1
|
Uppathi P, Rajakumari S, Saritha KV. Molecular Docking: An Emerging Tool for Target-Based Cancer Therapy. Crit Rev Oncog 2025; 30:1-13. [PMID: 39819431 DOI: 10.1615/critrevoncog.2024056533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
Molecular docking is a structure-based computational technique that plays a major role in drug discovery. Molecular docking enhances the efficacy of determining the metabolic interaction between two molecules, i.e., the small molecule (ligand) and the target molecule (protein), to find the best orientation of a ligand to its target molecule with minimal free energy in forming a stable complex. By stimulating drug-target interactions, docking helps identify small molecules that might inhibit cancer-promoting proteins, aiding in the development of novel targeted therapies. Molecular docking enables researchers to screen vast reorganization, identifying potential anti-cancer drugs with enhanced specificity and reduced toxicity. The growing importance of molecular docking underscores its potential to revolutionize cancer treatment by accelerating the identification of novel drugs and improving clinical outcomes. As a wide approach, this computational drug design technique can be considered more effective and timesaving than other cancer treatment methods. In this review, we showcase brief information on the role of molecular docking and its importance in cancer research for drug discovery and target identification. Therefore, in recent years, it can be concluded that molecular docking can be scrutinized as one of the novel strategies at the leading edge of cancer-targeting drug discovery.
Collapse
Affiliation(s)
| | - Suraj Rajakumari
- Department of Biotechnology, Sri Venkateswara University, Tirupati, AP-517502 India
| | | |
Collapse
|
2
|
Dhlamini KS, Selepe CT, Ramalapa B, Cele Z, Malatji K, Govender KK, Tshweu L, Ray SS. Dual Antimicrobial Activity of HTCC and Its Nanoparticles: A Synergistic Approach for Antibacterial and Antiviral Applications Through Combined In Silico and In Vitro Studies. Polymers (Basel) 2024; 16:2999. [PMID: 39518210 PMCID: PMC11548688 DOI: 10.3390/polym16212999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 10/12/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
N-(2-hydroxyl) propyl-3-trimethyl ammonium chitosan chloride (HTCC), a quaternized chitosan derivative, has been shown to exhibit a broad spectrum of antimicrobial activity, especially against bacteria and enveloped viruses. Despite this, molecular docking studies showing its atomic-level mechanisms against these microorganisms are scarce. Here, for the first time, we employed molecular docking analyses to investigate the potential antibacterial activity of HTCC against Staphylococcus aureus and its antiviral activity against human immunodeficiency virus 1 (HIV-1). According to the findings, HTCC exhibited promising antibacterial activity with high binding affinities; however, it had limited antiviral activity. To validate these theoretical outcomes, experimental studies were conducted. Different derivatives of HTCC were synthesized and characterized using NMR, XRD, FTIR, and DLS. The in vitro assays validated the potent antibacterial efficacy of HTCC against S. aureus, whereas the antiviral studies did not show good antiviral activity. However, our research also revealed a promising avenue for further exploration of the antimicrobial activity of HTCC nanoparticles (NPs), since, thus far, no studies have been conducted to show the antiviral activity of HTCC NPs against HIV-1. The nanosized HTCC exhibited superior antiviral performance compared to the parent polymers, with complete (100%) inhibition of HIV-1 viral activity at the highest tested concentration (0.33 mg/mL).
Collapse
Affiliation(s)
- Khanyisile S. Dhlamini
- Centre for Nanostructures and Advanced Materials, DSI-CSIR Nanotechnology Innovation Centre, Council for Scientific and Industrial Research, Pretoria 0001, South Africa; (K.S.D.); (C.T.S.); (B.R.); (Z.C.); (K.M.)
- Department of Chemical Sciences, University of Johannesburg, Doornfontein 2028, Johannesburg, South Africa;
| | - Cyril T. Selepe
- Centre for Nanostructures and Advanced Materials, DSI-CSIR Nanotechnology Innovation Centre, Council for Scientific and Industrial Research, Pretoria 0001, South Africa; (K.S.D.); (C.T.S.); (B.R.); (Z.C.); (K.M.)
- Department of Chemical Sciences, University of Johannesburg, Doornfontein 2028, Johannesburg, South Africa;
| | - Bathabile Ramalapa
- Centre for Nanostructures and Advanced Materials, DSI-CSIR Nanotechnology Innovation Centre, Council for Scientific and Industrial Research, Pretoria 0001, South Africa; (K.S.D.); (C.T.S.); (B.R.); (Z.C.); (K.M.)
- Material Science, Innovation and Modelling (MaSIM), Faculty of Natural and Agricultural Sciences, North-West University, Mmabatho 2735, South Africa
| | - Zamani Cele
- Centre for Nanostructures and Advanced Materials, DSI-CSIR Nanotechnology Innovation Centre, Council for Scientific and Industrial Research, Pretoria 0001, South Africa; (K.S.D.); (C.T.S.); (B.R.); (Z.C.); (K.M.)
| | - Kanyane Malatji
- Centre for Nanostructures and Advanced Materials, DSI-CSIR Nanotechnology Innovation Centre, Council for Scientific and Industrial Research, Pretoria 0001, South Africa; (K.S.D.); (C.T.S.); (B.R.); (Z.C.); (K.M.)
| | - Krishna K. Govender
- Department of Chemical Sciences, University of Johannesburg, Doornfontein 2028, Johannesburg, South Africa;
| | - Lesego Tshweu
- Centre for Nanostructures and Advanced Materials, DSI-CSIR Nanotechnology Innovation Centre, Council for Scientific and Industrial Research, Pretoria 0001, South Africa; (K.S.D.); (C.T.S.); (B.R.); (Z.C.); (K.M.)
- Material Science, Innovation and Modelling (MaSIM), Faculty of Natural and Agricultural Sciences, North-West University, Mmabatho 2735, South Africa
| | - Suprakas Sinha Ray
- Centre for Nanostructures and Advanced Materials, DSI-CSIR Nanotechnology Innovation Centre, Council for Scientific and Industrial Research, Pretoria 0001, South Africa; (K.S.D.); (C.T.S.); (B.R.); (Z.C.); (K.M.)
- Department of Chemical Sciences, University of Johannesburg, Doornfontein 2028, Johannesburg, South Africa;
| |
Collapse
|
3
|
Zeng J, Deng Q, Chen Z, Yan S, Dong Q, Zhang Y, Cui Y, Li L, He Y, Shi J. Recent development of VEGFR small molecule inhibitors as anticancer agents: A patent review (2021-2023). Bioorg Chem 2024; 146:107278. [PMID: 38484586 DOI: 10.1016/j.bioorg.2024.107278] [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: 12/12/2023] [Revised: 01/15/2024] [Accepted: 03/08/2024] [Indexed: 04/13/2024]
Abstract
VEGFR, a receptor tyrosine kinase inhibitor (TKI), is an important regulatory factor that promotes angiogenesis and vascular permeability. It plays a significant role in processes such as tumor angiogenesis, tumor cell invasion, and metastasis. VEGFR is mainly composed of three subtypes: VEGFR-1, VEGFR-2, and VEGFR-3. Among them, VEGFR-2 is the crucial signaling receptor for VEGF, which is involved in various pathological and physiological functions. At present, VEGFR-2 is closely related to a variety of cancers, such as non-small cell lung cancer (NSCLC), Hepatocellular carcinoma, Renal cell carcinoma, breast cancer, gastric cancer, glioma, etc. Consequently, VEGFR-2 serves as a crucial target for various cancer treatments. An increasing number of VEGFR inhibitors have been discovered to treat cancer, and they have achieved tremendous success in the clinic. Nevertheless, VEGFR inhibitors often exhibit severe cytotoxicity, resistance, and limitations in indications, which weaken the clinical therapeutic effect. In recent years, many small molecule inhibitors targeting VEGFR have been identified with anti-drug resistance, lower cytotoxicity, and better affinity. Here, we provide an overview of the structure and physiological functions of VEGFR, as well as some VEGFR inhibitors currently in clinical use. Also, we summarize the in vivo and in vitro activities, selectivity, structure-activity relationship, and therapeutic or preventive use of VEGFR small molecule inhibitors reported in patents in the past three years (2021-2023), thereby presenting the prospects and insights for the future development of targeted VEGFR inhibitors.
Collapse
Affiliation(s)
- Jing Zeng
- School of Food and Bioengineering, Xihua University, Chengdu, Sichuan 610039, China
| | - Qichuan Deng
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Zheng Chen
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Shuang Yan
- Sichuan University of Arts and Science, DaZhou 635000, China
| | - Qin Dong
- School of Food and Bioengineering, Xihua University, Chengdu, Sichuan 610039, China
| | - Yuyu Zhang
- School of Food and Bioengineering, Xihua University, Chengdu, Sichuan 610039, China
| | - Yuan Cui
- School of Food and Bioengineering, Xihua University, Chengdu, Sichuan 610039, China
| | - Ling Li
- School of Food and Bioengineering, Xihua University, Chengdu, Sichuan 610039, China; Chengdu University of Traditional Chinese Medicine State Key Laboratory of Southwestern Chinese Medicine Resources, Sichuan 611137, China.
| | - Yuxin He
- School of Food and Bioengineering, Xihua University, Chengdu, Sichuan 610039, China.
| | - Jianyou Shi
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China.
| |
Collapse
|
4
|
Abdalla Ali A, Mhamad SA, Hasan AH, Ahmad I, Abdullah SA, Jamil S, Patel H, Murugesan S, Jamalis J. Synthesis, biological evaluation and molecular modeling studies of modulated benzyloxychalcones as potential acetylcholinesterase inhibitors. J Biomol Struct Dyn 2024; 42:3604-3615. [PMID: 37293930 DOI: 10.1080/07391102.2023.2220032] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 05/09/2023] [Indexed: 06/10/2023]
Abstract
Acetylcholinesterase inhibitors (AChEIs) have become a significant target in the search for an efficient treatment of Alzheimer's disease. Chalcone-based compounds display a strong potency to hinder AChE. So, this study focused on the synthesis of a series of new chalcone derivatives with anti-cholinesterase potential and their structures were characterized based on spectroscopic methods including IR, 1H NMR, 13C NMR and HRMS. Chalcone derivatives were screened against AChE. Most of them exhibited potent inhibitory activity against AChE. Compound 11i showed the most potent activity toward acetylcholinesterase compared to the positive compound, Galantamine. Docking studies into the active site of the acetylcholinesterase enzyme ravealed the significant docking score of the synthesized compounds with docking score of -7.959 to -9.277 kcal/mol when compared to the co-crystallized ligand, Donepezil (-10.567 kcal/mol). The interaction's stability was further assessed using a conventional atomistic 100 ns dynamics simulation study, which revealed the conformational stability of representative compound 11i in the cavity of the acetylcholinesterase enzyme.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Arman Abdalla Ali
- Department of Chemistry, Faculty of Science, Universiti Teknologi Malaysia, Johor Bahru, Johor, Malaysia
- Tafan Preparatory School, General Directorate of Education of Sulaimani, Sulaimani, Kurdistan, Iraq
| | - Shakhawan Ahmad Mhamad
- Department of Chemistry, Faculty of Science, Universiti Teknologi Malaysia, Johor Bahru, Johor, Malaysia
- Department of Chemistry, College of Education, University of Sulaimani, Sulaimani, Kurdistan, Iraq
| | - Aso Hameed Hasan
- Department of Chemistry, Faculty of Science, Universiti Teknologi Malaysia, Johor Bahru, Johor, Malaysia
- Department of Chemistry, College of Science, University of Garmian, Kalar, Kurdistan, Iraq
| | - Iqrar Ahmad
- Department of Pharmaceutical Chemistry, Prof. Ravindra Nikam College of Pharmacy, Gondur, Dhule, Maharashtra, India
- Division of Computer Aided Drug Design, Department of Pharmaceutical Chemistry, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, Maharashtra, India
| | - Siti Awanis Abdullah
- Department of Chemistry, Faculty of Science, Universiti Teknologi Malaysia, Johor Bahru, Johor, Malaysia
| | - Shajarahtunnur Jamil
- Department of Chemistry, Faculty of Science, Universiti Teknologi Malaysia, Johor Bahru, Johor, Malaysia
| | - Harun Patel
- Division of Computer Aided Drug Design, Department of Pharmaceutical Chemistry, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, Maharashtra, India
| | - Sankaranarayanan Murugesan
- Medicinal Chemistry Research Laboratory, Birla Institute of Technology & Science Pilani (BITS Pilani), Pilani, Rajasthan, India
| | - Joazaizulfazli Jamalis
- Department of Chemistry, Faculty of Science, Universiti Teknologi Malaysia, Johor Bahru, Johor, Malaysia
| |
Collapse
|
5
|
Halder SK, Ahmad I, Shathi JF, Mim MM, Hassan MR, Jewel MJI, Dey P, Islam MS, Patel H, Morshed MR, Shakil MS, Hossen MS. A Comprehensive Study to Unleash the Putative Inhibitors of Serotype2 of Dengue Virus: Insights from an In Silico Structure-Based Drug Discovery. Mol Biotechnol 2024; 66:612-625. [PMID: 36307631 PMCID: PMC9616416 DOI: 10.1007/s12033-022-00582-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 10/03/2022] [Indexed: 11/16/2022]
Abstract
Dengue fever is a mosquito-borne disease that claims the lives of millions of people around the world. A number of factors like disease's non-specific symptoms, increased viral mutation, growing antiviral drug resistance due to reduced susceptibility, unavailability of an effective vaccine for dengue, weak immunity against the virus, and many more are involved. Dengue belongs to the Flaviviridae family of viruses. The two species of the vector transmitting dengue are Aedes aegypti and Aedes albopictus, with the former one being dominant. Serotypes 2 of dengue fever are spread to the human body and cause severe illness. Recently, dengue has imposed an aggressive effect synergistically with the COVID-19 pandemic. As a result, we concentrated our efforts on finding a potential therapeutic. For this, we chose natural compounds to fight dengue fever, which is currently regarded as successful among many drug therapies. Following this, we started the in silico experiment with 922 plant extracts as lead compounds to fight serotype 2. In this study, we used SwissADME for analyzing ligand drug-likeness, pkCSM for designing an ADMET profile, Autodock vina 4.2 and Swissdock tools for molecular docking, and finally Desmond for molecular dynamics simulation. Ultimately 45 were found effective against the 2'O methyltransferase protein of serotype 2. CHEMBL376820 was found as possible therapeutic candidates for inhibiting methyltransferase protein in this thorough analysis. Nevertheless, more in vitro and in vivo research are required to substantiate their potential therapeutic efficacy.
Collapse
Affiliation(s)
- Sajal Kumar Halder
- Department of Biochemistry and Molecular Biology, Jahangirnagar University, Savar, Dhaka 1342 Bangladesh
- Division of Computer Aided Drug Design, BioAid, Mirpur, Dhaka, 1216 Bangladesh
| | - Iqrar Ahmad
- Division of Computer Aided Drug Design, Department of Pharmaceutical Chemistry, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, Maharashtra 425405 India
| | - Jannatul Fardous Shathi
- Department of Biochemistry and Molecular Biology, Jahangirnagar University, Savar, Dhaka 1342 Bangladesh
- Division of Computer Aided Drug Design, BioAid, Mirpur, Dhaka, 1216 Bangladesh
| | - Maria Mulla Mim
- Department of Pharmacy, Jahangirnagar University, Savar, Dhaka 1342 Bangladesh
- Division of Computer Aided Drug Design, BioAid, Mirpur, Dhaka, 1216 Bangladesh
| | - Md Rakibul Hassan
- Department of Biochemistry, Gono Bishwabidyalay, Savar, Dhaka 1344 Bangladesh
- Division of Computer Aided Drug Design, BioAid, Mirpur, Dhaka, 1216 Bangladesh
| | - Md Johurul Islam Jewel
- Department of Biochemistry and Molecular Biology, Primeasia University, Banani, Dhaka 1213 Bangladesh
- Division of Computer Aided Drug Design, BioAid, Mirpur, Dhaka, 1216 Bangladesh
| | - Piyali Dey
- Department of Biochemistry and Molecular Biology, Primeasia University, Banani, Dhaka 1213 Bangladesh
- Division of Computer Aided Drug Design, BioAid, Mirpur, Dhaka, 1216 Bangladesh
| | - Md Sirajul Islam
- Department of Biochemistry and Molecular Biology, Primeasia University, Banani, Dhaka 1213 Bangladesh
- Division of Computer Aided Drug Design, BioAid, Mirpur, Dhaka, 1216 Bangladesh
| | - Harun Patel
- Division of Computer Aided Drug Design, Department of Pharmaceutical Chemistry, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, Maharashtra 425405 India
| | - Md Reaz Morshed
- Department of Biochemistry and Molecular Biology, Noakhali Science and Technology University, Noakhali, 3814 Bangladesh
| | - Md Salman Shakil
- Department of Mathematics and Natural Sciences, Brac University, Dhaka, 1212 Bangladesh
- Division of Computer Aided Drug Design, BioAid, Mirpur, Dhaka, 1216 Bangladesh
| | - Md Sakib Hossen
- Department of Biochemistry and Molecular Biology, Primeasia University, Banani, Dhaka 1213 Bangladesh
- Division of Computer Aided Drug Design, BioAid, Mirpur, Dhaka, 1216 Bangladesh
| |
Collapse
|
6
|
Owen AE, Chima CM, Ahmad I, Emori W, Agwamba EC, Cheng CR, Benjamin I, Patel H, Ahuekwe EF, Ojong MA, Ubah CB, Manicum ALE, Louis H. Antibacterial Potential of Trihydroxycyclohexa-2,4-Diene-1-Carboxylic Acid: Insight from DFT, Molecular Docking, and Molecular Dynamic Simulation. Polycycl Aromat Compd 2024; 44:2128-2151. [DOI: 10.1080/10406638.2023.2214280] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 05/07/2023] [Indexed: 09/21/2024]
Affiliation(s)
- Aniekan E. Owen
- Computational and Bio-Simulation Research Group, University of Calabar, Calabar, Nigeria
| | - Chioma M. Chima
- Computational and Bio-Simulation Research Group, University of Calabar, Calabar, Nigeria
- Department of Pure and Applied Chemistry, Faculty of Physical Sciences, University of Calabar, Calabar, Nigeria
| | - Iqrar Ahmad
- Department of Pharmaceutical Chemistry, Division of Computer-Aided Drug Design, R. C. Patel Institute of Pharmaceutical Education and Research, Maharashtra, India
| | - Wilfred Emori
- Department of Chemistry, Sichuan University of Science & Engineering, Zigong, Sichuan, P. R. China
| | - Ernest C. Agwamba
- Computational and Bio-Simulation Research Group, University of Calabar, Calabar, Nigeria
- Department of Microbiology, Covenant University, Otta, Nigeria
| | - Chun-Ru Cheng
- Department of Chemistry, Sichuan University of Science & Engineering, Zigong, Sichuan, P. R. China
| | - Innocent Benjamin
- Computational and Bio-Simulation Research Group, University of Calabar, Calabar, Nigeria
- Department of Microbiology, Faculty of Biological Sciences, University of Calabar, Calabar, Nigeria
| | - Harun Patel
- Department of Pharmaceutical Chemistry, Division of Computer-Aided Drug Design, R. C. Patel Institute of Pharmaceutical Education and Research, Maharashtra, India
| | - Eze F. Ahuekwe
- Department of Microbiology, Covenant University, Otta, Nigeria
| | - Mmefone A. Ojong
- Computational and Bio-Simulation Research Group, University of Calabar, Calabar, Nigeria
- Department of Microbiology, Faculty of Biological Sciences, University of Calabar, Calabar, Nigeria
| | - Chioma B. Ubah
- Computational and Bio-Simulation Research Group, University of Calabar, Calabar, Nigeria
- Department of Microbiology, Faculty of Biological Sciences, University of Calabar, Calabar, Nigeria
| | | | - Hitler Louis
- Computational and Bio-Simulation Research Group, University of Calabar, Calabar, Nigeria
- Department of Pure and Applied Chemistry, Faculty of Physical Sciences, University of Calabar, Calabar, Nigeria
| |
Collapse
|
7
|
Das RP, Sahoo S, Paidesetty SK, Ahmad I, Sahoo B, Jayabaskaran C, Patel H, Arakha M, Pradhan AK. Isolation, characterization, and multimodal evaluation of novel glycolipid biosurfactant derived from Bacillus species: A promising Staphylococcus aureus tyrosyl-tRNA synthetase inhibitor through molecular docking and MD simulations. Int J Biol Macromol 2024; 261:129848. [PMID: 38302032 DOI: 10.1016/j.ijbiomac.2024.129848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 01/16/2024] [Accepted: 01/28/2024] [Indexed: 02/03/2024]
Abstract
Glycolipid-based biosurfactants (BSs), known for their intriguing and diverse properties, represent a largely uncharted territory in the realm of potential biomedical applications. This field holds great promise yet remains largely unexplored. This investigation provides new insights into the isolation, characterization, and comprehensive biomedical assessment of a novel glycolipid biosurfactant derived from Bacillus species, meeting the growing demand for understanding its multifaceted impact on various biomedical issues. Within this framework, two glycolipids, BG2A and BG2B, emerged as the most proficient strains in biosurfactant (BS) production. The biosurfactants (BSs) ascertained as glycolipids via thin layer chromatography (TLC) exhibited antimicrobial activity against S. aureus and E. coli. Both isolates exhibited anticancer effects against cervical carcinoma cells and demonstrated significant anti-biofilm activity against V. cholerae. Moreover, molecular docking and molecular dynamics (MD) simulations were employed to explore their antimicrobial resistance properties against Tyrosyl-tRNA synthetase (TyrRS) of Staphylococcus aureus, a well-annotated molecular target. Characterization and interpretation using Fourier transform infrared spectroscopy (FTIR) and nuclear magnetic resonance spectroscopy (1H and 13C NMR) confirmed that the BSs produced by each strain were glycolipids. These findings suggest that the isolated BSs can serve as effective agents with antibiofilm, antimicrobial, antioxidant, and anticancer properties, in addition to their considerable antibacterial resistance attributes.
Collapse
Affiliation(s)
- Rohit Pritam Das
- Department of Bioengineering, Centre for Biotechnology, Siksha 'O' Anusandhan (Deemed to be University), 751030, Odisha, Bhubaneswar, India
| | - Subhadarsini Sahoo
- Department of Biochemistry, Indian Institute of Science, Bangalore, India
| | - Sudhir Kumar Paidesetty
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University), 751030 Bhubaneswar, Odisha, India
| | - Iqrar Ahmad
- Department of Pharmaceutical Chemistry, Prof. Ravindra Nikam College of Pharmacy, Gondur, Dhule 424002, Maharashtra, India
| | - Banishree Sahoo
- Department of Bioengineering, Centre for Biotechnology, Siksha 'O' Anusandhan (Deemed to be University), 751030, Odisha, Bhubaneswar, India
| | - C Jayabaskaran
- Department of Biochemistry, Indian Institute of Science, Bangalore, India
| | - Harun Patel
- Division of Computer Aided Drug Design, Department of Pharmaceutical Chemistry, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur 425405, Maharashtra, India
| | - Manoranjan Arakha
- Department of Bioengineering, Centre for Biotechnology, Siksha 'O' Anusandhan (Deemed to be University), 751030, Odisha, Bhubaneswar, India
| | - Arun Kumar Pradhan
- Department of Bioengineering, Centre for Biotechnology, Siksha 'O' Anusandhan (Deemed to be University), 751030, Odisha, Bhubaneswar, India.
| |
Collapse
|
8
|
Bouali N, Ahmad I, Patel H, Alhejaili EB, Hamadou WS, Badraoui R, Hadj Lajimi R, Alreshidi M, Siddiqui AJ, Adnan M, Abdulhakeem MA, Bazaid AS, Patel M, Saeed M, Snoussi M, Noumi E. GC-MS screening of the phytochemical composition of Ziziphus honey: ADME properties and in vitro/ in silico study of its antimicrobial activity. J Biomol Struct Dyn 2024; 42:1368-1380. [PMID: 37191027 DOI: 10.1080/07391102.2023.2205945] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 03/30/2023] [Indexed: 05/17/2023]
Abstract
A revival interest has been given to natural products as sources of phytocompounds to be used as alternative treatment against infectious diseases. In this context, we aimed to investigate the antimicrobial potential of Ziziphus honey (ZH) against twelve clinical bacterial strains and several yeasts and molds using in vitro and computational approaches. The well-diffusion assay revealed that ZH was able to induce growth inhibition of most Gram-positive and Gram-negative bacteria. The high mean growth inhibition zone (mGIZ) was recorded in E. coli (Clinical strain, 217), S. aureus followed by E. coli ATCC 10536 (mGIZ values: 41.00 ± 1 mm, 40.67 ± 0.57 mm, and 34.67 ± 0.57 mm, respectively). The minimal bactericidal concentrations (MBCs) and minimal fungicidal concentration values (MFCs) from approximately 266.33 mg/mL to over 532.65 mg/mL. Molecular docking results revealed that the identified compounds maltose, 2-furoic acid, isopropyl ester, 2,4-imidazolidinedione, 5-(2-methylpropyl)-(S)- and 3,4,5-trihydroxytoluene, S-Methyl-L-Cysteine, 2-Furancarboxylic acid, L-Valine-N-ethoxycarbonyl, Hexanoic acid, 3,5,5-trimethyl-, Methyl-beta-D-thiogalactoside, gamma-Sitosterol, d-Mannose, 4-O-Methylmannose, 2,4-Imidazolidinedione, 5-(2-methylpropyl)- (S) were found to have good affinity for targeted receptor, respectively. Through a 100-ns dynamic simulation research, binding interactions and stability between promising phytochemicals and the active residues of the studied enzymes were confirmed. The ADMET profiling of all identified compounds revealed that most of them could be qualified as biologically active with good absorption and permeation. Overall, the results highlighted the efficiency of ZH against the tested clinical pathogenic strains. The antimicrobial potential and the potency displayed by the identified compounds could imply their further pharmacological applications.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Nouha Bouali
- Department of Biology, University of Hail, College of Science, Ha'il, Saudi Arabia
| | - Iqrar Ahmad
- Department of Pharmaceutical Chemistry, Prof. Ravindra Nikam College of Pharmacy, Gondur, Dhule, Maharashtra, India
| | - Harun Patel
- Department of Pharmaceutical Chemistry, Prof. Ravindra Nikam College of Pharmacy, Gondur, Dhule, Maharashtra, India
| | | | - Walid Sabri Hamadou
- Department of Biology, University of Hail, College of Science, Ha'il, Saudi Arabia
- Research Unit: Molecular Biology of Leukemia and Lymphoma, Department of Biochemistry, University of Medecine of Sousse, Sousse, Tunisia
| | - Riadh Badraoui
- Department of Biology, University of Hail, College of Science, Ha'il, Saudi Arabia
- Section of Histology - Cytology, University of Medicine of Tunis, University of Tunis El Manar, Tunis, Tunisia
- Department of Histo-Embryology and Cytogenetics, Medicine Faculty of Sfax, University of Sfax, Sfax, Tunisia
| | - Ramzi Hadj Lajimi
- Department of Chemistry, College of Science, University of Ha'il, Ha'il, Saudi Arabia
- Laboratory of Water, Membranes and Environmental Biotechnologies, Center of Research and Water Technologies, Soliman, Tunisia
| | - Mousa Alreshidi
- Department of Biology, University of Hail, College of Science, Ha'il, Saudi Arabia
- Department of Medical Laboratory Science, College of Applied Medical Sciences, University of Hail, Hail, Saudi Arabia
| | - Arif Jamal Siddiqui
- Department of Biology, University of Hail, College of Science, Ha'il, Saudi Arabia
| | - Mohd Adnan
- Department of Biology, University of Hail, College of Science, Ha'il, Saudi Arabia
| | | | - Abdulrahman S Bazaid
- Department of Medical Laboratory Science, College of Applied Medical Sciences, University of Hail, Hail, Saudi Arabia
| | - Mitesh Patel
- Department of Biotechnology, Parul Institute of Applied Sciences and Centre of Research for Development, Parul University, Vadodara, India
| | - Mohd Saeed
- Department of Biology, University of Hail, College of Science, Ha'il, Saudi Arabia
| | - Mejdi Snoussi
- Department of Biology, University of Hail, College of Science, Ha'il, Saudi Arabia
- Laboratory of Genetics, Biodiversity and Valorisation of Bioressources, High Institute of Biotechnology University of Monastir, Monastir, Tunisia
| | - Emira Noumi
- Department of Biology, University of Hail, College of Science, Ha'il, Saudi Arabia
- Laboratory of Genetics, Biodiversity and Valorisation of Bioressources, High Institute of Biotechnology University of Monastir, Monastir, Tunisia
| |
Collapse
|
9
|
Gordon AT, Hosten EC, van Vuuren S, Ogunlaja AS. Copper(II)-photocatalyzed Hydrocarboxylation of Schiff bases with CO 2: antimicrobial evaluation and in silico studies of Schiff bases and unnatural α-amino acids. J Biomol Struct Dyn 2024:1-14. [PMID: 38192072 DOI: 10.1080/07391102.2024.2301765] [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: 08/11/2023] [Accepted: 12/30/2023] [Indexed: 01/10/2024]
Abstract
We synthesized and characterized two copper(II) complexes: [CuL2Cl]Cl and [CuL'2Cl]Cl, where L = 2,2'-bipyridine and L' = 4,4'-dimethyl-2,2'-bipyridine. We evaluated their photocatalytic hydrocarboxylation properties on a series of synthesized Schiff bases (SBs): (E)-1-(4-((5-bromo-2-hydroxybenzylidene)amino)phenyl)ethanone (SB1), (E)-N-(4-(dimethylamino)benzylidene)benzo[d]thiazol-2-amine (SB2), (E)-4-Bromo-2-((thiazol-2-ylimino)methyl)phenol (SB3), and (E)-4-((5-bromo-2-hydroxybenzylidene)amino)-1,5-dimethyl-2-phenyl-1H-pyrazol-3(2H)-one (SB4). Under mild photocatalytic reaction conditions (room temperature, 1 atm CO2, 30-watt Blue LED light), the derivatives of α-amino acids UAA1-4 were obtained with yields ranging from 5% to 44%. Experimental results demonstrated that [CuL2Cl]Cl exhibited superior photocatalytic efficiency compared to [CuL'2Cl]Cl, attributed to favourable electronic properties. In silico studies revealed strong binding strengths with E. faecalis DHFR (4M7U) for docked Schiff bases (SB) and unnatural α-amino acids (UAAs). In vitro studies further demonstrated significant antimicrobial and antifungal activity for SB2, SB3, and SB4, while none of the synthesized UAAs exhibited such properties, primarily due to the electronic and binding properties of these molecules.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Allen T Gordon
- Department of Chemistry, Nelson Mandela University, Port Elizabeth, South Africa
| | - Eric C Hosten
- Department of Chemistry, Nelson Mandela University, Port Elizabeth, South Africa
| | - Sandy van Vuuren
- Department of Pharmacy and Pharmacology, Faculty of Health Sciences, University of the Witwatersrand, Parktown, South Africa
| | - Adeniyi S Ogunlaja
- Department of Chemistry, Nelson Mandela University, Port Elizabeth, South Africa
| |
Collapse
|
10
|
Ayipo YO, Ahmad I, Chong CF, Zainurin NA, Najib SY, Patel H, Mordi MN. Carbazole derivatives as promising competitive and allosteric inhibitors of human serotonin transporter: computational pharmacology. J Biomol Struct Dyn 2024; 42:993-1014. [PMID: 37021485 DOI: 10.1080/07391102.2023.2198016] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 03/25/2023] [Indexed: 04/07/2023]
Abstract
The human serotonin transporters (hSERTs) are neurotransmitter sodium symporters of the aminergic G protein-coupled receptors, regulating the synaptic serotonin and neuropharmacological processes related to neuropsychiatric disorders, notably, depression. Selective serotonin reuptake inhibitors (SSRIs) such as fluoxetine and (S)-citalopram are competitive inhibitors of hSERTs and are commonly the first-line medications for major depressive disorder (MDD). However, treatment-resistance and unpleasant aftereffects constitute their clinical drawbacks. Interestingly, vilazodone emerged with polypharmacological (competitive and allosteric) inhibitions on hSERTs, amenable to improved efficacy. However, its application usually warrants adjuvant/combination therapy, another subject of critical adverse events. Thus, the discovery of alternatives with polypharmacological potentials (one-drug-multiple-target) and improved safety remains essential. In this study, carbazole analogues from chemical libraries were explored using docking and molecular dynamics (MD) simulation. Selectively, two IBScreen ligands, STOCK3S-30866 and STOCK1N-37454 predictively bound to the active pockets and expanded boundaries (extracellular vestibules) of the hSERTs more potently than vilazodone and (S)-citalopram. For instance, the two ligands showed docking scores of -9.52 and -9.59 kcal/mol and MM-GBSA scores of -92.96 and -65.66 kcal/mol respectively compared to vilazodone's respective scores of -7.828 and -59.27 against the central active site of the hSERT (PDB 7LWD). Similarly, the two ligands also docked to the allosteric pocket (PDB 5I73) with scores of -8.15 and -8.40 kcal/mol and MM-GBSA of -96.14 and -68.46 kcal/mol whereas (S)-citalopram has -6.90 and -69.39 kcal/mol respectively. The ligands also conferred conformational stability on the receptors during 100 ns MD simulations and displayed interesting ADMET profiles, representing promising hSERT modulators for MDD upon experimental validation.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Yusuf Oloruntoyin Ayipo
- Centre for Drug Research, Universiti Sains Malaysia, USM, Pulau Pinang, Malaysia
- Department of Chemistry and Industrial Chemistry, Kwara State University, Ilorin, Nigeria
| | - Iqrar Ahmad
- Department of Pharmaceutical Chemistry, Prof Ravindra Nikam College of Pharmacy, Dhule, Maharashtra, India
- Department of Pharmaceutical Chemistry, R C Patel Institute of Pharmaceutical Education and Research, Shirpur, Maharashtra, India
| | - Chien Fung Chong
- Centre for Drug Research, Universiti Sains Malaysia, USM, Pulau Pinang, Malaysia
- Department of Allied Health Sciences, Universiti Tunku Abdul Rahman, Kampar, Perak, Malaysia
| | - Nurul Amira Zainurin
- Faculty of Agro-Based Industry, Universiti Malaysia Kelantan, Jeli, Kelantan, Malaysia
| | - Sani Yahaya Najib
- Centre for Drug Research, Universiti Sains Malaysia, USM, Pulau Pinang, Malaysia
- Department of Pharmaceutical and Medicinal Chemistry, Bayero University, Kano, Nigeria
| | - Harun Patel
- Department of Pharmaceutical Chemistry, R C Patel Institute of Pharmaceutical Education and Research, Shirpur, Maharashtra, India
| | - Mohd Nizam Mordi
- Centre for Drug Research, Universiti Sains Malaysia, USM, Pulau Pinang, Malaysia
| |
Collapse
|
11
|
Benny F, Oh JM, Kumar S, Abdelgawad MA, Ghoneim MM, Abdel-Bakky MS, Kukerti N, Jose J, Kim H, Mathew B. Isatin-based benzyloxybenzene derivatives as monoamine oxidase inhibitors with neuroprotective effect targeting neurogenerative disease treatment. RSC Adv 2023; 13:35240-35250. [PMID: 38053684 PMCID: PMC10694828 DOI: 10.1039/d3ra07035b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 11/26/2023] [Indexed: 12/07/2023] Open
Abstract
Eighteen isatin-based benzyloxybenzaldehyde derivatives from three subseries, ISB, ISFB, and ISBB, were synthesized and their ability to inhibit monoamine oxidase (MAO) was evaluated. The inhibitory activity of all synthesized compounds was found to be more profound against MAO-B than MAO-A. Compound ISB1 most potently inhibited MAO-B with an IC50 of 0.124 ± 0.007 μM, ensued by ISFB1 (IC50 = 0.135 ± 0.002 μM). Compound ISFB1 most potently inhibited MAO-A with an IC50 of 0.678 ± 0.006 μM, ensued by ISBB3 (IC50 = 0.731 ± 0.028 μM), and had the highest selectivity index (SI) value (55.03). The three sub-parental compounds, ISB1, ISFB1, and ISBB1, had higher MAO-B inhibition than the other derivatives, indicating that the substitutions of the 5-H in the A-ring of isatin diminished the inhibition of MAO-A and MAO-B. Among these, ISB1 (para-benzyloxy group in the B-ring) displayed more significant MAO-B inhibition when compared to ISBB1 (meta-benzyloxy group in the B-ring). ISB1 and ISFB1 were identified to be competitive and reversible MAO-B inhibitors, having Ki values of 0.055 ± 0.010, and 0.069 ± 0.025 μM, respectively. Furthermore, in the parallel artificial membrane penetration assay, ISB1 and ISFB1 traversed the blood-brain barrier in the in vitro condition. Additionally, the current study found that ISB1 decreased rotenone-induced cell death in SH-SY5Y neuroblastoma cells. In docking and simulation studies, the hydrogen bonding formed by the imino nitrogen in ISB1 and the pi-pi stacking interaction of the phenyl ring in isatin significantly aided in the protein-ligand complex's stability, effectively inhibiting MAO-B. According to these observations, the MAO-B inhibitors ISB1 and ISFB1 were potent, selective, and reversible, making them conceivable therapies for neurological diseases.
Collapse
Affiliation(s)
- Feba Benny
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham AIMS Health Sciences Campus Kochi 682041 India
| | - Jong Min Oh
- Department of Pharmacy, Research Institute of Life Pharmaceutical Sciences, Sunchon National University Suncheon 57922 Republic of Korea
| | - Sunil Kumar
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham AIMS Health Sciences Campus Kochi 682041 India
| | - Mohamed A Abdelgawad
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University Sakaka 72341 Saudi Arabia
| | - Mohammed M Ghoneim
- Department of Pharmacy Practice, College of Pharmacy, AlMaarefa University Ad Diriyah 13713 Saudi Arabia
| | - Mohamed Sadek Abdel-Bakky
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University Buraydah 51452 Saudi Arabia
| | - Neelima Kukerti
- School of Pharmacy, Graphic Era Hill University Dehradun Uttarakhand 248002 India
| | - Jobin Jose
- Department of Pharmaceutics, NGSM Institute of Pharmaceutical Science, NITTE University Mangalore Karnataka 575018 India
| | - Hoon Kim
- Department of Pharmacy, Research Institute of Life Pharmaceutical Sciences, Sunchon National University Suncheon 57922 Republic of Korea
| | - Bijo Mathew
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham AIMS Health Sciences Campus Kochi 682041 India
| |
Collapse
|
12
|
Bharadwaj KK, Rabha B, Ahmad I, Mathew SP, Bhattacharjee CK, Jaganathan BG, Poddar S, Patel H, Subramaniyan V, Chinni SV, Ramachawolran G, Saleem R, Khalifa Ali EH, Abdel-Daim MM, Baishya D, Ghosh A. Rhamnetin, a nutraceutical flavonoid arrests cell cycle progression of human ovarian cancer (SKOV3) cells by inhibiting the histone deacetylase 2 protein. J Biomol Struct Dyn 2023; 42:13421-13436. [PMID: 38014451 DOI: 10.1080/07391102.2023.2275187] [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: 06/12/2023] [Accepted: 10/20/2023] [Indexed: 11/29/2023]
Abstract
Overexpression of HDAC 2 promotes cell proliferation in ovarian cancer. HDAC 2 is involved in chromatin remodeling, transcriptional repression, and the formation of condensed chromatin structures. Targeting HDAC 2 presents a promising therapeutic approach for correcting cancer-associated epigenetic abnormalities. Consequently, HDAC 2 inhibitors have evolved as an attractive class of anti-cancer agents. This work intended to investigate the anti-cancer abilities and underlying molecular mechanisms of Rhamnetin in human epithelial ovarian carcinoma cells (SKOV3), which remain largely unexplored. We employed various in vitro methods, including MTT, apoptosis study, cell cycle analysis, fluorescence microscopy imaging, and in vitro enzymatic HDAC 2 protein inhibition, to examine the chemotherapeutic sensitivity of Rhamnetin in SKOV3 cells. Additionally, we conducted in silico studies using molecular docking, MD simulation, MM-GBSA, DFT, and pharmacokinetic analysis to investigate the binding interaction mechanism within Rhamnetin and HDAC 2, alongside the compound's prospective as a lead candidate. The in vitro assay confirmed the cytotoxic effects of Rhamnetin on SKOV3 cells, through its inhibition of HDAC 2 activity. Rhamnetin, a nutraceutical flavonoid, halted at the G1 phase of the cell cycle and triggered apoptosis in SKOV3 cells. Furthermore, computational studies provided additional evidence of its stable binding to the HDAC 2 protein's binding site cavity. Based on our findings, we conclude that Rhamnetin effectively promotes apoptosis and mitigates the proliferation of SKOV3 cells through HDAC 2 inhibition. These results highlight Rhamnetin as a potential lead compound, opening a new therapeutic strategy for human epithelial ovarian cancer.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
| | - Bijuli Rabha
- Department of Bioengineering and Technology, Gauhati University, Guwahati, Assam, India
| | - Iqrar Ahmad
- Department of Pharmaceutical Chemistry, Prof. Ravindra Nikam College of Pharmacy, Dhule, Maharashtra, India
- Division of Computer Aided Drug Design, Department of Pharmaceutical Chemistry, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, Maharashtra, India
| | - Sam P Mathew
- Stem Cells and Cancer Biology Research Group, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| | | | - Bithiah Grace Jaganathan
- Stem Cells and Cancer Biology Research Group, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India
- Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| | - Snikdha Poddar
- Department of Bioengineering and Technology, Gauhati University, Guwahati, Assam, India
| | - Harun Patel
- Division of Computer Aided Drug Design, Department of Pharmaceutical Chemistry, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, Maharashtra, India
| | - Vetriselvan Subramaniyan
- Pharmacology Unit, Jeffrey Cheah School of Medicine and Health Sciences, MONASH University, Selangor, Malaysia
| | - Suresh V Chinni
- Department of Biochemistry, Faculty of Medicine, Bioscience, and Nursing, MAHSA University, Jenjarom, Selangor, Malaysia
| | - Gobinath Ramachawolran
- Department of Foundation, RCSI & UCD Malaysia Campus, Georgetown, Pulau Pinang, Malaysia
| | - Rasha Saleem
- Department of Laboratory Medicine, Faculty of Applied Medical Sciences, Al-Baha University, Al-Baha, Saudi Arabia
| | - Eman Hussain Khalifa Ali
- Department of Laboratory Medicine, Faculty of Applied Medical Sciences, Al-Baha University, Al-Baha, Saudi Arabia
| | - Mohamed M Abdel-Daim
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, Jeddah, Saudi Arabia
- Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| | - Debabrat Baishya
- Department of Bioengineering and Technology, Gauhati University, Guwahati, Assam, India
| | - Arabinda Ghosh
- Department of Computational Biology and Biotechnology, Mahapurasha Srimanta Sankaradeva Viswavidyalaya, Guwahati, India
| |
Collapse
|
13
|
Rahman MO, Ahmed SS. Anti-angiogenic potential of bioactive phytochemicals from Helicteres isora targeting VEGFR-2 to fight cancer through molecular docking and molecular dynamics simulation. J Biomol Struct Dyn 2023; 41:7447-7462. [PMID: 36099201 DOI: 10.1080/07391102.2022.2122568] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 09/02/2022] [Indexed: 10/14/2022]
Abstract
Cancer is one of the leading causes of death due to its very high rate of morbidity and mortality, and there is a constant demand of effective drugs for cancer therapy. Vascular endothelial growth factor receptor-2 (VEGFR-2) plays a significant role as central modulator of angiogenesis and is targeted frequently for developing anti-angiogenic agents to fight cancer. Helicteres isora L. (Malvaceae) is reported to possess diverse medicinal properties including anti-cancer potentials. In the current investigation, 38 bioactive phytochemicals of H. isora were screened virtually to evaluate their anti-angiogenic potentials targeting VEGFR-2. The study unveiled three potential candidates such as, Diosgenin (-9.8 Kcal//mol), Trifolin (-8.4 Kcal/mol) and Yohimbine (-8.1 Kcal/mol) that showed favorable pharmacokinetic, pharmacodynamic and toxicity properties with no significant side effects. Molecular dynamics simulation employing 100 ns revealed noteworthy structural stability and compactness for all the top three candidates. The MM/GBSA binding free energy estimation corroborated the docking interactions where Yohimbine (-30.47 Kcal/mol) scored better than Diosgenin (-27.54 Kcal/mol) and Trifolin (-29.58 Kcal/mol). Target class prediction revealed enzymes in most of the cases and some FDA approved drugs were found as structurally similar analogs for Trifolin and Yohimbine. These findings could lead to the development of novel and effective anti-angiogenic agents.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- M Oliur Rahman
- Department of Botany, Faculty of Biological Sciences, University of Dhaka, Dhaka, Bangladesh
| | - Sheikh Sunzid Ahmed
- Department of Botany, Faculty of Biological Sciences, University of Dhaka, Dhaka, Bangladesh
| |
Collapse
|
14
|
Samajdar S, Mondal P. In silico studies on the phytochemical components of Lagenaria siceraria targeting aromatase receptors against breast cancer. In Silico Pharmacol 2023; 11:19. [PMID: 37525849 PMCID: PMC10387019 DOI: 10.1007/s40203-023-00155-3] [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: 06/08/2023] [Accepted: 07/17/2023] [Indexed: 08/02/2023] Open
Abstract
In India, breast cancer is the most common cause of mortality for women and has the potential to spread to other body organs. As a transcription factor, interactions with the estrogen receptor (ER) alpha are primarily responsible for the development of malignant tumors. Aromatase inhibitors are the most often used treatment for ER(+) breast cancer. Various synthetic compounds have been developed over the years to block the aromatase receptor, however, the majority of them are hazardous and cause multidrug resistance. So, combating these natural drugs can be prioritized. The current study was conducted to investigate the anticancer potential of Lagenaria siceraria phytoconstituents against breast cancer target protein (PDB ID: 3EQM) based on a literature review. In this study, 34 Lagenaria siceraria ligands were chosen, and the structure of the human aromatase receptor was acquired from the protein data bank. For those natural chemicals, molecular docking, drug-likeness, toxicity, and molecular dynamics were used to evaluate and analyse their anti-breast cancer activity. Five substances, 2,3-Diphenyl quinoxaline, 17-Acetoxy pregnolone, Benzyl-d-glucoside, Ergostenol acetate, and Stigmast-7-en-3-ol, shown higher binding affinity than Tamoxifen, signaling their potential use in breast cancer treatment.
Collapse
Affiliation(s)
- Saptarshi Samajdar
- Department of Pharmaceutical Technology, Brainware University, 328, Ramkrishnapur Road, Kolkata, India
| | - Prasenjit Mondal
- Department of Pharmaceutical Technology, Brainware University, 328, Ramkrishnapur Road, Kolkata, India
| |
Collapse
|
15
|
Alminderej F, Ghannay S, Omer Elsamani M, Alhawday F, Albadri AEAE, Elbehairi SEI, Alfaifi MY, Kadri A, Aouadi K. In Vitro and In Silico Evaluation of Antiproliferative Activity of New Isoxazolidine Derivatives Targeting EGFR: Design, Synthesis, Cell Cycle Analysis, and Apoptotic Inducers. Pharmaceuticals (Basel) 2023; 16:1025. [PMID: 37513936 PMCID: PMC10384175 DOI: 10.3390/ph16071025] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 07/11/2023] [Accepted: 07/16/2023] [Indexed: 07/30/2023] Open
Abstract
A series of novel enantiopure isoxazolidine derivatives were synthesized and evaluated for their anticancer activities against three human cancer cell lines such as human breast carcinoma (MCF-7), human lung adenocarcinoma (A-549), and human ovarian carcinoma (SKOV3) by employing MTT assay. The synthesized compounds were characterized by NMR and elemental analysis. Results revealed that all the synthesized compounds displayed significant inhibition towards the tested cell lines. Among them, 2g and 2f, which differ only by the presence of an ester group at the C-3 position and small EDG (methyl) at the C-5 position of the phenyl ring (2g), were the most active derivatives in attenuating the growth of the three cells in a dose-dependent manner. The IC50 for 2g were 17.7 ± 1 µM (MCF-7), 12.1 ± 1.1 µM (A-549), and 13.9 ± 0.7 µM (SKOV3), and for 2f were 9.7 ± 1.3µM (MCF-7), 9.7 ± 0.7µM (A-549), and 6.5 ± 0.9µM (SKOV3), respectively, which were comparable to the standard drug, doxorubicin. The enzymatic inhibition of 2f and 2g against EGFR afforded good inhibitory activity with IC50 of 0.298 ± 0.007 μM and 0.484 ± 0.01 µM, respectively, close to the positive control, Afatinib. Compound 2f arrested the cell cycle in the S phase in MCF-7 and SKOV3 cells, and in the G2/M phase in the A549 cell; however, 2g induced G0/G1 phase cell cycle arrest, and inhibited the progression of the three cancer cells, together with significant apoptotic effects. The docking study of compounds 2f and 2g into EGFR ATP-active site revealed that it fits nicely with good binding affinity. The pharmacokinetic and drug-likeness scores revealed notable lead-like properties. At 100 ns, the dynamic simulation investigation revealed high conformational stability in the EGFR binding cavity.
Collapse
Affiliation(s)
- Fahad Alminderej
- Department of Chemistry, College of Science, Qassim University, Buraidah 51452, Saudi Arabia
| | - Siwar Ghannay
- Department of Chemistry, College of Science, Qassim University, Buraidah 51452, Saudi Arabia
| | - Mohamed Omer Elsamani
- Department of Chemistry, Faculty of Science and Arts of Baljurashi, Al-Baha University, P.O. Box 1988, Albaha 65527, Saudi Arabia
- Department of Food Science and Technology, Faculty of Sciences, Omdurman Islamic University, Omdurman P.O. Box 382, Sudan
| | - Fahad Alhawday
- Department of Chemistry, College of Science, Qassim University, Buraidah 51452, Saudi Arabia
| | - Abuzar E A E Albadri
- Department of Chemistry, College of Science, Qassim University, Buraidah 51452, Saudi Arabia
| | - Serag Eldin I Elbehairi
- Department of Biology, Faculty of Science, King Khalid University, Abha 9004, Saudi Arabia
- Cell Culture Laboratory, Egyptian Organization for Biological Products and Vaccines, VACSERA Holding Company, Giza 2311, Egypt
| | - Mohammad Y Alfaifi
- Department of Biology, Faculty of Science, King Khalid University, Abha 9004, Saudi Arabia
| | - Adel Kadri
- Department of Chemistry, Faculty of Science and Arts of Baljurashi, Al-Baha University, P.O. Box 1988, Albaha 65527, Saudi Arabia
- Department of Chemistry, Faculty of Science of Sfax, University of Sfax, B.P. 1171, Sfax 3000, Tunisia
| | - Kaïss Aouadi
- Department of Chemistry, College of Science, Qassim University, Buraidah 51452, Saudi Arabia
- Department of Chemistry, Faculty of Science of Sfax, University of Sfax, B.P. 1171, Sfax 3000, Tunisia
- Department of Chemistry, Laboratory of Heterocyclic Chemistry Natural Product and Reactivity/CHPNR, Faculty of Science of Monastir, University of Monastir, Avenue of the Environment, Monastir 5019, Tunisia
| |
Collapse
|
16
|
Abdullahi M, Uzairu A, Shallangwa GA, Mamza PA, Ibrahim MT, Ahmad I, Patel H. Structure-based drug design, molecular dynamics simulation, ADMET, and quantum chemical studies of some thiazolinones targeting influenza neuraminidase. J Biomol Struct Dyn 2023; 41:13829-13843. [PMID: 37158006 DOI: 10.1080/07391102.2023.2208225] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 02/11/2023] [Indexed: 05/10/2023]
Abstract
The genetic mutability of the influenza virus leads to the existence of drug-resistant strains which is dangerous, particularly with the lingering coronavirus disease (COVID-19). This necessitated the need for the search and discovery of more potential anti-influenza agents to avert future outbreaks. In furtherance of our previous in-silico studies on 5-benzyl-4-thiazolinones as anti-influenza neuraminidase (NA) inhibitors, molecule 11 was selected as the template scaffold for the structure-based drug design due to its good binding, pharmacokinetic profiling, and better NA inhibitory activity. As such, eighteen (18) new molecules (11a-r) were designed with better MolDock scores as compared with the template scaffold and the zanamivir reference drug. However, the dynamic stability of molecule 11a in the binding cavity of the NA target (3TI5) showed water-mediated hydrogen and hydrophobic bondings with the active residues such as Arg118, Ile149, Arg152, Ile222, Trp403, and Ile427 after the MD simulation for 100 ns. The drug-likeness and ADMET assessment of all designed molecules predicted non-violation of the stipulated thresholds of Lipinski's rule and good pharmacokinetic properties respectively. In addition, the quantum chemical calculations also suggested the significant chemical reactivity of molecules with their smaller band energy gap, high electrophilicity, high softness, and low hardness. The results obtained in this study proposed a reliable in-silico viewpoint for anti-influenza drug discovery and development.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Mustapha Abdullahi
- Faculty of Physical Sciences, Department of Chemistry, Ahmadu Bello University, Zaria, Kaduna State, Nigeria
- Faculty of Sciences, Department of Pure and Applied Chemistry, Kaduna State University, Kaduna, Kaduna State, Nigeria
| | - Adamu Uzairu
- Faculty of Physical Sciences, Department of Chemistry, Ahmadu Bello University, Zaria, Kaduna State, Nigeria
| | - Gideon Adamu Shallangwa
- Faculty of Physical Sciences, Department of Chemistry, Ahmadu Bello University, Zaria, Kaduna State, Nigeria
| | - Paul Andrew Mamza
- Faculty of Physical Sciences, Department of Chemistry, Ahmadu Bello University, Zaria, Kaduna State, Nigeria
| | - Muhammad Tukur Ibrahim
- Faculty of Physical Sciences, Department of Chemistry, Ahmadu Bello University, Zaria, Kaduna State, Nigeria
| | - Iqrar Ahmad
- Division of Computer Aided Drug Design, Department of Pharmaceutical Chemistry, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, Maharashtra, India
| | - Harun Patel
- Division of Computer Aided Drug Design, Department of Pharmaceutical Chemistry, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, Maharashtra, India
| |
Collapse
|
17
|
Sayed HM, Ramadan MA, Salem HH, Ahmad I, Patel H, Fayed MAA. Phytochemical Investigation, In Silico/In Vivo Analgesic, and Anti-inflammatory Assessment of the Egyptian Cassia occidentalis L. Steroids 2023; 196:109245. [PMID: 37141980 DOI: 10.1016/j.steroids.2023.109245] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 04/24/2023] [Accepted: 04/26/2023] [Indexed: 05/06/2023]
Abstract
Cassia occidentalis L., from Fabaceae family phytochemical screening, revealed several biologically active principles mainly flavonoids and anthraquinones. GLC analysis of the lipoidal matter afforded 12 hydrocarbons: 9-dodecyl-tetradecahydro-anthracene (48.97 %), 9-dodecyl-tetradecahydro-phenanthrene (14.43 %), and 6 sterols/triterpenes: iso-jaspisterol (11.99%) and fatty acids were palmitic acid (50 %), and Linoleic acid (16.06%). Column chromatography led to the isolation of fifteen compounds (1-15), elucidated using spectroscopic evidence. First report of undecanoic acid (4) from the family Fabaceae, while p-dimethyl amino-benzaldehyde (15) was first time isolated from a natural origin. Eight compounds isolated for the first time from C. occidentalis L.; β-amyrin (1), β-sitosterol (2), stigmasterol (3), camphor (5), lupeol (6), chrysin (7), pectolinargenin (8), and 1, 2, 5-trihydroxy anthraquinone (14) besides five known compounds previously isolated; apigenin (9), kaempferol (10), chrysophanol (11), physcion (12), and aloe-emodin (13). In-vivo evaluation of anti-inflammatory and analgesic effects of C. occidentalis L. extracts where the n-butanol and total extracts showed the highest activities. The percentage of the inhibitory effect of the n-butanol extract was 29.7 at a dose of 400 mg/Kg. Furthermore, identified phytoconstituents were docked into the active sites of enzymes nAChRs, COX-1, and COX-2 to evaluate binding affinity. Phyto-compounds Physcion, aloe-emodin, and chrysophanol were found to have a good affinity for targeted receptors compared to co-crystalized inhibitors, validating the analgesic and anti-inflammatory effects of the phytochemicals.
Collapse
Affiliation(s)
- Hanaa M Sayed
- Department of Pharmacognosy, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt
| | - Mahmoud A Ramadan
- Department of Pharmacognosy, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt
| | - Heba H Salem
- Department of Pharmacognosy, Faculty of Pharmacy, Northern Border University, Rafha Region, Saudi Arabia
| | - Iqrar Ahmad
- Division of Computer Aided Drug Design, Department of Pharmaceutical Chemistry, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur,425405, Maharashtra, India
| | - Harun Patel
- Division of Computer Aided Drug Design, Department of Pharmaceutical Chemistry, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur,425405, Maharashtra, India
| | - Marwa A A Fayed
- Department of Pharmacognosy, Faculty of Pharmacy, University of Sadat City, Sadat City 32897, Egypt.
| |
Collapse
|
18
|
Kumar S, Oh JM, Abdelgawad MA, Abourehab MA, Tengli AK, Singh AK, Ahmad I, Patel H, Mathew B, Kim H. Development of Isopropyl-Tailed Chalcones as a New Class of Selective MAO-B Inhibitors for the Treatment of Parkinson's Disorder. ACS OMEGA 2023; 8:6908-6917. [PMID: 36844523 PMCID: PMC9947953 DOI: 10.1021/acsomega.2c07694] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 01/30/2023] [Indexed: 06/16/2023]
Abstract
Thirteen isopropyl chalcones (CA1-CA13) were synthesized and evaluated for their inhibitory activity against monoamine oxidase (MAO). All compounds inhibited MAO-B more effectively than MAO-A. Compound CA4 most potently inhibited MAO-B with an IC50 value of 0.032 μM, similar to that of CA3 (IC50 = 0.035 μM) and with high selectivity index (SI) values for MAO-B over MAO-A (SI = 49.75 and 353.23, respectively). The -OH (CA4) or -F (CA3) group at the para position on the A ring provided higher MAO-B inhibition than that of the other substituents (-OH ≥ -F > -Cl > -Br > -OCH2CH3 > -CF3). On the other hand, compound CA10 most potently inhibited MAO-A with an IC50 value of 0.310 μM and effectively MAO-B (IC50 = 0.074 μM). The Br-containing thiophene substituent (CA10) instead of the A ring showed the highest MAO-A inhibition. In a kinetic study, K i values of compounds CA3 and CA4 for MAO-B were 0.076 ± 0.001 and 0.027 ± 0.002 μM, respectively, and that of CA10 for MAO-A was 0.016 ± 0.005 μM. A reversibility study showed that CA3 and CA4 were reversible inhibitors of MAO-B and CA10 was a reversible inhibitor of MAO-A. In docking and molecular dynamics, the hydroxyl group of CA4 and two hydrogen bonds contributed to the stability of the protein-ligand complex. These results suggest that CA3 and CA4 are potent reversible selective MAO-B inhibitors and can be used for the treatment of Parkinson's disease.
Collapse
Affiliation(s)
- Sunil Kumar
- Department
of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa
Vidyapeetham, AIMS Health Sciences Campus, Kochi 682 041, India
| | - Jong Min Oh
- Department
of Pharmacy, and Research Institute of Life Pharmaceutical Sciences, Sunchon National University, Suncheon 57922, Republic of Korea
| | - Mohamed A. Abdelgawad
- Department
of Pharmaceutical Chemistry, College of Pharmacy, Jouf University, Sakaka 72341, Saudi Arabia
- Pharmaceutical
Organic Chemistry Department, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt
| | - Mohammed A.S. Abourehab
- Department
of Pharmaceutics, College of Pharmacy, Umm
Al-Qura University, Makkah 21955, Saudi Arabia
| | - Anand Kumar Tengli
- Department
of Pharmaceutical Chemistry, JSS College
of Pharmacy, Mysuru 570015, India
| | - Ashutosh Kumar Singh
- Department
of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa
Vidyapeetham, AIMS Health Sciences Campus, Kochi 682 041, India
| | - Iqrar Ahmad
- Department
of Pharmaceutical Chemistry, Prof. Ravindra
Nikam College of Pharmacy, Gondur, Dhule 424002, Maharashtra, India
- Division
of Computer Aided Drug Design, Department of Pharmaceutical Chemistry, R. C. Patel Institute of Pharmaceutical Education
and Research, Shirpur 425405, Maharashtra, India
| | - Harun Patel
- Division
of Computer Aided Drug Design, Department of Pharmaceutical Chemistry, R. C. Patel Institute of Pharmaceutical Education
and Research, Shirpur 425405, Maharashtra, India
| | - Bijo Mathew
- Department
of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa
Vidyapeetham, AIMS Health Sciences Campus, Kochi 682 041, India
| | - Hoon Kim
- Department
of Pharmacy, and Research Institute of Life Pharmaceutical Sciences, Sunchon National University, Suncheon 57922, Republic of Korea
| |
Collapse
|
19
|
Zala AR, Rajani DP, Ahmad I, Patel H, Kumari P. Synthesis, characterization, molecular dynamic simulation, and biological assessment of cinnamates linked to imidazole/benzimidazole as a CYP51 inhibitor. J Biomol Struct Dyn 2023; 41:11518-11534. [PMID: 36691770 DOI: 10.1080/07391102.2023.2170918] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 12/26/2022] [Indexed: 01/25/2023]
Abstract
A class of 2-(1H-imidazol-1-yl)-1-phenylethyl cinnamates 6a-6j and 2-(1H-benzo[d]imidazol-1-yl)-1-phenylethyl cinnamates 7a-7j were synthesized, and their synthesis was validated using various spectroscopic techniques like IR, NMR, and Mass spectrometry. In addition, the compounds were assessed for in-vitro antibacterial against gram-positive and gram-negative strains and in-vitro antifungal against six different fungal strains. Compounds 6 g, 7 b, 7f, and 7 g exhibited significant activity against all bacterial strains ranging from MIC = 12.5-50 µg/mL, and compounds 6 g, 7 b, and 7 g exhibited considerable activity against all fungal strains ranging from MFC = 125-200 µg/mL. A molecular docking study indicated that compounds 6 g, 7 b, 7 g, and 7j could be lodged in the active pocket and inhibit C. albicans Sterol 14α-demethylase (CYP51) protein via various interactions, and these studies validate the antifungal results. Different parameters from the 100 ns MD simulation study are investigated to evaluate the dynamic stability of protein-ligand complexes. According to the MD simulation study, the proposed compounds effectively kept their molecular interaction and structural integrity within the C. albicans Sterol 14-demethylase. Compounds 6 g, 7 b, and 7 g are promising lead compounds in searching for novel antifungal drug-like molecules. Furthermore, in silico ADME indicates that these compounds possess drug-like physicochemical properties to be orally bioavailable.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Ajayrajsinh R Zala
- Department of Chemistry, S.V. National Institute of Technology, Surat, India
| | | | - Iqrar Ahmad
- Department of Pharmaceutical Chemistry, Prof. Ravindra Nikam College of Pharmacy, Gondur, India
| | - Harun Patel
- Division of Computer Aided Drug Design, Department of Pharmaceutical Chemistry, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, India
| | - Premlata Kumari
- Department of Chemistry, S.V. National Institute of Technology, Surat, India
| |
Collapse
|
20
|
Benjamin I, Louis H, O. Ekpen F, Gber TE, Gideon ME, Ahmad I, Unimuke TO, P. Akanimo N, Patel H, Eko IJ, Simon O, Agwamba EC, Ejiofor EU. Modeling the anti-Methicillin-Resistant Staphylococcus aureus (MRSA) Activity of (E)-6-chloro-N 2-phenyl-N 4-(4-Phenyl-5-(Phenyl Diazinyl)-2λ 3, 3 λ 2- Thiazol-2-yl)-1, 3, 5-Triazine-2,4- Diamine. Polycycl Aromat Compd 2022. [DOI: 10.1080/10406638.2022.2160773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Innocent Benjamin
- Computational and Bio-Simulation Research Group, University of Calabar, Calabar, Nigeria
- Department of Microbiology, Faculty of Biological Sciences, University of Calabar, Calabar, Nigeria
| | - Hitler Louis
- Computational and Bio-Simulation Research Group, University of Calabar, Calabar, Nigeria
- Department of Pure and Applied Chemistry, Faculty of Physical Sciences, University of Calabar, Calabar, Nigeria
| | - Francis O. Ekpen
- Computational and Bio-Simulation Research Group, University of Calabar, Calabar, Nigeria
| | - Terkumbur E. Gber
- Computational and Bio-Simulation Research Group, University of Calabar, Calabar, Nigeria
- Department of Pure and Applied Chemistry, Faculty of Physical Sciences, University of Calabar, Calabar, Nigeria
| | - Mathias E. Gideon
- Computational and Bio-Simulation Research Group, University of Calabar, Calabar, Nigeria
- Department of Pure and Applied Chemistry, Faculty of Physical Sciences, University of Calabar, Calabar, Nigeria
| | - Iqrar Ahmad
- Division of Computer Aided Drug Design, Department of Pharmaceutical Chemistry, R. C. Patel; Institute of Pharmaceutical Education and Research, Shirpur, India
| | - Tomsmith O. Unimuke
- Computational and Bio-Simulation Research Group, University of Calabar, Calabar, Nigeria
- Department of Pure and Applied Chemistry, Faculty of Physical Sciences, University of Calabar, Calabar, Nigeria
| | - Nyong P. Akanimo
- Computational and Bio-Simulation Research Group, University of Calabar, Calabar, Nigeria
| | - Harun Patel
- Division of Computer Aided Drug Design, Department of Pharmaceutical Chemistry, R. C. Patel; Institute of Pharmaceutical Education and Research, Shirpur, India
| | | | - Ojima Simon
- Computational and Bio-Simulation Research Group, University of Calabar, Calabar, Nigeria
| | - Ernest C. Agwamba
- Computational and Bio-Simulation Research Group, University of Calabar, Calabar, Nigeria
| | - Emmanuel U. Ejiofor
- Computational and Bio-Simulation Research Group, University of Calabar, Calabar, Nigeria
| |
Collapse
|
21
|
Noumi E, Ahmad I, Bouali N, Patel H, Ghannay S, ALrashidi AA, Abdulhakeem MA, Patel M, Ceylan O, Badraoui R, Mousa Elayyan AE, Adnan M, Kadri A, Snoussi M. Thymus musilii Velen. Methanolic Extract: In Vitro and In Silico Screening of Its Antimicrobial, Antioxidant, Anti-Quorum Sensing, Antibiofilm, and Anticancer Activities. Life (Basel) 2022; 13:62. [PMID: 36676011 PMCID: PMC9862435 DOI: 10.3390/life13010062] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/19/2022] [Accepted: 12/20/2022] [Indexed: 12/28/2022] Open
Abstract
Thymus musilii Velen. is a rare plant species cultivated in the Ha'il region (Saudi Arabia) under greenhouse conditions. In this work, we described, for the first time, the phytochemical composition, antimicrobial, antioxidant, anti-quorum sensing, and anticancer activities of T. musilii methanolic extract using both experimental and computational approaches. The obtained results showed the identification of eight small-like peptides and eighteen phyto-compounds by using high-resolution liquid chromatography-mass spectrometry (HR-LCMS) dominated mainly by compounds belonging to isoprenoid, fatty acyl, flavonoid, and alkaloid classes. The tested extracts exhibited high antifungal and antibacterial activity with the mean diameter of growth inhibition zones ranging from 12.33 ± 0.57 mm (Pseudomonas aeruginosa ATCC 27853) to 29.33 ± 1.15 mm (Candida albicans ATCC 10231). Low minimal inhibitory concentrations were recorded for the tested micro-organisms ranging from 0.781 mg/mL to 12.5 mg/mL. While higher doses were necessary to completely kill all tested bacterial and fungal strains. Thyme extract was able to scavenge DPPH•, ABTS•+, β-carotene, and FRAP free radicals, and the IC50 values were 0.077 ± 0.0015 mg/mL, 0.040 ± 0.011 mg/mL, 0.287 ± 0.012 mg/mL, and 0.106 ± 0.007 mg/mL, respectively. The highest percentage of swarming and swimming inhibition was recorded at 100 µg/mL with 39.73 ± 1.5% and 25.18 ± 1%, respectively. The highest percentage of biofilm inhibition was recorded at 10 mg/mL for S. typhimurium ATCC 14028 (53.96 ± 4.21%) and L. monocytogenes ATCC 7644 (49.54 ± 4.5 mg/mL). The in silico docking study revealed that the observed antimicrobial, antioxidant, and anticancer activities of the constituent compounds of T. musilii are thermodynamically feasible, notably, such as those of the tripeptides (Asn-Met-His, His-Cys-Asn, and Phe-His-Gln), isoprenoids (10-Hydroxyloganin), and diterpene glycosides (4-Ketoretinoic acid glucuronide).
Collapse
Affiliation(s)
- Emira Noumi
- Department of Biology, College of Science, University of Ha'il, P.O. Box 2440, Hail 81451, Saudi Arabia
- Laboratory of Genetics, Biodiversity and Valorization of Bio-Resources (LR11ES41), Higher Institute of Biotechnology of Monastir, University of Monastir, Avenue Tahar Haddad, BP74, Monastir 5000, Tunisia
| | - Iqrar Ahmad
- Department of Pharmaceutical Chemistry, Prof. Ravindra Nikam College of Pharmacy, Gondur, Dhule 424002, India
| | - Nouha Bouali
- Department of Biology, College of Science, University of Ha'il, P.O. Box 2440, Hail 81451, Saudi Arabia
| | - Harun Patel
- Division of Computer Aided Drug Design, Department of Pharmaceutical Chemistry, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur 425405, India
| | - Siwar Ghannay
- Department of Chemistry, College of Science, Qassim University, P.O. Box 6688, Buraidah 51452, Saudi Arabia
| | - Ayshah Aysh ALrashidi
- Department of Biology, College of Science, University of Ha'il, P.O. Box 2440, Hail 81451, Saudi Arabia
| | - Mohammad A Abdulhakeem
- Department of Biology, College of Science, University of Ha'il, P.O. Box 2440, Hail 81451, Saudi Arabia
| | - Mitesh Patel
- Centre of Research for Development, Department of Biotechnology, Parul Institute of Applied Sciences, Parul University, Vadodara 391760, India
| | - Ozgur Ceylan
- Ula Ali Kocman Vocational School, Mugla Sitki Kocman University, Mugla 48147, Turkey
| | - Riadh Badraoui
- Department of Biology, College of Science, University of Ha'il, P.O. Box 2440, Hail 81451, Saudi Arabia
- Department of Histo Embryology and Cytogenetics, Medicine Faculty of Sfax, University of Sfax, Road of Majida Boulia, Sfax 3029, Tunisia
| | - Afnan Elayyan Mousa Elayyan
- Department of Clinical Laboratory Science, College of Applied Sciences-Qurayyat, Jouf University, P.O. Box 2014, Sakaka 72388, Saudi Arabia
| | - Mohd Adnan
- Department of Biology, College of Science, University of Ha'il, P.O. Box 2440, Hail 81451, Saudi Arabia
| | - Adel Kadri
- Faculty of Science and Arts in Baljurashi, Albaha University, P.O. Box 1988, Albaha 65527, Saudi Arabia
- Department of Chemistry, Faculty of Science of Sfax, University of Sfax, B.P. 1171, Sfax 3000, Tunisia
| | - Mejdi Snoussi
- Department of Biology, College of Science, University of Ha'il, P.O. Box 2440, Hail 81451, Saudi Arabia
- Laboratory of Genetics, Biodiversity and Valorization of Bio-Resources (LR11ES41), Higher Institute of Biotechnology of Monastir, University of Monastir, Avenue Tahar Haddad, BP74, Monastir 5000, Tunisia
| |
Collapse
|
22
|
Desai NC, Joshi SB, Khasiya AG, Jadeja DJ, Mehta HK, Pandya M, Ahmad I, Patel H. Pyrazolo-imidazolidinones: Synthesis, antimicrobial assessment and molecular modelling studies by molecular mechanic and quantum mechanic approach. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.134000] [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]
|
23
|
Snoussi M, Ahmad I, Aljohani AMA, Patel H, Abdulhakeem MA, Alhazmi YS, Tepe B, Adnan M, Siddiqui AJ, Sarikurkcu C, Riadh B, De Feo V, Alreshidi M, Noumi E. Phytochemical Analysis, Antioxidant, and Antimicrobial Activities of Ducrosia flabellifolia: A Combined Experimental and Computational Approaches. Antioxidants (Basel) 2022; 11:2174. [PMID: 36358545 PMCID: PMC9686979 DOI: 10.3390/antiox11112174] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/27/2022] [Accepted: 10/28/2022] [Indexed: 09/02/2023] Open
Abstract
Ducrosia flabellifolia Boiss. is a rare desert plant known to be a promising source of bioactive compounds. In this paper, we report for the first time the phytochemical composition and biological activities of D. flabellifolia hydroalcoholic extract by using liquid chromatography-electrospray tandem mass spectrometry (ESI-MS/MS) technique. The results obtained showed the richness of the tested extract in phenols, tannins, and flavonoids. Twenty-three phytoconstituents were identified, represented mainly by chlorogenic acid, followed by ferulic acid, caffeic acid, and sinapic acid. The tested hydroalcoholic extract was able to inhibit the growth of all tested bacteria and yeast on agar Petri dishes at 3 mg/disc with mean growth inhibition zone ranging from 8.00 ± 0.00 mm for Enterococcus cloacae (E. cloacae) to 36.33 ± 0.58 mm for Staphylococcus epidermidis. Minimal inhibitory concentration ranged from 12.5 mg/mL to 200 mg/mL and the hydroalcoholic extract from D. flabellifolia exhibited a bacteriostatic and fungistatic character. In addition, D. flabellifolia hydroalcoholic extract possessed a good ability to scavenge different free radicals as compared to standard molecules. Molecular docking studies on the identified phyto-compounds in bacterial, fungal, and human peroxiredoxin 5 receptors were performed to corroborate the in vitro results, which revealed good binding profiles on the examined protein targets. A standard atomistic 100 ns dynamic simulation investigation was used to further evaluate the interaction stability of the promising phytocompounds, and the results showed conformational stability in the binding cavity. The obtained results highlighted the medicinal use of D. flabellifolia as source of bioactive compounds, as antioxidant, antibacterial, and antifungal agent.
Collapse
Affiliation(s)
- Mejdi Snoussi
- Department of Biology, College of Science, University of Hail, Ha’il 2440, Saudi Arabia
- Laboratory of Genetics, Biodiversity and Valorization of Bio-Resources (LR11ES41), Higher Institute of Biotechnology of Monastir, University of Monastir, Avenue Tahar Haddad, BP74, Monastir 5000, Tunisia
| | - Iqrar Ahmad
- Division of Computer Aided Drug Design, Department of Pharmaceutical Chemistry, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur 425405, Maharashtra, India
| | | | - Harun Patel
- Division of Computer Aided Drug Design, Department of Pharmaceutical Chemistry, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur 425405, Maharashtra, India
| | | | - Yasser S. Alhazmi
- Department of Biology, College of Science, University of Hail, Ha’il 2440, Saudi Arabia
| | - Bektas Tepe
- Department of Molecular Biology and Genetics, Faculty of Science and Literature, TR-79000 Kilis, Turkey
| | - Mohd Adnan
- Department of Biology, College of Science, University of Hail, Ha’il 2440, Saudi Arabia
| | - Arif J. Siddiqui
- Department of Biology, College of Science, University of Hail, Ha’il 2440, Saudi Arabia
| | - Cengiz Sarikurkcu
- Faculty of Pharmacy, Afyonkarahisar Health Sciences University, TR-03100 Afyonkarahisar, Turkey
| | - Badraoui Riadh
- Department of Biology, College of Science, University of Hail, Ha’il 2440, Saudi Arabia
- Section of Histology Cytology, Medicine Faculty of Tunis, University of Tunis El Manar, La Rabta 1007, Road Djebal Lakhdhar, Tunis 1007, Tunisia
- Department of HistoEmbryology and Cytogenetics, Medicine Faculty of Sfax, University of Sfax, Road of Majida Boulia, Sfax 3029, Tunisia
| | - Vincenzo De Feo
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132, Fisciano, 84084 Salerno, Italy
| | - Mousa Alreshidi
- Department of Biology, College of Science, University of Hail, Ha’il 2440, Saudi Arabia
- Molecular Diagnostics and Personalized Therapeutics Unit, University of Hail, Ha’il 2440, Saudi Arabia
| | - Emira Noumi
- Department of Biology, College of Science, University of Hail, Ha’il 2440, Saudi Arabia
| |
Collapse
|
24
|
Evaluation of oxindole derivatives as a potential anticancer agent against breast carcinoma cells: In vitro, in silico, and molecular docking study. Toxicol In Vitro 2022; 86:105517. [DOI: 10.1016/j.tiv.2022.105517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 11/08/2022] [Accepted: 11/10/2022] [Indexed: 11/16/2022]
|
25
|
Multifunctional Derivatives of Spiropyrrolidine Tethered Indeno-Quinoxaline Heterocyclic Hybrids as Potent Antimicrobial, Antioxidant and Antidiabetic Agents: Design, Synthesis, In Vitro and In Silico Approaches. Molecules 2022; 27:molecules27217248. [PMID: 36364077 PMCID: PMC9653804 DOI: 10.3390/molecules27217248] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 10/12/2022] [Accepted: 10/17/2022] [Indexed: 12/04/2022] Open
Abstract
To combat emerging antimicrobial-resistant microbes, there is an urgent need to develop new antimicrobials with better therapeutic profiles. For this, a series of 13 new spiropyrrolidine derivatives were designed, synthesized, characterized and evaluated for their in vitro antimicrobial, antioxidant and antidiabetic potential. Antimicrobial results revealed that the designed compounds displayed good activity against clinical isolated strains, with 5d being the most potent (MIC 3.95 mM against Staphylococcus aureus ATCC 25923) compared to tetracycline (MIC 576.01 mM). The antioxidant activity was assessed by trapping DPPH, ABTS and FRAP assays. The results suggest remarkable antioxidant potential of all synthesized compounds, particularly 5c, exhibiting the strongest activity with IC50 of 3.26 ± 0.32 mM (DPPH), 7.03 ± 0.07 mM (ABTS) and 3.69 ± 0.72 mM (FRAP). Tested for their α-amylase inhibitory effect, the examined analogues display a variable degree of α-amylase activity with IC50 ranging between 0.55 ± 0.38 mM and 2.19 ± 0.23 mM compared to acarbose (IC50 1.19 ± 0.02 mM), with the most active compounds being 5d, followed by 5c and 5j, affording IC50 of 0.55 ± 0.38 mM, 0.92 ± 0.10 mM, and 0.95 ± 0.14 mM, respectively. Preliminary structure–activity relationships revealed the importance of such substituents in enhancing the activity. Furthermore, the ADME screening test was applied to optimize the physicochemical properties and determine their drug-like characteristics. Binding interactions and stability between ligands and active residues of the investigated enzymes were confirmed through molecular docking and dynamic simulation study. These findings provided guidance for further developing leading new spiropyrrolidine scaffolds with improved dual antimicrobial and antidiabetic activities.
Collapse
|