors Prediction of Protein Arginine Methyltransferase-2 in Carcinoma: An Insight from Molecular Docking, ADMET Profiling and Molecular Dynamics Simulation Studies. Euroasian J Hepatogastroenterol 2024; 14:160-171. [PMID: 39802857 PMCID: PMC11714102 DOI: 10.5005/jp-journals-10018-1443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 08/13/2024] [Indexed: 01/16/2025] Open
Abstract
Objectives To predict and characterize the three-dimensional (3D) structure of protein arginine methyltransferase 2 (PRMT2) using homology modeling, besides, the identification of potent inhibitors for enhanced comprehension of the biological function of this protein arginine methyltransferase (PRMT) family protein in carcinogenesis. Materials and methods An in silico method was employed to predict and characterize the three-dimensional structure. The bulk of PRMTs in the PDB shares just a structurally conserved catalytic core domain. Consequently, it was determined that ligand compounds may be the source of co-crystallized complexes containing additional PRMTs. Possible PRMT2 inhibitor compounds are found by using S-adenosyl methionine (SAM), a methyl group donor, as a positive control. Results Protein arginine methyltransferases are associated with a range of physiological processes, including as splicing, proliferation, regulation of the cell cycle, differentiation, and signaling of DNA damage. These functional capacities are also related to carcinogenesis and metastasis-several forms of PRMT have been cited in the literature. These include PRMT-1, PRMT-2, and PRMT-5. Among these, the role of PRMT-2 has been shown in breast cancer and hepatocellular carcinoma. To gain more insights into the role of PRMT2 in cancer pathogenesis, we opted to characterize tertiary structure utilizing an in silico approach. The majority of PRMTs in the PDB have a structurally conserved catalytic core domain. Thus, ligand compounds were identified as a possible source of co-crystallized complexes of other PRMTs. The SAM, a methyl group donor, is used as a positive control in order to identify potential inhibitor compounds of PRMT2 by the virtual screening method. We hypothesized that an inhibitor for other PRMTs could alter PRMT2 activities. Out of 45 inhibitor compounds, we ultimately identified three potential inhibitor compounds based on the results of the pharmacokinetics and binding affinity studies. These compounds are identified as 3BQ (PubChem CID: 77620540), 6DX (PubChem CID: 124222721), and TDU (PubChem CID: 53346504). Their binding affinities are -8.5 kcal/mol, -8.1 kcal/mol, and -8.8 kcal/mol, respectively. These compounds will be further investigated to determine the binding stability and compactness using molecular dynamics simulations on a 100 ns time scale. In vitro and in vivo studies may be conducted with these three compounds, and we think that focusing on them might lead to the creation of a PRMT2 inhibitor. Conclusion Three strong inhibitory compounds that were non-carcinogenic also have drug-like properties. By using desirable parameters in root mean square deviation (RMSD), root mean square fluctuation (RMSF), radius of gyration (Rg), solvent accessible surface area (SASA), molecular surface area (MolSA), and intermolecular hydrogen bonding, complexes verified structural stability and compactness over the 100 ns time frame. How to cite this article Hossen MS, Islam MN, Pramanik MEA et al. Molecular Characterization and Potential Inhibitors Prediction of Protein Arginine Methyltransferase-2 (PRMT2) in Carcinoma: An Insight from Molecular Docking, ADMET Profiling and Molecular Dynamics Simulation Studies. Euroasian J Hepato-Gastroenterol 2024;14(2):160-171.
Collapse
Affiliation(s)
- Md Sahadot Hossen
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Shahjalal University of Science and Technology, Sylhet, Bangladesh
| | - Md Nur Islam
- National Laboratory of Biomacromolecules, Chinese Academy of Sciences Center for Excellence in Biomacromolecules, Institute of Biophysics, CAS, Beijing, People's Republic of China; Department of Pharmacy, Manarat International University, Gulshan, Dhaka, Bangladesh
| | - Md Enayet A Pramanik
- Department of Entomology, On-Farm Research Division, Bangladesh Agricultural Research Institute, Terokhadia, Rajshahi, Bangladesh
| | - Md Hasanur Rahman
- Department of Biotechnology and Genetic Engineering, Faculty of Life Sciences, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Bangladesh
| | - Md Al Amin
- Department of Biotechnology and Genetic Engineering, Faculty of Life Sciences, Mawlana Bhashani Science and Technology University, Tangail, Santosh, Bangladesh
| | - Saraban T Antora
- Department of Biochemistry and Molecular Biology, Faculty of Life Sciences, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Bangladesh
| | - Farzana S Sraboni
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Sciences, University of Rajshahi, Rajshahi, Bangladesh
| | - Rifah N Chowdhury
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka (DU), Mokarram Hussain Khundker Bhaban, University St, Dhaka, Bangladesh
| | - Nazia Farha
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Sciences, University of Chittagong, Chittagong, Bangladesh
| | - Amina A Sathi
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Sciences, University of Rajshahi, Rajshahi, Bangladesh
| | - Samia Sadaf
- Department of Biology, Development Biology Laboratory, Clarkson University, Potsdam, New York, United States of America
| | - Farjana Banna
- Department of Biotechnology and Genetic Engineering, Faculty of Science, Islamic University, Kushtia, Bangladesh
| | - Md Rezaul Karim
- Department of Pharmacy, School of Engineering, Science and Technology, Manarat International University, Gulshan, Dhaka, Bangladesh
| | - Nasrin Akter
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi, Bangladesh
| | - Md Royhan Gofur
- Department of Veterinary and Animal Sciences, University of Rajshahi, Rajshahi, Bangladesh
| | - Md Shariful Islam
- Department of Veterinary and Animal Sciences, University of Rajshahi, Rajshahi, Bangladesh
| | - M Morsed Z Miah
- Department of Hematology, Rajshahi Medical College, Rajshahi, Bangladesh
| | - Mira Akhter
- Department of Clinical Pathology, Rajshahi Medical College Hospital, Rajshahi, Bangladesh
| | - Md Shariful Islam
- Department of Ortho-Surgery, Rajshahi Medical College, Rajshahi, Bangladesh
| | - Md Sharif Hasan
- Department of Cardiology, Mymensingh Medical College Hospital, Mymensingh, Bangladesh
| | - Fahmida Fahmin
- Department of Paediatric, Mymensingh Medical College Hospital, Mymensingh, Bangladesh
| | - Mohammad M Rahman
- Department of Medicine, Rajshahi Medical College, Rajshahi, Bangladesh
| | - Prabir M Basak
- Department of Medicine, Rajshahi Medical College, Rajshahi, Bangladesh
| | - Amio K Sonnyashi
- Department of Medicine, Rajshahi Medical College, Rajshahi, Bangladesh
| | - Haimanti S Das
- Department of Virology, Rajshahi Medical College, Rajshahi, Bangladesh
| | - Mamun Al Mahtab
- Department of Interventional Hepatology, Bangabandhu Sheikh Mujib Medical University, Shahbagh, Dhaka, Bangladesh
| | - Sheikh MF Akbar
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, Ehime, Japan
| |
Collapse
|
8
|
Ding H, Xing F, Zou L, Zhao L. QSAR analysis of VEGFR-2 inhibitors based on machine learning, Topomer CoMFA and molecule docking. BMC Chem 2024; 18:59. [PMID: 38555462 PMCID: PMC10981835 DOI: 10.1186/s13065-024-01165-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 03/12/2024] [Indexed: 04/02/2024] Open
Abstract
VEGFR-2 kinase inhibitors are clinically approved drugs that can effectively target cancer angiogenesis. However, such inhibitors have adverse effects such as skin toxicity, gastrointestinal reactions and hepatic impairment. In this study, machine learning and Topomer CoMFA, which is an alignment-dependent, descriptor-based method, were employed to build structural activity relationship models of potentially new VEGFR-2 inhibitors. The prediction ac-curacy of the training and test sets of the 2D-SAR model were 82.4 and 80.1%, respectively, with KNN. Topomer CoMFA approach was then used for 3D-QSAR modeling of VEGFR-2 inhibitors. The coefficient of q2 for cross-validation of the model 1 was greater than 0.5, suggesting that a stable drug activity-prediction model was obtained. Molecular docking was further performed to simulate the interactions between the five most promising compounds and VEGFR-2 target protein and the Total Scores were all greater than 6, indicating that they had a strong hydrogen bond interactions were present. This study successfully used machine learning to obtain five potentially novel VEGFR-2 inhibitors to increase our arsenal of drugs to combat cancer.
Collapse
Affiliation(s)
- Hao Ding
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning, China
| | - Fei Xing
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning, China
| | - Lin Zou
- Medical College of Guangxi University, Nanning, 530004, Guangxi, China
| | - Liang Zhao
- Hepatobiliary and Splenic Surgery Ward, Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning, China.
| |
Collapse
|
9
|
Vijayakumar S, Kumar LL, Borkotoky S, Murali A. The Application of MD Simulation to Lead Identification, Vaccine Design, and Structural Studies in Combat against Leishmaniasis - A Review. Mini Rev Med Chem 2024; 24:1089-1111. [PMID: 37680156 DOI: 10.2174/1389557523666230901105231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 06/07/2023] [Accepted: 07/14/2023] [Indexed: 09/09/2023]
Abstract
Drug discovery, vaccine design, and protein interaction studies are rapidly moving toward the routine use of molecular dynamics simulations (MDS) and related methods. As a result of MDS, it is possible to gain insights into the dynamics and function of identified drug targets, antibody-antigen interactions, potential vaccine candidates, intrinsically disordered proteins, and essential proteins. The MDS appears to be used in all possible ways in combating diseases such as cancer, however, it has not been well documented as to how effectively it is applied to infectious diseases such as Leishmaniasis. As a result, this review aims to survey the application of MDS in combating leishmaniasis. We have systematically collected articles that illustrate the implementation of MDS in drug discovery, vaccine development, and structural studies related to Leishmaniasis. Of all the articles reviewed, we identified that only a limited number of studies focused on the development of vaccines against Leishmaniasis through MDS. Also, the PCA and FEL studies were not carried out in most of the studies. These two were globally accepted utilities to understand the conformational changes and hence it is recommended that this analysis should be taken up in similar approaches in the future.
Collapse
Affiliation(s)
| | | | - Subhomoi Borkotoky
- Department of Biotechnology, Invertis University, Bareilly, Uttar Pradesh, India
| | - Ayaluru Murali
- Department of Bioinformatics, Pondicherry University, Puducherry, India
| |
Collapse
|
10
|
Khwaza V, Oselusi SO, Morifi E, Nwamadi M, Hlope KS, Ndinteh DT, Matsebatlela TM, Oyedeji OO, Aderibigbe BA. Synthesis of Ursolic Acid-based Hybrids: In Vitro Antibacterial, Cytotoxicity Studies, In Silico Physicochemical and Pharmacokinetic Properties. RECENT ADVANCES IN ANTI-INFECTIVE DRUG DISCOVERY 2024; 19:232-253. [PMID: 38317466 DOI: 10.2174/0127724344272444231114103144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 10/04/2023] [Accepted: 10/11/2023] [Indexed: 02/07/2024]
Abstract
BACKGROUND There is a critical need for the discovery of novel and effective antibacterial or anticancer molecules. OBJECTIVES Amine-linked ursolic acid-based hybrid compounds were prepared in good yields in the range of 60-68%. METHODS Their molecular structures were successfully confirmed using different spectroscopic methods including 1H/13C NMR, UHPLC-HRMS and FTIR spectroscopy. The in vitro cytotoxicity of some of these hybrid molecules against three human tumour cells, such as MDA-MB23, MCF7, and HeLa was evaluated using the MTT colorimetric method. RESULT Their antibacterial efficacy was evaluated against eleven bacterial pathogens using a serial dilution assay. Majority of the bacterial strains were inhibited significantly by compounds 17 and 24, with the lowest MIC values in the range of 15.3-31.25 μg/mL. Compound 16 exhibited higher cytotoxicity against HeLa cells than ursolic acid, with an IC50 value of 43.64 g/mL. CONCLUSION The in vitro antibacterial activity and cytotoxicity of these hybrid compounds demonstrated that ursolic acid-based hybrid molecules are promising compounds. Further research into ursolic acid-based hybrid compounds is required.
Collapse
Affiliation(s)
- Vuyolwethu Khwaza
- Department of Chemistry, University of Fort Hare, Alice Campus, Alice, Eastern Cape, South Africa
| | - Samson Olaitan Oselusi
- School of Pharmacy, University of the Western Cape, Bellville, Cape Town 7535, South Africa
| | - Eric Morifi
- School of Chemistry, Mass Spectrometry Division, University of Witwatersrand, Johannesburg, South Africa
| | - Mutshinyalo Nwamadi
- Department of Chemistry, University of Johannesburg, Auckland Park Campus, Johannesburg, South Africa
| | - Kamogelo S Hlope
- Department of Biochemistry, Microbiology and Biotechnology, Faculty of Science and Agriculture, University of Limpopo, South Africa
| | - Derek Tantoh Ndinteh
- Department of Applied Chemistry, University of Johannesburg, Doornfontein Campus, Johannesburg, South Africa
| | - Thabe Moses Matsebatlela
- Department of Biochemistry, Microbiology and Biotechnology, Faculty of Science and Agriculture, University of Limpopo, South Africa
| | - Opeoluwa Oyehan Oyedeji
- Department of Chemistry, University of Fort Hare, Alice Campus, Alice, Eastern Cape, South Africa
| | - Blessing Atim Aderibigbe
- Department of Chemistry, University of Fort Hare, Alice Campus, Alice, Eastern Cape, South Africa
| |
Collapse
|
11
|
Ranjan P, Dubey VK. Novel chemical scaffold as potential drug against Leishmania donovani: Integrated computational and experimental approaches. J Cell Biochem 2023; 124:1404-1422. [PMID: 37566640 DOI: 10.1002/jcb.30455] [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: 06/06/2023] [Revised: 07/23/2023] [Accepted: 07/24/2023] [Indexed: 08/13/2023]
Abstract
In this study, we have screened a large number of Food and Drug Administration-approved compounds for novel anti-leishmanial molecules targeting the citrate synthase enzyme of the parasite. Based on their docking and molecular dynamic simulation statistics, five compounds were selected. These compounds followed Lipinski's rule of five. Additionally, in vitro, antileishmanial and cytotoxicity studies were performed. The three compounds, Abemaciclib, Bazedoxifene, and Vorapaxar, had shown effective anti-leishmanial activities with IC50 values of 0.92 ± 0.02, 0.65 ± 0.09, and 6.1 ± 0.91 against Leishmania donovani promastigote and with EC50 values of 1.52 ± 0.37, 2.11 ± 0.38, 10.4 ± 1.27 against intramacrophagic amastigote without significantly harming macrophage cells. Among them, from in silico and antileishmanial activities studies, Abemaciclib had been selected based on their less binding energy, good antileishmanial activities, and also a significant difference in their binding energy with human citrate synthase for cell death mechanistic studies using flow cytometry and a DNA fragmentation assay. The action of this compound resulted in an increased reactive oxygen species production, depolarization of mitochondrial membrane potential, DNA damage, and an increase in the sub-G1 cell population. These properties are the hallmarks of apoptosis which were further confirmed by apoptotic assay. Based on the above result, this anticancer compound Abemaciclib could be employed as a potential treatment option for leishmaniasis after further confirmation.
Collapse
Affiliation(s)
- Preeti Ranjan
- School of Biochemical Engineering, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh, India
| | - Vikash Kumar Dubey
- School of Biochemical Engineering, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh, India
| |
Collapse
|
12
|
Oyewusi HA, Wu YS, Safi SZ, Wahab RA, Hatta MHM, Batumalaie K. Molecular dynamics simulations reveal the inhibitory mechanism of Withanolide A against α-glucosidase and α-amylase. J Biomol Struct Dyn 2023; 41:6203-6218. [PMID: 35904027 DOI: 10.1080/07391102.2022.2104375] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 07/15/2022] [Indexed: 10/16/2022]
Abstract
Diabetes mellitus (DM) is a global chronic disease characterized by hyperglycemia and insulin resistance. The unsavory severe gastrointestinal side-effects of synthetic drugs to regulate hyperglycemia have warranted the search for alternative treatments to inhibit the carbohydrate digestive enzymes (e.g. α-amylase and α-glucosidase). Certain phytochemicals recently captured the scientific community's attention as carbohydrate digestive enzyme inhibitors due to their low toxicity and high efficacy, specifically the Withanolides-loaded extract of Withania somnifera. That said, the present study evaluated in silico the efficacy of Withanolide A in targeting both α-amylase and α-glucosidase in comparison to the synthetic drug Acarbose. Protein-ligand interactions, binding affinity, and stability were characterized using pharmacological profiling, high-end molecular docking, and molecular-dynamic simulation. Withanolide A inhibited the activity of α-glucosidase and α-amylase better, exhibiting good pharmacokinetic properties, absorption, and metabolism. Also, Withanolide A was minimally toxic, with higher bioavailability. Interestingly, Withanolide A bonded well to the active site of α-amylase and α-glucosidase, yielding the lowest binding free energy of -82.144 ± 10.671 kcal/mol and -102.1043 ± 11.231 kcal/mol compared to the Acarbose-enzyme complexes (-63.220 ± 13.283 kcal/mol and -82.148 ± 10.671 kcal/mol). Hence, the findings supported the therapeutic potential of Withanolide A as α-amylase and α-glucosidase inhibitor for DM treatment.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Habeebat Adekilekun Oyewusi
- Department of Biosciences, Faculty of Science, Universiti Teknologi Malaysia, Johor, Malaysia
- Enzyme Technology and Green Synthesis Group, Faculty of Science, Universiti Teknologi Malaysia, Johor, Malaysia
- Department of Science Technology, Biochemistry unit, The Federal Polytechnic, Ado Ekiti, Ekiti State, Nigeria
| | - Yuan-Seng Wu
- Centre for Virus and Vaccine Research, School of Medical and Life Sciences, Sunway University, Subang Jaya, Malaysia
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, Petaling Jaya, Malaysia
| | - Sher Zaman Safi
- IRCBM, COMSATS University Islamabad, Lahore Campus, Punjab, Pakistan
| | - Roswanira Abdul Wahab
- Enzyme Technology and Green Synthesis Group, Faculty of Science, Universiti Teknologi Malaysia, Johor, Malaysia
- Department of Chemistry, Faculty of Science, Universiti Teknologi Malaysia, Johor, Malaysia
| | | | - Kalaivani Batumalaie
- Department of Biomedical Science, Faculty of Health Sciences, Asia Metropolitan University, Johor Bahru, Johor, Malaysia
| |
Collapse
|
13
|
Sasidharan S, Radhakrishnan K, Lee JY, Saudagar P, Gosu V, Shin D. Molecular dynamics of the ERRγ ligand-binding domain bound with agonist and inverse agonist. PLoS One 2023; 18:e0283364. [DOI: doi.org/10.1371/journal.pone.0283364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/18/2023] Open
Abstract
Estrogen-related receptor gamma (ERRγ), the latest member of the ERR family, does not have any known reported natural ligands. Although the crystal structures of the apo, agonist-bound, and inverse agonist-bound ligand-binding domain (LBD) of ERRγ have been solved previously, their dynamic behavior has not been studied. Hence, to explore the intrinsic dynamics of the apo and ligand-bound forms of ERRγ, we applied long-range molecular dynamics (MD) simulations to the crystal structures of the apo and ligand-bound forms of the LBD of ERRγ. Using the MD trajectories, we performed hydrogen bond and binding free energy analysis, which suggested that the agonist displayed more hydrogen bonds with ERRγ than the inverse agonist 4-OHT. However, the binding energy of 4-OHT was higher than that of the agonist GSK4716, indicating that hydrophobic interactions are crucial for the binding of the inverse agonist. From principal component analysis, we observed that the AF-2 helix conformation at the C-terminal domain was similar to the initial structures during simulations, indicating that the AF-2 helix conformation is crucial with respect to the agonist or inverse agonist for further functional activity of ERRγ. In addition, we performed residue network analysis to understand intramolecular signal transduction within the protein. The betweenness centrality suggested that few of the amino acids are important for residue signal transduction in apo and ligand-bound forms. The results from this study may assist in designing better therapeutic compounds against ERRγ associated diseases.
Collapse
|
14
|
Pande M, Kundu D, Srivastava R. Selective vitamins as potential options for dietary therapeutic interventions: In silico and In vitro insights from mutant C terminal fragment of FGA. J Steroid Biochem Mol Biol 2023; 230:106290. [PMID: 36907427 DOI: 10.1016/j.jsbmb.2023.106290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 02/18/2023] [Accepted: 03/09/2023] [Indexed: 03/12/2023]
Abstract
We have used an integrated computational approach to explore the role of vitamin C and vitamin D in preventing aggregation of Fibrinogen A alpha-chain (FGActer) protein responsible for renal amyloidosis. We modelled structures of E524K / E526K mutants of FGActer protein and examined the potential interactions of these mutants with vitamin C and vitamin D3. Interaction of these vitamins at the amyloidogenic site may prevent the intermolecular interaction required for amyloid formation. The binding free energy values of vitamin C and vitamin D3 for E524K FGActer and E526K FGActer are - 67.12 ± 30.46 kJ/mole and - 79.45 ± 26.12 kJ/mol, respectively. Experimental studies using Congo red absorption, aggregation index studies and AFM imaging show encouraging results. The AFM images of E526K FGActer contained more extensive and higher protofibril aggregates, whereas, in the presence of vitamin D3, small monomeric and oligomeric aggregates were observed. Overall, the works provide interesting results about vitamin C and D role in preventing renal amyloidosis.
Collapse
Affiliation(s)
- Monu Pande
- Department of Biochemistry, Institute of Medical Science, Banaras Hindu University, Varanasi 221005, India
| | - Debanjan Kundu
- School of Biochemical Engineering, Indian Institute of Technology (BHU), Varanasi 221005, India
| | - Ragini Srivastava
- Department of Biochemistry, Institute of Medical Science, Banaras Hindu University, Varanasi 221005, India.
| |
Collapse
|
15
|
Çelik F, Aydın A, Bektaş Kİ, Güler Hİ, Nalçaoğlu A, Ünver Y. Biscarbazole Derivatives: Synthesis, Characterization, Antimicrobial and Antitumor Activity, and Molecular Docking Study. RUSS J GEN CHEM+ 2022. [DOI: 10.1134/s1070363222100279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
16
|
Ünver Y, Süleymanoğlu N, Ustabaş R, Güler Hİ, Bektaş E, Bektaş Kİ, Çelik F. New carbazol derivatives containing 1,2,4-triazole: Synthesis, characterization, DFT study, acetylcholinesterase activity and docking study. J INDIAN CHEM SOC 2022. [DOI: 10.1016/j.jics.2022.100690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
|
17
|
Santos ES, Silva PC, Sousa PSA, Aquino CC, Pacheco G, Teixeira LFLS, Araujo AR, Sousa FBM, Barros RO, Ramos RM, Rocha JA, Nicolau LAD, Medeiros JVR. Antiviral potential of diminazene aceturate against SARS-CoV-2 proteases using computational and in vitro approaches. Chem Biol Interact 2022; 367:110161. [PMID: 36116513 PMCID: PMC9476334 DOI: 10.1016/j.cbi.2022.110161] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 08/30/2022] [Accepted: 09/01/2022] [Indexed: 11/05/2022]
Abstract
Diminazene aceturate (DIZE), an antiparasitic, is an ACE2 activator, and studies show that activators of this enzyme may be beneficial for COVID-19, disease caused by SARS-CoV-2. Thus, the objective was to evaluate the in silico and in vitro affinity of diminazene aceturate against molecular targets of SARS-CoV-2. 3D structures from DIZE and the proteases from SARS-CoV-2, obtained through the Protein Data Bank and Drug Database (Drubank), and processed in computer programs like AutodockTools, LigPlot, Pymol for molecular docking and visualization and GROMACS was used to perform molecular dynamics. The results demonstrate that DIZE could interact with all tested targets, and the best binding energies were obtained from the interaction of Protein S (closed conformation −7.87 kcal/mol) and Mpro (−6.23 kcal/mol), indicating that it can act both by preventing entry and viral replication. The results of molecular dynamics demonstrate that DIZE was able to promote a change in stability at the cleavage sites between S1 and S2, which could prevent binding to ACE2 and fusion with the membrane. In addition, in vitro tests confirm the in silico results showing that DIZE could inhibit the binding between the spike receptor-binding domain protein and ACE2, which could promote a reduction in the virus infection. However, tests in other experimental models with in vivo approaches are needed.
Collapse
Affiliation(s)
- Esley S Santos
- Laboratory of Pharmacology of Inflammation and Gastrointestinal Disorders (LAFIDG), Post-graduation Program in Biotechnology, Parnaíba Delta Federal University, Parnaíba, PI, Brazil; Medicinal Plants Research Center (NPPM), Federal University of Piauí, Teresina, Brazil
| | - Priscila C Silva
- Laboratory of Pharmacology of Inflammation and Gastrointestinal Disorders (LAFIDG), Post-graduation Program in Biotechnology, Parnaíba Delta Federal University, Parnaíba, PI, Brazil
| | - Paulo S A Sousa
- Laboratory of Medicinal Chemistry and Biotechnology, QUIMEBIO, Federal University of Maranhão, São Bernardo, MA, Brazil; Biodiversity and Biotechnology Research Center, BIOTEC, Post-graduation Program in Biotechnology, Parnaíba Delta Federal University, Parnaíba, PI, Brazil
| | - Cristhyane C Aquino
- Postgraduate Program in Medical Sciences, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Gabriella Pacheco
- Laboratory of Pharmacology of Inflammation and Gastrointestinal Disorders (LAFIDG), Post-graduation Program in Biotechnology, Parnaíba Delta Federal University, Parnaíba, PI, Brazil; Medicinal Plants Research Center (NPPM), Federal University of Piauí, Teresina, Brazil
| | - Luiz F L S Teixeira
- Laboratory of Pharmacology of Inflammation and Gastrointestinal Disorders (LAFIDG), Post-graduation Program in Biotechnology, Parnaíba Delta Federal University, Parnaíba, PI, Brazil; Biodiversity and Biotechnology Research Center, BIOTEC, Post-graduation Program in Biotechnology, Parnaíba Delta Federal University, Parnaíba, PI, Brazil
| | - Alyne R Araujo
- Biodiversity and Biotechnology Research Center, BIOTEC, Post-graduation Program in Biotechnology, Parnaíba Delta Federal University, Parnaíba, PI, Brazil
| | - Francisca B M Sousa
- Laboratory of Pharmacology of Inflammation and Gastrointestinal Disorders (LAFIDG), Post-graduation Program in Biotechnology, Parnaíba Delta Federal University, Parnaíba, PI, Brazil
| | - Romulo O Barros
- Research Laboratory in Information Systems, Department of Information, Environment, Health and Food Production, Federal Institute of Piauí, LaPeSI/IFPI, Teresina, Piauí, Brazil
| | - Ricardo M Ramos
- Research Laboratory in Information Systems, Department of Information, Environment, Health and Food Production, Federal Institute of Piauí, LaPeSI/IFPI, Teresina, Piauí, Brazil
| | - Jefferson A Rocha
- Laboratory of Medicinal Chemistry and Biotechnology, QUIMEBIO, Federal University of Maranhão, São Bernardo, MA, Brazil; Biodiversity and Biotechnology Research Center, BIOTEC, Post-graduation Program in Biotechnology, Parnaíba Delta Federal University, Parnaíba, PI, Brazil
| | - Lucas A D Nicolau
- Laboratory of Pharmacology of Inflammation and Gastrointestinal Disorders (LAFIDG), Post-graduation Program in Biotechnology, Parnaíba Delta Federal University, Parnaíba, PI, Brazil; Biodiversity and Biotechnology Research Center, BIOTEC, Post-graduation Program in Biotechnology, Parnaíba Delta Federal University, Parnaíba, PI, Brazil
| | - Jand V R Medeiros
- Laboratory of Pharmacology of Inflammation and Gastrointestinal Disorders (LAFIDG), Post-graduation Program in Biotechnology, Parnaíba Delta Federal University, Parnaíba, PI, Brazil; Medicinal Plants Research Center (NPPM), Federal University of Piauí, Teresina, Brazil; Biodiversity and Biotechnology Research Center, BIOTEC, Post-graduation Program in Biotechnology, Parnaíba Delta Federal University, Parnaíba, PI, Brazil.
| |
Collapse
|
18
|
Modanwal S, Maurya AK, Mishra SK, Mishra N. Development of QSAR model using machine learning and molecular docking study of polyphenol derivatives against obesity as pancreatic lipase inhibitor. J Biomol Struct Dyn 2022:1-12. [PMID: 35947118 DOI: 10.1080/07391102.2022.2109753] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
Abstract
In developed countries and developing countries, obesity/overweight is considered a major problem, in fact, it is now recognized as a major metabolic disorder. Additionally, obesity is connected with other metabolic diseases, including cardiovascular disorders, type 2 diabetes, some types of cancer, etc. Therefore, the development of novel drugs/medications for obesity is essential. The best target for treating obesity is Pancreatic Lipase (PL), it breaks 50-70% triglycerides into monoglycerol and free fatty acids.The major aim of this in silico study is to generate a QSAR model by using Multiple Linear Regression (MLR) and to inhibit pancreatic lipase by polyphenol derivatives mainly flavonoids, plant secondary metabolites shows good inhibitory activity against PL, maybe with less unpleasant side effects.In this in silico study, a potent inhibitor was found through calculating drug likness, QSAR (Quantitative structure-activity relationship) and molecular docking. The docking was performed in Maestro 12.0 and the ADME (absorption, distribution, metabolism, and excretion) properties (drug-likeness) of compounds/ligands were predicted by the Qikprop module of Maestro 12.0. The QSAR model was developed to show the relationship between the chemical/structural properties and the compound's biological activity. We have found the best interaction between pancreatic lipase and flavonoids. The best docked compound is Epigallocatechin 3,5,-di-O-gallate with docking score -10.935 kcal/mol .All compounds also show drug-likeness activity.The developed model has satisfied all internal and external validation criteria and has square correlation coefficient (r2) 0.8649, which shows its predictive ability and has good acceptability, predictive ability, and statistical robustness.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Shristi Modanwal
- Chemistry Laboratory, Department of Applied Sciences, Indian Institute of Information Technology Allahabad, Prayagraj, Uttar Pradesh, India
| | - Akhilesh Kumar Maurya
- Chemistry Laboratory, Department of Applied Sciences, Indian Institute of Information Technology Allahabad, Prayagraj, Uttar Pradesh, India
| | - Saurav Kumar Mishra
- Chemistry Laboratory, Department of Applied Sciences, Indian Institute of Information Technology Allahabad, Prayagraj, Uttar Pradesh, India
| | - Nidhi Mishra
- Chemistry Laboratory, Department of Applied Sciences, Indian Institute of Information Technology Allahabad, Prayagraj, Uttar Pradesh, India
| |
Collapse
|
19
|
Investigation of potential inhibitor properties of violacein against HIV-1 RT and CoV-2 Spike RBD:ACE-2. World J Microbiol Biotechnol 2022; 38:161. [PMID: 35834025 PMCID: PMC9281270 DOI: 10.1007/s11274-022-03350-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 06/28/2022] [Indexed: 11/18/2022]
Abstract
A violacein-producing bacterium was isolated from a mud sample collected near a hot spring on Kümbet Plateau in Giresun Province and named the GK strain. According to the phylogenetic tree constructed using 16S rRNA gene sequence analysis, the GK strain was identified and named Janthinobacterium sp. GK. The crude violacein pigments were separated into three different bands on a TLC sheet. Then violacein and deoxyviolacein were purified by vacuum liquid column chromatography and identified by NMR spectroscopy. According to the inhibition studies, the HIV-1 RT inhibition rate of 1 mM violacein from the GK strain was 94.28% and the CoV-2 spike RBD:ACE2 inhibition rate of 2 mM violacein was 53%. In silico studies were conducted to investigate the possible interactions between violacein and deoxyviolacein and three reference molecules with the target proteins: angiotensin-converting enzyme 2 (ACE2), HIV-1 reverse transcriptase, and SARS-CoV-2 spike receptor binding domain. Ligand violacein binds strongly to the receptor ACE2, HIV-1 reverse transcriptase, and SARS-CoV-2 spike receptor binding domain with a binding energy of −9.94 kcal/mol, −9.32 kcal/mol, and −8.27 kcal/mol, respectively. Deoxyviolacein strongly binds to the ACE2, HIV-1 reverse transcriptase, and SARS-CoV-2 spike receptor binding domain with a binding energy of −10.38 kcal/mol, -9.50 kcal/mol, and −8.06 kcal/mol, respectively. According to these data, violacein and deoxyviolacein bind to all the receptors quite effectively. SARS-CoV-2 spike protein and HIV-1-RT inhibition studies with violacein and deoxyviolacein were performed for the first time in the literature.
Collapse
|
20
|
Lee S, Wong AR, Yang AWH, Hung A. Interaction of compounds derived from the Chinese medicinal formula Huangqi Guizhi Wuwu Tang with stroke-related numbness and weakness targets: An in-silico docking and molecular dynamics study. Comput Biol Med 2022; 146:105568. [DOI: 10.1016/j.compbiomed.2022.105568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/15/2022] [Accepted: 04/25/2022] [Indexed: 11/03/2022]
|
21
|
Raj S, Sasidharan S, Tripathi T, Saudagar P. Biofunctionalized Chrysin-conjugated gold nanoparticles neutralize Leishmania parasites with high efficacy. Int J Biol Macromol 2022; 205:211-219. [PMID: 35183598 DOI: 10.1016/j.ijbiomac.2022.02.047] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 02/02/2022] [Accepted: 02/09/2022] [Indexed: 12/18/2022]
Abstract
Current treatments for leishmaniasis involve various drugs, including miltefosine and amphotericin B, which are associated with several side effects and high costs. Long-term use of these drugs may lead to the development of resistance, thereby reducing their efficiency. Chrysin (CHY) is a well-known, non-toxic flavonoid with antioxidant, antiviral, anti-inflammatory, anti-cancer, hepatoprotective, and neuroprotective properties. Recently we have shown that CHY targets the MAP kinase 3 enzyme of Leishmania and neutralizes the parasite rapidly. However, CHY is associated with low bioavailability, poor absorption, and rapid excretion issues, limiting its usage. In this study, we developed and tested a novel CHY-gold nanoformulation with improved efficacy against the parasites. The reducing power of CHY was utilized to reduce and conjugate with gold nanoparticles. Gold nanoparticles, which are already known for their anti-leishmanial properties, along with conjugated CHY, exhibited a decreased parasite burden in mammalian macrophages. Our findings showed that this biofunctionalized nanoformulation could be used as a potential therapeutic tool against leishmaniasis.
Collapse
Affiliation(s)
- Shweta Raj
- Department of Biotechnology, National Institute of Technology-Warangal, Warangal 506004, India
| | - Santanu Sasidharan
- Department of Biotechnology, National Institute of Technology-Warangal, Warangal 506004, India
| | - Timir Tripathi
- Molecular and Structural Biophysics Laboratory, Department of Biochemistry, North-Eastern Hill University, Shillong 793022, India
| | - Prakash Saudagar
- Department of Biotechnology, National Institute of Technology-Warangal, Warangal 506004, India.
| |
Collapse
|
22
|
Jomhori M, Mosaddeghi H. Molecular modeling of natural and synthesized inhibitors against SARS-CoV-2 spike glycoprotein. RESEARCH ON BIOMEDICAL ENGINEERING 2022. [PMCID: PMC7779244 DOI: 10.1007/s42600-020-00122-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Purpose Viral diseases increasingly endanger the world public health because of the transient efficacy of antiviral therapies. The novel coronavirus disease 2019 (COVID-19) has been recently identified as caused by a new type of coronaviruses. This type of coronavirus binds to the human receptor through the Spike glycoprotein (S) Receptor Binding Domain (RBD). The spike protein is found in inaccessible (closed) or accessible (open) conformations in which the accessible conformation causes severe infection. Thus, this receptor is a significant target for antiviral drug design. Methods An attempt was made to recognize 111 natural and synthesized compounds in order to utilize them against SARS-CoV-2 spike glycoprotein to inhibit Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) using simulation methods, such as molecular docking. The FAF-Drugs3, Pan-Assay Interference Compounds (PAINS), ADME (absorption, distribution, metabolism, excretion) databases along with Lipinski’s rules were used to evaluate the drug-like properties of the identified ligands. In order to analyze and identify the residues critical in the docking process of the spike glycoprotein, the interactions of proposed ligands with both conformations of the spike glycoprotein was simulated. Results The results showed that among the available ligands, seven ligands had significant interactions with the binding site of the spike glycoprotein, in which angiotensin-converting enzyme 2 (ACE2) is bounded. Out of seven candidate molecules, six ligands exhibited drug-like characteristics. The results also demonstrated that fluorophenyl and propane groups of ligands had optimal interactions with the binding site of the spike glycoprotein. Conclusion According to the results, our findings indicated the ability of six ligands to prevent the binding of the SARS-CoV-2 spike glycoprotein to its cognate receptor, providing novel compounds for the treatment of COVID-19. Supplementary Information The online version contains supplementary material available at 10.1007/s42600-020-00122-3.
Collapse
Affiliation(s)
| | - Hamid Mosaddeghi
- Department of Chemistry, Isfahan University of Technology, Isfahan, 84156-83111 Iran
| |
Collapse
|
23
|
Designing a multilayer film via machine learning of scientific literature. Sci Rep 2022; 12:930. [PMID: 35042971 PMCID: PMC8766440 DOI: 10.1038/s41598-022-05010-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 01/04/2022] [Indexed: 12/23/2022] Open
Abstract
Scientists who design chemical substances often use materials informatics (MI), a data-driven approach with either computer simulation or artificial intelligence (AI). MI is a valuable technique, but applying it to layered structures is difficult. Most of the proposed computer-aided material search techniques use atomic or molecular simulations, which are limited to small areas. Some AI approaches have planned layered structures, but they require a physical theory or abundant experimental results. There is no universal design tool for multilayer films in MI. Here, we show a multilayer film can be designed through machine learning (ML) of experimental procedures extracted from chemical-coating articles. We converted material names according to International Union of Pure and Applied Chemistry rules and stored them in databases for each fabrication step without any physicochemical theory. Compared with experimental results which depend on authors, experimental protocol is superiority at almost unified and less data loss. Connecting scientific knowledge through ML enables us to predict untrained film structures. This suggests that AI imitates research activity, which is normally inspired by other scientific achievements and can thus be used as a general design technique.
Collapse
|
24
|
Kaur P, Goyal N. Pathogenic role of mitogen activated protein kinases in protozoan parasites. Biochimie 2021; 193:78-89. [PMID: 34706251 DOI: 10.1016/j.biochi.2021.10.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 09/29/2021] [Accepted: 10/21/2021] [Indexed: 01/18/2023]
Abstract
Protozoan parasites with complex life cycles have high mortality rates affecting billions of human lives. Available anti-parasitic drugs are inadequate due to variable efficacy, toxicity, poor patient compliance and drug-resistance. Hence, there is an urgent need for the development of safer and better chemotherapeutics. Mitogen Activated Protein Kinases (MAPKs) have drawn much attention as potential drug targets. This review summarizes unique structural and functional features of MAP kinases and their possible role in pathogenesis of obligate intracellular protozoan parasites namely, Leishmania, Trypanosoma, Plasmodium and Toxoplasma. It also provides an overview of available knowledge concerning the target proteins of parasite MAPKs and the need to understand and unravel unknown interaction network(s) of MAPK(s).
Collapse
Affiliation(s)
- Pavneet Kaur
- Division of Biochemistry and Structural Biology, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, Uttar Pradesh, India
| | - Neena Goyal
- Division of Biochemistry and Structural Biology, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, Uttar Pradesh, India.
| |
Collapse
|
25
|
Singh SK, Singh S, Singh R. Targeting novel coronavirus SARS-CoV-2 spike protein with phytoconstituents of Momordica charantia. J Ovarian Res 2021; 14:126. [PMID: 34579761 PMCID: PMC8474883 DOI: 10.1186/s13048-021-00872-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 08/27/2021] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Infections by the SARS-CoV-2 virus causing COVID-19 are presently a global emergency. The current vaccination effort may reduce the infection rate, but strain variants are emerging under selection pressure. Thus, there is an urgent need to find drugs that treat COVID-19 and save human lives. Hence, in this study, we identified phytoconstituents of an edible vegetable, Bitter melon (Momordica charantia), that affect the SARS-CoV-2 spike protein. METHODS Components of Momordica charantia were tested to identify the compounds that bind to the SARS-CoV-2 spike protein. An MTiOpenScreen web-server was used to perform docking studies. The Lipinski rule was utilized to evaluate potential interactions between the drug and other target molecules. PyMol and Schrodinger software were used to identify the hydrophilic and hydrophobic interactions. Surface plasmon resonance (SPR) was employed to assess the interaction between an extract component (erythrodiol) and the spike protein. RESULTS Our in-silico evaluations showed that phytoconstituents of Momordica charantia have a low binding energy range, -5.82 to -5.97 kcal/mol. A docking study revealed two sets of phytoconstituents that bind at the S1 and S2 domains of SARS-CoV-2. SPR showed that erythrodiol has a strong binding affinity (KD = 1.15 μM) with the S2 spike protein of SARS-CoV-2. Overall, docking, ADME properties, and SPR displayed strong interactions between phytoconstituents and the active site of the SARS-CoV-2 spike protein. CONCLUSION This study reveals that phytoconstituents from bitter melon are potential agents to treat SARS-CoV-2 viral infections due to their binding to spike proteins S1 and S2.
Collapse
Affiliation(s)
- Santosh Kumar Singh
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, 720 Westview Drive SW, Atlanta, GA 30310 USA
- Cancer Health Equity Institute, Morehouse School of Medicine, 720 Westview Drive SW, Atlanta, GA 30310 USA
| | - Shailesh Singh
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, 720 Westview Drive SW, Atlanta, GA 30310 USA
- Cancer Health Equity Institute, Morehouse School of Medicine, 720 Westview Drive SW, Atlanta, GA 30310 USA
| | - Rajesh Singh
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, 720 Westview Drive SW, Atlanta, GA 30310 USA
- Cancer Health Equity Institute, Morehouse School of Medicine, 720 Westview Drive SW, Atlanta, GA 30310 USA
| |
Collapse
|
26
|
Panwar U, Singh SK. In silico virtual screening of potent inhibitor to hamper the interaction between HIV-1 integrase and LEDGF/p75 interaction using E-pharmacophore modeling, molecular docking, and dynamics simulations. Comput Biol Chem 2021; 93:107509. [PMID: 34153658 DOI: 10.1016/j.compbiolchem.2021.107509] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 05/11/2021] [Indexed: 02/07/2023]
Abstract
The rapid increase of HIV-1 infection throughout the globe has a high demand for a superior drug with lesser side effects. LEDGF/p75, the human Lens Epithelium-Derived Growth Factor is identified as a promising cellular cofactor with integrase in facilitating the viral replication in an early stage by acting as a tethering factor in the pre-integration to the chromatin. Therefore, the present study was designed to identify a potent inhibitor by applying an E-pharmacophore based virtual screening, molecular docking, and dynamics simulation approaches. Finally, ZINC22077550 and ZINC32124441 were best identified potent molecules with the efficient binding affinity, strong hydrogen bonding, and acceptable pharmacological properties to hamper the interaction between integrase and LEDGF/p75. Further, the DFT and MDS studies were also analyzed, and shown a favorable energetic state and dynamic stability then reference compound. In conclusion, we suggest that these findings could be novel therapeutics in the future and may increase the lifespan of individuals suffering from viral infection.
Collapse
Affiliation(s)
- Umesh Panwar
- Computer Aided Drug Design and Molecular Modelling Lab, Department of Bioinformatics, Alagappa University, Karaikudi, 630 004, Tamil Nadu, India
| | - Sanjeev Kumar Singh
- Computer Aided Drug Design and Molecular Modelling Lab, Department of Bioinformatics, Alagappa University, Karaikudi, 630 004, Tamil Nadu, India.
| |
Collapse
|
27
|
Qureshi S, Khandelwal R, Madhavi M, Khurana N, Gupta N, Choudhary SK, Suresh RA, Hazarika L, Srija CD, Sharma K, Hindala MR, Hussain T, Nayarisseri A, Singh SK. A Multi-target Drug Designing for BTK, MMP9, Proteasome and TAK1 for the Clinical Treatment of Mantle Cell Lymphoma. Curr Top Med Chem 2021; 21:790-818. [PMID: 33463471 DOI: 10.2174/1568026621666210119112336] [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] [Received: 10/23/2020] [Revised: 12/18/2020] [Accepted: 12/24/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Mantle cell lymphoma (MCL) is a type of non-Hodgkin lymphoma characterized by the mutation and overexpression of the cyclin D1 protein by the reciprocal chromosomal translocation t(11;14)(q13:q32). AIM The present study aims to identify potential inhibition of MMP9, Proteasome, BTK, and TAK1 and determine the most suitable and effective protein target for the MCL. METHODOLOGY Nine known inhibitors for MMP9, 24 for proteasome, 15 for BTK and 14 for TAK1 were screened. SB-3CT (PubChem ID: 9883002), oprozomib (PubChem ID: 25067547), zanubrutinib (PubChem ID: 135565884) and TAK1 inhibitor (PubChem ID: 66760355) were recognized as drugs with high binding capacity with their respective protein receptors. 41, 72, 102 and 3 virtual screened compounds were obtained after the similarity search with compound (PubChem ID:102173753), PubChem compound SCHEMBL15569297 (PubChem ID:72374403), PubChem compound SCHEMBL17075298 (PubChem ID:136970120) and compound CID: 71814473 with best virtual screened compounds. RESULT MMP9 inhibitors show commendable affinity and good interaction profile of compound holding PubChem ID:102173753 over the most effective established inhibitor SB-3CT. The pharmacophore study of the best virtual screened compound reveals its high efficacy based on various interactions. The virtual screened compound's better affinity with the target MMP9 protein was deduced using toxicity and integration profile studies. CONCLUSION Based on the ADMET profile, the compound (PubChem ID: 102173753) could be a potent drug for MCL treatment. Similar to the established SB-3CT, the compound was non-toxic with LD50 values for both the compounds lying in the same range.
Collapse
Affiliation(s)
- Shahrukh Qureshi
- In silico Research Laboratory, Eminent Biosciences, Mahalakshmi Nagar, Indore - 452010, Madhya Pradesh, India
| | - Ravina Khandelwal
- In silico Research Laboratory, Eminent Biosciences, Mahalakshmi Nagar, Indore - 452010, Madhya Pradesh, India
| | - Maddala Madhavi
- Department of Zoology, Nizam College, Osmania University, Hyderabad - 500001, Telangana State, India
| | - Naveesha Khurana
- In silico Research Laboratory, Eminent Biosciences, Mahalakshmi Nagar, Indore - 452010, Madhya Pradesh, India
| | - Neha Gupta
- In silico Research Laboratory, Eminent Biosciences, Mahalakshmi Nagar, Indore - 452010, Madhya Pradesh, India
| | - Saurav K Choudhary
- In silico Research Laboratory, Eminent Biosciences, Mahalakshmi Nagar, Indore - 452010, Madhya Pradesh, India
| | - Revathy A Suresh
- In silico Research Laboratory, Eminent Biosciences, Mahalakshmi Nagar, Indore - 452010, Madhya Pradesh, India
| | - Lima Hazarika
- In silico Research Laboratory, Eminent Biosciences, Mahalakshmi Nagar, Indore - 452010, Madhya Pradesh, India
| | - Chillamcherla D Srija
- In silico Research Laboratory, Eminent Biosciences, Mahalakshmi Nagar, Indore - 452010, Madhya Pradesh, India
| | - Khushboo Sharma
- In silico Research Laboratory, Eminent Biosciences, Mahalakshmi Nagar, Indore - 452010, Madhya Pradesh, India
| | - Mali R Hindala
- In silico Research Laboratory, Eminent Biosciences, Mahalakshmi Nagar, Indore - 452010, Madhya Pradesh, India
| | - Tajamul Hussain
- Center of Excellence in Biotechnology Research, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Anuraj Nayarisseri
- In silico Research Laboratory, Eminent Biosciences, Mahalakshmi Nagar, Indore - 452010, Madhya Pradesh, India
| | - Sanjeev K Singh
- Computer Aided Drug Designing and Molecular Modeling Lab, Department of Bioinformatics, Alagappa University, Karaikudi-630 003, Tamil Nadu, India
| |
Collapse
|
28
|
Efstathiou A, Smirlis D. Leishmania Protein Kinases: Important Regulators of the Parasite Life Cycle and Molecular Targets for Treating Leishmaniasis. Microorganisms 2021; 9:microorganisms9040691. [PMID: 33801655 PMCID: PMC8066228 DOI: 10.3390/microorganisms9040691] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 03/21/2021] [Accepted: 03/24/2021] [Indexed: 02/07/2023] Open
Abstract
Leishmania is a protozoan parasite of the trypanosomatid family, causing a wide range of diseases with different clinical manifestations including cutaneous, mucocutaneous and visceral leishmaniasis. According to WHO, one billion people are at risk of Leishmania infection as they live in endemic areas while there are 12 million infected people worldwide. Annually, 0.9-1.6 million new infections are reported and 20-50 thousand deaths occur due to Leishmania infection. As current chemotherapy for treating leishmaniasis exhibits numerous drawbacks and due to the lack of effective human vaccine, there is an urgent need to develop new antileishmanial therapy treatment. To this end, eukaryotic protein kinases can be ideal target candidates for rational drug design against leishmaniasis. Eukaryotic protein kinases mediate signal transduction through protein phosphorylation and their inhibition is anticipated to be disease modifying as they regulate all essential processes for Leishmania viability and completion of the parasitic life cycle including cell-cycle progression, differentiation and virulence. This review highlights existing knowledge concerning the exploitation of Leishmania protein kinases as molecular targets to treat leishmaniasis and the current knowledge of their role in the biology of Leishmania spp. and in the regulation of signalling events that promote parasite survival in the insect vector or the mammalian host.
Collapse
|
29
|
Pande M, Kundu D, Srivastava R. Vitamin C and Vitamin D3 show strong binding with the amyloidogenic region of G555F mutant of Fibrinogen A alpha-chain associated with renal amyloidosis: proposed possible therapeutic intervention. Mol Divers 2021; 26:939-949. [PMID: 33710477 DOI: 10.1007/s11030-021-10205-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 02/22/2021] [Indexed: 12/14/2022]
Abstract
G555F mutant of Fibrinogen A alpha-chain (FGA) is reported to be associated with kidney amyloidosis. In the current study, we have modelled the G555F mutant and examined the mutation's effect on the structural and functional level. We have also docked Vitamin C and D3 on the mutant's amyloidogenic region to identify if these vitamins can bind amyloidogenic regions. Further, we analyzed if they could prevent or modulate amyloid formation by stopping critical interactions in amyloidogenic regions in FGA. We used the wild type FGA model protein as a control. Our docking and molecular dynamics simulation results indicate stronger Vitamin D3 binding than Vitamin C to the amyloidogenic region of the mutant protein. The RMSD, radius of gyration, and RMSF values were higher for the G555F mutant than the FGA wild type protein. Overall, the results support these vitamins' potential as a therapeutic and anti-amyloidogenic agent for FGA renal amyloidosis.
Collapse
Affiliation(s)
- Monu Pande
- Department of Biochemistry, Institute of Medical Science, Banaras Hindu University, Varanasi, 221005, India
| | - Debanjan Kundu
- School of Biochemical Engineering, Indian Institute of Technology (BHU), Varanasi, 221005, India
| | - Ragini Srivastava
- Department of Biochemistry, Institute of Medical Science, Banaras Hindu University, Varanasi, 221005, India.
| |
Collapse
|
30
|
Nath P, Goyal A. Structure and dynamics analysis of multi-domain putative β-1,4-glucosidase of family 3 glycoside hydrolase (PsGH3) from Pseudopedobacter saltans. J Mol Model 2021; 27:106. [PMID: 33694107 PMCID: PMC7945971 DOI: 10.1007/s00894-021-04721-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 03/01/2021] [Indexed: 11/30/2022]
Abstract
Structure and conformational behaviour of a putative β-1,4-glucosidase of glycoside hydrolase family 3 (PsGH3) from Pseudopedobacter saltans was predicted by using in-silico tools. PsGH3 modeled structure constructed using Phyre2 displayed multidomain architecture comprising an N-terminal (β/α)8-fold domain followed by (α/β)6-sandwich domain, PA14 domain, and a C-terminal domain resembling an immunoglobulin fold. Ramachandran plot displayed 99.3% of amino acids in the allowed region and 0.7% residues in the disallowed region. Multiple sequence alignment (MSA) and structure superposition of PsGH3 with other homologues from GH3 family revealed the conserved residues, Asp274 and Glu624 present in loops LA and LB, respectively originating from N-terminal domain act as catalytic residues. The volume and area calculated for PsGH3 displayed a deep active-site conformation comparable with its homologues, β-1,4-glucosidases (GH3) of Kluyveromyces marxianus and Streptomyces venezuelae. Molecular dynamic (MD) simulation of PsGH3 structure for 80 ns suggested stable and compact structure. Molecular docking studies revealed deeper active site conformation of PsGH3 that could house larger cellooligosaccharides up to 7° of polymerization (DP7). The amino acid residues, Ala86, Leu88, Cys275, Pro483, Phe493, Asn417, Asn491, Pro492, and Leu495 created a binding pocket near the catalytic cleft, crucial for ligand binding. MD simulation of PsGH3 in the presence of cellooligosaccharides, viz., cellobiose and celloheptaose showed stability in terms of RMSD, Rg, and SASA values till 80 ns. The calculation of average number of hydrogen bond (H-bond), interaction energy, and binding free energy confirmed the stronger binding affinity of the larger cellooligosaccharides such as celloheptaose in the binding cavity of PsGH3.
Collapse
Affiliation(s)
- Priyanka Nath
- Carbohydrate Enzyme Biotechnology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India
| | - Arun Goyal
- Carbohydrate Enzyme Biotechnology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India.
| |
Collapse
|
31
|
Rahman F, Tabrez S, Ali R, Alqahtani AS, Ahmed MZ, Rub A. Molecular docking analysis of rutin reveals possible inhibition of SARS-CoV-2 vital proteins. J Tradit Complement Med 2021; 11:173-179. [PMID: 33520682 PMCID: PMC7825826 DOI: 10.1016/j.jtcme.2021.01.006] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 01/12/2021] [Accepted: 01/16/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND AND AIM COVID-19 emerged by the end of 2019 in Wuhan, China. It spreaded and became a public health emergency all over the world by mid of April 2020. Flavonoids are specialized metabolites that have antimicrobial properties including anti-viral activity. Rutin, a medicinally important flavonoid belongs to one of the best natural antioxidant classes. It has antiprotozoal, antibacterial, and antiviral properties. Keeping the antimicrobial potential of rutin in mind, we studied its role in the inhibition of essential proteins of SARS-CoV-2 including main protease (Mpro), RNA-dependent RNA polymerase (RdRp), papain-like protease (PLpro), and spike (S)-protein through different in silico approaches. EXPERIMENTAL PROCEDURE Molecular docking, inhibition constant, hydrogen bond calculations, and ADMET-properties prediction were performed using different softwares. RESULTS AND CONCLUSION Molecular docking study showed significant binding of rutin with Mpro, RdRp, PLpro, and S-proteins of SARS-CoV-2. Out of these four proteins, Mpro exhibited the strongest binding affinity with the least binding energy (-8.9 kcal/mol) and stabilized through hydrogen bonds with bond lengths ranging from 1.18 Å to 3.17 Å as well as hydrophobic interactions. The predicted ADMET and bioactivity showed its optimal solubility, non-toxic, and non-carcinogenic properties. The values of the predicted inhibitory constant of the rutin with SARS-CoV-2 vital proteins ranged between 5.66 μM and 6.54 μM which suggested its promising drug candidature. This study suggested rutin alone or in combination as a dietary supplement may be used to fight against COVID-19 after detailed in vitro and in vivo studies.
Collapse
Affiliation(s)
- Fazlur Rahman
- Infection and Immunity Lab (414), Department of Biotechnology, Jamia Millia Islamia (A Central University), New Delhi, 110025, India
| | - Shams Tabrez
- Infection and Immunity Lab (414), Department of Biotechnology, Jamia Millia Islamia (A Central University), New Delhi, 110025, India
| | - Rahat Ali
- Infection and Immunity Lab (414), Department of Biotechnology, Jamia Millia Islamia (A Central University), New Delhi, 110025, India
| | - Ali S. Alqahtani
- King Saud University College of Pharmacy, Department of Pharmacognosy, Riyadh, 11451, Saudi Arabia
| | - Mohammad Z. Ahmed
- King Saud University College of Pharmacy, Department of Pharmacognosy, Riyadh, 11451, Saudi Arabia
| | - Abdur Rub
- Infection and Immunity Lab (414), Department of Biotechnology, Jamia Millia Islamia (A Central University), New Delhi, 110025, India
| |
Collapse
|
32
|
Alıcı H, Demir K. Investigation of the stability and the helix-tail interaction of sCT and its various charged mutants based on comparative molecular dynamics simulations. Chem Phys 2021. [DOI: 10.1016/j.chemphys.2020.111057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
33
|
Tabrez S, Rahman F, Ali R, Alouffi AS, Alshehri BM, Alshammari FA, Alaidarous MA, Banawas S, Bin Dukhyil AA, Rub A. Assessment of the Antileishmanial Potential of Cassia fistula Leaf Extract. ACS OMEGA 2021; 6:2318-2327. [PMID: 33521470 PMCID: PMC7841934 DOI: 10.1021/acsomega.0c05629] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 12/28/2020] [Indexed: 05/14/2023]
Abstract
Cassia fistula has a wide array of biologically active and therapeutically important class of compounds. Leishmania donovani important drug targets, sterol 24-c methyltransferase (LdSMT), trypanothione reductase (LdTR), pteridine reductase (LdPTR1), and nucleoside hydrolase (LdNH), were modelled, and molecular docking was performed against the abundant phytochemicals of its leaf extract. Molecular docking results provided the significant prima facie evidence of the leaf extract to have antileishmanial potential. To confirm this, we performed in vitro antileishmanial and cytotoxicity assays. Methanolic extract of C. fistula leaves showed growth inhibition and proliferation of L. donovani promastigote with an IC50 value of 43.31 ± 4.202 μg/mL. It also inhibited the growth of intra-macrophagic amastigotes with an IC50 value of 80.76 ± 3.626 μg/mL. C. fistula extract was found cytotoxic at a very high concentration on human macrophages (CC50 = 626 ± 39 μg/mL). Annexin V/propidium iodide (PI) staining assay suggested partial apoptosis induction in parasites by C. fistula to exert its antileishmanial activity. Here, for the first time, we have shown the antileishmanial potential of C. fistula leaves. Overall, our results could open new insight for an affordable and natural antileishmanial with high efficacy and less toxicity.
Collapse
Affiliation(s)
- Shams Tabrez
- Infection
and Immunity Lab (414), Department of Biotechnology, Jamia Millia Islamia (A Central University), New Delhi 110025, India
| | - Fazlur Rahman
- Infection
and Immunity Lab (414), Department of Biotechnology, Jamia Millia Islamia (A Central University), New Delhi 110025, India
| | - Rahat Ali
- Infection
and Immunity Lab (414), Department of Biotechnology, Jamia Millia Islamia (A Central University), New Delhi 110025, India
| | - Abdulaziz S. Alouffi
- King
Abdulaziz City for Science and Technology, P.O. Box 6086, Riyadh 11442, Saudi Arabia
| | - Bader Mohammed Alshehri
- Department
of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Al Majmaah 11952, Saudi Arabia
| | - Fahdah Ayed Alshammari
- College
of Sciences and Literature Microbiology, Northern Border University, P.O. Box 1321, Arar 73222, Saudi Arabia
| | - Mohammed A. Alaidarous
- Department
of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Al Majmaah 11952, Saudi Arabia
- Health
and Basic Sciences Research Center, Majmaah
University, Al Majmaah 15341, Saudi Arabia
| | - Saeed Banawas
- Department
of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Al Majmaah 11952, Saudi Arabia
- Health
and Basic Sciences Research Center, Majmaah
University, Al Majmaah 15341, Saudi Arabia
- Department
of Biomedical Sciences, Oregon State University, Corvallis, Oregon 97331, United States
| | - Abdul Aziz Bin Dukhyil
- Department
of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Al Majmaah 11952, Saudi Arabia
- Health
and Basic Sciences Research Center, Majmaah
University, Al Majmaah 15341, Saudi Arabia
| | - Abdur Rub
- Infection
and Immunity Lab (414), Department of Biotechnology, Jamia Millia Islamia (A Central University), New Delhi 110025, India
| |
Collapse
|
34
|
Cynaroside inhibits Leishmania donovani UDP-galactopyranose mutase and induces reactive oxygen species to exert antileishmanial response. Biosci Rep 2021; 41:227423. [PMID: 33367614 PMCID: PMC7805024 DOI: 10.1042/bsr20203857] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 12/11/2020] [Accepted: 12/18/2020] [Indexed: 12/23/2022] Open
Abstract
Cynaroside, a flavonoid, has been shown to have antibacterial, antifungal and anticancer activities. Here, we evaluated its antileishmanial properties and its mechanism of action through different in silico and in vitro assays. Cynaroside exhibited antileishmanial activity in time- and dose-dependent manner with 50% of inhibitory concentration (IC50) value of 49.49 ± 3.515 µM in vitro. It inhibited the growth of parasite significantly at only 20 µM concentration when used in combination with miltefosine, a standard drug which has very high toxicity. It also inhibited the intra-macrophagic parasite significantly at low doses when used in combination with miltefosine. It showed less toxicity than the existing antileishmanial drug, miltefosine at similar doses. Propidium iodide staining showed that cynaroside inhibited the parasites in G0/G1 phase of cell cycle. 2,7-dichloro dihydro fluorescein diacetate (H2DCFDA) staining showed cynaroside induced antileishmanial activity through reactive oxygen species (ROS) generation in parasites. Molecular-docking studies with key drug targets of Leishmania donovani showed significant inhibition. Out of these targets, cynaroside showed strongest affinity with uridine diphosphate (UDP)-galactopyranose mutase with −10.4 kcal/mol which was further validated by molecular dynamics (MD) simulation. The bioactivity, ADMET (absorption, distribution, metabolism, excretion and toxicity) properties, Organisation for Economic Co-operation and Development (OECD) chemical classification and toxicity risk prediction showed cynaroside as an enzyme inhibitor having sufficient solubility and non-toxic properties. In conclusion, cynaroside may be used alone or in combination with existing drug, miltefosine to control leishmaniasis with less cytotoxicity.
Collapse
|
35
|
Alıcı H. In silico analysis: structural insights about inter-protofilaments interactions for α-synuclein (50–57) fibrils and its familial mutation. MOLECULAR SIMULATION 2020. [DOI: 10.1080/08927022.2020.1786084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Affiliation(s)
- Hakan Alıcı
- Department of Physics, Zonguldak Bülent Ecevit University, Zonguldak, Turkey
| |
Collapse
|
36
|
Pandey P, Rane JS, Chatterjee A, Kumar A, Khan R, Prakash A, Ray S. Targeting SARS-CoV-2 spike protein of COVID-19 with naturally occurring phytochemicals: an in silico study for drug development. J Biomol Struct Dyn 2020; 39:6306-6316. [PMID: 32698689 PMCID: PMC7441770 DOI: 10.1080/07391102.2020.1796811] [Citation(s) in RCA: 144] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Spike glycoprotein, a class I fusion protein harboring the surface of SARS-CoV-2
(SARS-CoV-2S), plays a seminal role in the viral infection starting from recognition of
the host cell surface receptor, attachment to the fusion of the viral envelope with the
host cells. Spike glycoprotein engages host Angiotensin-converting enzyme 2 (ACE2)
receptors for entry into host cells, where the receptor recognition and attachment of
spike glycoprotein to the ACE2 receptors is a prerequisite step and key determinant of the
host cell and tissue tropism. Binding of spike glycoprotein to the ACE2 receptor triggers
a cascade of structural transitions, including transition from a metastable pre-fusion to
a post-fusion form, thereby allowing membrane fusion and internalization of the virus.
From ancient times people have relied on naturally occurring substances like
phytochemicals to fight against diseases and infection. Among these phytochemicals,
flavonoids and non-flavonoids have been the active sources of different anti-microbial
agents. We performed molecular docking studies using 10 potential naturally occurring
compounds (flavonoids/non-flavonoids) against the SARS-CoV-2 spike protein and compared
their affinity with an FDA approved repurposed drug hydroxychloroquine (HCQ). Further, our
molecular dynamics (MD) simulation and energy landscape studies with fisetin, quercetin,
and kamferol revealed that these molecules bind with the hACE2-S complex with low binding
free energy. The study provided an indication that these molecules might have the
potential to perturb the binding of hACE2-S complex. In addition, ADME analysis also
suggested that these molecules consist of drug-likeness property, which may be further
explored as anti-SARS-CoV-2 agents. Communicated by Ramaswamy H. Sarma
Collapse
Affiliation(s)
- Preeti Pandey
- Department of Chemistry & Biochemistry, University of Oklahoma, OK, USA
| | - Jitendra Subhash Rane
- Department of Biosciences & Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Aroni Chatterjee
- Indian Council of Medical Research (ICMR)-Virus Research Laboratory, NICED, Kolkata, India
| | - Abhijeet Kumar
- Department of Chemistry, Mahatma Gandhi Central University, Motihari, India
| | - Rajni Khan
- Motihari College of Engineering, Motihari, India
| | - Amresh Prakash
- Amity Institute of Integrative Sciences and Health, Amity University Haryana, Gurgaon, India
| | - Shashikant Ray
- Department of Biotechnology, Mahatma Gandhi Central University, Motihari, India
| |
Collapse
|
37
|
Computational design of a potential multi-epitope subunit vaccine using immunoinformatics to fight Ebola virus. INFECTION GENETICS AND EVOLUTION 2020; 85:104464. [PMID: 32681997 DOI: 10.1016/j.meegid.2020.104464] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 06/23/2020] [Accepted: 07/12/2020] [Indexed: 01/11/2023]
Abstract
Ebola virus (EBOV) is a rare but fatal disease that has been a burden to mankind for over 40 years. EBOV exhibits several symptoms including severe bleeding, organ failure and if left untreated causes death. It is assumed that fruit bats of the Pteropodidae family are natural hosts for the virus. Over the years, there has been no effective vaccine that can confer immunity to this virus. Considering the necessity of a vaccine against EBOV, this study to develop a multi-epitope subunit vaccine for the EBOV using the immunoinformatics approach was conducted. The construct was designed using structural and non-structural proteins of EBOV. Class I and Class II MHC epitopes were predicted and linked along with β defensin and compatible linkers. B-cell linear epitopes were also assessed and the physiological parameters of the vaccine were determined. The vaccine was capable of administration to humans and also is capable of an immune response. The vaccine was modeled further and affinity towards the TLR4 receptor was studied by docking and simulation for 20 ns. The trajectory analysis high affinity between the vaccine and the construct with an average hydrogen bond of 18. For ease of purification, the vaccine construct was ligated into pET28a(+) vector with His-tag. Concluding from the results, the vaccine construct has the potentiality to help develop immunity against the Ebola virus. Furthermore, experimental and immunological investigations will be required to verify the feasibility of the multi-epitope subunit construct as a commercial vaccine.
Collapse
|
38
|
Althobiti HA, Zabin SA. New Schiff bases of 2-(quinolin-8-yloxy)acetohydrazide and their Cu(ii), and Zn(ii) metal complexes: their in vitro antimicrobial potentials and in silico physicochemical and pharmacokinetics properties. OPEN CHEM 2020. [DOI: 10.1515/chem-2020-0085] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
AbstractThe purpose of this work was to prepare Schiff base ligands containing quinoline moiety and using them for preparing Cu(ii) and Zn(ii) complexes. Four bidentate Schiff base ligands (SL1–SL4) with quinoline hydrazine scaffold and a series of mononuclear Cu(ii) and Zn(ii) complexes were successfully prepared and characterized. The in vitro antibacterial and antifungal potential experimentation revealed that the ligands exhibited moderate antibacterial activity against the Gram-positive bacterial types and were inactive against the Gram-negative bacteria and the fungus strains. The metal complexes showed some enhancement in the activity against the Gram-positive bacterial strains and were inactive against the Gram-negative bacteria and the fungus strains similar to the parent ligands. The complex [Cu(SL1)2] was the most toxic compound against both Gram-positive S. aureus and E. faecalis bacteria. The in silico physicochemical investigation revealed that the ligand SL4 showed highest in silico absorption (82.61%) and the two complexes [Cu(SL4)2] and [Zn(SL4)2] showed highest in silico absorption with 56.23% for both compounds. The in silico pharmacokinetics predictions showed that the ligands have high gastrointestinal (GI) absorption and the complexes showed low GI absorption. The ligands showed a good bioavailability score of 0.55 where the complexes showed moderate to poor bioavailability.
Collapse
Affiliation(s)
- Hanan A. Althobiti
- Chemistry Department, Faculty of Science, Albaha University, Al-Baha, Saudi Arabia
| | - Sami A. Zabin
- Chemistry Department, Faculty of Science, Albaha University, Al-Baha, Saudi Arabia
| |
Collapse
|
39
|
Raj S, Sasidharan S, Balaji SN, Saudagar P. An overview of biochemically characterized drug targets in metabolic pathways of Leishmania parasite. Parasitol Res 2020; 119:2025-2037. [DOI: 10.1007/s00436-020-06736-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 05/28/2020] [Indexed: 12/18/2022]
|
40
|
Raj S, Saha G, Sasidharan S, Dubey VK, Saudagar P. Biochemical characterization and chemical validation of Leishmania MAP Kinase-3 as a potential drug target. Sci Rep 2019; 9:16209. [PMID: 31700105 PMCID: PMC6838069 DOI: 10.1038/s41598-019-52774-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 10/22/2019] [Indexed: 12/16/2022] Open
Abstract
Protozoan parasites of the Leishmania genus have evolved unique signaling pathways that can sense various environmental changes and trigger stage differentiation for survival and host infectivity. MAP kinase (MAPK) plays a critical role in various cellular activities like cell differentiation, proliferation, stress regulation, and apoptosis. The Leishmania donovani MAPK3 (LdMAPK3) is involved in the regulation of flagella length and hence plays an important role in disease transmission. Here, we reported the gene cloning, protein expression, biochemical characterizations, inhibition studies and cell proliferation assay of LdMAPK3. The recombinant purified LdMAPK3 enzyme obeys the Michaelis-Menten equation with Km and Vmax of LdMAPK3 was found to be 20.23 nM and 38.77 ± 0.71 nmoles ATP consumed/mg LdMAPK3/min respectively. The maximum kinase activity of LdMAPK3 was recorded at 35 °C and pH 7. The in-vitro inhibition studies with two natural inhibitors genistein (GEN) and chrysin (CHY) was evaluated against LdMAPK3. The Ki value for GEN and CHY were found to be 3.76 ± 0.28 µM and Ki = 8.75 ± 0.11 µM respectively. The IC50 value for the compounds, GEN and CHY against L. donovani promastigotes were calculated as 9.9 µg/mL and 13 µg/mL respectively. Our study, therefore, reports LdMAPK3 as a new target for therapeutic approach against leishmaniasis.
Collapse
Affiliation(s)
- Shweta Raj
- Department of Biotechnology, National Institute of Technology-Warangal, Telangana State, 506004, India
| | - Gundappa Saha
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India
| | - Santanu Sasidharan
- Department of Biotechnology, National Institute of Technology-Warangal, Telangana State, 506004, India
| | - Vikash Kumar Dubey
- School of Biochemical Engineering, Indian Institute of Technology-Banaras Hindu University, Uttar Pradesh, 221005, India
| | - Prakash Saudagar
- Department of Biotechnology, National Institute of Technology-Warangal, Telangana State, 506004, India.
| |
Collapse
|