1
|
Priyanka, Rani P, Ranolia A, Kiran, Joshi G, Singh S, Kumar R, Wahajuddin M, Kumar P, Singh D, Sindhu J. Design and synthesis of new 1,4-naphthoquinones appended sulfenylated thiazoles as cyclooxygenase II inhibitors: Exploring the utility in the development of anticancer agents. Bioorg Chem 2025; 161:108537. [PMID: 40334422 DOI: 10.1016/j.bioorg.2025.108537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 04/05/2025] [Accepted: 04/25/2025] [Indexed: 05/09/2025]
Abstract
Clinical evaluations revealed the direct role of chronic inflammation in cancer progression. The cyclooxygenase (COX) pathway is particularly important among the various pathways involved in inflammation. There are two COX isoforms: COX-1 and COX-2. The importance of drug discovery lies in selectively inhibiting COX-2, an enzyme expressed during inflammation, unlike COX-1, which is constitutively active. The inhibition of COX-1 is correlated further with gastric ulcers. However, recently approved COX-2 inhibitors have intricate cardiotoxicity, thus creating an utmost need for new COX-2 inhibitors. Considering this, in our present research, we rationally designed and synthesized a series (4a-4q) of 1,4-naphthoquinones appended sulfenylated thiazoles using molecular hybridization approach under metal-free conditions. These synthetics were explored for their anticancer potential against three cell lines, and the hits portraying anticancer effects were further tested against COX isoforms. Among all, compounds 4d and 4f were found to be potent anticancer leads, exhibiting selective inhibition of COX-2 and, concomitantly, the lipoxygenase (LOX) pathway, both of which share the same substrate, arachidonic acid. Both compounds were found to reduce oxidative stress and induce cancer cell death via apoptosis pathway. The experimental outcome was further corroborated using in silico techniques, including density field theory (DFT), Molecular docking, and dynamics.
Collapse
Affiliation(s)
- Priyanka
- Department of Chemistry, COBS&H, CCS Haryana Agricultural University, Hisar 125004, India
| | - Payal Rani
- Department of Chemistry, COBS&H, CCS Haryana Agricultural University, Hisar 125004, India
| | - Anju Ranolia
- Department of Chemistry, COBS&H, CCS Haryana Agricultural University, Hisar 125004, India
| | - Kiran
- Department of Chemistry, COBS&H, CCS Haryana Agricultural University, Hisar 125004, India
| | - Gaurav Joshi
- Department of Pharmaceutical Sciences, Chauras Campus, HNB Garhwal University (A Central University), Srinagar, Uttarakhand 246174, India; Institute of Cancer Therapeutics, School of Pharmacy and Medical Sciences, University of Bradford, Bradford, West Yorkshire BD7 1BD, United Kingdom.
| | - Snigdha Singh
- Department of Chemistry, University of Delhi, 110007, India
| | - Roshan Kumar
- Department of Microbiology, Central University of Punjab, VPO-Ghudda, Punjab 151401, India; Department of Microbiology, Graphic Era (Deemed to be University) Clement Town, Dehradun 248002, India
| | - Muhammad Wahajuddin
- Institute of Cancer Therapeutics, School of Pharmacy and Medical Sciences, University of Bradford, Bradford, West Yorkshire BD7 1BD, United Kingdom
| | - Parvin Kumar
- Department of Chemistry, Kurukshetra University, Kurukshetra 136119, India
| | - Devender Singh
- Department of Chemistry, Maharshi Dayanand University, Rohtak 124001, India
| | - Jayant Sindhu
- Department of Chemistry, COBS&H, CCS Haryana Agricultural University, Hisar 125004, India.
| |
Collapse
|
2
|
Huwaimel B, Younes KM, Alsaab HO, Alzahrani RM, Alobaida A, Abouzied AS. Enhancing thermal stability of pectinase using thermal titration molecular dynamics and density functional theory approach. J Biomol Struct Dyn 2025:1-18. [PMID: 40395215 DOI: 10.1080/07391102.2025.2505100] [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: 01/18/2025] [Accepted: 05/07/2025] [Indexed: 05/22/2025]
Abstract
Pectinase, an enzyme primarily produced from Aspergillus niger, is essential in various industrial applications. However, the enzyme's functionality at high temperatures is challenging, restricting its effectiveness and potential uses. Therefore, the present study investigated the potential of peptide binding to enhance the thermal stability of pectinase. Thermal titration molecular dynamics (MD) simulations were performed at 300, 320, 340 and 360 K to identify regions susceptible to thermal fluctuations. Based on these results, 235,200 peptide sequences were screened to target the detected unstable regions. Machine learning models predicted the peptide activity and 12 promising peptide-protein complexes were identified using docking. Binding free energy calculations showed pep-10 (-19.4 kcal/mol), pep-8 (-17.97 kcal/mol), pep-12 (-15.25 kcal/mol) and pep-6 (-9.86 kcal/mol) as the most promising candidates to improve the thermal stability. Density functional theory calculations showed that pep-12 had the lowest energy of -2365. MD simulations at 360 K for 100 ns demonstrated that pep-12 maintained the most stable conformation with root mean square deviation (0.2-0.25 nm) compared to other peptides. Quantum mechanics/molecular mechanics hybrid approach to examine the mechanism of the pep-12 complex with Pectinase. The outcomes of this study suggested that pep-12 is the most potential candidate for enhancing pectinase thermal stability.
Collapse
Affiliation(s)
- Bader Huwaimel
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Ha'il, Ha'il, Saudi Arabia
- Medical and Diagnostic Research Center, University of Ha'il, Ha'il, Saudi Arabia
| | - Kareem M Younes
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Ha'il, Ha'il, Saudi Arabia
| | - Hashem O Alsaab
- Department of Pharmaceutics and Industrial Pharmacy, College of Pharmacy, Taif University, Taif, Saudi Arabia
| | - Rami M Alzahrani
- Department of Pharmaceutics and Industrial Pharmacy, College of Pharmacy, Taif University, Taif, Saudi Arabia
| | - Ahmed Alobaida
- Department of Pharmaceutics, College of Pharmacy, University of Ha'il, Ha'il, Saudi Arabia
| | - Amr S Abouzied
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Ha'il, Ha'il, Saudi Arabia
| |
Collapse
|
3
|
Gu J, Sim BR, Li J, Yu Y, Qin L, Wu L, Liu H, Xu Y, Zhao YL, Nie Y. Coevolution-based protein engineering of alcohol dehydrogenase at distal sites enables enzymatic compatibility with substrate diversity and stereoselectivity. Int J Biol Macromol 2025; 306:141233. [PMID: 39993679 DOI: 10.1016/j.ijbiomac.2025.141233] [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: 09/04/2024] [Revised: 01/16/2025] [Accepted: 02/16/2025] [Indexed: 02/26/2025]
Abstract
Chiral alcohols with various substituents and functional groups are attractive synthesizers in many fields. Biocatalysts have attracted great interest for their use in " sustainable chemistry". However, substrate specificity of enzymes limits their widespread use as "generalists" in biocatalysis. In addition, engineering enzymes for simultaneously improving catalytic efficiency and stereoselectivity for structurally diverse substrates is a contemporary challenge. Inspired by naturally occurring coevolution of residues dedicated to a particular function and clustered together in space, we applied coevolution-based engineering to the alcohol dehydrogenase CpRCR from Candida parapsilosis to identify distal sites which can synergistically improve the catalytic properties of diverse substrates. Five variants were developed by clustering the coupling strength and structure of coevolutionary sites which showed improved (up to 28-fold) catalytic efficiency with high stereoselectivity toward 16 structurally diverse substrates (aryl ketones, heterocyclic ketones and β-ketoesters). In particular, for substrate 2-acetylpyridine, the specific activity of K191L/D216H is 12-fold higher than the previously reported highest activity of alcohol dehydrogenase. Theses distal mutations do not directly modify the active center but rather modulate catalytic capacity in various allosteric ways favoring substrate diversity. This study provides a broadly applicable strategy for protein engineering and expanded the applications of biocatalyst on value-added chemicals.
Collapse
Affiliation(s)
- Jie Gu
- Lab of Brewing Microbiology and Applied Enzymology, School of Biotechnology and Key Laboratory of Industrial Biotechnology of Ministry of Education, Jiangnan University, Wuxi 214122, China
| | - Byu Ri Sim
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic and Developmental Sciences, MOE-LSB & MOE-LSC, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China; Department of Biochemistry, University of Toronto, Ontario M5S 3H6, Canada
| | - Jiarui Li
- Lab of Brewing Microbiology and Applied Enzymology, School of Biotechnology and Key Laboratory of Industrial Biotechnology of Ministry of Education, Jiangnan University, Wuxi 214122, China
| | - Yangqing Yu
- Lab of Brewing Microbiology and Applied Enzymology, School of Biotechnology and Key Laboratory of Industrial Biotechnology of Ministry of Education, Jiangnan University, Wuxi 214122, China
| | - Lei Qin
- Lab of Brewing Microbiology and Applied Enzymology, School of Biotechnology and Key Laboratory of Industrial Biotechnology of Ministry of Education, Jiangnan University, Wuxi 214122, China
| | - Lunjie Wu
- Lab of Brewing Microbiology and Applied Enzymology, School of Biotechnology and Key Laboratory of Industrial Biotechnology of Ministry of Education, Jiangnan University, Wuxi 214122, China
| | - Huan Liu
- Lab of Brewing Microbiology and Applied Enzymology, School of Biotechnology and Key Laboratory of Industrial Biotechnology of Ministry of Education, Jiangnan University, Wuxi 214122, China
| | - Yan Xu
- Lab of Brewing Microbiology and Applied Enzymology, School of Biotechnology and Key Laboratory of Industrial Biotechnology of Ministry of Education, Jiangnan University, Wuxi 214122, China; State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Yi-Lei Zhao
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic and Developmental Sciences, MOE-LSB & MOE-LSC, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Yao Nie
- Lab of Brewing Microbiology and Applied Enzymology, School of Biotechnology and Key Laboratory of Industrial Biotechnology of Ministry of Education, Jiangnan University, Wuxi 214122, China.
| |
Collapse
|
4
|
Rai GP, Shanker A. The coevolutionary landscape of drug resistance in epidermal growth factor receptor: A cancer perspective. Comput Biol Med 2025; 189:110001. [PMID: 40073493 DOI: 10.1016/j.compbiomed.2025.110001] [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: 09/25/2024] [Revised: 03/03/2025] [Accepted: 03/04/2025] [Indexed: 03/14/2025]
Abstract
Epidermal growth factor receptor (EGFR), the first receptor tyrosine kinase, plays a critical role in neoplastic metastasis, angiogenesis, tumor invasion, and apoptosis, making it a prime target for treating non-small cell lung cancer (NSCLC). Although tyrosine kinase inhibitors (TKIs) have shown high efficacy and promise for cancer patients, resistance to these drugs often develops within a year due to alterations. The present study investigates the compensatory alterations in EGFR to understand the evolutionary process behind drug resistance. Our findings reveal that coevolutionary alterations expand the drug-binding pocket; leading to reduced drug efficacy and suggested that such changes significantly influence the structural adaptation of the EGFR against these drugs. Analysis such as root mean square deviation (RMSD), root mean square fluctuation (RMSF), solvent accessible surface area (SASA), principal component analysis (PCA), and free energy landscape (FEL) demonstrated that structures of wild EGFR docked with gefitinib are more stable which suggests its susceptibility towards drug than coevolution dependent double mutant. The findings were supported by MM-GBSA binding affinity analysis. The insights from this study highlighted the evolution-induced structural changes which contributes to drug resistance in EGFR and may certainly aid in designing more effective drugs.
Collapse
Affiliation(s)
- Gyan Prakash Rai
- Department of Bioinformatics, Central University of South Bihar, Gaya, Bihar, 824236, India
| | - Asheesh Shanker
- Department of Bioinformatics, Central University of South Bihar, Gaya, Bihar, 824236, India.
| |
Collapse
|
5
|
Bhattacharjee A, Kar S, Ojha PK. Ligand-based cheminformatics and free energy-inspired molecular simulations for prioritizing and optimizing G-protein coupled receptor kinase-6 (GRK6) inhibitors in multiple myeloma treatment. Comput Biol Chem 2025; 115:108347. [PMID: 39824142 DOI: 10.1016/j.compbiolchem.2025.108347] [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: 10/26/2024] [Revised: 01/02/2025] [Accepted: 01/04/2025] [Indexed: 01/20/2025]
Abstract
Multiple myeloma (MM) is the second most frequently diagnosed hematological malignancy, presenting limited treatment options with no curative potential and significant drug resistance. Recent studies involving genetic knockdown established the crucial role of GRK6 in upholding the viability of MM cells, emphasizing the need to identify potential inhibitors. Computational exploration of GRK6 inhibitors has not been attempted previously. Herein, the present study reports a multilayered lead prioritization and optimization framework using chemometrics and molecular simulations. 2D QSAR studies revealed that hydrogen bonding and polar interactions enhanced GRK6 inhibitory activity, while increased electron accessibility posed a risk of off-target effects. The pharmacophore hypothesis (DDHRRR_1) featured two hydrogen bond donors, one hydrophobic region, and three aromatic rings, laying the foundation for the 3D QSAR models. Hydrophobic groups, such as pyridine and pyrazole, were shown to enhance inhibition, while smaller groups, like ethyl and hydroxyl, reduced activity. 12,557 DrugBank compounds were screened using the developed chemometric models and molecular docking in tandem, which led to the identification of 7 potential parent leads for subsequent QSAR-guided structural optimizations. 350 lead analogs were generated and the top 4 were further analyzed using molecular docking, ADMET, molecular dynamics, and metadynamics analysis based on Principal Component Analysis (PCA), Probability Density Function (PDF), and Free Energy Landscapes (FEL). Upon cumulative retrospection, we propose a novel analog of DB07168 (DB07168-A13) (docking score: -11.2 kcal/mol, MM-GBSA binding energy: -55.2 kcal/mol) as the most promising GRK6 inhibitor, warranting further in vitro validation, for addressing prospective therapeutic intervention in MM.
Collapse
Affiliation(s)
- Arnab Bhattacharjee
- Drug Discovery and Development Laboratory (DDD Lab), Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India
| | - Supratik Kar
- Chemometrics and Molecular Modeling Laboratory, Department of Chemistry and Physics, Kean University, 1000 Morris Avenue, Union, NJ 07083, USA
| | - Probir Kumar Ojha
- Drug Discovery and Development Laboratory (DDD Lab), Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India.
| |
Collapse
|
6
|
Er-Rajy M, El Fadili M, Zarougui S, Mujwar S, Aloui M, Zerrouk M, Hammouti B, Rhazi L, Sabbahi R, Alanazi MM, Azzaoui K, Salghi R, Elhallaoui M. Design and evaluation of novel triazole derivatives as potential anti-gout inhibitors: a comprehensive molecular modeling study. Front Chem 2025; 13:1518777. [PMID: 40115054 PMCID: PMC11922854 DOI: 10.3389/fchem.2025.1518777] [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: 10/28/2024] [Accepted: 01/30/2025] [Indexed: 03/22/2025] Open
Abstract
Introduction Gout is the most common inflammatory arthritis, characterized by hyperuricemia, tophus formation, joint disease, and kidney stones. Uric acid, the final byproduct of purine catabolism, is eliminated via the kidneys and digestive system. Xanthine oxidase (XO) catalyzes the conversion of hypoxanthine and xanthine into uric acid, making XO inhibitors crucial for treating hyperuricemia and gout. Currently, three XO inhibitors are clinically used, showing significant efficacy. A molecular modeling study on triazole derivatives aims to identify novel XO inhibitors using 3D-QSAR, molecular docking, MD simulations, ADMET analysis, and DFT calculations. These computational approaches facilitate drug discovery while reducing research costs. Methods Our work focuses on a series of synthesized anti-xanthine oxidase inhibitors, aiming to develop new inhibitors. A computational study was carried out to identify the xanthine oxidase inhibitory structural features of a series of triazole inhibitors using computational method. Results A model based on CoMFA and CoMSIA/SEA has been built to predict new triazole derivatives. Discussion The optimal model established from CoMFA and CoMSIA/SEA was successfully evaluated for its predictive capability. Visualization of the contour maps of both models showed that modifying the substituents plays a key role in enhancing the biological activity of anti-gout inhibitors. Molecular docking results for complexes N°8-3NVY and N°22-3NVY showed scores of -7.22 kcal/mol and -8.36 kcal/mol, respectively, indicating substantial affinity for the enzyme. Complex N°8-3NVY forms two hydrogen bonds with SER 69 and ASN 71, three alkyl bonds with ALA 70, LEU 74, and ALA 75, and one Pi-Pi T-shaped bond with PHE 68. Complex N°22-3NVY forms three hydrogen bonds with HIS 99, ARG 29, and ILE 91, and one halogen bond with LEU 128 at 3.60 Å. A MD study revealed that the N°22-3NVY complex remained highly stable throughout the simulation. Therefore, we proposed six new molecules, their anti-gout inhibitory activities were predicted using two models, and they were evaluated for Lipinski's rule, and ADMET properties. The results show that both Pred 4 and Pred 5 have better pharmacokinetic properties than the height potent molecule in the studied series, making these two compounds valuable candidates for new anti-gout drugs. Subsequently, using DFT study to evaluate the chemical reactivity properties of these two proposed compounds, the energy gap results revealed that both molecules exhibit moderate chemical stability and reactivity.
Collapse
Affiliation(s)
- Mohammed Er-Rajy
- LIMAS Laboratory, Faculty of Sciences Dhar El Mahraz, Sidi Mohamed Ben Abdellah University, Fez, Morocco
- Euromed University of Fes, UMF, Fez, Morocco
| | - Mohamed El Fadili
- LIMAS Laboratory, Faculty of Sciences Dhar El Mahraz, Sidi Mohamed Ben Abdellah University, Fez, Morocco
| | - Sara Zarougui
- LIMAS Laboratory, Faculty of Sciences Dhar El Mahraz, Sidi Mohamed Ben Abdellah University, Fez, Morocco
| | - Somdutt Mujwar
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Mourad Aloui
- LIMAS Laboratory, Faculty of Sciences Dhar El Mahraz, Sidi Mohamed Ben Abdellah University, Fez, Morocco
| | - Mohammed Zerrouk
- Engineering Laboratory of Organometallic, Molecular Materials and Environment, Faculty of Sciences, Sidi Mohamed Ben Abdellah University, Fez, Morocco
| | | | - Larbi Rhazi
- Institut Polytechnique UniLaSalle, Université d'Artois, Beauvais, France
| | - Rachid Sabbahi
- Research Team in Science and Technology, Higher School of Technology, Ibn Zohr University, Laayoune, Morocco
| | - Mohammed M Alanazi
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Khalil Azzaoui
- Engineering Laboratory of Organometallic, Molecular Materials and Environment, Faculty of Sciences, Sidi Mohamed Ben Abdellah University, Fez, Morocco
- Laboratory of Industrial Engineering, Energy and the Environment (LI3E) SUPMTI, Rabat, Morocco
| | - Rachid Salghi
- Laboratory of Applied Chemistry and Environment, National School of Applied Sciences, University Ibn Zohr, Agadir, Morocco
| | - Menana Elhallaoui
- LIMAS Laboratory, Faculty of Sciences Dhar El Mahraz, Sidi Mohamed Ben Abdellah University, Fez, Morocco
| |
Collapse
|
7
|
Nandan PK, Sivaraman J. Elucidating the therapeutic potential of indazole derivative bindarit against K-ras receptor: An in-silico analysis using molecular dynamics exploration. Biochem Biophys Rep 2025; 41:101913. [PMID: 39867680 PMCID: PMC11758134 DOI: 10.1016/j.bbrep.2024.101913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 12/27/2024] [Accepted: 12/30/2024] [Indexed: 01/28/2025] Open
Abstract
Ras gene is frequently mutated in cancer. Among different subtypes of Ras gene, K-Ras mutation occurs in nearly 30 % of human cancers. K-Ras mutation, specifically K-Ras (G12D) mutation is prevalent in cancers like lung, colon and pancreatic cancer. During cancer occurrence, mutant Ras remain in activated form (GTP bound state) for cancer cell proliferation. In the quest for a potential K-Ras inhibitor, nitrogen-containing indazole derivatives can show promise as inhibitors, as they have numerous therapeutic properties like anti-inflammatory, anti-viral and anti-tumor. Furthermore, among various indazole derivatives, "Bindarit" is an important therapeutic compound which could have potential inhibitory action against K-Ras due to its structural resemblance with reference compound "Benzimidazole". So, the current study is an attempt to find out the inhibitory effect of Bindarit against K-Ras activation by binding to a pocket which is adjacent to the switch I/II regions of the K-Ras receptor. AutoDock tool was used to investigate the binding affinity of protein ligand interaction and GROMACS package was utilised to assess their interactions in a dynamic setting. Bindarit shows better binding affinity than reference with binding energy of -7.3 kcal/mol. Upon ligand binding conformational changes take place, which could lead to the loss of GTPase activity. Consequently, further downstream signalling of the K-Ras pathway would be blocked and this could lead to the inhibition of K-Ras dependent cancer cell proliferation. However, further validation of present study can be done through experimental assay such as cytotoxic and protein expression analysis.
Collapse
Affiliation(s)
- Parmar Keshri Nandan
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore, India
| | - Jayanthi Sivaraman
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore, India
| |
Collapse
|
8
|
Yan X, Feng B, Song H, Wang L, Wang Y, Sun Y, Cai X, Rong Y, Wang X, Wang Y. Identification and mechanistic study of piceatannol as a natural xanthine oxidase inhibitor. Int J Biol Macromol 2025; 293:139231. [PMID: 39732228 DOI: 10.1016/j.ijbiomac.2024.139231] [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: 10/04/2024] [Revised: 12/20/2024] [Accepted: 12/24/2024] [Indexed: 12/30/2024]
Abstract
Natural Xanthine oxidase (XOD) inhibitors represent promising therapeutic agents for hyperuricemia (HUA) treatment due to their potent efficacy and favorable safety profiles. This study involved the construction of a comprehensive database of 315 XOD inhibitors and development of 28 machine learning-based QSAR models. The ChemoPy light gradient boosting machine model exhibited the best performance (AUC = 0.9371 and MCC = 0.7423). This model identified three potential XOD inhibitors from the FooDB database: daphnetin, 7-hydroxycoumarin, and piceatannol. Molecular docking and dynamics simulations revealed favorable interactions, with piceatannol showing a remarkable stability through hydrogen bonding and hydrophobic interactions. ADME predictions suggested that all three compounds possess desirable drug-like properties and safety characteristics. Subsequent in vitro enzyme inhibition assays validated computational predictions, with piceatannol exhibiting the strongest inhibitory activity (IC50 = 8.80 ± 0.05 μM). Multispectroscopic analyses revealed that piceatannol-XOD binding was predominantly mediated by hydrogen bonding and van der Waals forces, which induced conformational changes characterized by decreased α-helical content and increased proportions of β-sheets, β-turns, and random coils. This study presents an efficient strategy for the identification of natural XOD inhibitors, elucidates the molecular mechanism of piceatannol-mediated XOD inhibition, and establishes a foundation for its therapeutic application in HUA treatment.
Collapse
Affiliation(s)
- Xinxu Yan
- College of Food Science, Northeast Agricultural University, Harbin 150030, Heilongjiang, China; Western Agricultural Research Center, Chinese Academy of Agricultural Sciences, Changji 831100, PR China
| | - Baolong Feng
- Center for Education Technology, Northeast Agricultural University, Harbin 150030, Heilongjiang, China
| | - Hongjie Song
- College of Food Science, Northeast Agricultural University, Harbin 150030, Heilongjiang, China
| | - Lili Wang
- College of Food Science, Northeast Agricultural University, Harbin 150030, Heilongjiang, China
| | - Yehui Wang
- College of Food Science, Northeast Agricultural University, Harbin 150030, Heilongjiang, China
| | - Yulin Sun
- College of Food Science, Northeast Agricultural University, Harbin 150030, Heilongjiang, China
| | - Xiaoshuang Cai
- College of Food Science, Northeast Agricultural University, Harbin 150030, Heilongjiang, China
| | - Yating Rong
- College of Food Science, Northeast Agricultural University, Harbin 150030, Heilongjiang, China
| | - Xibo Wang
- College of Food Science, Northeast Agricultural University, Harbin 150030, Heilongjiang, China.
| | - Yutang Wang
- Institute of Agro-Products Processing Science and Technology, Chinese Academy of Agricultural Sciences/Key Laboratory of Agro-Products Processing, Ministry of Agriculture, Beijing 100193, China; Western Agricultural Research Center, Chinese Academy of Agricultural Sciences, Changji 831100, PR China.
| |
Collapse
|
9
|
Cabrera M, Armando R, Czarnowski I, Chinestrad P, Blanco R, Zinni A, Gómez D, Mengual Gómez DL, Menna PL. CADD-based discovery of novel oligomeric modulators of PKM2 with antitumor activity in aggressive human glioblastoma models. Heliyon 2025; 11:e42238. [PMID: 39959478 PMCID: PMC11830341 DOI: 10.1016/j.heliyon.2025.e42238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 01/22/2025] [Accepted: 01/22/2025] [Indexed: 02/18/2025] Open
Abstract
Pyruvate kinase isoform M2 (PKM2) is a multifunctional enzyme capable of transitioning between monomeric, dimeric, and tetrameric states, with its oligomeric equilibrium playing a pivotal role in tumour progression and survival. The unique exon ten at the dimer-dimer interface represents an attractive target for isoform-specific modulation, offering opportunities for disrupting this equilibrium and altering tumour cell dynamics. This study identifies a novel druggable pocket at the PKM2 dimer interface through conformational analysis. This pocket was exploited in a virtual screening of a large small-molecule library, identifying two promising candidates, C599 and C998. Both compounds exhibited dose-dependent antiproliferative effects in glioblastoma cell lines and induced apoptosis, as evidenced by caspase 3/7 activation. These effects were directly linked to their inhibition of PKM2 enzymatic activity, validating the proposed mechanism of action in their rational design. ADMET studies further highlighted their strong potential as lead PKM2 inhibitors for GBM treatment. Molecular dynamics (MD) simulations and post-MD analyses, including Dynamic Cross-Correlation Maps (DCCM), Probability Density Function (PDF), and Free Energy Landscape (FEL), confirmed the stability of the protein-ligand interactions and highlighted critical residues at the dimer-dimer interface. The Steered MD simulations demonstrated the high affinity of the compounds for PKM2, as evidenced by the requirement of high rupture forces to induce an unbinding event. These results highlight the potential of the compounds as oligomeric modulators of PKM2. These findings position C599 and C998 as promising lead compounds for antitumor applications. Future studies will focus on optimising these candidates and assessing their efficacy in vivo glioblastoma models, reassuring the thoroughness of our research and the potential for further advancements.
Collapse
Affiliation(s)
- Maia Cabrera
- Laboratorio de Farmacología Molecular, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Argentina
| | - Romina Armando
- Unidad de Oncología Molecular, Centro de Oncología Molecular y Traslacional, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Argentina
| | - Ian Czarnowski
- Laboratorio de Farmacología Molecular, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Argentina
| | - Patricio Chinestrad
- Laboratorio de Farmacología Molecular, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Argentina
| | - Ramiro Blanco
- Laboratorio de Farmacología Molecular, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Argentina
| | - Alejandra Zinni
- Laboratorio de Farmacología Molecular, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Argentina
| | - Daniel Gómez
- Unidad de Oncología Molecular, Centro de Oncología Molecular y Traslacional, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Argentina
| | - Diego L. Mengual Gómez
- Unidad de Oncología Molecular, Centro de Oncología Molecular y Traslacional, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Argentina
| | - Pablo Lorenzano Menna
- Laboratorio de Farmacología Molecular, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Argentina
| |
Collapse
|
10
|
Sehra GE, Azam S, Ahmad S, Ali A, Khan I, Ullah A, Waqas M, Rehman N, Absar M, Alshammari A, Albekairi NA, Wei DQ. Elucidating the resistance mechanisms and binding pattern of novel Oxa-48-like carbapenemases covalent inhibitors: A hybrid experimental and in silico approach. J Mol Struct 2025; 1321:140073. [DOI: 10.1016/j.molstruc.2024.140073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2025]
|
11
|
Bai L, Xiang Y, Shen M, Han Y, Li P, Zuo Z, Li Y. Design, synthesis and activity evaluation of novel quinazolinone compounds as TRPC5 inhibitors. Bioorg Chem 2025; 155:108147. [PMID: 39817997 DOI: 10.1016/j.bioorg.2025.108147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 12/31/2024] [Accepted: 01/05/2025] [Indexed: 01/18/2025]
Abstract
The TRPC5 channel plays an important role in regulating various physiological processes, which is related to various human diseases, especially psychiatric and kidney diseases. Although the TRPC5 channel is one of the essential potential target, no drugs against TRPC5 channels have been granted in the market to date. In this study, based on the structure of hit compound ph1, we further synthesied 49 compounds of novel quinazolinone and heterocyclic fusion pyrimidinone derivatives, and their activities were evaluated by electrophysiological assays. After extensive screening, 21 compounds exhibited significant TRPC5 inhibitory activity, and compounds ph8 and ph14 displayed strong inhibitory with IC50 of 1.28 and 2.16 μM, respectively. These identified potential TRPC5 inhibitor may provide lead compounds and experimental evidence for the development of novel TRPC5 inhibitors with potential treatment for anxiety, depression, and progressive kidney disease.
Collapse
Affiliation(s)
- Longhui Bai
- School of Pharmacology Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024 China; State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201 China; University of the Chinese Academy of Sciences, Beijing 100049 China
| | - Yu Xiang
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023 China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203 China
| | - Meiling Shen
- School of Pharmacology Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024 China; State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201 China; University of the Chinese Academy of Sciences, Beijing 100049 China
| | - Yujun Han
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023 China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203 China
| | - Penghua Li
- School of Pharmacology Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024 China; State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201 China; University of the Chinese Academy of Sciences, Beijing 100049 China
| | - Zhili Zuo
- School of Pharmacology Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024 China; State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201 China; University of the Chinese Academy of Sciences, Beijing 100049 China.
| | - Yang Li
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023 China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203 China; University of the Chinese Academy of Sciences, Beijing 100049 China; National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040 China.
| |
Collapse
|
12
|
Mei Y, Yu Z, Gong Y, Meng R, Ma X, Li H, Li J, Jiang J, Peng Z, Li Y, Song D. Lycorine esters exert anti-HCoV-OC43 effect through reversibly acylating cysteine residue in the nsp 12 NiRAN domain. Bioorg Chem 2025; 154:108086. [PMID: 39721145 DOI: 10.1016/j.bioorg.2024.108086] [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: 09/20/2024] [Revised: 12/06/2024] [Accepted: 12/18/2024] [Indexed: 12/28/2024]
Abstract
By introducing ester warheads into the hydroxyl groups in lycorine (1), three types of lycorine mono-ester or di-ester analogues were synthesized and evaluated for their antiviral activities against HCoV-OC43. Most of them showed higher selective indexes (SI) than 1, up to nearly 14 times. Using compound 6b as a probe, we firstly demonstrated that lycorine esters directly targeted nidovirus RdRp-associated nucleotidyltransferase (NiRAN) domain in the non-structural protein 12 (nsp 12) by reversibly acylating Cys12 to induce the shrink of NiRAN pocket and block the viral replication, different from the known RdRp inhibitors. Meanwhile, the reversible acylation mode of lycorine esters guaranteed the higher SI values and long-acting effects of its kind. Thus, in addition to acting as prodrugs, ester compounds with a highly acylating warhead can be used as covalent probes to explore the detailed mode of action and improve the safety window. Compound 6b has been identified as a new reversible covalent RdRp allosteric inhibitor for further investigation.
Collapse
Affiliation(s)
- Yuheng Mei
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100050, China
| | - Zhihui Yu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100050, China
| | - Yue Gong
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100050, China
| | - Runze Meng
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100050, China
| | - Xican Ma
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100050, China
| | - Hu Li
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100050, China
| | - Jiayu Li
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100050, China
| | - Jiandong Jiang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100050, China
| | - Zonggen Peng
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100050, China
| | - Yinghong Li
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100050, China.
| | - Danqing Song
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100050, China.
| |
Collapse
|
13
|
Wu X, Liang J, Meng L, Wang B, Liu B, Jin Y. Towards novel small-molecule inhibitors blocking PD-1/PD-L1 pathway: From explainable machine learning models to molecular dynamics simulation. Int J Biol Macromol 2024; 282:136325. [PMID: 39414203 DOI: 10.1016/j.ijbiomac.2024.136325] [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: 08/09/2024] [Revised: 09/29/2024] [Accepted: 10/03/2024] [Indexed: 10/18/2024]
Abstract
Molecular design of small-molecule inhibitors targeting programmed cell death-1 (PD-1)/programmed cell death ligand-1 (PD-L1) pathway has been recognized as an active research area by the clinical success of cancer immunotherapy. In recent years, using machine learning (ML) methods to accelerate drug design have been confirmed. However, the black box character of ML methods makes model interpretation and ligands optimization obscured. Herein, five explainable ML models were constructed by integrating five ML models with the SHAP method, where these ML models were pretrained with >4000 molecules and their R2 ranged from 0.835 to 0.86 on test set. Subsequently, the explainable ML models were employed to identify the relationship between fragments and bio-activity of a small molecule inhibitor BMS-1166, leading to the modification of BMS-1166 into 60 novel compounds. After consensus docking and ADMET test, 3 small molecules (C27, C52 and C54) with better docking scores and lower toxicity than BMS-1166 were screened out further. Finally, the improved binding affinity of C27, C52 to the PD-L1 dimer was validated by the MD simulation. Overall, this work proposed an efficient protocol on the basis of explainable ML models for designing small-molecule inhibitors targeting PD-1/PD-L1 pathway in a rational way.
Collapse
Affiliation(s)
- Xiaoyan Wu
- College of Materials and Energy, South China Agricultural University, Guangzhou 510630, China
| | - Jingyi Liang
- College of Materials and Energy, South China Agricultural University, Guangzhou 510630, China
| | - Luming Meng
- College of Materials and Energy, South China Agricultural University, Guangzhou 510630, China
| | - Bingfeng Wang
- College of Materials and Energy, South China Agricultural University, Guangzhou 510630, China.
| | - Boping Liu
- College of Materials and Energy, South China Agricultural University, Guangzhou 510630, China.
| | - Yulong Jin
- College of Materials and Energy, South China Agricultural University, Guangzhou 510630, China.
| |
Collapse
|
14
|
Dhiman S, Gupta S, Kashaw SK, Chtita S, Kaya S, Almehizia AA, Asati V. Discovery of novel pyrrolo[2,3-d]pyrimidine derivatives as anticancer agents: virtual screening and molecular dynamic studies. SAR AND QSAR IN ENVIRONMENTAL RESEARCH 2024; 35:993-1025. [PMID: 39607421 DOI: 10.1080/1062936x.2024.2432009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 11/14/2024] [Indexed: 11/29/2024]
Abstract
CDK/Cyclins are dysregulated in several human cancers. Recent studies showed inhibition of CDK4/6 was responsible for controlling cell cycle progression and cancer cell growth. In the present study, atom-based and field-based 3D-QSAR, virtual screening, molecular docking and molecular dynamics studies were done for the development of novel pyrrolo[2,3-d]pyrimidine (P2P) derivatives as anticancer agents. The developed models showed good Q2 and r2 values for atom-based 3D-QSAR, which were equal to 0.7327 and 0.8939, whereas for field-based 3D-QSAR the values were 0.8552 and 0.6255, respectively. Molecular docking study showed good-binding interactions with amino acid residues such as VAL-101, HIE-100, ASP-104, ILE-19, LYS-147 and GLU-99, important for CDK4/6 inhibitory activity by using PDB ID: 5L2S. Pharmacophore hypothesis (HHHRR_1) was used in the screening of ZINC database. The top scored ZINC compound ZINC91325512 showed binding interactions with amino acid residues VAL-101, ILE-19, and LYS-147. Enumeration study revealed that the screened compound R1 showed binding interactions with VAL 101 and GLN 149 residues. Furthermore, the Molecular dynamic study showed compound R1, ZINC91325512 and ZINC04000264 having RMSD values of 1.649, 1.733 and 1.610 Å, respectively. These ZINC and enumerated compounds may be used for the development of novel pyrrolo[2,3-d]pyrimidine derivatives as anticancer agent.
Collapse
Affiliation(s)
- S Dhiman
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga, India
- Department of Pharmaceutical Analysis, NIPER, Hajipur, India
| | - S Gupta
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga, India
| | - S K Kashaw
- Department of Pharmaceutical Sciences, Dr. Harassing Gour Central University, Sagar, India
| | - S Chtita
- Laboratory of Analytical and Molecular Chemistry, Faculty of Sciences Ben M'Sik, Hassan II University of Casablanca, Casablanca, Morocco
| | - S Kaya
- Health Services Vocational School, Department of Pharmacy, Sivas Cumhuriyet University, Sivas, Turkey
| | - A A Almehizia
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - V Asati
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga, India
| |
Collapse
|
15
|
Suo Q, Wang J, Wu N, Geng L, Zhang Q, Yue Y. Discovery of a novel nanomolar angiotensin-I converting enzyme inhibitory peptide with unusual binding mechanisms derived from Chlorella pyrenoidosa. Int J Biol Macromol 2024; 280:135873. [PMID: 39307496 DOI: 10.1016/j.ijbiomac.2024.135873] [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: 08/11/2024] [Revised: 09/17/2024] [Accepted: 09/19/2024] [Indexed: 09/26/2024]
Abstract
Chlorella pyrenoidosa (C. pyrenoidosa) has been cultivated in large quantities and proven to be antihypertensive when consumed orally. However, the antihypertensive peptides derived from C. pyrenoidosa remains scarce. In this study, trypsin was chosen to prepare the hydrolysate of C. pyrenoidosa, which was then fractionated by column chromatography. And ninety-nine peptides were identified by LC-MS/MS, after which 10 peptides were chosen by docking-based virtual screening and demonstrated their ability to inhibit ACE. Among them, LVAKA (LV-5) had the lowest IC50 (26.66 μM). LV-5, LKKAP, and PGLRP were identified as non-competitive ACE inhibitory peptides with significant stability under extreme pH and high temperatures conditions. Both in silico and in-vitro simulated gastrointestinal digestion revealed that these three peptides could release ACE inhibitory peptide fragments upon digestion. Sequence optimization of LV-5 led to the discovery of LRAKA (LR-5), which was identified as a novel nanomolar ACE peptide with an IC50 of 350 nM in-vitro and a potent antihypertensive peptide in-vivo. Moreover, molecular dynamic simulation indicated that LR-5 interacted with an unconventional binding site on ACE. These findings underscore the potential of Chlorella as a source of antihypertensive peptides and suggest a promising future for the use of Chlorella-derived peptides in hypertension management.
Collapse
Affiliation(s)
- Qishan Suo
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China; Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, China; University of Chinese Academy of Sciences, Beijing, China
| | - Jing Wang
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China; Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, China
| | - Ning Wu
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China; Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, China
| | - Lihua Geng
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China; Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, China
| | - Quanbin Zhang
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China; Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, China; University of Chinese Academy of Sciences, Beijing, China
| | - Yang Yue
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China; Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, China.
| |
Collapse
|
16
|
Liang JJ, Pitsillou E, Lau HLY, Mccubbery CP, Gan H, Hung A, Karagiannis TC. Utilization of the EpiMed Coronabank Chemical Collection to identify potential SARS-CoV-2 antivirals: in silico studies targeting the nsp14 ExoN domain and PL pro naphthalene binding site. J Mol Graph Model 2024; 131:108803. [PMID: 38815531 DOI: 10.1016/j.jmgm.2024.108803] [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: 12/08/2023] [Revised: 05/22/2024] [Accepted: 05/24/2024] [Indexed: 06/01/2024]
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) genome encodes 29 proteins including four structural, 16 nonstructural (nsps), and nine accessory proteins (https://epimedlab.org/sars-cov-2-proteome/). Many of these proteins contain potential targetable sites for the development of antivirals. Despite the widespread use of vaccinations, the emergence of variants necessitates the investigation of new therapeutics and antivirals. Here, the EpiMed Coronabank Chemical Collection (https://epimedlab.org/crl/) was utilized to investigate potential antivirals against the nsp14 exoribonuclease (ExoN) domain. Molecular docking was performed to evaluate the binding characteristics of our chemical library against the nsp14 ExoN site. Based on the initial screen, trisjuglone, ararobinol, corilagin, and naphthofluorescein were identified as potential lead compounds. Molecular dynamics (MD) simulations were subsequently performed, with the results highlighting the stability of the lead compounds in the nsp14 ExoN site. Protein-RNA docking revealed the potential for the lead compounds to disrupt the interaction with RNA when bound to the ExoN site. Moreover, hypericin, cyanidin-3-O-glucoside, and rutin were previously identified as lead compounds targeting the papain-like protease (PLpro) naphthalene binding site. Through performing MD simulations, the stability and interactions of lead compounds with PLpro were further examined. Overall, given the critical role of the exonuclease activity of nsp14 in ensuring viral fidelity and the multifunctional role of PLpro in viral pathobiology and replication, these nsps represent important targets for antiviral drug development. Our databases can be utilized for in silico studies, such as the ones performed here, and this approach can be applied to other potentially druggable SARS-CoV-2 protein targets.
Collapse
Affiliation(s)
- Julia J Liang
- Epigenomic Medicine Laboratory at prospED Polytechnic, Carlton, VIC, 3053, Australia; School of Science, STEM College, RMIT University, Melbourne, VIC, 3001, Australia; Epigenetics in Human Health and Disease Program, Baker Heart and Diabetes Institute, 75 Commercial Road, Prahran, VIC, 3004, Australia
| | - Eleni Pitsillou
- Epigenomic Medicine Laboratory at prospED Polytechnic, Carlton, VIC, 3053, Australia; School of Science, STEM College, RMIT University, Melbourne, VIC, 3001, Australia
| | - Hannah L Y Lau
- Epigenomic Medicine Laboratory at prospED Polytechnic, Carlton, VIC, 3053, Australia; Department of Microbiology and Immunology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Cian P Mccubbery
- Epigenomic Medicine Laboratory at prospED Polytechnic, Carlton, VIC, 3053, Australia; Department of Microbiology and Immunology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Hockxuen Gan
- Epigenomic Medicine Laboratory at prospED Polytechnic, Carlton, VIC, 3053, Australia; Department of Microbiology and Immunology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Andrew Hung
- School of Science, STEM College, RMIT University, Melbourne, VIC, 3001, Australia
| | - Tom C Karagiannis
- Epigenomic Medicine Laboratory at prospED Polytechnic, Carlton, VIC, 3053, Australia; Epigenetics in Human Health and Disease Program, Baker Heart and Diabetes Institute, 75 Commercial Road, Prahran, VIC, 3004, Australia; Department of Microbiology and Immunology, The University of Melbourne, Parkville, VIC, 3010, Australia; Department of Clinical Pathology, The University of Melbourne, Parkville, VIC, 3010, Australia.
| |
Collapse
|
17
|
Baidya SK, Banerjee S, Ghosh B, Jha T, Adhikari N. Pinpointing prime structural attributes of potential MMP-2 inhibitors comprising alkyl/arylsulfonyl pyrrolidine scaffold: a ligand-based molecular modelling approach validated by molecular dynamics simulation analysis. SAR AND QSAR IN ENVIRONMENTAL RESEARCH 2024; 35:665-692. [PMID: 39193767 DOI: 10.1080/1062936x.2024.2389822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 08/02/2024] [Indexed: 08/29/2024]
Abstract
MMP-2 overexpression is strongly related to several diseases including cancer. However, none of the MMP-2 inhibitors have been marketed as drug candidates due to various adverse effects. Here, a set of sulphonyl pyrrolidines was subjected to validation of molecular modelling followed by binding mode analysis to explore the crucial structural features required for the discovery of promising MMP-2 inhibitors. This study revealed the importance of hydroxamate as a potential zinc-binding group compared to the esters. Importantly, hydrophobic and sterical substituents were found favourable at the terminal aryl moiety attached to the sulphonyl group. The binding interaction study revealed that the S1' pocket of MMP-2 similar to 'a basketball passing through a hoop' allows the aryl moiety for proper fitting and interaction at the active site to execute potential MMP-2 inhibition. Again, the sulphonyl pyrrolidine moiety can be a good fragment necessary for MMP-2 inhibition. Moreover, some novel MMP-2 inhibitors were also reported. They showed the significance of the 3rd position substitution of the pyrrolidine ring to produce interaction inside S2' pocket. The current study can assist in the design and development of potential MMP-2 inhibitors as effective drug candidates for the management of several diseases including cancers in the future.
Collapse
Affiliation(s)
- S K Baidya
- Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, India
| | - S Banerjee
- Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, India
| | - B Ghosh
- Epigenetic Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad, India
| | - T Jha
- Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, India
| | - N Adhikari
- Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, India
| |
Collapse
|
18
|
Premkumar T, Sajitha Lulu S. Targeting key players in Alzheimer's disease: bioactive compounds from Moringa oleifera, Desmodium gangeticum, and Centella asiatica as potential therapeutics. J Biomol Struct Dyn 2024:1-23. [PMID: 38887054 DOI: 10.1080/07391102.2024.2335300] [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: 12/23/2023] [Accepted: 03/20/2024] [Indexed: 06/20/2024]
Abstract
Alzheimer's Disease (AD) is one of the critical reasons for dementia around the world, with a huge number of cases being reported every year. The breakdown of Amyloid Precursor Protein (APP) plays a crucial role in AD development. The Beta-site APP Cleaving Enzyme 1 (BACE1) is a highly significant proteolytic enzyme found to be critically involved in the APP breakdown process and generates beta-amyloid plaques in the extracellular neuronal membrane. In this study, we have used natural compounds with cognitive and neuroprotective activities from three plants, Centella asiatica, Moringa oleifera, and Desmodium gangeticum to inhibit the activity of BACE1. We have identified nine compounds out of 73 compounds filtered out from the three plants showing high affinity with the catalytic dyad region of BACE1 through molecular docking studies. Interestingly, the 200 ns molecular dynamics simulation study further confirmed the stability of the complexes formed between 9 compounds and the BACE1 protein. Furthermore, the free energy calculations also revealed these complexes possess favorable energies. Astilbin, Delphinidin 3-glucoside, and kaempferol 7-O-glucoside showed good binding affinity and structural stability when compared to other compounds and the control CNP520. Following a preliminary screening, the Astilbin compound was chosen based on the grounds of binding affinity, ADMET Properties, Hbond formation, Molecular Dynamic simulation, and MM-PBSA studies. A subsequent 1microsecond molecular dynamics simulation was conducted for the Astilbin complex. Through microsecond simulation, it was found that Astilbin alters BACE1's behavior and induces conformational rearrangements. Thus, this study opens a gateway to inhibit the activity of BACE1 protein through Astilbin thereby disclosing the possibility of managing Alzheimer's Disease.
Collapse
Affiliation(s)
- T Premkumar
- Integrative Multiomics Laboratory, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - S Sajitha Lulu
- Integrative Multiomics Laboratory, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| |
Collapse
|
19
|
Cañizares-Carmenate Y, Perera-Sardiña Y, Marrero-Ponce Y, Díaz-Amador R, Torrens F, Castillo-Garit JA. Ligand and structure-based discovery of phosphorus-containing compounds as potential metalloproteinase inhibitors. SAR AND QSAR IN ENVIRONMENTAL RESEARCH 2024; 35:219-240. [PMID: 38380444 DOI: 10.1080/1062936x.2024.2314103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 01/29/2024] [Indexed: 02/22/2024]
Abstract
In this study, a methodology is proposed, combining ligand- and structure-based virtual screening tools, for the identification of phosphorus-containing compounds as inhibitors of zinc metalloproteases. First, we use Dragon molecular descriptors to develop a Linear Discriminant Analysis classification model, which is widely validated according to the OECD principles. This model is simple, robust, stable and has good discriminating power. Furthermore, it has a defined applicability domain and it is used for virtual screening of the DrugBank database. Second, docking experiments are carried out on the identified compounds that showed good binding energies to the enzyme thermolysin. Considering the potential toxicity of phosphorus-containing compounds, their toxicological profile is evaluated according to Protox II. Of the five molecules evaluated, two show carcinogenic and mutagenic potential at small LD50, not recommended as drugs, while three of them are classified as non-toxic, and could constitute a starting point for the development of new vasoactive metalloprotease inhibitor drugs. According to molecular dynamics simulation, two of them show stable interactions with the active site maintaining coordination with the metal. A high agreement is evident between QSAR, docking and molecular dynamics results, demonstrating the potentialities of the combination of these tools.
Collapse
Affiliation(s)
- Y Cañizares-Carmenate
- Unit of Computer-Aided Molecular ''Biosilico" Discovery and Bioinformatic Research (CAMD-BIR Unit), Departamento de Farmacia, Facultad de Química-Farmacia, Universidad Central ''Marta Abreu" de Las Villas, Santa Clara, Cuba
| | - Y Perera-Sardiña
- Departamento de Ciencias Básicas Biomédicas, Facultad de Ciencias de la Salud, Universidad de Talca, Talca, Chile
| | - Y Marrero-Ponce
- Grupo de Medicina Molecular Y Traslacional (MeM & T), Escuela de Medicina, Universidad San Francisco de Quito, Edificio de Especialidades Médicas, Quito, Ecuador
| | - R Díaz-Amador
- Laboratorio de Bioinformática y Química Computacional, Escuela de Química y Farmacia, Facultad de Medicina, Universidad Católica de Maule, Maule, Chile
| | - F Torrens
- Institut Universitari de Ciència Molecular, Universitat de València, Edifici d'Instituts de Paterna, València, Spain
| | - J A Castillo-Garit
- Instituto Universitario de Investigación y Desarrollo Tecnológico (IDT), Universidad Tecnológica Metropolitana, Santiago, Chile
| |
Collapse
|
20
|
Dhiman P, Yadav N, Auti PS, Jaswal S, Singh G, Mehan S, Ghosh B, Paul AT, Monga V. Discovery of thiazolidinedione-based pancreatic lipase inhibitors as anti-obesity agents: synthesis, in silico studies and pharmacological investigations. J Biomol Struct Dyn 2024:1-23. [PMID: 38315459 DOI: 10.1080/07391102.2024.2310799] [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: 10/05/2023] [Accepted: 01/20/2024] [Indexed: 02/07/2024]
Abstract
A series of new 2,5-disubstituted arylidene derivatives of thiazolidinedione (16a-e, 17a-d, 18a-c) designed using molecular hybridization approach were synthesized, structurally characterized, and explored for their anti-obesity potential via inhibition of Pancreatic Lipase (PL). Compound 18a presented the most potent PL inhibitory activity with IC50 = 2.71 ± 0.31 µM, as compared to the standard drug, Orlistat (IC50 = 0.99 µM). Kinetic study revealed reversible competitive mode of enzyme inhibition by compound 18a with an inhibitory constant value of 1.19 µM. The most promising compound 18a revealed satisfactory binding mode within the active site of the target protein (human PL, PDB ID: 1LPB). Also, MM/PBSA binding free energy and molecular dynamics (MD) simulation analysis were performed for the most promising compound 18a, which showed potent inhibition according to the results of in vitro studies. Furthermore, a stable conformation of the 1LPB-ligand suggested the stability of this compound in the dynamic environment. The ADME and toxicity analysis of the compounds were examined using web-based online platforms. Results of in vivo studies confirmed the anti-obesity efficacy of compound 18a, wherein oral treatment with compound 18a (30 mg/kg) resulted in a significant reduction in the body weight, BMI, Lee index, feed intake (in Kcal), body fat depots and serum triglycerides. Compound 18a significantly decreased the levels of serum total cholesterol (TC) to 128.6 ± 0.59 mg/dl and serum total triglycerides (TG) to 95.73 ± 0.67 mg/dl as compared to the HFD control group. The present study identified disubstituted TZD derivatives as a new promising class of anti-obesity agents.
Collapse
Affiliation(s)
- Prashant Dhiman
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga, India
| | - Nisha Yadav
- Laboratory of Natural Product Chemistry, Department of Pharmacy, Birla Institute of Technology and Science, Pilani (BITS Pilani), Pilani, India
| | - Prashant S Auti
- Laboratory of Natural Product Chemistry, Department of Pharmacy, Birla Institute of Technology and Science, Pilani (BITS Pilani), Pilani, India
| | - Shalini Jaswal
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga, India
| | - Gurpreet Singh
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga, India
| | - Sidharth Mehan
- Department of Pharmacology, ISF College of Pharmacy, Moga, India
| | - Balaram Ghosh
- Epigenetic Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad, India
| | - Atish T Paul
- Laboratory of Natural Product Chemistry, Department of Pharmacy, Birla Institute of Technology and Science, Pilani (BITS Pilani), Pilani, India
| | - Vikramdeep Monga
- Drug Design and Molecular Synthesis Laboratory, Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, VPO-Ghudda, Bathinda, India
| |
Collapse
|