1
|
Sowa ST, Kücükdisli M, Mostinski Y, Schaller DA, Vinagreiro CS, Cirillo D, Bosetti C, Brinch SA, van Laar K, Wegert A, Leenders RGG, Krauss S, Waaler J, Volkamer A, Lehtiö L, Nazaré M. Deconstruction of Dual-Site Tankyrase Inhibitors Provides Insights into Binding Energetics and Suggests Critical Hotspots for Ligand Optimization. J Med Chem 2025; 68:7263-7279. [PMID: 40134122 PMCID: PMC11997991 DOI: 10.1021/acs.jmedchem.4c02845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 03/03/2025] [Accepted: 03/07/2025] [Indexed: 03/27/2025]
Abstract
Designing inhibitors is a complex task that requires a deep understanding of protein-ligand interactions and their dynamics. Ligands often interact with multiple binding subsites, with noncovalent interactions affecting binding affinity. Conformational changes and plasticity of both, the ligand and the protein influence binding energetics. We investigated the tankyrase ADP-ribosyltransferase as a promising drug target regulating many cellular pathways. Despite the existence of diverse tankyrase inhibitors, their binding energetics and contributions of flexible cryptic subpockets to binding affinity remain elusive. To examine these aspects, we deconstructed inhibitors to key fragments, dissected their energetic contribution to the affinity, and determined their binding mode by X-ray crystallography. Varying ligand efficiencies of the deconstructed, pocket-binding fragments revealed the cryptic nature of subpockets. These insights enabled us to redesign inhibitors with novel linkers, the observed key area for optimization, increasing the potency in enzymatic and cell-based assays by 7.5-fold and 6.2-fold compared to the parent ligand.
Collapse
Affiliation(s)
- Sven T. Sowa
- Faculty
for Biochemistry and Molecular Medicine & Biocenter Oulu, University of Oulu, Aapistie 7, 90220 Oulu, Finland
| | - Murat Kücükdisli
- Medicinal
Chemistry, Leibniz-Forschungsinstitut für
Molekulare Pharmakologie (FMP), Campus Berlin Buch, Robert-Roessle-Str. 10, 13125 Berlin, Germany
| | - Yelena Mostinski
- Medicinal
Chemistry, Leibniz-Forschungsinstitut für
Molekulare Pharmakologie (FMP), Campus Berlin Buch, Robert-Roessle-Str. 10, 13125 Berlin, Germany
| | - David A. Schaller
- In Silico
Toxicology and Structural Bioinformatics, Institute of Physiology, Charité Universitätsmedizin Berlin, Virchowweg 6, 10117 Berlin, Germany
| | - Carolina S. Vinagreiro
- Medicinal
Chemistry, Leibniz-Forschungsinstitut für
Molekulare Pharmakologie (FMP), Campus Berlin Buch, Robert-Roessle-Str. 10, 13125 Berlin, Germany
| | - Davide Cirillo
- Medicinal
Chemistry, Leibniz-Forschungsinstitut für
Molekulare Pharmakologie (FMP), Campus Berlin Buch, Robert-Roessle-Str. 10, 13125 Berlin, Germany
| | - Chiara Bosetti
- Faculty
for Biochemistry and Molecular Medicine & Biocenter Oulu, University of Oulu, Aapistie 7, 90220 Oulu, Finland
| | - Shoshy Alam Brinch
- Oslo
University Hospital, P.O. Box 4950, Nydalen, 0424 Oslo, Norway
- Hybrid
Technology Hub—Centre of Excellence, Institute of Basic Medical
Sciences, University of Oslo, 0317 Oslo, Norway
| | - Kirsten van Laar
- Symeres
Netherlands B.V., Kerkenbos
1013, 6546 BB Nijmegen, The Netherlands
| | - Anita Wegert
- Symeres
Netherlands B.V., Kerkenbos
1013, 6546 BB Nijmegen, The Netherlands
| | | | - Stefan Krauss
- Oslo
University Hospital, P.O. Box 4950, Nydalen, 0424 Oslo, Norway
- Hybrid
Technology Hub—Centre of Excellence, Institute of Basic Medical
Sciences, University of Oslo, 0317 Oslo, Norway
| | - Jo Waaler
- Oslo
University Hospital, P.O. Box 4950, Nydalen, 0424 Oslo, Norway
- Hybrid
Technology Hub—Centre of Excellence, Institute of Basic Medical
Sciences, University of Oslo, 0317 Oslo, Norway
| | - Andrea Volkamer
- In Silico
Toxicology and Structural Bioinformatics, Institute of Physiology, Charité Universitätsmedizin Berlin, Virchowweg 6, 10117 Berlin, Germany
- Data
Driven Drug Design, Faculty of Mathematics and Computer Sciences, Saarland University, 66123 Saarbrücken, Germany
| | - Lari Lehtiö
- Faculty
for Biochemistry and Molecular Medicine & Biocenter Oulu, University of Oulu, Aapistie 7, 90220 Oulu, Finland
| | - Marc Nazaré
- Medicinal
Chemistry, Leibniz-Forschungsinstitut für
Molekulare Pharmakologie (FMP), Campus Berlin Buch, Robert-Roessle-Str. 10, 13125 Berlin, Germany
| |
Collapse
|
2
|
Orr AA, Uwakweh AO, Li X, Karanji AK, Hoag SW, Deredge DJ, MacKerell AD. Mapping the distribution and affinities of ligand interaction sites on human serum albumin. Biophys J 2025:S0006-3495(25)00170-5. [PMID: 40134214 DOI: 10.1016/j.bpj.2025.03.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 03/10/2025] [Accepted: 03/20/2025] [Indexed: 03/27/2025] Open
Abstract
Ligands in many instances interact with a protein at multiple sites with a range of affinities. In this study, ligand-protein interaction sites on human serum albumin (HSA) are mapped using the site-identification by ligand competitive saturation (SILCS)-Biologics approach in conjunction with hydrogen-deuterium exchange (HDX)-mass spectrometry (MS) experiments. Ligands studied include known HSA binders, ibuprofen and ketoprofen, and compounds arginine, alanine, sucrose, and trehalose, excipients used in therapeutic formulations of protein-based drugs. In addition, the impact of excipient binding to HSA on its stability is investigated through temperature-ramp stability studies monitoring solution viscosity. For the studied ligands, interactions that correspond to known drug-binding sites (DSs) are identified. These include previously identified ibuprofen and ketoprofen interaction sites as well as additional sites and, in the case of the excipients, the ligands are shown to also bind at previously unidentified interaction sites, termed excipient sites (ESs) with 20 or more sites identified for the studied compounds. HDX-MS titrations were used to determine dissociation constants for a subset of the interaction sites for ibuprofen, ketoprofen, arginine, and sucrose, which exhibited Kd values in the low micromolar to millimolar range in satisfactory agreement with SILCS-Biologics predicted affinities, validating the computational approach to identify both high- and low-affinity interaction sites. The stability studies indicate the excipients offer protection at low excipient/protein ratios up to 66 with destabilization occurring at ratios above 132 with the exception of sucrose at the t0 time point, indicating that the more favorable affinities of sucrose seen in the SILCS-Biologics and HDX-MS analyses contribute to protein stabilization. These results indicate that ligands can bind to large numbers of interaction sites on proteins, with those interactions having implications for the development of formulations for therapeutic proteins.
Collapse
Affiliation(s)
- Asuka A Orr
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland Baltimore, Baltimore, Maryland; SilcsBio LLC, Baltimore, Maryland
| | - Agbo-Oma Uwakweh
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland Baltimore, Baltimore, Maryland
| | - Xun Li
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland Baltimore, Baltimore, Maryland
| | - Ahmad Kiani Karanji
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland Baltimore, Baltimore, Maryland
| | - Stephen W Hoag
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland Baltimore, Baltimore, Maryland
| | - Daniel J Deredge
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland Baltimore, Baltimore, Maryland.
| | - Alexander D MacKerell
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland Baltimore, Baltimore, Maryland.
| |
Collapse
|
3
|
Grineva OV. Direct Determination of Ratios of All Conformations and Their Lifetimes for Small Flexible Molecules from Molecular Dynamics Simulations: 1,3-Propanediol in an Aqueous Environment. Molecules 2025; 30:1285. [PMID: 40142060 PMCID: PMC11946693 DOI: 10.3390/molecules30061285] [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: 01/24/2025] [Revised: 03/02/2025] [Accepted: 03/11/2025] [Indexed: 03/28/2025] Open
Abstract
For the first time in the course of molecular dynamics modeling of a liquid, the conformations of each of the small flexible molecules present in the system were fixed at short (1 ps) time intervals. This allowed the establishment of the ratios between various individual conformations and their families and determination of the average lifetimes of both individual conformations and families. As an example, data are presented for modeling boxes with different numbers of molecules (800, 2700, and 6400) for an aqueous solution with 1 mol. % 1,3-propanediol at 298.15 K and 1 atm. The results of the conformational analysis turned out to be very close for systems with different numbers of molecules and with different choices of initial conformations. For the systems under investigation, the tTTg conformation, which does not have intramolecular hydrogen bond, predominated (37-39%), and the total fractions of all conformations in the TT family were 74-76%. Only 0.4-0.5% of 1,3-propanediol molecules had conformations with the possible formation of intramolecular hydrogen bond, although the most stable conformers of free 1,3-propanediol molecules exhibit such a bond. The average lifetimes of each individual conformation did not exceed 7 ps in simulated systems, while the maximum lifetimes reached 60 ps. The average lifetimes of the main chain vary from ~110 ps in TT family to ~12 ps in GG' family, in which the conformations tend to have intramolecular hydrogen bonds. It was found that calculations for an individual 1,3-propanediol molecule at the MP2/aug-cc-pVDZ or MP2/aug-cc-pVTZ theoretical levels lead to 22 conformers both in vacuum and by using the PCM model for implicit aqueous solvation (at the MP2/aug-cc-pVDZ level) and that such solvation reduces the energy difference between the conformers.
Collapse
Affiliation(s)
- Olga V Grineva
- Chemistry Department, Moscow M. V. Lomonosov State University, 119991 Moscow, Russia
| |
Collapse
|
4
|
Sethi G, Hwang JH, Krishna R. Structure based exploration of potential lead molecules against the extracellular cysteine protease (EcpA) of Staphylococcus epidermidis: a therapeutic halt. J Biomol Struct Dyn 2024; 42:9167-9183. [PMID: 37615425 DOI: 10.1080/07391102.2023.2250455] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 08/15/2023] [Indexed: 08/25/2023]
Abstract
Nosocomial infection caused by Staphylococcus epidermidis is one of the most widely spread diseases affecting the world's population. No strategies have been developed to overcome this infection and inhibit its spread in immunocompromised patients or patients with indwelling medical devices. EcpA is an extracellular cysteine protease protein involved in biofilm formation on medical devices. Thus, blocking this mechanism may be viable for developing a drug against S. epidermidis. The current research aimed to find new, potent inhibitors that could stop the S. epidermidis EcpA protein from functioning. This study attempted to identify the most promising drug candidates using structure-based virtual screening (SBVS) from libraries of natural ligands. The top-scored molecules were shortlisted based on their IC50 values and pharmacophore properties and further validated through density functional theory (DFT) studies. We found five inhibitors using virtual screening, and the results indicate that these drugs had the highest energy binding potential towards the EcpA targets when compared to the reference molecule E-64, a known cysteine protease inhibitor. In order to evaluate the binding conformational stability of protein-ligand complexes, molecular dynamics (MD) simulations were performed in triplicate for 100 ns, revealing the significant stability of anticipated molecules at the docked site. Furthermore, principal component analysis and binding free energy calculations were performed to understand the dynamics and stability of the complexes. The current study indicated that these compounds looked to be suitable novel inhibitors of the EcpA protein and pave the path for further discovery of novel inhibitors of EcpA.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Guneswar Sethi
- Department of Bioinformatics, Pondicherry University, Puducherry, India
- Department of Predictive Toxicology, Korea Institute of Toxicology (KIT), Daejeon, Republic of Korea
| | - Jeong Ho Hwang
- Animal Model Research Group, Korea Institute of Toxicology, Jeonguep, Republic of Korea
| | - Ramadas Krishna
- Department of Bioinformatics, Pondicherry University, Puducherry, India
| |
Collapse
|
5
|
Kataria A, Srivastava A, Singh DD, Haque S, Han I, Yadav DK. Systematic computational strategies for identifying protein targets and lead discovery. RSC Med Chem 2024; 15:2254-2269. [PMID: 39026640 PMCID: PMC11253860 DOI: 10.1039/d4md00223g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 05/10/2024] [Indexed: 07/20/2024] Open
Abstract
Computational algorithms and tools have retrenched the drug discovery and development timeline. The applicability of computational approaches has gained immense relevance owing to the dramatic surge in the structural information of biomacromolecules and their heteromolecular complexes. Computational methods are now extensively used in identifying new protein targets, druggability assessment, pharmacophore mapping, molecular docking, the virtual screening of lead molecules, bioactivity prediction, molecular dynamics of protein-ligand complexes, affinity prediction, and for designing better ligands. Herein, we provide an overview of salient components of recently reported computational drug-discovery workflows that includes algorithms, tools, and databases for protein target identification and optimized ligand selection.
Collapse
Affiliation(s)
- Arti Kataria
- Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH) Hamilton MT 59840 USA
| | - Ankit Srivastava
- Laboratory of Neurological Infections and Immunity, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH) Hamilton MT 59840 USA
| | - Desh Deepak Singh
- Amity Institute of Biotechnology, Amity University Rajasthan Jaipur India
| | - Shafiul Haque
- Research and Scientific Studies Unit, College of Nursing and Health Sciences, Jazan University Jazan-45142 Saudi Arabia
| | - Ihn Han
- Plasma Bioscience Research Center, Applied Plasma Medicine Center, Department of Electrical & Biological Physics, Kwangwoon University Seoul 01897 Republic of Korea +82 32 820 4948
| | - Dharmendra Kumar Yadav
- Department of Biologics, College of Pharmacy, Gachon University Hambakmoeiro 191, Yeonsu-gu Incheon 21924 Republic of Korea
| |
Collapse
|
6
|
Islam S, Parves MR, Islam MJ, Ali MA, Efaz FM, Hossain MS, Ullah MO, Halim MA. Structural and functional effects of the L84S mutant in the SARS-COV-2 ORF8 dimer based on microsecond molecular dynamics study. J Biomol Struct Dyn 2024; 42:5770-5787. [PMID: 37403295 DOI: 10.1080/07391102.2023.2228919] [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: 01/10/2023] [Accepted: 06/17/2023] [Indexed: 07/06/2023]
Abstract
The L84S mutation has been observed frequently in the ORF8 protein of SARS-CoV-2, which is an accessory protein involved in various important functions such as virus propagation, pathogenesis, and evading the immune response. However, the specific effects of this mutation on the dimeric structure of ORF8 and its impacts on interactions with host components and immune responses are not well understood. In this study, we performed one microsecond molecular dynamics (MD) simulation and analyzed the dimeric behavior of the L84S and L84A mutants in comparison to the native protein. The MD simulations revealed that both mutations caused changes in the conformation of the ORF8 dimer, influenced protein folding mechanisms, and affected the overall structural stability. In particular, the 73YIDI76 motif has found to be significantly affected by the L84S mutation, leading to structural flexibility in the region connecting the C-terminal β4 and β5 strands. This flexibility might be responsible for virus immune modulation. The free energy landscape (FEL) and principle component analysis (PCA) have also supported our investigation. Overall, the L84S and L84A mutations affect the ORF8 dimeric interfaces by reducing the frequency of protein-protein interacting residues (Arg52, Lys53, Arg98, Ile104, Arg115, Val117, Asp119, Phe120, and Ile121) in the ORF8 dimer. Our findings provide detail insights for further research in designing structure-based therapeutics against the SARS-CoV-2.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Shafiqul Islam
- Division of Infectious disease and Division of Computer Aided Drug Design, The Red-Green Research Centre, Dhaka, Bangladesh
| | - Md Rimon Parves
- Division of Infectious disease and Division of Computer Aided Drug Design, The Red-Green Research Centre, Dhaka, Bangladesh
| | - Md Jahirul Islam
- Division of Infectious disease and Division of Computer Aided Drug Design, The Red-Green Research Centre, Dhaka, Bangladesh
| | - Md Ackas Ali
- Department of Chemistry and Biochemistry, Kennesaw State University, Kennesaw, GA, USA
| | - Faiyaz Md Efaz
- Division of Infectious disease and Division of Computer Aided Drug Design, The Red-Green Research Centre, Dhaka, Bangladesh
| | - Md Shahadat Hossain
- Division of Infectious disease and Division of Computer Aided Drug Design, The Red-Green Research Centre, Dhaka, Bangladesh
| | - M Obayed Ullah
- Division of Infectious disease and Division of Computer Aided Drug Design, The Red-Green Research Centre, Dhaka, Bangladesh
| | - Mohammad A Halim
- Department of Chemistry and Biochemistry, Kennesaw State University, Kennesaw, GA, USA
| |
Collapse
|
7
|
Hadi N, Drioiche A, Bouchra EM, Baammi S, Abdelaziz Shahat A, Tagnaout I, Radi M, Remok F, Bouzoubaa A, Zair T. Phytochemical Analysis and Evaluation of Antioxidant and Antimicrobial Properties of Essential Oils and Seed Extracts of Anethum graveolens from Southern Morocco: In Vitro and In Silico Approach for a Natural Alternative to Synthetic Preservatives. Pharmaceuticals (Basel) 2024; 17:862. [PMID: 39065713 PMCID: PMC11280095 DOI: 10.3390/ph17070862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 06/21/2024] [Accepted: 06/25/2024] [Indexed: 07/28/2024] Open
Abstract
Anethum graveolens is an aromatic plant traditionally used as an antispasmodic and carminative. The objective of this study is to analyze the chemical composition of the essential oils and extracts obtained from seeds gathered in Errachidia, southern Morocco. Additionally, the antioxidant and antimicrobial properties of these oils and extracts will be evaluated. GC-MS analysis of the EO isolated by hydrodistillation revealed that its main compounds were E-anethole (38.13%), estragole (29.32%), fenchone (17.21%), and α-pinene (7.37%). The phenolic components were extracted using the methods of decoction and Soxhlet. The assay of the phenolic compounds showed that A. graveolens seeds contained considerable amounts of polyphenols, flavonoids, and condensed tannins, with variable levels depending on the extract analyzed. HPLC/UV-ESI-MS analyses performed on the decoction revealed a structural diversity of the molecules present in this extract, the most important of which were umbelliferone (12.35%), 3-hydroxyflavone (11.23%), rosmanol (8.95%), biotin (8.36%), emmotin H (4.91%), and coumarin (4.21%). The antioxidant activity, as determined by three techniques (DPPH•, FRAP, and CAT), demonstrated that the essential oils (EOs) and extracts had a potent capacity to counteract detrimental free radicals, control the generation of reactive oxygen species, and mitigate oxidative damages. The antimicrobial activity of the Eos and extracts was carried out in a liquid medium against five strains (E. cloacae, K. pneumoniae, E. coli, S. aureus, and S. epidermidis) and four candidiasis (C. albicans, C. dubliniensis, C. tropicalis, and C. parapsilosis) and Aspergillus niger. The results showed the effectiveness of the EOs compared to the aqueous, ethanolic, and decoction extracts against most of the microorganisms tested. In addition, the ethanolic extract showed antifungal activity that was distinguished from that of the other extracts. The antimicrobial efficacy of the essential oils under study can primarily be attributed to the synergistic interactions among its three principal constituents (E-anethole, estragole, and fenchone). Furthermore, molecular docking and molecular dynamics simulation results reveal significant interactions and stability between the selected bioactive compounds and different target proteins involved in antimicrobial and antioxidant activities. Compounds like 3-hydroxyflavone, emmotin H, trans-caftaric acid, methyl rosmarinate, 1-caffeoyl-beta-D-glucose, and kaempferol exhibited better binding energies with the explored proteins, indicating their potential as antimicrobial and antioxidant agents. Finally, our findings emphasize the significance of A. graveolens seeds as a promising reservoir of advantageous health compounds that can serve as organic substitutes for the presently employed synthetic preservatives.
Collapse
Affiliation(s)
- Nadia Hadi
- Research Team of Chemistry of Bioactive Molecules and the Environment, Laboratory of Innovative Materials and Biotechnology of Natural Resources, Faculty of Sciences, Moulay Ismaïl University, B.P. 11201, Zitoune, Meknes 50070, Morocco; (N.H.); (E.M.B.); (I.T.); (M.R.); (F.R.); (A.B.)
| | - Aziz Drioiche
- Research Team of Chemistry of Bioactive Molecules and the Environment, Laboratory of Innovative Materials and Biotechnology of Natural Resources, Faculty of Sciences, Moulay Ismaïl University, B.P. 11201, Zitoune, Meknes 50070, Morocco; (N.H.); (E.M.B.); (I.T.); (M.R.); (F.R.); (A.B.)
| | - El Moumen Bouchra
- Research Team of Chemistry of Bioactive Molecules and the Environment, Laboratory of Innovative Materials and Biotechnology of Natural Resources, Faculty of Sciences, Moulay Ismaïl University, B.P. 11201, Zitoune, Meknes 50070, Morocco; (N.H.); (E.M.B.); (I.T.); (M.R.); (F.R.); (A.B.)
| | - Soukayna Baammi
- Bioinformatics Laboratory, College of Computing, Mohammed VI Polytechnic University, Ben Guerir 43150, Morocco;
| | | | - Imane Tagnaout
- Research Team of Chemistry of Bioactive Molecules and the Environment, Laboratory of Innovative Materials and Biotechnology of Natural Resources, Faculty of Sciences, Moulay Ismaïl University, B.P. 11201, Zitoune, Meknes 50070, Morocco; (N.H.); (E.M.B.); (I.T.); (M.R.); (F.R.); (A.B.)
| | - Mohamed Radi
- Research Team of Chemistry of Bioactive Molecules and the Environment, Laboratory of Innovative Materials and Biotechnology of Natural Resources, Faculty of Sciences, Moulay Ismaïl University, B.P. 11201, Zitoune, Meknes 50070, Morocco; (N.H.); (E.M.B.); (I.T.); (M.R.); (F.R.); (A.B.)
| | - Fidaous Remok
- Research Team of Chemistry of Bioactive Molecules and the Environment, Laboratory of Innovative Materials and Biotechnology of Natural Resources, Faculty of Sciences, Moulay Ismaïl University, B.P. 11201, Zitoune, Meknes 50070, Morocco; (N.H.); (E.M.B.); (I.T.); (M.R.); (F.R.); (A.B.)
| | - Amal Bouzoubaa
- Research Team of Chemistry of Bioactive Molecules and the Environment, Laboratory of Innovative Materials and Biotechnology of Natural Resources, Faculty of Sciences, Moulay Ismaïl University, B.P. 11201, Zitoune, Meknes 50070, Morocco; (N.H.); (E.M.B.); (I.T.); (M.R.); (F.R.); (A.B.)
| | - Touriya Zair
- Research Team of Chemistry of Bioactive Molecules and the Environment, Laboratory of Innovative Materials and Biotechnology of Natural Resources, Faculty of Sciences, Moulay Ismaïl University, B.P. 11201, Zitoune, Meknes 50070, Morocco; (N.H.); (E.M.B.); (I.T.); (M.R.); (F.R.); (A.B.)
| |
Collapse
|
8
|
Bahena Culhuac E, Bello M. Unveiling the Mechanisms of EGCG-p53 Interactions through Molecular Dynamics Simulations. ACS OMEGA 2024; 9:20066-20085. [PMID: 38737068 PMCID: PMC11080030 DOI: 10.1021/acsomega.3c10523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 03/24/2024] [Accepted: 03/27/2024] [Indexed: 05/14/2024]
Abstract
Green tea consumption is associated with protective and preventive effects against various types of cancer. These effects are attributed to polyphenols, particularly epigallocatechin-3-gallate (EGCG). EGCG acts by directly inhibiting tumor suppressor protein p53. The binding mechanism by which EGCG inhibits p53 activity is associated with residues Trp23-Lys24 and Pro47-Thr55 within the p53 N-terminal domain (NTD). However, the structural and thermodynamic aspects of the interaction between EGCG and p53 are poorly understood. Therefore, based on crystallographic data, we combine docking, molecular dynamics (MD) simulations, and molecular mechanics generalized Born surface area approaches to explore the intricacies of the EGCG-p53 binding mechanism. A triplicate microsecond MD simulation for each system is initially performed to capture diverse p53 NTD conformations. From the start, the most populated cluster of the second run (R2-1) stands out due to a unique opening between Trp23 and Trp53. During MD simulations, this conformation allows EGCG to sustain a high level of stability and affinity while interacting with both regions of interest and deepening the binding pocket. Structural analysis emphasizes the significance of pyrogallol motifs in EGCG binding. Therefore, the conformational shift in this gap is pivotal, enabling EGCG to impede p53 interactions and manifest its anticancer properties. These findings enhance the present comprehension of the anticancer properties of green tea polyphenols and pave the way for future therapeutic developments.
Collapse
Affiliation(s)
- Erick Bahena Culhuac
- Laboratorio
de Diseño y Desarrollo de Nuevos Fármacos e Innovación
Biotecnológica, Escuela Superior de Medicina, Instituto Politécnico Nacional, Ciudad de México 11340, Mexico
- Universidad
Autónoma del Estado de México Facultad de Ciencias, Toluca 50000, Mexico
| | - Martiniano Bello
- Laboratorio
de Diseño y Desarrollo de Nuevos Fármacos e Innovación
Biotecnológica, Escuela Superior de Medicina, Instituto Politécnico Nacional, Ciudad de México 11340, Mexico
| |
Collapse
|
9
|
Saravanan V, Ahammed I, Bhattacharya A, Bhattacharya S. Uncovering allostery and regulation in SORCIN through molecular dynamics simulations. J Biomol Struct Dyn 2024; 42:1812-1825. [PMID: 37098805 DOI: 10.1080/07391102.2023.2202772] [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: 02/02/2023] [Accepted: 04/08/2023] [Indexed: 04/27/2023]
Abstract
Soluble resistance-related calcium-binding protein or Sorcin is an allosteric, calcium-binding Penta-EF hand (PEF) family protein implicated in multi-drug resistant cancers. Sorcin is known to bind chemotherapeutic molecules such as Doxorubicin. This study uses in-silico molecular dynamics simulations to explore the dynamics and allosteric behavior of Sorcin in the context of Ca2+ uptake and Doxorubicin binding. The results show that Ca2+ binding induces large, but reversible conformational changes in the Sorcin structure which manifest as rigid body reorientations that preserve the local secondary structure. A reciprocal allosteric handshake centered around the EF5 hand is found to be key in Sorcin dimer formation and stabilization. Binding of Doxorubicin results in rearrangement of allosteric communities which disrupts long-range allosteric information transfer from the N-terminal domain to the middle lobe. However, this binding does not result in secondary structure destabilization. Sorcin does not appear to have a distinct Ca2+ activated mode of Doxorubicin binding.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Vinnarasi Saravanan
- Department of Chemical Engineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Ijas Ahammed
- Department of Chemical Engineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Akash Bhattacharya
- Visiting Assistant Professor of Physics, St. Mary's University, San Antonio, Texas, USA
| | - Swati Bhattacharya
- Department of Chemical Engineering, Indian Institute of Technology Bombay, Mumbai, India
| |
Collapse
|
10
|
Sahoo S, Lee HK, Shin D. Structure-based virtual screening and molecular dynamics studies to explore potential natural inhibitors against 3C protease of foot-and-mouth disease virus. Front Vet Sci 2024; 10:1340126. [PMID: 38298458 PMCID: PMC10827980 DOI: 10.3389/fvets.2023.1340126] [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: 11/21/2023] [Accepted: 12/27/2023] [Indexed: 02/02/2024] Open
Abstract
Foot-and-mouth disease (FMD) is a highly infectious animal disease caused by foot-and-mouth disease virus (FMDV) and primarily infects cloven-hoofed animals such as cattle, sheep, goats, and pigs. It has become a significant health concern in global livestock industries because of diverse serotypes, high mutation rates, and contagious nature. There is no specific antiviral treatment available for FMD. Hence, based on the importance of 3C protease in FMDV viral replication and pathogenesis, we have employed a structure-based virtual screening method by targeting 3C protease with a natural compounds dataset (n = 69,040) from the InterBioScreen database. Virtual screening results identified five potential compounds, STOCK1N-62634, STOCK1N-96109, STOCK1N-94672, STOCK1N-89819, and STOCK1N-80570, with a binding affinity of -9.576 kcal/mol, -8.1 kcal/mol, -7.744 kcal/mol, -7.647 kcal/mol, and - 7.778 kcal/mol, respectively. The compounds were further validated through physiochemical properties and density functional theory (DFT). Subsequently, the comparative 300-ns MD simulation of all five complexes exhibited overall structural stability from various MD analyses such as root mean square deviation (RMSD), root mean square fluctuation (RMSF), radius of gyration (Rg), solvent accessible surface area (SASA), H-bonds, principal component analysis (PCA), and free energy landscape (FEL). Furthermore, MM-PBSA calculation suggests that all five compounds, particularly STOCK1N-62634, STOCK1N-96109, and STOCK1N-94672, can be considered as potential inhibitors because of their strong binding affinity toward 3C protease. Thus, we hope that these identified compounds can be studied extensively to develop natural therapeutics for the better management of FMD.
Collapse
Affiliation(s)
- Sthitaprajna Sahoo
- Department of Agricultural Convergence Technology, Jeonbuk National University, Jeonju, Republic of Korea
| | - Hak-Kyo Lee
- Department of Agricultural Convergence Technology, Jeonbuk National University, Jeonju, Republic of Korea
- Department of Animal Biotechnology, Jeonbuk National University, Jeonju, Republic of Korea
| | - Donghyun Shin
- Department of Agricultural Convergence Technology, Jeonbuk National University, Jeonju, Republic of Korea
| |
Collapse
|
11
|
Coste A, Slejko E, Zavadlav J, Praprotnik M. Developing an Implicit Solvation Machine Learning Model for Molecular Simulations of Ionic Media. J Chem Theory Comput 2024; 20:411-420. [PMID: 38118122 PMCID: PMC10782447 DOI: 10.1021/acs.jctc.3c00984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 12/04/2023] [Accepted: 12/04/2023] [Indexed: 12/22/2023]
Abstract
Molecular dynamics (MD) simulations of biophysical systems require accurate modeling of their native environment, i.e., aqueous ionic solution, as it critically impacts the structure and function of biomolecules. On the other hand, the models should be computationally efficient to enable simulations of large spatiotemporal scales. Here, we present the deep implicit solvation model for sodium chloride solutions that satisfies both requirements. Owing to the use of the neural network potential, the model can capture the many-body potential of mean force, while the implicit water treatment renders the model inexpensive. We demonstrate our approach first for pure ionic solutions with concentrations ranging from physiological to 2 M. We then extend the model to capture the effective ion interactions in the vicinity and far away from a DNA molecule. In both cases, the structural properties are in good agreement with all-atom MD, showcasing a general methodology for the efficient and accurate modeling of ionic media.
Collapse
Affiliation(s)
- Amaury Coste
- Laboratory
for Molecular Modeling, National Institute of Chemistry, Ljubljana SI-1001, Slovenia
| | - Ema Slejko
- Laboratory
for Molecular Modeling, National Institute of Chemistry, Ljubljana SI-1001, Slovenia
- Department
of Physics, Faculty of Mathematics and Physics, University of Ljubljana, Ljubljana SI-1000, Slovenia
| | - Julija Zavadlav
- Professorship
of Multiscale Modeling of Fluid Materials, TUM School of Engineering
and Design, Technical University of Munich, Garching Near Munich DE-85748, Germany
| | - Matej Praprotnik
- Laboratory
for Molecular Modeling, National Institute of Chemistry, Ljubljana SI-1001, Slovenia
- Department
of Physics, Faculty of Mathematics and Physics, University of Ljubljana, Ljubljana SI-1000, Slovenia
| |
Collapse
|
12
|
AlRawashdeh S, Barakat KH. Applications of Molecular Dynamics Simulations in Drug Discovery. Methods Mol Biol 2024; 2714:127-141. [PMID: 37676596 DOI: 10.1007/978-1-0716-3441-7_7] [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] [Indexed: 09/08/2023]
Abstract
In the current drug development process, molecular dynamics (MD) simulations have proven to be very useful. This chapter provides an overview of the current applications of MD simulations in drug discovery, from detecting protein druggable sites and validating drug docking outcomes to exploring protein conformations and investigating the influence of mutations on its structure and functions. In addition, this chapter emphasizes various strategies to improve the conformational sampling efficiency in molecular dynamics simulations. With a growing computer power and developments in the production of force fields and MD techniques, the importance of MD simulations in helping the drug development process is projected to rise significantly in the future.
Collapse
Affiliation(s)
- Sara AlRawashdeh
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - Khaled H Barakat
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
13
|
Jovičić SM. Enzyme ChE, cholinergic therapy and molecular docking: Significant considerations and future perspectives. Int J Immunopathol Pharmacol 2024; 38:3946320241289013. [PMID: 39367568 PMCID: PMC11526157 DOI: 10.1177/03946320241289013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 09/18/2024] [Indexed: 10/06/2024] Open
Abstract
Enzyme Che plays an essential role in cholinergic and non-cholinergic functions. It is present in the fertilized/unfertilized eggs and sperm of different species. Inclusion criteria for data collection from electronic databases NCBI and Google Scholar are enzyme AChE/BChE, cholinergic therapy, genomic organization and gene transcription, enzyme structure, biogenesis, transport, processing and localization, molecular signaling and biological function, polymorphism and influencing factors. Enzyme Che acts as a signaling receptor during hematopoiesis, protein adhesion, amyloid fiber formation, neurite outgrowth, bone development, and maturation, explaining the activity out of synaptic neurotransmission. Polymorphism in the Che genes correlates to various diseases and diverse drug responses. In particular, change accompanies cancer, neurodegenerative, and cardiovascular disease. Literature knowledge indicates the importance of Che inhibitors that influence biochemical and molecular pathways in disease treatment, genomic organization, gene transcription, structure, biogenesis, transport, processing, and localization of Che enzyme. Enzyme Che polymorphism changes indicate the possibility of efficient and new inhibitor drug target mechanisms in diverse research areas.
Collapse
Affiliation(s)
- Snežana M Jovičić
- Department of Genetics, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
14
|
Plau J, Morgan CE, Fedorov Y, Banerjee S, Adams DJ, Blaner WS, Yu EW, Golczak M. Discovery of Nonretinoid Inhibitors of CRBP1: Structural and Dynamic Insights for Ligand-Binding Mechanisms. ACS Chem Biol 2023; 18:2309-2323. [PMID: 37713257 PMCID: PMC10591915 DOI: 10.1021/acschembio.3c00402] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 09/01/2023] [Indexed: 09/16/2023]
Abstract
The dysregulation of retinoid metabolism has been linked to prevalent ocular diseases including age-related macular degeneration and Stargardt disease. Modulating retinoid metabolism through pharmacological approaches holds promise for the treatment of these eye diseases. Cellular retinol-binding protein 1 (CRBP1) is the primary transporter of all-trans-retinol (atROL) in the eye, and its inhibition has recently been shown to protect mouse retinas from light-induced retinal damage. In this report, we employed high-throughput screening to identify new chemical scaffolds for competitive, nonretinoid inhibitors of CRBP1. To understand the mechanisms of interaction between CRBP1 and these inhibitors, we solved high-resolution X-ray crystal structures of the protein in complex with six selected compounds. By combining protein crystallography with hydrogen/deuterium exchange mass spectrometry, we quantified the conformational changes in CRBP1 caused by different inhibitors and correlated their magnitude with apparent binding affinities. Furthermore, using molecular dynamic simulations, we provided evidence for the functional significance of the "closed" conformation of CRBP1 in retaining ligands within the binding pocket. Collectively, our study outlines the molecular foundations for understanding the mechanism of high-affinity interactions between small molecules and CRBPs, offering a framework for the rational design of improved inhibitors for this class of lipid-binding proteins.
Collapse
Affiliation(s)
- Jacqueline Plau
- Department
of Pharmacology, Small Molecule Drug Development Core Facility, Department of Genetics, and Cleveland Center
for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, Ohio 44106, United States
| | - Christopher E. Morgan
- Department
of Pharmacology, Small Molecule Drug Development Core Facility, Department of Genetics, and Cleveland Center
for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, Ohio 44106, United States
- Department
of Chemistry, Thiel College, Greenville, Pennsylvania 16125, United States
| | - Yuriy Fedorov
- Department
of Pharmacology, Small Molecule Drug Development Core Facility, Department of Genetics, and Cleveland Center
for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, Ohio 44106, United States
| | - Surajit Banerjee
- Department
of Chemistry and Chemical Biology, Cornell
University, Ithaca, New York 14850, United States
- Northeastern
Collaborative Access Team, Argonne National
Laboratory, Argonne, Illinois 60439, United States
| | - Drew J. Adams
- Department
of Pharmacology, Small Molecule Drug Development Core Facility, Department of Genetics, and Cleveland Center
for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, Ohio 44106, United States
| | - William S. Blaner
- Department
of Medicine, College of Physicians and Surgeons, Columbia University, New York, New York 10032, United States
| | - Edward W. Yu
- Department
of Pharmacology, Small Molecule Drug Development Core Facility, Department of Genetics, and Cleveland Center
for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, Ohio 44106, United States
| | - Marcin Golczak
- Department
of Pharmacology, Small Molecule Drug Development Core Facility, Department of Genetics, and Cleveland Center
for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, Ohio 44106, United States
| |
Collapse
|
15
|
Muthusamy K, Ramasamy G, Ravikumar C, Natesan S, Muthurajan R, Uthandi S, Kalyanasundaram K, Tiwari V. Exploring bixin from Bixa orellana L. seeds: quantification and in silico insights into its anti-cancer potential. J Biomol Struct Dyn 2023; 42:12244-12258. [PMID: 37837422 DOI: 10.1080/07391102.2023.2268202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 10/03/2023] [Indexed: 10/16/2023]
Abstract
Bixin, the key pigment of Bixa orellana L., is an apo-carotenoid found in the seed arils. The present study aimed to quantitatively determine the bixin content of seeds and explore its anti-cancer activity through in silico studies. The bixin content from the seeds of the local genotype, TNMTP8, quantified by RP-HPLC was 4.58 mg per gram. The prediction of pharmacological activity suggested that bixin may serve as a BRAF, MMP9, TNF expression inhibitors, and TP53 expression enhancer. According to molecular docking analysis, bixin interacted with eight different skin cancer targets and had the lowest binding energy compared to the standard drug, 5-fluorouracil. The binding score between bixin and the targets ranged from -4.7 to -8.7 kcal/mol. The targets BRAF and SIRT3 interacted well with bixin, with binding energies as low as -8.3 and -8.7 kcal/mol, respectively. Hence, the dynamic behavior of these two docked complexes throughout a 500 ns trajectory run was investigated further. The Root Mean Square Deviation (RMSD), Root Mean Square Fluctuation (RMSF) values, and total contacts as a function of time recorded during scrutiny suggest that both complexes were stable. This was validated by post-molecular dynamics analysis using Molecular Mechanics Generalized Born Surface Area (MM-GBSA). Principal component analysis (PCA) was used to analyze the significant differences in motion exhibited by BRAF-Bixin and SIRT3-Bixin. The results showed that bixin is a promising source for potential treatment interventions in skin cancer therapies.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Kaviyapriya Muthusamy
- Centre for Plant Molecular Biology and Biotechnology, Tamil Nadu Agricultural University, Coimbatore, India
| | - Gnanam Ramasamy
- Centre for Plant Molecular Biology and Biotechnology, Tamil Nadu Agricultural University, Coimbatore, India
| | - Caroline Ravikumar
- Centre for Plant Molecular Biology and Biotechnology, Tamil Nadu Agricultural University, Coimbatore, India
| | - Senthil Natesan
- Centre for Plant Molecular Biology and Biotechnology, Tamil Nadu Agricultural University, Coimbatore, India
| | - Raveendran Muthurajan
- Centre for Plant Molecular Biology and Biotechnology, Tamil Nadu Agricultural University, Coimbatore, India
| | - Sivakumar Uthandi
- Biocatalysts Laboratory, Department of Agricultural Microbiology, Tamil Nadu Agricultural University, Coimbatore, India
| | - Kumaran Kalyanasundaram
- Department of Forest Biology and Tree Improvement, Forest College and Research Institute, Tamil Nadu Agricultural University, Coimbatore, India
| | - Vikas Tiwari
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore, India
| |
Collapse
|
16
|
Liu C, Kutchukian P, Nguyen ND, AlQuraishi M, Sorger PK. A Hybrid Structure-Based Machine Learning Approach for Predicting Kinase Inhibition by Small Molecules. J Chem Inf Model 2023; 63:5457-5472. [PMID: 37595065 PMCID: PMC10498990 DOI: 10.1021/acs.jcim.3c00347] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Indexed: 08/20/2023]
Abstract
Kinases have been the focus of drug discovery programs for three decades leading to over 70 therapeutic kinase inhibitors and biophysical affinity measurements for over 130,000 kinase-compound pairs. Nonetheless, the precise target spectrum for many kinases remains only partly understood. In this study, we describe a computational approach to unlocking qualitative and quantitative kinome-wide binding measurements for structure-based machine learning. Our study has three components: (i) a Kinase Inhibitor Complex (KinCo) data set comprising in silico predicted kinase structures paired with experimental binding constants, (ii) a machine learning loss function that integrates qualitative and quantitative data for model training, and (iii) a structure-based machine learning model trained on KinCo. We show that our approach outperforms methods trained on crystal structures alone in predicting binary and quantitative kinase-compound interaction affinities; relative to structure-free methods, our approach also captures known kinase biochemistry and more successfully generalizes to distant kinase sequences and compound scaffolds.
Collapse
Affiliation(s)
- Changchang Liu
- Laboratory
of Systems Pharmacology, Department of Systems Biology, Harvard Program
in Therapeutic Science, Harvard Medical
School, Boston, Massachusetts 02115, United States
| | - Peter Kutchukian
- Novartis
Institutes for Biomedical Research, Cambridge, Massachusetts 02139, United States
| | - Nhan D. Nguyen
- Pritzker
School of Molecular Engineering, University
of Chicago, Chicago, Illinois 60637, United
States
| | - Mohammed AlQuraishi
- Department
of Systems Biology, Columbia University, New York, New York 10032, United States
| | - Peter K. Sorger
- Laboratory
of Systems Pharmacology, Department of Systems Biology, Harvard Program
in Therapeutic Science, Harvard Medical
School, Boston, Massachusetts 02115, United States
| |
Collapse
|
17
|
Gao M, Kang D, Liu N, Liu Y. In Silico Discovery of Small-Molecule Inhibitors Targeting SARS-CoV-2 Main Protease. Molecules 2023; 28:5320. [PMID: 37513194 PMCID: PMC10383128 DOI: 10.3390/molecules28145320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 06/29/2023] [Accepted: 07/05/2023] [Indexed: 07/30/2023] Open
Abstract
The COVID-19 pandemic has caused severe health threat globally, and novel SARS-Cov-2 inhibitors are urgently needed for antiviral treatment. The main protease (Mpro) of the virus is one of the most effective and conserved targets for anti-SARS-CoV-2 drug development. In this study, we utilized a molecular docking-based virtual screening approach against the conserved catalytic site to identify small-molecule inhibitors of SARS-CoV-2 Mpro. Further biological evaluation helped us identify two compounds, AF-399/40713777 and AI-942/42301830, with moderate inhibitory activity. Besides that, the in silico data, including molecular dynamics (MD) simulation, binding free energy calculations, and AMDET profiles, suggested that these two hits could serve as the starting point for the future development of COVID-19 intervention treatments.
Collapse
Affiliation(s)
- Menghan Gao
- School of Pharmacy and Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, 6699 Qingdao Road, Jinan 250117, China
- NHC Key Laboratory of Biotechnology Drugs, Shandong Academy of Medical Sciences, 6699 Qingdao Road, Jinan 250117, China
- Key Lab for Rare & Uncommon Diseases of Shandong Province, 6699 Qingdao Road, Jinan 250117, China
| | - Dongwei Kang
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, Jinan 250012, China
| | - Na Liu
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, Jinan 250012, China
| | - Yanna Liu
- School of Pharmacy and Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, 6699 Qingdao Road, Jinan 250117, China
- NHC Key Laboratory of Biotechnology Drugs, Shandong Academy of Medical Sciences, 6699 Qingdao Road, Jinan 250117, China
- Key Lab for Rare & Uncommon Diseases of Shandong Province, 6699 Qingdao Road, Jinan 250117, China
| |
Collapse
|
18
|
Mehta CC, Rohit S, Patel S, Bhatt HG. New molecular insights for 4 H-1,2,4-triazole derivatives as inhibitors of tankyrase and Wnt-signaling antagonist: a molecular dynamics simulation study. J Biomol Struct Dyn 2023; 41:13496-13508. [PMID: 36755438 DOI: 10.1080/07391102.2023.2175376] [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: 09/27/2022] [Accepted: 01/27/2023] [Indexed: 02/10/2023]
Abstract
Tankyrase (TNKS) enzymes remained central biotargets to treat Wnt-driven colorectal cancers. The success of Olaparib posited the druggability of PARP family enzymes depending on their role in tumor proliferation. In this work, an MD-simulation-based comparative assessment of the protein-ligand interactions using the best-docked poses of three selected compounds (two of the designed and previously synthesized molecules obtained through molecular docking and one reported TNKS inhibitor) was performed for a 500 ns period. The PDB:ID-7KKP and 3U9H were selected for TNKS1 and TNKS2, respectively. The Molecular Mechanics Generalized Born Surface Area (MM-GBSA) based binding energy data exhibited stronger binding of compound-15 (average values of -102.92 and -104.32 kcal/mol for TNKS1 and TNKS2, respectively) as compared to compound-22 (average values of -82.99 and -85.68 kcal/mol for TNKS1 and TNKS2, respectively) and the reported compound-32 (average values of -81.89 and -74.43 kcal/mol for TNKS1 and TNKS2, respectively). Compound-15 and compound-22 exhibited comparable or superior binding to both receptors forming stable complexes when compared to that of compound-32 upon examining their MD trajectories. The key contributors were hydrophobic stacking and optimum hydrogen bonding allowing these molecules to occupy the adenosine pocket by interfacing D-loop residues. The results of bond distance analysis, radius of gyration, root mean square deviation, root mean square fluctuation, snapshots at different time intervals, LUMO-HUMO energy differences, electrostatic potential calculations, and binding free energy suggested better binding efficiency for compound-15 to TNKS enzymes. The computed physicochemical and ADMET properties of compound-15 were encouraging and could be explored further for drug development.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Chirag C Mehta
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, India
| | | | - Saumya Patel
- Department of Botany, Bioinformatics and Climate Change Impacts Management, Gujarat University, Ahmedabad, India
| | - Hardik G Bhatt
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, India
| |
Collapse
|
19
|
Darmadi D, Lindarto D, Siregar J, Widyawati T, Rusda M, Amin MM, Yusuf F, Eyanoer PC, Lubis M, Rey I. Study of the Molecular Dynamics Stability in the Inhibitory Interaction of Tenofovir Disoproxil Fumarate against CTLA-4 in Chronic Hepatitis B Patients. Med Arch 2023; 77:227-230. [PMID: 37700917 PMCID: PMC10495148 DOI: 10.5455/medarh.2023.77.227-230] [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: 04/26/2023] [Accepted: 05/24/2023] [Indexed: 09/14/2023] Open
Abstract
Background Tenofovir disoproxil fumarate (TDF) is a first-line nucleotide analog (NA) drug for hepatitis B therapy. Long-term NA therapy increases peripheral T cell levels to enhance antiviral response, while CTLA-4 inhibits the activation. Objective This study analyzed the interaction between TDF and CTLA-4 through molecular docking. Methods Target protein and ligand data mining were performed, and proteins were prepared by removing water molecules in the Discovery Studio 2019 software. The energy minimization was performed on ligands using Pyrx v.0.9.8 software. Protein-ligand docking was performed using Autodock Vina integrated with Pyrx v.09.8. Meanwhile, the docking of proteins was accomplished using the Haddock server. The BioVia Discovery Studio 2019 software visualized the interaction between the compound and the docked protein. Molecular dynamics simulations were carried out using the YASARA Dynamic program developed by Biosciences GmbH. Results TDF ligand has good and stable inhibitory activity against the CTLA-4/B7-1 and CTLA4/B7-2 complexes. TDF docking has been shown to initiate conformational changes, indicating the ligand's inhibitory activity. The significant conformational changes based on superimposition results were shown by the CTLA-4/TDF/B7-2 and CTLA-4/B7-1/TDF complexes. TDF in all ligands undergoes bonding and displacement of binding sites. Conclusion Treatment with TDF was predicted to have inhibitory activity against CTLA-4, especially in its complex form with B7-1 and B7-2.
Collapse
Affiliation(s)
- Darmadi Darmadi
- Philosophy Doctor in Medicine Program, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
- Department of Internal Medicine, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| | - Dharma Lindarto
- Department of Internal Medicine, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| | - Jelita Siregar
- Department of Clinical Pathology, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| | - Tri Widyawati
- Department of Pharmacology and Therapeutic, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
- Master Program in Tropical Medicine, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| | - Muhammad Rusda
- Department of Obstetrics and Gynecology, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| | - Mustafa Mahmud Amin
- Department of Psychiatry, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| | - Fauzi Yusuf
- Department of Internal Medicine, Faculty of Medicine, Universitas Syiah Kuala, Banda Aceh, Indonesia
| | - Putri Chairani Eyanoer
- Department of Public Health, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| | - Masrul Lubis
- Department of Internal Medicine, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| | - Imelda Rey
- Department of Internal Medicine, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| |
Collapse
|
20
|
Kalhor S, Fattahi A. Design of ionic liquids containing glucose and choline as drug carriers, finding the link between QM and MD studies. Sci Rep 2022; 12:21941. [PMID: 36535965 PMCID: PMC9763358 DOI: 10.1038/s41598-022-25963-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 12/07/2022] [Indexed: 12/23/2022] Open
Abstract
Designing drug delivery systems for therapeutic compounds whose receptors are located in the cytosol of cells is challenging as a bilayer cell membrane is negatively charged. The newly designed drug delivery systems should assist the mentioned drugs in passing the membrane barriers and achieving their targets. This study concentrated on developing novel ionic liquids (ILs) that interact effectively with cell membranes. These ILs are based on glucose-containing choline and are expected to be non-toxic. The binding energies of the known pharmaceutically active ionic liquids were calculated at the B3LYP/6-311++G(d,p) level in the gas phase and compared with those of our newly designed carbohydrate-based ionic liquids. Subsequently, we employed MD simulations to obtain information about the interactions of these known and designed ILs with the cell membrane. In our approach, we adopted QM and MD studies and illustrated that there could be a link between the QM and MD results.
Collapse
Affiliation(s)
- Sepideh Kalhor
- Department of Chemistry, Sharif University of Technology, Tehran, Iran
| | - Alireza Fattahi
- Department of Chemistry, Sharif University of Technology, Tehran, Iran.
| |
Collapse
|
21
|
Fludarabine, a Potential DNA-Dependent RNA Polymerase Inhibitor, as a Prospective Drug against Monkeypox Virus: A Computational Approach. Pharmaceuticals (Basel) 2022; 15:ph15091129. [PMID: 36145351 PMCID: PMC9504824 DOI: 10.3390/ph15091129] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 08/26/2022] [Accepted: 08/29/2022] [Indexed: 12/16/2022] Open
Abstract
Monkeypox is a zoonotic contagious disease that has recently re-emerged in different countries worldwide. Due to the lack of an effective treatment that eliminates the virus, there is an urgent need to find effective drugs to stop the spread of the multi-country outbreak. The current study aimed to use computational methods to quickly identify potentially effective drugs against the Monkeypox virus (MPXV). Three MPXV proteins were targeted in this study due to their essential role in viral replication (a DNA-Dependent RNA Polymerase subunit (A6R)), a protein involved in cell entry (D8L), and a protein catalyzing the envelopment of intracellular mature virus particles (F13L). We virtually screened a library of 1615 FDA-approved compounds, utilizing different in-silico approaches including computational modeling, molecular docking, molecular dynamic (MD) simulation, and MM-GBSA. The compound Fludarabine was found to have the best docking score (−7.53 kcal/mol) in relation to the MPXV A6R protein. Additionally, Fludarabine showed in-silico activity on the D8L and F13L proteins. During the whole period of the 100 ns MD simulation, the complex of A6R and Fludarabine exhibited the best stability. This stability was reflected in a good score of MM-GBSA, with an average value of −44.62 kcal/mole in a range between −53.26 and −35.49 and a low value of standard deviation (3.76). Furthermore, Fludarabine blocked efficiently the Asn175 residue which has an important role in the attachment of the virus to a host cell. The results of this study recommend more in vitro studies on this compound, as a starting point to develop a novel treatment against MPXV.
Collapse
|