51
|
Adekunle YA, Samuel BB, Oluyemi WM, Adewumi AT, Mosebi S, Nahar L, Fatokun AA, Sarker SD. Oleanolic acid purified from the stem bark of Olax subscorpioidea Oliv. inhibits the function and catalysis of human 17 β-hydroxysteroid dehydrogenase 1. J Biomol Struct Dyn 2024:1-14. [PMID: 39485270 DOI: 10.1080/07391102.2024.2423173] [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: 01/20/2024] [Accepted: 06/04/2024] [Indexed: 11/03/2024]
Abstract
Cancer is a leading cause of global death. Medicinal plants have gained increasing attention in cancer drug discovery. In this study, the stem bark extract of Olax subscorpioidea, which is used in ethnomedicine to treat cancer, was subjected to phytochemical investigation leading to the isolation of oleanolic acid (OA). The structure was elucidated by 1-dimensional and 2-dimensional nuclear magnetic resonance spectroscopic (NMR) data, and by comparing its data with previously reported data. Molecular docking was used to investigate the interactions of OA with nine selected cancer-related protein targets. OA docked well with human 17β-hydroxysteroid dehydrogenase type-1 (17βHSD1), caspase-3, and epidermal growth factor receptor tyrosine kinase (binding affinities: -9.8, -9.3, and -9.1 kcal/mol, respectively). OA is a triterpenoid compound with structural similarity to steroids. This similarity with the substrates of 17βHSD1 gives the inhibitor candidate an excellent opportunity to bind to 17βHSD1. The structural and functional dynamics of OA-17βHSD1 were investigated by molecular dynamics simulations at 240 ns. Molecular mechanics/Poisson-Boltzmann surface area (MMPBSA) studies showed that OA had a binding free energy that is comparable with that of vincristine (-52.76, and -63.56 kcal/mol, respectively). The average C-α root mean square of deviation (RMSD) value of OA (1.69 Å) compared with the unbound protein (2.01 Å) indicated its high stability at the protein's active site. The binding energy and stability at the active site of 17βHSD1 recorded in this study indicate that OA exhibited profound inhibitory potential. OA could be a good scaffold for developing new anti-breast cancer drugs.
Collapse
Affiliation(s)
- Yemi A Adekunle
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Ibadan, Ibadan, Oyo State, Nigeria
- Centre for Natural Products Discovery, School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
- Department of Pharmaceutical and Medicinal Chemistry, College of Pharmacy, Afe Babalola University, Ado-Ekiti, Ekiti State, Nigeria
| | - Babatunde B Samuel
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Ibadan, Ibadan, Oyo State, Nigeria
| | - Wande M Oluyemi
- Department of Pharmaceutical and Medicinal Chemistry, College of Pharmacy, Afe Babalola University, Ado-Ekiti, Ekiti State, Nigeria
| | - Adeniyi T Adewumi
- Department of Life and Consumer Sciences, University of South Africa, Florida, South Africa
| | - Salerwe Mosebi
- Department of Life and Consumer Sciences, University of South Africa, Florida, South Africa
| | - Lutfun Nahar
- Laboratory of Growth Regulators, Palacký University and Institute of Experimental Botany, The Czech Academy of Sciences, Olomouc, Czech Republic
| | - Amos A Fatokun
- Centre for Natural Products Discovery, School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
| | - Satyajit D Sarker
- Centre for Natural Products Discovery, School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
| |
Collapse
|
52
|
Fakih TM. Molecularly imprinted polymer-based sensors for identification volatile compounds in pharmaceutical products: in silico rational design. J Biomol Struct Dyn 2024; 42:9639-9649. [PMID: 37643016 DOI: 10.1080/07391102.2023.2252090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 08/21/2023] [Indexed: 08/31/2023]
Abstract
The present study aimed to strategically design a Molecularly Imprinted Polymer (MIP) with selective extraction capabilities for volatile compounds found in pork. These specific volatile compounds, such as 3-methyl-1-butanol, 1-nonanal, octanal, hexanal, 2-pentyl-furan, 1-penten-3-one, N-morpholinomethyl-isopropyl-sulfide, methyl butyrate, and (E,E)-2,4-decadienal, are primarily responsible for the distinctive aroma and flavor characteristics associated with pork. Molecular dynamics simulations were employed to investigate the stability of the pre-polymerization system, simulating the interactions between the volatile compounds as templates, 4-hydroxyethyl methacrylate (HEMA) as monomers, and ethylene glycol dimethacrylate (EGDMA) as crosslinkers. Computational simulations revealed that the optimal mole ratio of 1:4:20 for templates, monomers, and crosslinkers resulted in the most favorable functional radial distribution and exhibited the strongest interactions. To validate the computational findings, additional analyses were performed utilizing Molecular Mechanics Poisson-Boltzmann Surface Area (MM-PBSA), radial distribution function (RDF), and hydrogen bond (HBond) occupancy. The calculated binding free energy demonstrated that all template molecules were capable to bind with both the monomers and crosslinkers, including 1-penten-3-one and N-morpholinomethyl-isopropyl-sulfide displaying the strongest interactions, with values of -12,674 kJ/mol and -11,646 kJ/mol, respectively. The congruence between the results obtained from the molecular simulation analyses highlights the crucial role of molecular dynamics simulations in the study and development of MIP for the analysis of marker compounds present in pork.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Taufik Muhammad Fakih
- Department of Pharmacy, Faculty of Mathematics and Natural Sciences, Universitas Islam Bandung, Bandung, Indonesia
| |
Collapse
|
53
|
Ibrahim MAA, Ali SSM, Abdelrahman AHM, Abdeljawaad KAA, Sidhom PA, Sayed SRM, El-Tayeb MA, Paré PW, Hegazy MEF. Naturally Occurring Plant-Based Anticancerous Candidates as Potential ERK2 Inhibitors: In-Silico Database Mining and Molecular Dynamics Simulations. Chem Biodivers 2024; 21:e202401238. [PMID: 39075025 DOI: 10.1002/cbdv.202401238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 07/27/2024] [Accepted: 07/29/2024] [Indexed: 07/31/2024]
Abstract
The evolutionarily conserved extracellular signal-regulated kinase 2 (ERK2) is involved in regulating cellular signaling in both normal and pathological conditions. ERK2 expression is critical for human development, while hyperactivation is a major factor in tumor progression. Up to now, there have been no approved inhibitors that target ERK2, and as such, here we report on screening of a naturally occurring plant-based anticancerous compound-activity-target (NPACT) database for prospective ERK2 inhibitors. More than 1,500 phytochemicals were screened using in-silico molecular docking and molecular dynamics (MD) approaches. NPACT compounds with a docking score lower than a co-crystallized LHZ inhibitor (calc. -10.5 kcal/mol) were subjected to MD simulations. Binding energies (ΔGbinding) of inhibitor-ERK2 complexes over the MD course were estimated using an MM-GBSA approach. Based on MM-GBSA//100 ns MD simulations, the steroid zhankuic acid C (NPACT01034) demonstrated greater binding affinity against ERK2 protein than LHZ, with ΔGbinding values of -50.0 and -47.7 kcal/mol, respectively. Structural and energetical analyses throughout the MD course demonstrated stabilization of zhankuic acid C complexed with ERK2 protein. The anticipated ADMET properties of zhankuic acid C indicated minimal toxicity. Moreover, in-silico evaluation of fourteen ERK2 inhibitors in clinical trials demonstrated the higher binding affinity of zhankuic acid C towards ERK2 protein.
Collapse
Affiliation(s)
- Mahmoud A A Ibrahim
- Computational Chemistry Laboratory, Chemistry Department, Faculty of Science, Minia University, Minia, 61519, Egypt
- School of Health Sciences, Westville Campus, University of KwaZulu-Natal, Durban, 4000, South Africa
| | - Sara S M Ali
- Computational Chemistry Laboratory, Chemistry Department, Faculty of Science, Minia University, Minia, 61519, Egypt
| | - Alaa H M Abdelrahman
- Computational Chemistry Laboratory, Chemistry Department, Faculty of Science, Minia University, Minia, 61519, Egypt
| | - Khlood A A Abdeljawaad
- Computational Chemistry Laboratory, Chemistry Department, Faculty of Science, Minia University, Minia, 61519, Egypt
| | - Peter A Sidhom
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta, 31527, Egypt
| | - Shaban R M Sayed
- Department of Botany and Microbiology, College of Science, King Saud University, P.O. Box 2455, Riyadh, 11451, Saudi Arabia
| | - Mohamed A El-Tayeb
- Department of Botany and Microbiology, College of Science, King Saud University, P.O. Box 2455, Riyadh, 11451, Saudi Arabia
| | - Paul W Paré
- Department of Chemistry & Biochemistry, Texas Tech University, Lubbock, TX, 79409, USA
| | - Mohamed-Elamir F Hegazy
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, Mainz, 55128, Germany
| |
Collapse
|
54
|
El Alaouy MA, Alaqarbeh M, Ouabane M, Zaki H, ElBouhi M, Badaoui H, Moukhliss Y, Sbai A, Maghat H, Lakhlifi T, Bouachrine M. Computational Prediction of 3,5-Diaryl-1H-Pyrazole and spiropyrazolines derivatives as potential acetylcholinesterase inhibitors for alzheimer disease treatment by 3D-QSAR, molecular docking, molecular dynamics simulation, and ADME-Tox. J Biomol Struct Dyn 2024; 42:9547-9560. [PMID: 37655700 DOI: 10.1080/07391102.2023.2252116] [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: 09/03/2022] [Accepted: 08/20/2023] [Indexed: 09/02/2023]
Abstract
The efficacy of 40 synthesized variants of 3,5-diaryl-1H-pyrazole and spiropyrazoline' derivatives as acetylcholinesterase inhibitors is verified using a quantitative three-dimensional structure-activity relationship (3D-QSAR) by comparative molecular field analysis (CoMFA) and molecular similarity index analysis (CoMSIA) models. In this research, different field models proved that CoMSIA/SE model is the best model with high predictive power compared to several models (Qved2 = O.65; R2 = 0.980; R2test = 0.727). Also, contour maps produced by CoMSIA/SE model have been employed to prove the key structural needs of the activity. Consequently, six new compounds have been generated. Among these compounds, M4 and M5 were the most active but remained toxic and had poor absorption capacities. While the M1, M2, M3 and M6 remained highly active while respecting ADMET's characteristics. Molecular docking results showed compound M2 better with acetylcholinesterase than compound 22. The interactions are classical hydrogen bonding with residues TYR:124, TYR:72, and SER:293, which play a critical role in the biological activity as AChE inhibitors. MD results confirmed the docking results and showed that compound M2 had satisfactory stability with (ΔGbinding = -151.225 KJ/mol) in the active site of AChE receptor compared with compound 22 (ΔGbinding = -133.375 KJ/mol). In addition, both compounds had good stability regarding RMSD, Rg, and RMSF. The previous results show that the newly designed compound M2 is more active in the active site of AChE receptor than compound 22.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Moulay Ahfid El Alaouy
- Molecular Chemistry and Natural Substances Laboratory, Faculty of Science, University Moulay Ismail, Meknes, Morocco
| | | | - Mohamed Ouabane
- Molecular Chemistry and Natural Substances Laboratory, Faculty of Science, University Moulay Ismail, Meknes, Morocco
| | - Hanane Zaki
- BIO Laboratory, EST Khenifra, Sultan Moulay Slimane University Beni Mellal, Morocco
| | - Mohamed ElBouhi
- Molecular Chemistry and Natural Substances Laboratory, Faculty of Science, University Moulay Ismail, Meknes, Morocco
| | - Hassan Badaoui
- Molecular Chemistry and Natural Substances Laboratory, Faculty of Science, University Moulay Ismail, Meknes, Morocco
| | - Youness Moukhliss
- Molecular Chemistry and Natural Substances Laboratory, Faculty of Science, University Moulay Ismail, Meknes, Morocco
| | - Abdelouahid Sbai
- Molecular Chemistry and Natural Substances Laboratory, Faculty of Science, University Moulay Ismail, Meknes, Morocco
| | - Hamid Maghat
- Molecular Chemistry and Natural Substances Laboratory, Faculty of Science, University Moulay Ismail, Meknes, Morocco
| | - Tahar Lakhlifi
- Molecular Chemistry and Natural Substances Laboratory, Faculty of Science, University Moulay Ismail, Meknes, Morocco
| | - Mohammed Bouachrine
- Molecular Chemistry and Natural Substances Laboratory, Faculty of Science, University Moulay Ismail, Meknes, Morocco
- EST Khenifra, Sultan Moulay Sliman University, Benimellal, Morocco
| |
Collapse
|
55
|
Ancy I, Penislusshiyan S, Ameen F, Chitra L. Microsecond Molecular Dynamics Simulation to Gain Insight Into the Binding of MRTX1133 and Trametinib With KRAS G12D Mutant Protein for Drug Repurposing. J Mol Recognit 2024; 37:e3103. [PMID: 39318275 DOI: 10.1002/jmr.3103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 08/09/2024] [Accepted: 09/03/2024] [Indexed: 09/26/2024]
Abstract
The Kirsten Rat Sarcoma (KRAS) G12D mutant protein is a primary driver of pancreatic ductal adenocarcinoma, necessitating the identification of targeted drug molecules. Repurposing of drugs quickly finds new uses, speeding treatment development. This study employs microsecond molecular dynamics simulations to unveil the binding mechanisms of the FDA-approved MEK inhibitor trametinib with KRASG12D, providing insights for potential drug repurposing. The binding of trametinib was compared with clinical trial drug MRTX1133, which demonstrates exceptional activity against KRASG12D, for better understanding of interaction mechanism of trametinib with KRASG12D. The resulting stable MRTX1133-KRASG12D complex reduces root mean square deviation (RMSD) values, in Switch I and II domains, highlighting its potential for inhibiting KRASG12D. MRTX1133's robust interaction with Tyr64 and disruption of Tyr96-Tyr71-Arg68 network showcase its ability to mitigate the effects of the G12D mutation. In contrast, trametinib employs a distinctive binding mechanism involving P-loop, Switch I and II residues. Extended simulations to 1 μs reveal sustained network interactions with Tyr32, Thr58, and GDP, suggesting a role of trametinib in maintaining KRASG12D in an inactive state and impede the further cell signaling. The decomposition binding free energy values illustrate amino acids' contributions to binding energy, elucidating ligand-protein interactions and molecular stability. The machine learning approach reveals that van der Waals interactions among the residues play vital role in complex stability and the potential amino acids involved in drug-receptor interactions of each complex. These details provide a molecular-level understanding of drug binding mechanisms, offering essential knowledge for further drug repurposing and potential drug discovery.
Collapse
Affiliation(s)
- Iruthayaraj Ancy
- Research and Development Center, Bioinnov Solutions LLP, Salem, Tamil Nadu, India
| | | | - Fuad Ameen
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Loganathan Chitra
- Research and Development Center, Bioinnov Solutions LLP, Salem, Tamil Nadu, India
- Department of Prosthodontics and Implantology, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai, India
| |
Collapse
|
56
|
Vasconcelos D, Pina A, Habib N, Sousa S. In silico analysis of aptamer-RNA conjugate interactions with human transferrin receptor. Biophys Chem 2024; 314:107308. [PMID: 39208499 DOI: 10.1016/j.bpc.2024.107308] [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: 03/28/2024] [Revised: 08/02/2024] [Accepted: 08/09/2024] [Indexed: 09/04/2024]
Abstract
The human transmembrane protein Transferrin Receptor-1 is regarded as a promising target for the systemic delivery of therapeutic agents, particularly of nucleic acid therapeutics, such as short double stranded RNAs. This ubiquitous receptor is involved in cellular iron uptake, keeping intracellular homeostasis. It is overexpressed in multiple cancer cell types and is internalized via clathrin-mediated endocytosis. In previous studies, a human transferrin receptor-1 RNA aptamer, identified as TR14 ST1-3, was shown to be capable of effectively internalizing into cells in culture and to deliver small, double stranded RNAs in vitro and in vivo, via systemic administration. To understand, at the molecular level, the aptamer binding to the receptor and the impact of conjugation with the therapeutic RNA, a multi-level in silico protocol was employed, including protein-aptamer docking, molecular dynamics simulations and free energy calculations. The competition for the binding pocket, between the aptamer and the natural ligand human Transferrin, was also evaluated. The results show that the aptamer binds to the same region as Transferrin, with residues from the helical domain showing a critical role. Moreover, the conjugation to the therapeutic RNA, was shown not to affect aptamer binding. Overall, this study provides an atomic-level understanding of aptamer association to human Transferrin Receptor-1 and of its conjugation with a short model-therapeutic RNA, providing also important clues for futures studies aiming to deliver other oligonucleotide-based therapeutics via Transferrin Receptor.
Collapse
Affiliation(s)
- Daniel Vasconcelos
- Department of Surgery and Cancer, Imperial College London, London W12 0NN, UK; Apterna Ltd., London SW1P 2PN, UK; Center for Drug Discovery and Innovative Medicines (MedInUP), University of Porto, 4200-319 Porto, Portugal
| | - André Pina
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal; UCIBIO - Applied Molecular Biosciences Unit, BioSIM - Department of Biomedicine, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Nagy Habib
- Department of Surgery and Cancer, Imperial College London, London W12 0NN, UK; Apterna Ltd., London SW1P 2PN, UK
| | - Sérgio Sousa
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal; UCIBIO - Applied Molecular Biosciences Unit, BioSIM - Department of Biomedicine, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal.
| |
Collapse
|
57
|
Çankaya N, Kebiroğlu MH, Temüz MM. A comprehensive study of experimental and theoretical characterization and in silico toxicity analysis of new molecules. Drug Chem Toxicol 2024; 47:1226-1240. [PMID: 38757531 DOI: 10.1080/01480545.2024.2353724] [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: 01/10/2024] [Accepted: 05/04/2024] [Indexed: 05/18/2024]
Abstract
In this study, for the first time in the literature, a 2-(3-methoxyphenylamino)-2-oxoethyl acrylate (3MPAEA) molecule was synthesized in two steps, and a 2-chloro-N-(3-methoxyphenyl)acetamide (m-acetamide) was obtained in the first step. Experimental results were obtained using FTIR, 1H, and 13C NMR spectroscopy methods for m-acetamide and 3MPAEA compounds created in the laboratory environment and compared with theoretical results. Band gap (BG) energy, chemical hardness, electronegativity, chemical potential, and electrophilicity index were calculated. With vibration spectroscopic analysis, atom-molecule vibrations of the theoretical and experimental peaks of the spectrum were observed. The locations of C and H atoms were determined by nuclear magnetic resonance spectroscopy. The green, blue, and red regions of the potential energy map (MEP) map were examined. Some observed that the energy thermal, heat capacity, and entropy graphs increased in direct proportion to increasing the temperature in Kelvin, which is known as thermochemistry. The changes in the rotation, translation, and vibration of the molecule as its temperature increased were examined. When the thermochemistry surface map was examined, some observed that the temperature was high in the middle binding site of the molecules. Covalent interactions were graphed using the non-covalent interactions (NCIs) calculation method. In silico toxicity studies were carried out for m-acetamide and 3MPAEA molecules: fathead minnow LC50 (96 h), Daphnia magna LC50 (48 h), Tetrahymena pyriformis IGC50 (48 h), oral rat LD50, water solubility, bioconcentration factor, developmental toxicity, mutation, normal boiling point, flash point, melting point, density, thermal conductivity, viscosity, vapor pressure, etc. parameters were investigated.
Collapse
Affiliation(s)
- Nevin Çankaya
- Vocational School of Health Services, Usak University, Usak, Turkey
| | - Mehmet Hanifi Kebiroğlu
- Department of Opticianry, Darende Bekir Ilicak Vocational School, Malatya Turgut Ozal University, Malatya, Turkey
| | - Mehmet Mürşit Temüz
- Department of Chemistry, Faculty of Science, Firat University, Elazığ, Turkey
| |
Collapse
|
58
|
Vishwakarma KK, Kolthur US, Venkatramani R. Multiple Functional Protein-Protein Interaction Interfaces Allosterically Regulate ATP-Binding in Cyclin-Dependent Kinase-1. Proteins 2024; 92:1329-1342. [PMID: 39012208 DOI: 10.1002/prot.26729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 05/30/2024] [Accepted: 06/27/2024] [Indexed: 07/17/2024]
Abstract
The ATP-dependent phosphorylation activity of cyclin-dependent kinase 1 (CDK1), an essential enzyme for cell cycle progression, is regulated by interactions with Cyclin-B, substrate, and Cks proteins. We have recently shown that active site acetylation in CDK1 abrogated binding to Cyclin-B which posits an intriguing long-range communication between the catalytic site and the protein-protein interaction (PPI) interface. Now, we demonstrate a general allosteric link between the CDK1 active site and all three of its PPI interfaces through atomistic molecular dynamics (MD) simulations. Specifically, we examined ATP binding free energies to CDK1 in native nonacetylated (K33wt) and acetylated (K33Ac) forms as well as the acetyl-mimic K33Q and the acetyl-null K33R mutant forms, which are accessible in vitro. In agreement with experiments, ATP binding is stronger in K33wt relative to the other three perturbed states. Free energy decomposition reveals, in addition to expected local changes, significant and selective nonlocal entropic responses to ATP binding/perturbation of K33 from theαC -helix, activation loop (A-loop), andαG -α H segments in CDK1 which interface with Cyclin-B, substrate, and Cks proteins, respectively. Statistical analysis reveals that while entropic responses of protein segments to active site perturbations are on average correlated with their dynamical changes, such correlations are lost in about 9%-48% of the dataset depending on the segment. Besides proving the bi-directional communication between the active site and the CDK1:Cyclin-B interface, our study uncovers a hitherto unknown mode of ATP binding regulation by multiple PPI interfaces in CDK1.
Collapse
Affiliation(s)
| | - Ullas Seetharam Kolthur
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
- Tata Institute of Fundamental Research, Hyderabad, India
| | - Ravindra Venkatramani
- Department of Chemical Sciences, Tata Institute of Fundamental Research, Mumbai, India
| |
Collapse
|
59
|
Çoban V, Çankaya N, Azarkan SY. New oxomethacrylate and acetamide: synthesis, characterization, and their computational approaches: molecular docking, molecular dynamics, and ADME analyses. Drug Chem Toxicol 2024; 47:1175-1184. [PMID: 38745411 DOI: 10.1080/01480545.2024.2349651] [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: 01/16/2024] [Accepted: 04/25/2024] [Indexed: 05/16/2024]
Abstract
The compounds 2-chloro-N-(3-methoxyphenyl)acetamide (m-acetamide) and 2-(3-methoxyphenylamino)-2-oxoethyl methacrylate (3MPAEMA) were synthesized in this study for the first time in the literature. FTIR, 1H, and 13C NMR spectroscopic techniques were used to characterize it. Subsequently, computational techniques were used to assess various ADME factors, such as drug-likeness properties, bioavailability score, and adherence to Lipinski's rule. Finally, molecular docking experiments were conducted with the human topoisomerase α2 (TOP2A) protein to verify and validate the reliability and stability of the docking procedure. The results of the docking scores, which quantify binding affinity, indicated that these derivatives exhibited a stronger affinity for TOP2A.
Collapse
Affiliation(s)
- Verda Çoban
- Graduate Education Institute, Uşak University, Uşak, Turkey
| | - Nevin Çankaya
- Vocational School of Health Services, Uşak University, Uşak, Turkey
| | - Serap Yalçın Azarkan
- Department of Medical Pharmacology, Faculty of Medicine, Kırsehir Ahi Evran University, Kırşehir, Turkey
| |
Collapse
|
60
|
Liu J, Wu W, Hu J, Zhao S, Chang Y, Chen Q, Li Y, Tang J, Zhang Z, Wu X, Jiao S, Xiao H, Zhang Q, Du J, Zhao J, Ye K, Huang M, Xu J, Zhou H, Zheng J, Sun P. Novel benzothiazole derivatives target the Gac/Rsm two-component system as antibacterial synergists against Pseudomonas aeruginosa infections. Acta Pharm Sin B 2024; 14:4934-4961. [PMID: 39664420 PMCID: PMC11628855 DOI: 10.1016/j.apsb.2024.08.002] [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: 03/21/2024] [Revised: 08/01/2024] [Accepted: 08/01/2024] [Indexed: 12/13/2024] Open
Abstract
The management of antibiotic-resistant, bacterial biofilm infections in skin wounds poses an increasingly challenging clinical scenario. Pseudomonas aeruginosa infection is difficult to eradicate because of biofilm formation and antibiotic resistance. In this study, we identified a new benzothiazole derivative compound, SN12 (IC50 = 43.3 nmol/L), demonstrating remarkable biofilm inhibition at nanomolar concentrations in vitro. In further activity assays and mechanistic studies, we formulated an unconventional strategy for combating P. aeruginosa-derived infections by targeting the two-component (Gac/Rsm) system. Furthermore, SN12 slowed the development of ciprofloxacin and tobramycin resistance. By using murine skin wound infection models, we observed that SN12 significantly augmented the antibacterial effects of three widely used antibiotics-tobramycin (100-fold), vancomycin (200-fold), and ciprofloxacin (1000-fold)-compared with single-dose antibiotic treatments for P. aeruginosa infection in vivo. The findings of this study suggest the potential of SN12 as a promising antibacterial synergist, highlighting the effectiveness of targeting the two-component system in treating challenging bacterial biofilm infections in humans.
Collapse
Affiliation(s)
- Jun Liu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, College of Pharmacy, Jinan University, Guangzhou 510632, China
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Wenfu Wu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Jiayi Hu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Siyu Zhao
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Yiqun Chang
- Faculty of Medicine and Health, the University of Sydney, Sydney NSW 2006, Australia
| | - Qiuxian Chen
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Yujie Li
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Jie Tang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Zhenmeng Zhang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Xiao Wu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Shumeng Jiao
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Haichuan Xiao
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Qiang Zhang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Jiarui Du
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Jianfu Zhao
- Department of Oncology, the First Affiliated Hospital of Jinan University Guangzhou, Guangzhou 510632, China
| | - Kaihe Ye
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Meiyan Huang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Jun Xu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Haibo Zhou
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Junxia Zheng
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Pinghua Sun
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, College of Pharmacy, Jinan University, Guangzhou 510632, China
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, School of Pharmacy, Shihezi University, Shihezi 832003, China
- Department of Oncology, the First Affiliated Hospital of Jinan University Guangzhou, Guangzhou 510632, China
| |
Collapse
|
61
|
Yau MQ, Liew CWY, Toh JH, Loo JSE. A head-to-head comparison of MM/PBSA and MM/GBSA in predicting binding affinities for the CB 1 cannabinoid ligands. J Mol Model 2024; 30:390. [PMID: 39480515 DOI: 10.1007/s00894-024-06189-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 10/23/2024] [Indexed: 11/02/2024]
Abstract
CONTEXT The substantial increase in the number of active and inactive-state CB1 receptor experimental structures has provided opportunities for CB1 drug discovery using various structure-based drug design methods, including the popular end-point methods for predicting binding free energies-Molecular Mechanics/Poisson-Boltzmann Surface Area (MM/PBSA) and Molecular Mechanics/Generalized Born Surface Area (MM/GBSA). In this study, we have therefore evaluated the performance of MM/PBSA and MM/GBSA in calculating binding free energies for CB1 receptor. Additionally, with both MM/PBSA and MM/GBSA being known for their highly individualized performance, we have evaluated the effects of various simulation parameters including the use of energy minimized structures, choice of solute dielectric constant, inclusion of entropy, and the effects of the five GB models. Generally, MM/GBSA provided higher correlations than MM/PBSA (rMM/GBSA = 0.433 - 0.652 vs. rMM/PBSA = 0.100 - 0.486) regardless of the simulation parameters, while also offering faster calculations. Improved correlations were observed with the use of molecular dynamics ensembles compared with energy minimized structures and larger solute dielectric constants. Incorporation of entropic terms led to unfavorable results for both MM/PBSA and MM/GBSA for a majority of the dataset, while the evaluation of the various GB models exerted a varying effect on both the datasets. The findings obtained in this study demonstrate the utility of MM/PBSA and MM/GBSA in predicting binding free energies for the CB1 receptor, hence providing a useful benchmark for their applicability in the endocannabinoid system as well as other G protein-coupled receptors. METHODS The study utilized the docked dataset (Induced Fit Docking with Glide XP scoring function) from Loo et al., consisting of 46 ligands-23 agonists and 23 antagonists. The equilibrated structures from Loo et al. were subjected to 30 ns production simulations using GROMACS 2018 at 300 K and 1 atm with the velocity rescaling thermostat and the Parinello-Rahman barostat. AMBER ff99SB*-ILDN was used for the proteins, General Amber Force Field (GAFF) was used for the ligands, and Slipids parameters were used for lipids. MM/PBSA and MM/GBSA binding free energies were then calculated using gmx_MMPBSA. The solute dielectric constant was varied between 1, 2, and 4 to study the effect of different solute dielectric constants on the performance of MM/PB(GB)SA. The effect of entropy on MM/PB(GB)SA binding free energies was evaluated using the interaction entropy module implemented in gmx_MMPBSA. Five GB models, GBHCT, GBOBC1, GBOBC2, GBNeck, and GBNeck2, were evaluated to study the effect of the choice of GB models in the performance of MM/GBSA. Pearson correlation coefficients were used to measure the correlation between experimental and predicted binding free energies.
Collapse
Affiliation(s)
- Mei Qian Yau
- School of Pharmacy, Faculty of Health and Medical Sciences, Taylor's University, No. 1 Jalan Taylors 47500 Subang Jaya, Selangor, Malaysia.
- Digital Health and Medical Advancement Impact Lab, Taylor's University, No. 1 Jalan Taylors, 47500 Subang Jaya, Selangor, Malaysia.
| | - Clarence W Y Liew
- School of Pharmacy, Faculty of Health and Medical Sciences, Taylor's University, No. 1 Jalan Taylors 47500 Subang Jaya, Selangor, Malaysia
| | - Jing Hen Toh
- School of Pharmacy, Faculty of Health and Medical Sciences, Taylor's University, No. 1 Jalan Taylors 47500 Subang Jaya, Selangor, Malaysia
| | - Jason S E Loo
- School of Pharmacy, Faculty of Health and Medical Sciences, Taylor's University, No. 1 Jalan Taylors 47500 Subang Jaya, Selangor, Malaysia
- Digital Health and Medical Advancement Impact Lab, Taylor's University, No. 1 Jalan Taylors, 47500 Subang Jaya, Selangor, Malaysia
| |
Collapse
|
62
|
Zhang J, Zhao H, Zhou Q, Yang X, Qi H, Zhao Y, Yang L. Discovery of Cyclic Peptide Inhibitors Targeted on TNFα-TNFR1 from Computational Design and Bioactivity Verification. Molecules 2024; 29:5147. [PMID: 39519786 PMCID: PMC11547827 DOI: 10.3390/molecules29215147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 10/23/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
Activating tumor necrosis factor receptor 1 (TNFR1) with tumor necrosis factor alpha (TNFα) is one of the key pathological mechanisms resulting in the exacerbation of rheumatoid arthritis (RA) immune response. Despite various types of drugs being available for the treatment of RA, a series of shortcomings still limits their application. Therefore, developing novel peptide drugs that target TNFα-TNFR1 interaction is expected to expand therapeutic drug options. In this study, the detailed interaction mechanism between TNFα and TNFR1 was elucidated, based on which, a series of linear peptides were initially designed. To overcome its large conformational flexibility, two different head-to-tail cyclization strategies were adopted by adding a proline-glycine (GP) or cysteine-cysteine (CC) to form an amide or disulfide bond between the N-C terminal. The results indicate that two cyclic peptides, R1_CC4 and α_CC8, exhibit the strongest binding free energies. α_CC8 was selected for further optimization using virtual mutations through in vitro activity and toxicity experiments due to its optimal biological activity. The L16R mutant was screened, and its binding affinity to TNFR1 was validated using ELISA assays. This study designed a novel cyclic peptide structure with potential anti-inflammatory properties, possibly bringing an additional choice for the treatment of RA in the future.
Collapse
Affiliation(s)
- Jiangnan Zhang
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; (J.Z.); (H.Z.); (Q.Z.); (X.Y.); (H.Q.)
| | - Huijian Zhao
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; (J.Z.); (H.Z.); (Q.Z.); (X.Y.); (H.Q.)
| | - Qianqian Zhou
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; (J.Z.); (H.Z.); (Q.Z.); (X.Y.); (H.Q.)
| | - Xiaoyue Yang
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; (J.Z.); (H.Z.); (Q.Z.); (X.Y.); (H.Q.)
| | - Haoran Qi
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; (J.Z.); (H.Z.); (Q.Z.); (X.Y.); (H.Q.)
| | - Yongxing Zhao
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; (J.Z.); (H.Z.); (Q.Z.); (X.Y.); (H.Q.)
- Henan Key Laboratory of Nanomedicine for Targeting Diagnosis and Treatment, Zhengzhou 450001, China
| | - Longhua Yang
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; (J.Z.); (H.Z.); (Q.Z.); (X.Y.); (H.Q.)
| |
Collapse
|
63
|
Lietz S, Sommer A, Sokolowski LM, Kling C, Rodríguez Alfonso AA, Preising N, Alpízar-Pedraza D, King J, Streit L, Schröppel B, van Erp R, Barth E, Schneider M, Münch J, Michaelis J, Ständker L, Wiese S, Barth H, Pulliainen AT, Scanlon K, Ernst K. Alpha-1 antitrypsin inhibits pertussis toxin. J Biol Chem 2024; 300:107950. [PMID: 39481600 DOI: 10.1016/j.jbc.2024.107950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 10/07/2024] [Accepted: 10/09/2024] [Indexed: 11/02/2024] Open
Abstract
Pertussis (whooping cough) is a vaccine-preventable but re-emerging, highly infectious respiratory disease caused by Bordetella pertussis. There are currently no effective treatments for pertussis, complicating care for nonvaccinated individuals, especially newborns. Disease manifestations are predominantly caused by pertussis toxin (PT), a pivotal virulence factor classified as an ADP-ribosylating AB-type protein toxin. In this work, an unbiased approach using peptide libraries, bioassay-guided fractionation and mass spectrometry revealed α1-antitrypsin (α1AT) as a potent PT inhibitor. Biochemistry-, cell culture-, and molecular modeling-based in vitro experimentation demonstrated that the α1AT mode of action is based on blocking PT-binding to the host target cell surface. In the infant mouse model of severe pertussis, α1AT expression was reduced upon infection. Further, systemic administration of α1AT significantly reduced B. pertussis-induced leukocytosis, which is a hallmark of infant infection and major risk factor for fatal pertussis. Taken together our data demonstrates that α1AT is a novel PT inhibitor and that further evaluation and development of α1AT as a therapeutic agent for pertussis is warranted. Importantly, purified α1AT is already in use clinically as an intravenous augmentation therapy for those with genetic α1AT deficiency and could be repurposed to clinical management of pertussis.
Collapse
Affiliation(s)
- Stefanie Lietz
- Institute of Experimental and Clinical Pharmacology, Toxicology and Pharmacology of Natural Products, Ulm University Medical Center, Ulm, Germany
| | - Anja Sommer
- Institute of Experimental and Clinical Pharmacology, Toxicology and Pharmacology of Natural Products, Ulm University Medical Center, Ulm, Germany
| | - Lena-Marie Sokolowski
- Institute of Experimental and Clinical Pharmacology, Toxicology and Pharmacology of Natural Products, Ulm University Medical Center, Ulm, Germany
| | - Carolin Kling
- Institute of Experimental and Clinical Pharmacology, Toxicology and Pharmacology of Natural Products, Ulm University Medical Center, Ulm, Germany
| | - Armando A Rodríguez Alfonso
- Core Facility Functional Peptidomics, Faculty of Medicine, Ulm University, Ulm, Germany; Core Unit Mass Spectrometry and Proteomics, Faculty of Medicine, Ulm University, Ulm, Germany
| | - Nico Preising
- Core Facility Functional Peptidomics, Faculty of Medicine, Ulm University, Ulm, Germany
| | - Daniel Alpízar-Pedraza
- Biochemistry and Molecular Biology Department, Center for Pharmaceutical Research and Development, Nuevo Vedado, Cuba
| | - Jaylyn King
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Lisa Streit
- Institute of Biophysics, Ulm University, Ulm, Germany
| | - Bernd Schröppel
- Internal Medicine Clinic, Nephrology Section Core Facility, Ulm University Medical Center, Ulm, Germany
| | - Rene van Erp
- Internal Medicine Clinic, Nephrology Section Core Facility, Ulm University Medical Center, Ulm, Germany
| | - Eberhard Barth
- Anesthesiology an Intensive Medicine Clinic, Ulm University Medical Center, Ulm, Germany
| | - Marion Schneider
- Anesthesiology an Intensive Medicine Clinic, Ulm University Medical Center, Ulm, Germany
| | - Jan Münch
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | | | - Ludger Ständker
- Core Facility Functional Peptidomics, Faculty of Medicine, Ulm University, Ulm, Germany
| | - Sebastian Wiese
- Core Unit Mass Spectrometry and Proteomics, Faculty of Medicine, Ulm University, Ulm, Germany
| | - Holger Barth
- Institute of Experimental and Clinical Pharmacology, Toxicology and Pharmacology of Natural Products, Ulm University Medical Center, Ulm, Germany
| | | | - Karen Scanlon
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Katharina Ernst
- Institute of Experimental and Clinical Pharmacology, Toxicology and Pharmacology of Natural Products, Ulm University Medical Center, Ulm, Germany.
| |
Collapse
|
64
|
Zhen J, Zhang C, Huang T, Xie L, Yan Y, Yan S, Zhang J, Huang H, Xie J. Drug repurposing: An antidiabetic drug Ipragliflozin as Mycobacterium tuberculosis sirtuin-like protein inhibitor that synergizes with anti-tuberculosis drug isoniazid. Int J Biol Macromol 2024; 282:137003. [PMID: 39481722 DOI: 10.1016/j.ijbiomac.2024.137003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 10/26/2024] [Accepted: 10/26/2024] [Indexed: 11/02/2024]
Abstract
The surge of drug-resistant Mycobacterium tuberculosis (DR-TB) impedes the World Health Organization's efforts in ending TB and calls for new therapeutic formulations. M. tuberculosis sirtuin-like protein Rv1151c is a bifunctional enzyme with both deacetylation and desuccinylation activities, which plays an important role in M. tuberculosis drug resistance and stress responses. Thus, it appears to be a promising target for the development of new TB therapeutics. In this study, we screened 31,057 ligand compounds from seven compound libraries in silico to identify inhibitors of Rv1151c. Ipragliflozin can bind to Rv1151c and interact stably. Ipragliflozin can change the acylation level of M. tuberculosis by inhibiting Rv1151c and effectively inhibit the growth of M. tuberculosis H37Rv and M. smegmatis. It can potentiate the first-front anti-TB drug isoniazid. As an antidiabetic drug, Ipragliflozin can be potentially included in the regimen to treat diabetes-tuberculosis comorbidity.
Collapse
Affiliation(s)
- Junfeng Zhen
- Institute of Modern Biopharmaceuticals, School of Life Sciences, Southwest University, Chongqing, China
| | - Chao Zhang
- National Clinical Laboratory on Tuberculosis, Beijing Key Laboratory on Drug-Resistant Tuberculosis, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Tingting Huang
- Institute of Modern Biopharmaceuticals, School of Life Sciences, Southwest University, Chongqing, China
| | - Longxiang Xie
- Institute of Biomedical Informatics, Cell Signal Transduction Laboratory, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Huaihe Hospital, Henan University, Kaifeng, China
| | - Yaru Yan
- Institute of Modern Biopharmaceuticals, School of Life Sciences, Southwest University, Chongqing, China
| | - Shuangquan Yan
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Jinghan Zhang
- Institute of Modern Biopharmaceuticals, School of Life Sciences, Southwest University, Chongqing, China
| | - Hairong Huang
- National Clinical Laboratory on Tuberculosis, Beijing Key Laboratory on Drug-Resistant Tuberculosis, Beijing Chest Hospital, Capital Medical University, Beijing, China.
| | - Jianping Xie
- Institute of Modern Biopharmaceuticals, School of Life Sciences, Southwest University, Chongqing, China.
| |
Collapse
|
65
|
Chen X, Xu S, Chu B, Guo J, Zhang H, Sun S, Song L, Feng XQ. Applying Spatiotemporal Modeling of Cell Dynamics to Accelerate Drug Development. ACS NANO 2024; 18:29311-29336. [PMID: 39420743 DOI: 10.1021/acsnano.4c12599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Cells act as physical computational programs that utilize input signals to orchestrate molecule-level protein-protein interactions (PPIs), generating and responding to forces, ultimately shaping all of the physiological and pathophysiological behaviors. Genome editing and molecule drugs targeting PPIs hold great promise for the treatments of diseases. Linking genes and molecular drugs with protein-performed cellular behaviors is a key yet challenging issue due to the wide range of spatial and temporal scales involved. Building predictive spatiotemporal modeling systems that can describe the dynamic behaviors of cells intervened by genome editing and molecular drugs at the intersection of biology, chemistry, physics, and computer science will greatly accelerate pharmaceutical advances. Here, we review the mechanical roles of cytoskeletal proteins in orchestrating cellular behaviors alongside significant advancements in biophysical modeling while also addressing the limitations in these models. Then, by integrating generative artificial intelligence (AI) with spatiotemporal multiscale biophysical modeling, we propose a computational pipeline for developing virtual cells, which can simulate and evaluate the therapeutic effects of drugs and genome editing technologies on various cell dynamic behaviors and could have broad biomedical applications. Such virtual cell modeling systems might revolutionize modern biomedical engineering by moving most of the painstaking wet-laboratory effort to computer simulations, substantially saving time and alleviating the financial burden for pharmaceutical industries.
Collapse
Affiliation(s)
- Xindong Chen
- Institute of Biomechanics and Medical Engineering, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, China
- BioMap, Beijing 100144, China
| | - Shihao Xu
- Institute of Biomechanics and Medical Engineering, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, China
| | - Bizhu Chu
- School of Pharmacy, Shenzhen University, Shenzhen 518055, China
- Medical School, Shenzhen University, Shenzhen 518055, China
| | - Jing Guo
- Department of Medical Oncology, Xiamen Key Laboratory of Antitumor Drug Transformation Research, The First Affiliated Hospital of Xiamen University, Xiamen 361000, China
| | - Huikai Zhang
- Institute of Biomechanics and Medical Engineering, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, China
| | - Shuyi Sun
- Institute of Biomechanics and Medical Engineering, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, China
| | - Le Song
- BioMap, Beijing 100144, China
| | - Xi-Qiao Feng
- Institute of Biomechanics and Medical Engineering, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, China
| |
Collapse
|
66
|
Yang J, Fu B, Gou R, Lin X, Wu H, Xue W. Molecular Mechanism-Driven Discovery of Novel Small Molecule Inhibitors against Drug-Resistant SARS-CoV-2 M pro Variants. J Chem Inf Model 2024; 64:7998-8009. [PMID: 39387184 DOI: 10.1021/acs.jcim.4c01206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Under the selective pressure of nirmatrelvir, a peptidomimetic covalent drug targeting SARS-CoV-2 Mpro, various drug-resistant mutations on Mpro have been acquired in vitro. Among the mutations, L50F and E166V, along with the combination of L50F and E166V, are particularly representative and pose considerable obstacles to the effective treatment of COVID-19. Our previous study identified NMI-001 and NMI-002 as novel nonpeptide inhibitors that target SARS-CoV-2 Mpro, possessing unique scaffolds and binding modes different from those of nirmatrelvir. In view of these findings, we proposed a drug design strategy aimed at rapidly identifying inhibitors that can combat mutation-induced drug resistance. Initially, molecular dynamics (MD) simulation was employed to investigate the binding mechanisms of NMI-001 and NMI-002 against the three drug-resistant mutants (Mpro_L50F, Mpro_E166V, and Mpro_L50F+E166V). Then, we conducted two phases of high-throughput virtual screening. In the first phase, NMI-001 served as a template to perform scaffold hopping-based similarity search in a library of 15,742,661 compounds. In the second phase, 968 compounds exhibiting similarity to NMI-001 were evaluated via molecular docking and MD simulations. Six compounds that may be effective against at least one mutant were identified, and five compounds were procured for conducting in vitro assays. Finally, the compound Z1557501297 (NMI-003) exhibiting inhibitory effects against the E166V (IC50 = 27.81 ± 2.65 μM) and L50F+E166V (IC50 = 8.78 ± 0.74 μM) mutants was discovered. The binding modes referring to NMI-003-Mpro_E166V and NMI-003-Mpro_L50F+E166V were further elucidated at the atomic level. In summary, NMI-003 reported herein is the first compound with activity against E166V and L50F+E166V, which provides a good starting point to design novel antiviral drugs for the treatment of drug-resistant SARS-CoV-2.
Collapse
Affiliation(s)
- Jingyi Yang
- School of Pharmaceutical Sciences, Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Chongqing University, Chongqing 401331, China
| | - Beibei Fu
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Rongpei Gou
- School of Pharmaceutical Sciences, Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Chongqing University, Chongqing 401331, China
| | - Xiaoyuan Lin
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Haibo Wu
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Weiwei Xue
- School of Pharmaceutical Sciences, Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Chongqing University, Chongqing 401331, China
| |
Collapse
|
67
|
Kamel EM, Aba Alkhayl FF, Alqhtani HA, Bin-Jumah M, Lamsabhi AM. Dynamic interactions and inhibitory mechanisms of Artemisia annua terpenoids with carbonic anhydrase IX. Int J Biol Macromol 2024; 282:136982. [PMID: 39471916 DOI: 10.1016/j.ijbiomac.2024.136982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 09/24/2024] [Accepted: 10/25/2024] [Indexed: 11/01/2024]
Abstract
This study evaluates the inhibitory potential of terpenoids isolated from Artemisia annua against carbonic anhydrase IX (CAIX), a crucial enzyme overexpressed in hypoxic tumor environments. Employing a multidisciplinary approach, we utilized in vitro assays, enzyme kinetics, molecular docking, and molecular dynamics (MD) simulations to comprehensively assess the efficacy of these compounds. Among the terpenoids tested, manool emerged as the most potent inhibitor, exhibiting the lowest IC50 value of 160.2 ± 15.2 nM. This was followed by labda-8(17),12-diene-15,16-dial with an IC50 of 297.9 ± 8.84 nM. Enzyme kinetics revealed a mixed inhibition mode for both compounds. Molecular docking aligned well with in vitro data, showing extensive polar and hydrophobic interactions within the CAIX binding site. Further insights were gained through 300 ns MD simulations, which highlighted the dynamic interactions and stability of these complexes. Manool demonstrated the most significant stabilization of CAIX, as evidenced by favorable RMSD, Rg, SASA profiles, and the strongest hydrogen bonding interactions. Additionally, MM/PBSA calculations confirmed manool's superior binding affinity. These findings underscore the therapeutic potential of manool as a potent CAIX inhibitor, providing a foundation for the development of effective anticancer agents targeting hypoxic tumor environments. ADMET analysis revealed favorable pharmacokinetic profiles for the terpenoids, with manool demonstrating high lipophilicity and BBB permeability, though potential CYP-mediated interactions were noted.
Collapse
Affiliation(s)
- Emadeldin M Kamel
- Chemistry Department, Faculty of Science, Beni-Suef University, Beni-Suef 62514, Egypt.
| | - Faris F Aba Alkhayl
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, 51452 Buraydah, Saudi Arabia
| | - Haifa A Alqhtani
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, P.O. BOX 84428, Riyadh 11671, Saudi Arabia
| | - May Bin-Jumah
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, P.O. BOX 84428, Riyadh 11671, Saudi Arabia
| | - Al Mokhtar Lamsabhi
- Departamento de Química, Módulo 13, Universidad Autónoma de Madrid, Campus de Excelencia UAM-CSIC Cantoblanco, 28049 Madrid, Spain; Institute for Advanced Research in Chemical Sciences (IAdChem), Universidad Autónoma de Madrid, 28049 Madrid, Spain.
| |
Collapse
|
68
|
Shionoya K, Park JH, Ekimoto T, Takeuchi JS, Mifune J, Morita T, Ishimoto N, Umezawa H, Yamamoto K, Kobayashi C, Kusunoki A, Nomura N, Iwata S, Muramatsu M, Tame JRH, Ikeguchi M, Park SY, Watashi K. Structural basis for hepatitis B virus restriction by a viral receptor homologue. Nat Commun 2024; 15:9241. [PMID: 39455604 PMCID: PMC11511851 DOI: 10.1038/s41467-024-53533-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
Macaque restricts hepatitis B virus (HBV) infection because its receptor homologue, NTCP (mNTCP), cannot bind preS1 on viral surface. To reveal how mNTCP loses the viral receptor function, we here solve the cryo-electron microscopy structure of mNTCP. Superposing on the human NTCP (hNTCP)-preS1 complex structure shows that Arg158 of mNTCP causes steric clash to prevent preS1 from embedding onto the bile acid tunnel of NTCP. Cell-based mutation analysis confirms that only Gly158 permitted preS1 binding, in contrast to robust bile acid transport among mutations. As the second determinant, Asn86 on the extracellular surface of mNTCP shows less capacity to restrain preS1 from dynamic fluctuation than Lys86 of hNTCP, resulting in unstable preS1 binding. Additionally, presence of long-chain conjugated-bile acids in the tunnel induces steric hindrance with preS1 through their tailed-chain. This study presents structural basis in which multiple sites in mNTCP constitute a molecular barrier to strictly restrict HBV.
Collapse
Affiliation(s)
- Kaho Shionoya
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, Japan
- Graduate School of Science and Technology, Tokyo University of Science, Chiba, Japan
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo, Japan
| | - Jae-Hyun Park
- Drug Design Laboratory, Graduate School of Medical Life Science, Yokohama City University, Kanagawa, Japan
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| | - Toru Ekimoto
- Computational Life Science Laboratory, Graduate School of Medical Life Science, Yokohama City University, Kanagawa, Japan
| | - Junko S Takeuchi
- Center for Clinical Sciences, National Center for Global Health and Medicine, Tokyo, Japan
| | - Junki Mifune
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo, Japan
| | - Takeshi Morita
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo, Japan
| | - Naito Ishimoto
- Drug Design Laboratory, Graduate School of Medical Life Science, Yokohama City University, Kanagawa, Japan
| | - Haruka Umezawa
- Drug Design Laboratory, Graduate School of Medical Life Science, Yokohama City University, Kanagawa, Japan
| | - Kenichiro Yamamoto
- Computational Life Science Laboratory, Graduate School of Medical Life Science, Yokohama City University, Kanagawa, Japan
| | - Chisa Kobayashi
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, Japan
- Graduate School of Science and Technology, Tokyo University of Science, Chiba, Japan
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo, Japan
| | - Atsuto Kusunoki
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo, Japan
| | - Norimichi Nomura
- Department of Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - So Iwata
- Department of Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- RIKEN SPring-8 Center, Hyogo, Japan
| | - Masamichi Muramatsu
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, Japan
- Department of Infectious Disease Research, Foundation for Biomedical Research and Innovation at Kobe, Hyogo, Japan
| | - Jeremy R H Tame
- Drug Design Laboratory, Graduate School of Medical Life Science, Yokohama City University, Kanagawa, Japan
| | - Mitsunori Ikeguchi
- Computational Life Science Laboratory, Graduate School of Medical Life Science, Yokohama City University, Kanagawa, Japan
- HPC- and AI-driven Drug Development Platform Division, Center for Computational Science, RIKEN, Kanagawa, Japan
| | - Sam-Yong Park
- Drug Design Laboratory, Graduate School of Medical Life Science, Yokohama City University, Kanagawa, Japan.
| | - Koichi Watashi
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, Japan.
- Graduate School of Science and Technology, Tokyo University of Science, Chiba, Japan.
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo, Japan.
| |
Collapse
|
69
|
Rapacciuolo P, Finamore C, Giorgio CD, Fiorillo B, Massa C, Urbani G, Marchianò S, Bordoni M, Cassiano C, Morretta E, Spinelli L, Lupia A, Moraca F, Biagioli M, Sepe V, Monti MC, Catalanotti B, Fiorucci S, Zampella A. Design, Synthesis, and Pharmacological Evaluation of Dual FXR-LIFR Modulators for the Treatment of Liver Fibrosis. J Med Chem 2024; 67:18334-18355. [PMID: 39382988 DOI: 10.1021/acs.jmedchem.4c01651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/11/2024]
Abstract
Although multiple approaches have been suggested, treating mild-to-severe fibrosis in the context of metabolic dysfunction associated with liver disease (MASLD) remains a challenging area in drug discovery. Pathogenesis of liver fibrosis is multifactorial, and pathogenic mechanisms are deeply intertwined; thus, it is well accepted that future treatment requires the development of multitarget modulators. Harnessing the 3,4,5-trisubstituted isoxazole scaffold, previously described as a key moiety in Farnesoid X receptor (FXR) agonism, herein we report the discovery of a novel class of hybrid molecules endowed with dual activity toward FXR and the leukemia inhibitory factor receptor (LIFR). Up to 27 new derivatives were designed and synthesized. The pharmacological characterization of this series resulted in the identification of 3a as a potent FXR agonist and LIFR antagonist with excellent ADME properties. In vitro and in vivo characterization identified compound 3a as the first-in-class hybrid LIFR inhibitor and FXR agonist that protects against the development of acute liver fibrosis and inflammation.
Collapse
Affiliation(s)
- Pasquale Rapacciuolo
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano, 49, Naples I-80131, Italy
| | - Claudia Finamore
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano, 49, Naples I-80131, Italy
| | - Cristina Di Giorgio
- Department of Medicine and Surgery, University of Perugia, Piazza L. Severi, 1, Perugia 06132, Italy
| | - Bianca Fiorillo
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano, 49, Naples I-80131, Italy
| | - Carmen Massa
- Department of Medicine and Surgery, University of Perugia, Piazza L. Severi, 1, Perugia 06132, Italy
| | - Ginevra Urbani
- Department of Medicine and Surgery, University of Perugia, Piazza L. Severi, 1, Perugia 06132, Italy
| | - Silvia Marchianò
- Department of Medicine and Surgery, University of Perugia, Piazza L. Severi, 1, Perugia 06132, Italy
| | - Martina Bordoni
- Department of Medicine and Surgery, University of Perugia, Piazza L. Severi, 1, Perugia 06132, Italy
| | - Chiara Cassiano
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano, 49, Naples I-80131, Italy
| | - Elva Morretta
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano, 49, Naples I-80131, Italy
| | - Lucio Spinelli
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano, 49, Naples I-80131, Italy
| | - Antonio Lupia
- Department of Life and Environmental Sciences, University of Cagliari, Via Università, 40, Cagliari 09124, Italy
| | - Federica Moraca
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano, 49, Naples I-80131, Italy
| | - Michele Biagioli
- Department of Medicine and Surgery, University of Perugia, Piazza L. Severi, 1, Perugia 06132, Italy
| | - Valentina Sepe
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano, 49, Naples I-80131, Italy
| | - Maria Chiara Monti
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano, 49, Naples I-80131, Italy
| | - Bruno Catalanotti
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano, 49, Naples I-80131, Italy
| | - Stefano Fiorucci
- Department of Medicine and Surgery, University of Perugia, Piazza L. Severi, 1, Perugia 06132, Italy
| | - Angela Zampella
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano, 49, Naples I-80131, Italy
| |
Collapse
|
70
|
Remtulla R, Das SK, Levin LA. In Silico Modeling of Myelin Oligodendrocyte Glycoprotein Disulfide Bond Reduction by Phosphine-Borane Complexes. Pharmaceuticals (Basel) 2024; 17:1417. [PMID: 39598329 PMCID: PMC11597587 DOI: 10.3390/ph17111417] [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: 09/20/2024] [Revised: 10/11/2024] [Accepted: 10/13/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND Neurodegenerative diseases can cause vision loss by damaging retinal ganglion cells in the optic nerve. Novel phosphine-borane compounds (PBs) can protect these cells from oxidative stress via the reduction of disulfide bonds. However, the specific targets of these compounds are unknown. Proteomic evidence suggests that myelin oligodendrocyte glycoprotein (MOG) is a potential target. MOG is of significant interest due to its role in anti-MOG optic neuritis syndrome. METHODS We used in silico modeling to explore the structural consequences of cleaving the extracellular domain MOG disulfide bond, both in isolation and in complex with anti-MOG antibodies. The potential binding of PBs to this bond was examined using molecular docking. RESULTS Cleaving the disulfide bond of MOG altered the structure of MOG dimers and reduced their energetic favorability by 46.13 kcal/mol. The energy profiles of anti-MOG antibody complexes were less favorable when the disulfide bond of MOG was reduced in the monomeric state by 55.21 kcal/mol, but the reverse was true in the dimeric state. PBs exhibited reducing capabilities with the MOG extracellular disulfide bond, with this best-scoring compound binding with an energy of -28.54 kcal/mol to the MOG monomer and -24.97 kcal/mol to the MOG dimer. CONCLUSIONS These findings suggest that PBs can affect the structure of MOG dimers and the formation of antibody complexes by reducing the MOG disulfide bond. Structural changes in MOG could have implications for neurodegenerative diseases and anti-MOG syndrome.
Collapse
Affiliation(s)
- Raheem Remtulla
- Department of Ophthalmology and Visual Sciences, McGill University, Montreal, QC H3A 0G4, Canada;
| | - Sanjoy Kumar Das
- Division of Experimental Medicine, McGill University, Montreal, QC H3A 0G4, Canada;
- Drug Discovery Core, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Leonard A. Levin
- Department of Ophthalmology and Visual Sciences, McGill University, Montreal, QC H3A 0G4, Canada;
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 0G4, Canada
| |
Collapse
|
71
|
Ancajas CMF, Oyedele AS, Butt CM, Walker AS. Advances, opportunities, and challenges in methods for interrogating the structure activity relationships of natural products. Nat Prod Rep 2024; 41:1543-1578. [PMID: 38912779 PMCID: PMC11484176 DOI: 10.1039/d4np00009a] [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: 02/27/2024] [Indexed: 06/25/2024]
Abstract
Time span in literature: 1985-early 2024Natural products play a key role in drug discovery, both as a direct source of drugs and as a starting point for the development of synthetic compounds. Most natural products are not suitable to be used as drugs without further modification due to insufficient activity or poor pharmacokinetic properties. Choosing what modifications to make requires an understanding of the compound's structure-activity relationships. Use of structure-activity relationships is commonplace and essential in medicinal chemistry campaigns applied to human-designed synthetic compounds. Structure-activity relationships have also been used to improve the properties of natural products, but several challenges still limit these efforts. Here, we review methods for studying the structure-activity relationships of natural products and their limitations. Specifically, we will discuss how synthesis, including total synthesis, late-stage derivatization, chemoenzymatic synthetic pathways, and engineering and genome mining of biosynthetic pathways can be used to produce natural product analogs and discuss the challenges of each of these approaches. Finally, we will discuss computational methods including machine learning methods for analyzing the relationship between biosynthetic genes and product activity, computer aided drug design techniques, and interpretable artificial intelligence approaches towards elucidating structure-activity relationships from models trained to predict bioactivity from chemical structure. Our focus will be on these latter topics as their applications for natural products have not been extensively reviewed. We suggest that these methods are all complementary to each other, and that only collaborative efforts using a combination of these techniques will result in a full understanding of the structure-activity relationships of natural products.
Collapse
Affiliation(s)
| | | | - Caitlin M Butt
- Department of Chemistry, Vanderbilt University, Nashville, TN, USA.
| | - Allison S Walker
- Department of Chemistry, Vanderbilt University, Nashville, TN, USA.
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
72
|
Alruhaimi RS, Kamel EM, Alnasser SM, Alzoghaibi MA, Lamsabhi AM, Mahmoud AM. Mechanistic insights into carbonic anhydrase IX inhibition by coumarins from Calendula officinalis: in vitro and in silico approaches. RSC Adv 2024; 14:33602-33618. [PMID: 39444941 PMCID: PMC11497074 DOI: 10.1039/d4ra05984k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Accepted: 10/14/2024] [Indexed: 10/25/2024] Open
Abstract
Given the critical role of carbonic anhydrase IX (CA IX) in various pathological conditions, there is a significant demand for new inhibitors to enhance patient outcomes and clinical management. In this study, we examined the inhibitory effectiveness of five coumarins derived from Calendula officinalis against CA IX using in vitro assays and computational modeling. Among the coumarins tested, xeroboside and isobaisseoside were identified as the most potent inhibitors. Kinetic studies indicated that xeroboside and isobaisseoside exhibit a mixed inhibition mode. Molecular docking analysis showed that the tested coumarins exhibit binding affinities and extensive polar interactions with CA IX. These coumarins demonstrated significant hydrophobic interactions and occupied the same binding site as acetazolamide (AAZ). Molecular dynamics (MD) indicated that xeroboside and isobaisseoside exhibited consistent trajectories and notable energy stabilization during their interaction with CA IX. MM/PBSA calculations showed that xeroboside displayed the lowest binding free energy (-27.26 ± 2.48 kJ mol-1). Potential Energy Landscape (PEL) analysis revealed distinct and stable conformational states for the CA IX-ligand complexes, with xeroboside exhibiting the most stable and lowest energy configuration. These computational findings are consistent with the experimental results, highlighting the potential efficacy of xeroboside and isobaisseoside as CA IX inhibitors. In conclusion, Calendula officinalis-derived coumarins are promising candidates as effective CA IX inhibitors.
Collapse
Affiliation(s)
- Reem S Alruhaimi
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University Riyadh 11671 Saudi Arabia
| | - Emadeldin M Kamel
- Organic Chemistry Department, Faculty of Science, Beni-Suef University Beni-Suef 62514 Egypt
| | - Sulaiman M Alnasser
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University Qassim 51452 Saudi Arabia
| | - Mohammed A Alzoghaibi
- Physiology Department, College of Medicine, King Saud University Riyadh 11461 Saudi Arabia
| | - Al Mokhtar Lamsabhi
- Departamento de Química, Universidad Autónoma de Madrid, Campus de Excelencia UAM-CSIC Cantoblanco Módulo 13 Madrid 28049 Spain
- Institute for Advanced Research in Chemical Sciences (IAdChem), Universidad Autónoma de Madrid Madrid 28049 Spain
| | - Ayman M Mahmoud
- Department of Life Sciences, Faculty of Science and Engineering, Manchester Metropolitan University Manchester M1 5GD UK
- Molecular Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University Beni-Suef 62514 Egypt
| |
Collapse
|
73
|
Lian YE, Wang M, Ma L, Yi W, Liao S, Gao H, Zhou Z. Identification of Novel PPARγ Partial Agonists Based on Virtual Screening Strategy: In Silico and In Vitro Experimental Validation. Molecules 2024; 29:4881. [PMID: 39459249 PMCID: PMC11509912 DOI: 10.3390/molecules29204881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 10/08/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
Thiazolidinediones (TZDs) including rosiglitazone and pioglitazone function as peroxisome proliferator-activated receptor gamma (PPARγ) full agonists, which have been known as a class to be among the most effective drugs for the treatment of type 2 diabetes mellitus (T2DM). However, side effects of TZDs such as fluid retention and weight gain are associated with their full agonistic activities toward PPARγ induced by the AF-2 helix-involved "locked" mechanism. Thereby, this study aimed to obtain novel PPARγ partial agonists without direct interaction with the AF-2 helix. Through performing virtual screening of the Targetmol L6000 Natural Product Library and utilizing molecular dynamics (MD) simulation, as well as molecular mechanics Poisson-Boltzmann surface area (MM-PBSA) analysis, four compounds including tubuloside b, podophyllotoxone, endomorphin 1 and paliperidone were identified as potential PPARγ partial agonists. An in vitro TR-FRET competitive binding assay showed podophyllotoxone displayed the optimal binding affinity toward PPARγ among the screened compounds, exhibiting IC50 and ki values of 27.43 µM and 9.86 µM, respectively. Further cell-based transcription assays were conducted and demonstrated podophyllotoxone's weak agonistic activity against PPARγ compared to that of the PPARγ full agonist rosiglitazone. These results collectively demonstrated that podophyllotoxone could serve as a PPARγ partial agonist and might provide a novel candidate for the treatment of various diseases such as T2DM.
Collapse
Affiliation(s)
| | | | | | - Wei Yi
- School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | | | - Hui Gao
- School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Zhi Zhou
- School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| |
Collapse
|
74
|
Kumar P, Purohit R. Driving forces and large scale affinity calculations for piperine/γ-cyclodxetrin complexes: Mechanistic insights from umbrella sampling simulation and DFT calculations. Carbohydr Polym 2024; 342:122350. [PMID: 39048216 DOI: 10.1016/j.carbpol.2024.122350] [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: 01/27/2024] [Revised: 05/13/2024] [Accepted: 05/29/2024] [Indexed: 07/27/2024]
Abstract
Piperine (PiP), a bioactive molecule, exhibits numerous health benefits and is frequently employed as a co-delivery agent with various phytomedicines (e.g., curcumin) to enhance their bioavailability. This is attributed to PiP's inhibitory activity against drug-metabolizing proteins, notably CYP3A4. Nevertheless, PiP encounters solubility challenges addressed in this study using cyclodextrins (CDs). Specifically, γ-CD and its derivatives, Hydroxypropyl-γ-CD (HP-γ-CD), and Octakis (6-O-sulfo)-γ-CD (Octakis-S-γ-CD), were employed to form supramolecular complexes with PiP. The conformational space of the complexes was assessed through 1 μs molecular dynamics simulations and umbrella sampling. Additionally, quantum mechanical calculations using wB97X-D dispersion-corrected DFT functional and 6-311 + G(d,p) basis set were conducted on the complexes to examine the thermodynamics and kinetic stability. Results indicated that Octakis-S-γ-CD exhibits superior host capabilities for PiP, with the most favorable complexation energy (-457.05 kJ/mol), followed by HP-γ-CD (-249.16 kJ/mol). Furthermore, two conformations of the Octakis-S-γ-CD/PiP complex were explored to elucidate the optimal binding orientation of PiP within the binding pocket of Octakis-S-γ-CD. Supramolecular chemistry relies significantly on non-covalent interactions. Therefore, our investigation extensively explores the critical atoms involved in these interactions, elucidating the influence of substituted groups on the stability of inclusion complexes. This comprehensive analysis contributes to emphasizing the γ-CD derivatives with improved host capacity.
Collapse
Affiliation(s)
- Pramod Kumar
- Structural Bioinformatics Lab, CSIR-Institute of Himalayan Bioresource Technology (CSIR-IHBT), Palampur, HP 176061, India; Biotechnology division, CSIR-IHBT, Palampur, HP 176061, India; Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Rituraj Purohit
- Structural Bioinformatics Lab, CSIR-Institute of Himalayan Bioresource Technology (CSIR-IHBT), Palampur, HP 176061, India; Biotechnology division, CSIR-IHBT, Palampur, HP 176061, India; Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
75
|
Xu K, Wang Z, Xiang S, Tang R, Deng Q, Ge J, Jiang Z, Yang K, Hou T, Sun H. Characterizing the Cooperative Effect of PROTAC Systems with End-Point Binding Free Energy Calculation. J Chem Inf Model 2024; 64:7666-7678. [PMID: 39361611 DOI: 10.1021/acs.jcim.4c01227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/05/2024]
Abstract
Proteolytic targeting chimeras (PROTACs), as an emerging type of drug, function by proximity-based modalities that narrow the distance between a target protein and the E3 ubiquitin ligase to facilitate the ubiquitination labeling of the target protein for degradation. Although it is evidenced that the cooperativity of the PROTAC ternary interaction is one of the key factors affecting the degradation rate of a target protein, PROTAC design utilizing this indicator is still challenging because of the complicated/flexible interactions in a target-PROTAC-E3 ternary system. Therefore, developing reliable and practicable computational methods is of great interest for PROTAC design. Hence, in this study, we investigate the feasibility of using the end-point binding free energy calculation method, represented by molecular mechanics/Poisson-Boltzmann (generalized-Born) surface area (MM/PB(GB)SA), for characterizing cooperativity (including the stabilization and hook effects) of the PROTAC systems. The result shows that MM/GBSA is a good predictor in characterizing these effects under a relatively long molecular dynamics adjustment (50-100 ns) and low dielectric constant (εin = 1), with the Pearson correlation coefficient (rp) > 0.5 and 0.6 for the stabilization and hook effect, respectively. This study provides a feasible strategy for characterizing the cooperativity of the PROTAC systems, facilitating the rational design of PROTAC molecules.
Collapse
Affiliation(s)
- Kexin Xu
- Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009 Jiangsu, P. R. China
| | - Zhe Wang
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058 Zhejiang, P. R. China
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, P. R. China
| | - Sutong Xiang
- Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009 Jiangsu, P. R. China
| | - Rongfan Tang
- Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009 Jiangsu, P. R. China
| | - Qirui Deng
- Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009 Jiangsu, P. R. China
| | - Jingxuan Ge
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058 Zhejiang, P. R. China
| | - Zhiliang Jiang
- Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009 Jiangsu, P. R. China
| | - Kaimo Yang
- Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009 Jiangsu, P. R. China
| | - Tingjun Hou
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058 Zhejiang, P. R. China
| | - Huiyong Sun
- Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009 Jiangsu, P. R. China
| |
Collapse
|
76
|
Qian R, Xue J, Xu Y, Huang J. Alchemical Transformations and Beyond: Recent Advances and Real-World Applications of Free Energy Calculations in Drug Discovery. J Chem Inf Model 2024; 64:7214-7237. [PMID: 39360948 DOI: 10.1021/acs.jcim.4c01024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Computational methods constitute efficient strategies for screening and optimizing potential drug molecules. A critical factor in this process is the binding affinity between candidate molecules and targets, quantified as binding free energy. Among various estimation methods, alchemical transformation methods stand out for their theoretical rigor. Despite challenges in force field accuracy and sampling efficiency, advancements in algorithms, software, and hardware have increased the application of free energy perturbation (FEP) calculations in the pharmaceutical industry. Here, we review the practical applications of FEP in drug discovery projects since 2018, covering both ligand-centric and residue-centric transformations. We show that relative binding free energy calculations have steadily achieved chemical accuracy in real-world applications. In addition, we discuss alternative physics-based simulation methods and the incorporation of deep learning into free energy calculations.
Collapse
Affiliation(s)
- Runtong Qian
- Westlake AI Therapeutics Lab, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang 310024, China
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, 18 Shilongshan Road, Hangzhou, Zhejiang 310024, China
| | - Jing Xue
- Westlake AI Therapeutics Lab, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang 310024, China
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, 18 Shilongshan Road, Hangzhou, Zhejiang 310024, China
| | - You Xu
- Westlake AI Therapeutics Lab, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang 310024, China
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, 18 Shilongshan Road, Hangzhou, Zhejiang 310024, China
| | - Jing Huang
- Westlake AI Therapeutics Lab, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang 310024, China
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, 18 Shilongshan Road, Hangzhou, Zhejiang 310024, China
| |
Collapse
|
77
|
Shi T, Chang W, Wei X, Kong Y, Wei Y. Based on Molecular Docking, Molecular Dynamics Simulation and MM/PB(GB)SA to Study Potential Inhibitors of PRRSV-Nsp4. Proteins 2024. [PMID: 39392104 DOI: 10.1002/prot.26754] [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: 06/11/2024] [Revised: 09/02/2024] [Accepted: 09/26/2024] [Indexed: 10/12/2024]
Abstract
Porcine reproductive and respiratory syndrome (PRRS) is one of the most serious infectious immunosuppressive diseases in the world. The nonstructural protein Nsp4 can be used as an ideal target for anti-PRRSV replication inhibitors. However, little is known about potential inhibitors that target Nsp4 to affect PRRSV replication. The purpose of this study was to screen potential natural inhibitors that affect PRRSV replication by inhibiting Nsp4. Five compounds with strong binding affinity to Nsp4 were selected by structure-based molecular docking method. The complexes of naringin dihydrochalcone (NDC), agathisflavone (AGT), and amentoflavone (AMF) with Nsp4 were stable throughout the molecular dynamics simulation. According to MM/PBSA analysis, the free energies of binding of NDC, AGT, and AMF to Nsp4 were less than-30 Kcal/mol. In conclusion, these three compounds are worthy of further investigation as novel inhibitors of PRRSV. This study provides a theoretical basis for the development of anti-PRRSV natural drugs.
Collapse
Affiliation(s)
- Tianyu Shi
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, People's Republic of China
| | - Wenzhou Chang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, People's Republic of China
| | - Xinyu Wei
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, People's Republic of China
| | - Yiling Kong
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, People's Republic of China
| | - Ying Wei
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, People's Republic of China
- Laboratory of Functional Microbiology and Animal Health, Henan University of Science and Technology, Luoyang, People's Republic of China
| |
Collapse
|
78
|
Liao J, Sergeeva AP, Harder ED, Wang L, Sampson JM, Honig B, Friesner RA. A Method for Treating Significant Conformational Changes in Alchemical Free Energy Simulations of Protein-Ligand Binding. J Chem Theory Comput 2024; 20:8609-8623. [PMID: 39331379 PMCID: PMC11513859 DOI: 10.1021/acs.jctc.4c00954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/28/2024]
Abstract
Relative binding free energy (RBFE) simulation is a rigorous approach to the calculation of quantitatively accurate binding free energy values for protein-ligand binding in which a reference binder is gradually converted to a target binder through alchemical transformation during the simulation. The success of such simulations relies on being able to accurately sample the correct conformational phase space for each alchemical state; however, this becomes a challenge when a significant conformation change occurs between the reference and target binder-receptor complexes. Increasing the simulation time and using enhanced sampling methods can be helpful, but effects can be limited, especially when the free energy barrier between conformations is high or when the correct target complex conformation is difficult to find and maintain. Current RBFE protocols seed the reference complex structure into every alchemical window of the simulation. In our study, we describe an improved protocol in which the reference structure is seeded into the first half of the alchemical windows, and the target structure is seeded into the second half of the alchemical windows. By applying information about the relevant correct end point conformations to different simulation windows from the beginning, the need for large barrier crossings or simulation prediction of the correct structures during an alchemical simulation is in many cases obviated. In the diverse cases we examine below, the simulations yielded free energy predictions that are satisfactory as compared to experiment and superior to running the simulations utilizing the conventional protocol. The method is straightforward to implement for publicly available FEP workflows.
Collapse
Affiliation(s)
- Junzhuo Liao
- Department of Chemistry, Columbia University, New York, NY 10027, USA
| | - Alina P. Sergeeva
- Department of Systems Biology, Columbia University Medical Center, New York, NY 10032, USA
| | - Edward D. Harder
- Life Sciences Software, Schrödinger, Inc., New York, NY 10036, USA
| | - Lingle Wang
- Life Sciences Software, Schrödinger, Inc., New York, NY 10036, USA
| | - Jared M. Sampson
- Life Sciences Software, Schrödinger, Inc., New York, NY 10036, USA
| | - Barry Honig
- Department of Systems Biology, Columbia University Medical Center, New York, NY 10032, USA
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY 10032, USA
- Department of Medicine, Columbia University, New York, NY 10032
- Zuckerman Mind Brain and Behavior Institute, Columbia University, New York, NY 10027, USA
| | | |
Collapse
|
79
|
Irfan A, Zahoor AF, Boulaamane Y, Javed S, Hameed H, Maurady A, Muhammed MT, Ahmad S, Al-Mutairi AA, Shahzadi I, Al-Hussain SA, Zaki MEA. Computational exploration of acefylline derivatives as MAO-B inhibitors for Parkinson's disease: insights from molecular docking, DFT, ADMET, and molecular dynamics approaches. Front Chem 2024; 12:1449165. [PMID: 39439933 PMCID: PMC11493617 DOI: 10.3389/fchem.2024.1449165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 09/04/2024] [Indexed: 10/25/2024] Open
Abstract
Monoamine oxidase B (MAO-B) plays a pivotal role in the deamination process of monoamines, encompassing crucial neurotransmitters like dopamine and norepinephrine. The heightened interest in MAO-B inhibitors emerged after the revelation that this enzyme could potentially catalyze the formation of neurotoxic compounds from endogenous and exogenous sources. Computational screening methodologies serve as valuable tools in the quest for novel inhibitors, enhancing the efficiency of this pursuit. In this study, 43 acefylline derivatives were docked against the MAO-B enzyme for their chemotherapeutic potential and binding affinities that yielded GOLD fitness scores ranging from 33.21 to 75.22. Among them, five acefylline derivatives, namely, MAO-B14, MAO-B15, MAO-B16, MAO-B20, and MAO-B21, displayed binding affinities comparable to the both standards istradefylline and safinamide. These derivatives exhibited hydrogen-bonding interactions with key amino acids Phe167 and Ile197/198, suggesting their strong potential as MAO-B inhibitors. Finally, molecular dynamics (MD) simulations were conducted to evaluate the stability of the examined acefylline derivatives over time. The simulations demonstrated that among the examined acefylline derivatives and standards, MAO-B21 stands out as the most stable candidate. Density functional theory (DFT) studies were also performed to optimize the geometries of the ligands, and molecular docking was conducted to predict the orientations of the ligands within the binding cavity of the protein and evaluate their molecular interactions. These results were also validated by simulation-based binding free energies via the molecular mechanics energies combined with generalized Born and surface area solvation (MM-GBSA) method. However, it is necessary to conduct in vitro and in vivo experiments to confirm and validate these findings in future studies.
Collapse
Affiliation(s)
- Ali Irfan
- Department of Chemistry, Government College University Faisalabad, Faisalabad, Pakistan
| | - Ameer Fawad Zahoor
- Department of Chemistry, Government College University Faisalabad, Faisalabad, Pakistan
| | - Yassir Boulaamane
- Laboratory of Innovative Technologies, National School of Applied Sciences of Tangier, Abdelmalek Essaadi University, Tetouan, Morocco
| | - Sadia Javed
- Department of Biochemistry, Government College University Faisalabad, Faisalabad, Pakistan
| | - Huma Hameed
- Faculty of Pharmaceutical Sciences, University of Central Punjab, Lahore, Pakistan
| | - Amal Maurady
- Laboratory of Innovative Technologies, National School of Applied Sciences of Tangier, Abdelmalek Essaadi University, Tetouan, Morocco
| | - Muhammed Tilahun Muhammed
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Suleyman Demirel University, Isparta, Türkiye
| | - Sajjad Ahmad
- Department of Health and Biological Sciences, Abasyn University Peshawar, Peshawar, Pakistan
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, Lebanon
- Department of Natural Sciences, Lebanese American University, Beirut, Lebanon
| | - Aamal A. Al-Mutairi
- Department of Chemistry, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Irum Shahzadi
- Department of Chemistry, Government College University Faisalabad, Faisalabad, Pakistan
| | - Sami A. Al-Hussain
- Department of Chemistry, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Magdi E. A. Zaki
- Department of Chemistry, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| |
Collapse
|
80
|
Pawnikar S, Magenheimer BS, Joshi K, Nevarez-Munoz E, Haldane A, Maser RL, Miao Y. Activation of polycystin-1 signaling by binding of stalk-derived peptide agonists. eLife 2024; 13:RP95992. [PMID: 39373641 PMCID: PMC11458180 DOI: 10.7554/elife.95992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/08/2024] Open
Abstract
Polycystin-1 (PC1) is the protein product of the PKD1 gene whose mutation causes autosomal dominant Polycystic Kidney Disease (ADPKD). PC1 is an atypical G protein-coupled receptor (GPCR) with an autocatalytic GAIN domain that cleaves PC1 into extracellular N-terminal and membrane-embedded C-terminal (CTF) fragments. Recently, activation of PC1 CTF signaling was shown to be regulated by a stalk tethered agonist (TA), resembling the mechanism observed for adhesion GPCRs. Here, synthetic peptides of the first 9- (p9), 17- (p17), and 21-residues (p21) of the PC1 stalk TA were shown to re-activate signaling by a stalkless CTF mutant in human cell culture assays. Novel Peptide Gaussian accelerated molecular dynamics (Pep-GaMD) simulations elucidated binding conformations of p9, p17, and p21 and revealed multiple specific binding regions to the stalkless CTF. Peptide agonists binding to the TOP domain of PC1 induced close TOP-putative pore loop interactions, a characteristic feature of stalk TA-mediated PC1 CTF activation. Additional sequence coevolution analyses showed the peptide binding regions were consistent with covarying residue pairs identified between the TOP domain and the stalk TA. These insights into the structural dynamic mechanism of PC1 activation by TA peptide agonists provide an in-depth understanding that will facilitate the development of therapeutics targeting PC1 for ADPKD treatment.
Collapse
Affiliation(s)
- Shristi Pawnikar
- Center for Computational Biology and Department of Molecular Biosciences, University of KansasLawrenceUnited States
| | - Brenda S Magenheimer
- Clinical Laboratory Sciences, University of Kansas Medical CenterKansas CityUnited States
- The Jared Grantham Kidney Institute, University of Kansas Medical CenterKansas CityUnited States
| | - Keya Joshi
- Department of Pharmacology and Computational Medicine Program, University of North CarolinaChapel HillUnited States
| | - Ericka Nevarez-Munoz
- Clinical Laboratory Sciences, University of Kansas Medical CenterKansas CityUnited States
| | - Allan Haldane
- Department of Physics, and Center for Biophysics and Computational Biology, Temple UniversityPhiladelphiaUnited States
| | - Robin L Maser
- Clinical Laboratory Sciences, University of Kansas Medical CenterKansas CityUnited States
- The Jared Grantham Kidney Institute, University of Kansas Medical CenterKansas CityUnited States
- Department of Biochemistry and Molecular Biology, University of Kansas Medical CenterKansas CityUnited States
| | - Yinglong Miao
- Department of Pharmacology and Computational Medicine Program, University of North CarolinaChapel HillUnited States
| |
Collapse
|
81
|
Boopathi S, Garduño-Juárez R. A Small Molecule Impedes the Aβ 1-42 Tetramer Neurotoxicity by Preserving Membrane Integrity: Microsecond Multiscale Simulations. ACS Chem Neurosci 2024; 15:3496-3512. [PMID: 39292558 DOI: 10.1021/acschemneuro.4c00383] [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: 09/20/2024] Open
Abstract
Amyloid-β (Aβ1-42) peptides aggregated into plaques deposited in the brain are the main hallmark of Alzheimer's disease (AD), a social and economic burden worldwide. In this context, insoluble Aβ1-42 fibrils are the main components of plaques. The recent trials that used approved AD drugs show that they can remove the fibrils from AD patients' brains, but they did not halt the course of the disease. Mounting evidence envisages that the soluble Aβ1-42 oligomers' interactions with the neuronal membrane trigger higher cell death than Aβ1-42 fibril interactions. Developing a compound that can alleviate the oligomer's toxicity is one of the most demanding tasks for curing the disease. We performed two molecular dynamics (MD) simulations in an explicit solvent model. In the first case, 55-μs of multiscale all-atom (AA)/coarse-grained (CG) MD simulations were carried out to decipher the impact of a previously described small anti-Aβ molecule, termed M30 (2-octahydroisoquinolin-2(1H)-ylethanamine), on an Aβ1-42 tetramer structure in close contact with a DMPC bilayer. In the second case, 15-μs AA/CG MD simulations were performed to rationalize the dynamics between Aβ1-42 and Aβ1-42-M30 tetramer complexes embedded in DMPC. On the membrane bilayer, we found that the Aβ1-42 tetramer penetrates the bilayer surface due to unrestricted conformational flexibility and many contacts with the membrane phosphate groups. In contrast, no Aβ1-42-M30 tetramer penetration was observed during the entire course of the simulation. In the case of the membrane-embedded Aβ1-42 tetramer, the integrity of the bottom bilayer leaflet was severely affected by the interactions between the negatively charged phosphate groups and the positively charged residues of the Aβ1-42 tetramer, resulting in a deep tetramer penetration into the bilayer hydrophobic region. These contacts were not observed in the case of the membrane-embedded Aβ1-42-M30 tetramer. It was noted that M30 molecules bind to Aβ1-42 tetramer through hydrogen bonds, resulting in a conformational stable Aβ1-42-M30 complex. The associated complex has reduced conformational changes and an enhanced rigidity that prevents the tetramer dissociation by interfering with the tetramer-membrane contacts. Our findings suggest that the M30 molecules could bind to Aβ1-42 tetramer resulting in a rigid structure, and that such complexes do not significantly perturb the membrane bilayer organization. These observations support the in vitro and in vivo experimental evidence that the M30 molecules prevent synaptotocity, improving AD-affected mice memory.
Collapse
Affiliation(s)
- Subramanian Boopathi
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai 600036, India
- Instituto de Ciencias Físicas, Universidad Nacional Autónoma de México, Cuernavaca 62210, México
| | - Ramón Garduño-Juárez
- Instituto de Ciencias Físicas, Universidad Nacional Autónoma de México, Cuernavaca 62210, México
| |
Collapse
|
82
|
Kamal IM, Das A, Datta S, Paul K, Chakrabarti S, Chakrabarti S, Bhattacharjee P. A Novel spice-antioxidant-based nano-vehicle as a putative green alternative of synthetic AChE inhibitor drugs. J Biomol Struct Dyn 2024; 42:8813-8830. [PMID: 37639732 DOI: 10.1080/07391102.2023.2248274] [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: 05/24/2023] [Accepted: 08/08/2023] [Indexed: 08/31/2023]
Abstract
The present treatment for Alzheimer's disease (AD) involves well known synthetic acetylcholine esterase (AChE) inhibitor drugs which besides having short duration of action also have deleterious impact on human health. Therefore, there is a need for natural plant-based biomolecule(s) with potential AChE inhibition activity (ies). The aim of the work is to design a spice-based nano-vehicle as a novel green alternative of synthetic AD drugs by nanoencapsulating a solvent-less supercritical CO2 extract of small cardamom seeds (SCE) having a synergistic consortium of five antioxidant molecules, using polyethylene glycol and emulsifiers, selected based on Absorption, Distribution, Metabolism, Excretion, and Toxicity (ADMET) analyses. Ellman's assay and enzyme inhibition kinetics of the antioxidant molecules as well as the extract and its nanoliposomal formulation (SCE-NL) were performed, followed by rigorous molecular docking and dynamics studies using MM-PBSA and umbrella sampling. The antioxidants exhibited significant AChE inhibition in vitro, individually with 1, 8-cineole having the least IC50 value of 65.53 ± 0.05 µg/mL. . Although SCE-NL had higher IC50 value (575.67 ± 0.5 µg/mL) vis-à-vis that of rivastigmine (67.52 ± 0.02 µg/mL), it is safer for usage being 'green'.The Lineweaver-Burk plots (Vmax ∼1.04 mM/min) revealed competitive mode(s) of inhibition of AChE with each of these antioxidants. Binding energy analyses suggested very good binding free energies and stable docking/binding complexes (between the antioxidants and AChE). This study has delivered a nanoliposomal vehicle of food antioxidants as a putative 'green' alternative of synthetic AChE inhibitor drugs.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Izaz Monir Kamal
- Division of Structural Biology and Bioinformatics, CSIR-Indian Institute of Chemical Biology, TRUE Campus, Kolkata, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Anamika Das
- Department of Food Technology and Biochemical Engineering, Jadavpur University, Kolkata, India
| | - Sanjukta Datta
- Department of Food Technology and Biochemical Engineering, Jadavpur University, Kolkata, India
| | - Kaninika Paul
- Department of Food Technology and Biochemical Engineering, Jadavpur University, Kolkata, India
| | - Sasanka Chakrabarti
- Department of Biochemistry and Central Research Cell, M M Institute of Medical Sciences & Research, M M (Deemed to be) University, Ambala, India
| | - Saikat Chakrabarti
- Division of Structural Biology and Bioinformatics, CSIR-Indian Institute of Chemical Biology, TRUE Campus, Kolkata, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Paramita Bhattacharjee
- Department of Food Technology and Biochemical Engineering, Jadavpur University, Kolkata, India
| |
Collapse
|
83
|
Saha S, Ghosh M. Computational exploration of natural compounds targeting Staphylococcus aureus: inhibiting AgrA promoter binding for antimicrobial intervention. J Biomol Struct Dyn 2024; 42:8256-8267. [PMID: 37578046 DOI: 10.1080/07391102.2023.2246566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 07/30/2023] [Indexed: 08/15/2023]
Abstract
Staphylococcus aureus is a highly virulent nosocomial pathogen that poses a significant threat to individuals exposed to healthcare settings. Due to its sophisticated machinery for producing virulence factors, S. aureus can cause severe and potentially fatal infections in humans. This study focuses on the response regulator AgrA, which plays a crucial role in regulating the production of virulence factors in S. aureus. The objective is to identify natural compounds that can inhibit the binding of AgrA to its promoter site, thus inhibiting the expression of virulence genes. To achieve this, a pharmacophore model was generated using known drugs and applied to screen the ZINC natural product database. The resulting compounds were subjected to molecular docking-based virtual screening against the C-terminal DNA binding domain of AgrA. Three compounds, namely ZINC000077269178, ZINC000051012304, and ZINC000004266026, were shortlisted based on their strong affinity for key residues involved in DNA binding and transcription initiation. Subsequently, the unbound and ligand-bound complexes were subjected to a 200 ns molecular dynamics simulation to assess their conformational stability. Various analyses, including RMSD, RMSF, Rg, SASA, Principal Component Analysis, and Gibbs free energy landscape, were conducted on the simulation trajectory. The RMSD profile indicated similar fluctuations in both bound and unbound structures, while the Rg profile demonstrated the compactness of the protein without any unfolding during the simulation. Furthermore, Principal component analysis revealed that ligand binding reduced the overall atomic motion of the protein whereas free energy landscape suggested the energy variations obtained in complexes.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Subhadip Saha
- Department of Biotechnology, National Institute of Technology Durgapur, Durgapur, India
| | - Monidipa Ghosh
- Department of Biotechnology, National Institute of Technology Durgapur, Durgapur, India
| |
Collapse
|
84
|
Puri S, Kirad S, Muzaffar-Ur-Rehman M, Mandal SK, Sharma PK, Sankaranarayanan M, Deepa PR. Lipogenic stearoyl-CoA desaturase-1 (SCD1) targeted virtual screening for chemical inhibitors: molecular docking / dynamics simulation and in vitro assessment of anti-NAFLD efficacy. RSC Adv 2024; 14:31797-31808. [PMID: 39380655 PMCID: PMC11459445 DOI: 10.1039/d4ra06037g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 10/01/2024] [Indexed: 10/10/2024] Open
Abstract
Amidst rising global prevalence of metabolic syndrome, the associated risk of non-alcoholic fatty liver disease (NAFLD) is also rapidly increasing. The pathogenesis of NAFLD starts with fat accumulation and progresses through inflammation and fibrotic sequel, often involving complex molecular mechanisms involving de novo lipogenesis. Stearoyl-CoA desaturase 1 (SCD1) enzyme, expressed in liver and adipose tissue, converts saturated fatty acids to monounsaturated fatty acids (MUFAs), contributing to triglyceride and cholesterol ester formation. In this study, potential SCD1 inhibitors were screened using the ZINC database of curated medically-approved drugs by virtual screening, molecular docking, and molecular dynamics simulations. The top-scoring five ligands with strong binding affinity against SCD1 were ZINC000003831151 > ZINC000001540998 > ZINC000003830713 > ZINC000000897251 > ZINC000002005305, which showed stable protein-ligand complexation and favorable pharmacokinetic attributes. The top ligand, Montelukast, was experimentally validated for its pharmacological efficacy in an in vitro cell culture model of steatosis (NAFLD). Montelukast showed a dose-dependent decrease in hepatic fat accumulation, reduced levels of free radicals, and lowered oxidative stress (P < 0.05). These outcomes suggest Montelukast to be a potential SCD1 inhibitor, with anti-NAFLD efficacy. These findings open new avenues for therapeutic development of the top 5 ligands in metabolic disorders involving SCD1.
Collapse
Affiliation(s)
- Sonakshi Puri
- Biochemistry and Enzyme Biotechnology Lab, Department of Biological Sciences, Birla Institute of Technology and Science Pilani, Pilani Campus Pilani-333031 Rajasthan India +91 1596 255881
| | - Shivani Kirad
- Medicinal Chemistry Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus Pilani-333031 Rajasthan India +91-1596-255717
| | - Mohammed Muzaffar-Ur-Rehman
- Medicinal Chemistry Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus Pilani-333031 Rajasthan India +91-1596-255717
| | - Sumit Kumar Mandal
- Biochemistry and Enzyme Biotechnology Lab, Department of Biological Sciences, Birla Institute of Technology and Science Pilani, Pilani Campus Pilani-333031 Rajasthan India +91 1596 255881
| | - Pankaj Kumar Sharma
- Biochemistry and Enzyme Biotechnology Lab, Department of Biological Sciences, Birla Institute of Technology and Science Pilani, Pilani Campus Pilani-333031 Rajasthan India +91 1596 255881
| | - Murugesan Sankaranarayanan
- Medicinal Chemistry Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus Pilani-333031 Rajasthan India +91-1596-255717
| | - P R Deepa
- Biochemistry and Enzyme Biotechnology Lab, Department of Biological Sciences, Birla Institute of Technology and Science Pilani, Pilani Campus Pilani-333031 Rajasthan India +91 1596 255881
| |
Collapse
|
85
|
Phan TV, Nguyen VTV, Le MT, Nguyen BGD, Vu TT, Thai KM. Identification of efflux pump inhibitors for Pseudomonas aeruginosa MexAB-OprM via ligand-based pharmacophores, 2D-QSAR, molecular docking, and molecular dynamics approaches. Mol Divers 2024; 28:3295-3311. [PMID: 37919619 DOI: 10.1007/s11030-023-10758-9] [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: 10/04/2023] [Accepted: 10/24/2023] [Indexed: 11/04/2023]
Abstract
Efflux pumps have been reported as one of the significant mechanisms by which bacteria evade the effects of multiple antibiotics. The tripartite efflux pump MexAB-OprM in Pseudomonas aeruginosa is one of the most significant multidrug efflux systems due to its broad resistance to antibiotics such as chloramphenicol, fluoroquinolones, lipophilic β-lactam antibiotics, nalidixic acid, novobiocin, rifampicin, and tetracycline. A promising strategy to overcome this resistance mechanism is to combine antibiotics with efflux pump inhibitors (EPIs), which can increase their intracellular concentration to enhance their biological activities. Based on 143 EPIs with chemically diverse skeletons, the 3D pharmacophore and 2D-QSAR modelings were developed and used for the virtual screening on 9.2 million compounds including ZINC15, DrugBank, and Traditional Chinese Medicine databases to identify new EPIs. The molecular docking was also performed to evaluate the binding affinity of potential EPIs to the distal-binding pocket of MexB and resulted in 611 potential EPIs. The structure-activity relationship analyses suggested that nitrogen heterocyclic compounds, piperazine and pyridine scaffolds, and amide derivatives are the most favorable chemically features for MexAB inhibitory activities. The results from molecular dynamics analysis in 100 ns indicated that ZINC009296881 and ZINC009200074 were the most potential MexB inhibitors with strong binding affinity to the distal pocket and MM/GBSA ∆Gbind values of - 38.97 and - 30.19 kcal mol-1, respectively. The predicted pharmacokinetic properties and toxicity of these compounds indicated their potential oral drugs.
Collapse
Affiliation(s)
- Thien-Vy Phan
- Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, 700000, Vietnam
- Faculty of Pharmacy, Nguyen Tat Thanh University, Ho Chi Minh City, 700000, Vietnam
| | - Vu-Thuy-Vy Nguyen
- Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, 700000, Vietnam
- Faculty of Pharmacy, Nguyen Tat Thanh University, Ho Chi Minh City, 700000, Vietnam
| | - Minh-Tri Le
- Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, 700000, Vietnam
- School of Medicine, Vietnam National University Ho Chi Minh City, Linh Trung Ward., Thu Duc Dist, Ho Chi Minh City, 700000, Vietnam
| | | | - Thanh-Thao Vu
- Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, 700000, Vietnam
| | - Khac-Minh Thai
- Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, 700000, Vietnam.
| |
Collapse
|
86
|
Sharma A, Chandrashekar CR, Krishna S, Sowdhamini R. Computational Analysis of the Accumulation of Mutations in Therapeutically Important RNA Viral Proteins During Pandemics with Special Emphasis on SARS-CoV-2. J Mol Biol 2024; 436:168716. [PMID: 39047897 DOI: 10.1016/j.jmb.2024.168716] [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: 04/07/2024] [Revised: 07/06/2024] [Accepted: 07/18/2024] [Indexed: 07/27/2024]
Abstract
Single stranded RNA viruses are primary causative agents for pandemics, causing extensive morbidity and mortality worldwide. A pivotal question in pandemic preparedness and therapeutic intervention is what are the specific mutations which are more likely to emerge during such global health crises? This study aims to identify markers for mutations with the highest probability of emergence in these pandemics, focusing on the SARS-CoV-2 spike protein, an essential and therapeutically significant viral protein, starting from sequence information from the onset of the pandemic until July 2022. Quite consistently, we observed that emerged mutations tended to demonstrate a high genetic score, which reflects high similarity of the type of codon required for translation between an amino acid and to the mutated one. Further, this pattern is also observed in therapeutically significant proteins of other ssRNA pandemic viruses, including influenza (HA, NA), spike proteins of Ebola, envelope of Dengue and Chikungunya. We propose that the genetic score serves as an initial indicator, preceding the actual impact of the mutation on viral fitness. Finally, we developed a comprehensive computational pipeline to further explore and predict the subsequent effects of mutations on viral fitness. We believe that our pipeline can narrow down and predict future mutations in therapeutically important viral proteins during a pandemic.
Collapse
Affiliation(s)
- Abhishek Sharma
- National Centre for Biological Science, GKVK Campus, Bengaluru 560065, India
| | - C R Chandrashekar
- National Centre for Biological Science, GKVK Campus, Bengaluru 560065, India
| | - Sudhir Krishna
- National Centre for Biological Science, GKVK Campus, Bengaluru 560065, India
| | - Ramanathan Sowdhamini
- Molecular Biophysics Unit, Indian Institute of Science, Banagalore 560012, India; Institute of Bioinformatics and Applied Biotechnology, Bangalore, 560100, India.
| |
Collapse
|
87
|
Singothu S, Devsani N, Jahidha Begum P, Maddi D, Bhandari V. Molecular docking and molecular dynamics studies of natural products unravel potential inhibitors against OmpA of Acinetobacter baumannii. J Biomol Struct Dyn 2024; 42:9064-9075. [PMID: 37646649 DOI: 10.1080/07391102.2023.2250446] [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: 05/02/2023] [Accepted: 08/12/2023] [Indexed: 09/01/2023]
Abstract
Emerging antimicrobial resistance has highlighted the need to design more effective antibiotics to treat deadly bacterial infections. Acinetobacter baumannii's outer membrane protein A (OmpA) is a critical virulence component involved in biofilm formation, immunomodulation, and antibiotic resistance, which characterizes it as a potential therapeutic target. The present study aimed to screen the natural product database (>1,00,000) to identify the potential inhibitor against OmpA. Molecular docking studies revealed that 10 compounds had good docking scores (≤ -7 kcal/mol) compared to the reported inhibitor epiestriol (-3.079). Further, these 10 compounds were subjected to ADME analysis and MMGBSA analysis. Based on MMGBSA results, we selected 5 compounds [NP-1 (MolPort-039-337-117), NP-5(MolPort-019-932-973), NP-6 (MolPort-005-948-336), NP-8(MolPort-042-673-978) and NP-9(MolPort-042-673-766)] with high binding affinity. Molecular dynamics simulation found that NP-5, NP-8, and NP-9 were stable after analysing their RMSD, RMSF, the radius of gyration, and hydrogen interactions of complexes. Our study revealed that NP-5, NP-8, and NP-9 bind perfectly with OmpA and can act as its potential inhibitors. The results of this study imply that the identified inhibitors have the potential for further investigation.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Siva Singothu
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, Hyderabad, India
| | - Namrata Devsani
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, Hyderabad, India
| | - Pathan Jahidha Begum
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, Hyderabad, India
| | - Dhanashri Maddi
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, Hyderabad, India
| | - Vasundhra Bhandari
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, Hyderabad, India
| |
Collapse
|
88
|
Shahpouri P, Mehralitabar H, Kheirabadi M, Kazemi Noureini S. Potential suppression of multidrug-resistance-associated protein 1 by coumarin derivatives: an insight from molecular docking and MD simulation studies. J Biomol Struct Dyn 2024; 42:9184-9200. [PMID: 37667877 DOI: 10.1080/07391102.2023.2250456] [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: 04/24/2023] [Accepted: 08/15/2023] [Indexed: 09/06/2023]
Abstract
Human MRP1 protein plays a vital role in cancer multidrug resistance. Coumarins show promising pharmacological properties. Virtual screening, ADMET, molecular docking and molecular dynamics (MD) simulations were utilized as pharmacoinformatic tools to identify potential MRP1 inhibitors among coumarin derivatives. Using in silico ADMET, 50 hits were further investigated for their selectivity toward the nucleotide-binding domains (NBDs) of MRP1 using molecular docking. Accordingly, coumarin, its symmetrical ketone derivative Lig. No. 4, and Reversan were candidates for focused docking study with the NBDs domains compared with ATP. The result indicates that Lig. No. 4, with the best binding score, interacts with NBDs via hydrogen bonds with residues: GLN713, LYS684, GLY683, CYS682 in NBD1, and GLY1432, GLY771, SER769 and GLN1374 in NBD2, which mostly overlap with ATP binding residues. Moreover, doxorubicin (Doxo) was docked to the transmembrane domains (TMDs) active site of MRP1. Doxo interaction with TMDs was subjected to MD simulation in the NBDs free and occupied with Lig. No. 4 states. The results showed that Doxo interacts more strongly with TMD residues in inward facing feature of TMDs helices. However, when Lig. No. 4 exists in NBDs, Doxo interactions are different, and TMD helices show more outward-facing conformation. This result may suggest a partial competitive inhibition mechanism for the Lig. No. 4 on MRP1 compared with ATP. So, it may inhibit active complex formation by interfering with ATP entrance to NBDs and locking MRP1 conformation in outward-facing mode. This study suggests a valuable coumarin derivative that can be further investigated for potent MRP1 inhibitors.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Parisa Shahpouri
- Department of Biology, Faculty of Science, Hakim Sabzevari University, Sabzevar, Iran
| | - Havva Mehralitabar
- Department of Biology, Faculty of Science, Hakim Sabzevari University, Sabzevar, Iran
| | - Mitra Kheirabadi
- Department of Biology, Faculty of Science, Hakim Sabzevari University, Sabzevar, Iran
| | | |
Collapse
|
89
|
Asmare MM, Yun SI. E-pharmacophore and deep learning based high throughput virtual screening for identification of CDPK1 inhibitors of Cryptosporidium parvum. Comput Biol Chem 2024; 112:108172. [PMID: 39191165 DOI: 10.1016/j.compbiolchem.2024.108172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 08/07/2024] [Accepted: 08/12/2024] [Indexed: 08/29/2024]
Abstract
Cryptosporidiosis, a prevalent gastrointestinal illness worldwide, is caused by the protozoan parasite Cryptosporidium parvum. Calcium-dependent protein kinase 1 (CpCDPK1), crucial for the parasite's life cycle, serves as a promising drug target due to its role in regulating invasion and egress from host cells. While potent Pyrazolopyrimidine analogs have been identified as candidate hit molecules, they exhibit limitations in inhibiting Cryptosporidium growth in cell culture, prompting exploration of alternative scaffolds. Leveraging the most potent compound, RM-1-95, co-crystallized with CpCDPK1, an E-pharmacophore model was generated and validated alongside a deep learning model trained on known CpCDPK1 compounds. These models facilitated screening Enamine's 2 million HTS compound library for novel CpCDPK1 inhibitors. Subsequent hierarchical docking prioritized hits, with final selections subjected to Quantum polarized docking for accurate ranking. Results from docking studies and MD simulations highlighted similarities in interactions between the cocrystallized ligand RM-1-95 and identified hit molecules, indicating comparable inhibitory potential against CpCDPK1. Furthermore, assessing metabolic stability through Cytochrome 450 site of metabolism prediction offered crucial insights for drug design, optimization, and regulatory approval processes.
Collapse
Affiliation(s)
- Misgana Mengistu Asmare
- Department of Agricultural Convergence Technology, College of Agriculture and Life Science, Jeonbuk National University, 567 Baekje-daero, Deokjin-gu, Jeonju-si, Jeollabuk-do 54896, Republic of Korea.
| | - Soon-Il Yun
- Department of Agricultural Convergence Technology, College of Agriculture and Life Science, Jeonbuk National University, 567 Baekje-daero, Deokjin-gu, Jeonju-si, Jeollabuk-do 54896, Republic of Korea; Department of Food Science and Technology, College of Agriculture and Life Sciences, Jeonbuk National University, 567 Baekje-daero, Deokjin-gu, Jeonju-si, Jeollabuk-do 54896, Republic of Korea.
| |
Collapse
|
90
|
Chen Y, Zhang J, Lu J, Shi H, Lan P, Wang W, Ma G, Wei X, Wang X, Yu H. Computational simulations uncover enantioselective metabolism of chiral triazole fungicides by human CYP450 enzymes: A case study of tebuconazole. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 284:116865. [PMID: 39137461 DOI: 10.1016/j.ecoenv.2024.116865] [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: 03/19/2024] [Revised: 07/19/2024] [Accepted: 08/09/2024] [Indexed: 08/15/2024]
Abstract
Tebuconazole (TEB), a prominent chiral triazole fungicide, has been extensively utilized for plant pathogen control globally. Despite experimental evidence of TEB metabolism in mammals, the enantioselectivity in the biotransformation of R- and S-TEB enantiomers by specific CYP450s remains elusive. In this work, integrated in silico simulations were employed to unveil the binding interactions and enantioselective metabolic fate of TEB enantiomers within human CYP1A2, 2B6, 2E1, and 3A4. Molecular dynamics (MD) simulations clearly delineated the binding specificity of R- and S-TEB to the four CYP450s, crucially determining their differences in metabolic activity and enantioselectivity. The primary driving force for robust ligand binding was identified as van der Waals interactions with CYP450s, particularly involving the hydrophobic residues. Mechanistic insights derived from quantum mechanics/molecular mechanics (QM/MM) calculations established C2-methyl hydroxylation as the predominant route of R-/S-TEB metabolism, while C6-hydroxylation and triazol epoxidation were deemed kinetically infeasible pathways. Specifically, the resulting hydroxy-R-TEB metabolite primarily originates from R-TEB biotransformation by 1A2, 2E1 and 3A4, whereas hydroxy-S-TEB is preferentially produced by 2B6. These findings significantly contribute to our comprehension of the binding specificity and enantioselective metabolic fate of chiral TEB by CYP450s, potentially informing further research on human health risk assessment associated with TEB exposure.
Collapse
Affiliation(s)
- Yewen Chen
- College of Geography and Environmental Sciences, Zhejiang Normal University, Jinhua 321004, PR China
| | - Jing Zhang
- College of Geography and Environmental Sciences, Zhejiang Normal University, Jinhua 321004, PR China
| | - Jiayu Lu
- College of Geography and Environmental Sciences, Zhejiang Normal University, Jinhua 321004, PR China
| | - Huifang Shi
- College of Geography and Environmental Sciences, Zhejiang Normal University, Jinhua 321004, PR China
| | - Pengfei Lan
- College of Geography and Environmental Sciences, Zhejiang Normal University, Jinhua 321004, PR China
| | - Wei Wang
- College of Geography and Environmental Sciences, Zhejiang Normal University, Jinhua 321004, PR China
| | - Guangcai Ma
- College of Geography and Environmental Sciences, Zhejiang Normal University, Jinhua 321004, PR China.
| | - Xiaoxuan Wei
- College of Geography and Environmental Sciences, Zhejiang Normal University, Jinhua 321004, PR China
| | - Xueyu Wang
- College of Geography and Environmental Sciences, Zhejiang Normal University, Jinhua 321004, PR China
| | - Haiying Yu
- College of Geography and Environmental Sciences, Zhejiang Normal University, Jinhua 321004, PR China.
| |
Collapse
|
91
|
Lin Y, Cai Y, Li H, Li L, Jiang Z, Ni H. Efficiency enhancement in Aspergillus niger α-L-rhamnosidase reverse hydrolysis by using a tunnel site rational design strategy. Enzyme Microb Technol 2024; 180:110484. [PMID: 39079223 DOI: 10.1016/j.enzmictec.2024.110484] [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: 04/19/2024] [Revised: 06/08/2024] [Accepted: 07/14/2024] [Indexed: 09/15/2024]
Abstract
There has been ongoing interest in improving the efficiency of glycoside hydrolase for synthesizing glycoside compounds through protein engineering, given the potential applications of glycoside compounds. In this study, a strategy of modifying the substrate access tunnel was proposed to enhance the efficiency of reverse hydrolysis catalyzed by Aspergillus niger α-L-rhamnosidase. Analysis of the tunnel dynamics identified Tyr299 as a key modifiable residue in the substrate access tunnel. The location of Tyr299 was near the enzyme surface and at the outermost end of the substrate access tunnel, suggested its role in substrate recognition and throughput. Based on the properties of side chains, six mutants were designed and expressed by Pichia pastoris. Compared to WT, the reverse hydrolysis efficiencies of mutants Y299P and Y299W were increased by 21.3 % and 11.1 %, respectively. The calculation results of binding free energy showed that the binding free energy was inversely proportional to the reverse hydrolysis efficiency. Further, when binding free energy levels were comparable, the mutants with shorter side chains displayed a higher reverse hydrolysis efficiency. These results proved that substrate access tunnel modification was an effective method to improve the reverse hydrolysis efficacy of α-L-rhamnosidase and also provided new insights for modifying other glycoside hydrolases.
Collapse
Affiliation(s)
- Yanling Lin
- College of Food and Biological Engineering, Jimei University, Xiamen 361021, China
| | - Yuchen Cai
- College of Food and Biological Engineering, Jimei University, Xiamen 361021, China
| | - Han Li
- College of Food and Biological Engineering, Jimei University, Xiamen 361021, China
| | - Lijun Li
- College of Food and Biological Engineering, Jimei University, Xiamen 361021, China; Fujian Provincial Key Laboratory of Food Microbiology and Enzyme Engineering, Xiamen 361021, China; Research Center of Food Biotechnology of Xiamen City, Xiamen 361021, China.
| | - Zedong Jiang
- College of Food and Biological Engineering, Jimei University, Xiamen 361021, China; Fujian Provincial Key Laboratory of Food Microbiology and Enzyme Engineering, Xiamen 361021, China; Research Center of Food Biotechnology of Xiamen City, Xiamen 361021, China
| | - Hui Ni
- College of Food and Biological Engineering, Jimei University, Xiamen 361021, China; Fujian Provincial Key Laboratory of Food Microbiology and Enzyme Engineering, Xiamen 361021, China; Research Center of Food Biotechnology of Xiamen City, Xiamen 361021, China; Xiamen Ocean Vocational College, Xiamen 361021, China
| |
Collapse
|
92
|
Tassone G, Maramai S, Paolino M, Lamponi S, Poggialini F, Dreassi E, Petricci E, Alcaro S, Pozzi C, Romeo I. Exploiting the bile acid binding protein as transporter of a Cholic Acid/Mirin bioconjugate for potential applications in liver cancer therapy. Sci Rep 2024; 14:22514. [PMID: 39341955 PMCID: PMC11439058 DOI: 10.1038/s41598-024-73636-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 09/19/2024] [Indexed: 10/01/2024] Open
Abstract
Bioconjugation is one of the most promising strategies to improve drug delivery, especially in cancer therapy. Biomolecules such as bile acids (BAs) have been intensively explored as carriers, due to their peculiar physicochemical properties and biocompatibility. BAs trafficking is regulated by intracellular lipid-binding proteins and their transport in the liver can be studied using chicken liver Bile Acid-Binding Proteins (cL-BABPs) as a reference model. Therefore, we conceived the idea of developing a BA-conjugate with Mirin, an exonuclease inhibitor of Mre11 endowed with different anticancer activities, to direct its transport to the liver. Following computational analysis of various BAs in complex with cL-BABP, we identified cholic acid (CA) as the most promising candidate as carrier, leading to the synthesis of a novel bioconjugate named CA-M11. As predicted by computational data and confirmed by X-ray crystallographic studies, CA-M11 was able to accommodate into the binding pocket of BABP. Hence, it can enter BAs trafficking in the hepatic compartment and here release Mirin. The effect of CA-M11, evaluated in combination with varying concentrations of Doxorubicin on HepG2 cell line, demonstrated a significant increase in cell mortality compared to the use of the cytotoxic drug or Mirin alone, thus highlighting chemo-sensitizing properties. The promising results regarding plasma stability for CA-M11 validate its potential as a valuable agent or adjuvant for hepatic cancer therapy.
Collapse
Affiliation(s)
- Giusy Tassone
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100, Siena, Italy
| | - Samuele Maramai
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100, Siena, Italy.
| | - Marco Paolino
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100, Siena, Italy
| | - Stefania Lamponi
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100, Siena, Italy
| | - Federica Poggialini
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100, Siena, Italy
| | - Elena Dreassi
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100, Siena, Italy
| | - Elena Petricci
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100, Siena, Italy
| | - Stefano Alcaro
- Department of Health Science, Università "Magna Graecia" di Catanzaro, Campus "S. Venuta", Viale Europa, 88100, Catanzaro, Italy
- Net4Science Academic Spin-Off, Università "Magna Graecia" di Catanzaro, Campus "S. Venuta", Viale Europa, 88100, Catanzaro, Italy
| | - Cecilia Pozzi
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100, Siena, Italy.
- Consorzio Interuniversitario Risonanze Magnetiche di Metallo Proteine (CIRMMP), Via Sacconi 6, 50019, Sesto Fiorentino (FI), Italy.
| | - Isabella Romeo
- Department of Health Science, Università "Magna Graecia" di Catanzaro, Campus "S. Venuta", Viale Europa, 88100, Catanzaro, Italy
- Net4Science Academic Spin-Off, Università "Magna Graecia" di Catanzaro, Campus "S. Venuta", Viale Europa, 88100, Catanzaro, Italy
| |
Collapse
|
93
|
Zhang N, Dhumal D, Kuo SH, Lew SQ, Patil PD, Taher R, Vaidya S, Galanakou C, Elkihel A, Oh MW, Chong SY, Marson D, Zheng J, Rouvinski O, Abolarin WO, Pricl S, Lau GW, Lee LTO, Peng L. Targeting the phosphatidylglycerol lipid: An amphiphilic dendrimer as a promising antibacterial candidate. SCIENCE ADVANCES 2024; 10:eadn8117. [PMID: 39321303 PMCID: PMC11423894 DOI: 10.1126/sciadv.adn8117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 08/19/2024] [Indexed: 09/27/2024]
Abstract
The rapid emergence and spread of multidrug-resistant bacterial pathogens require the development of antibacterial agents that are robustly effective while inducing no toxicity or resistance development. In this context, we designed and synthesized amphiphilic dendrimers as antibacterial candidates. We report the promising potent antibacterial activity shown by the amphiphilic dendrimer AD1b, composed of a long hydrophobic alkyl chain and a tertiary amine-terminated poly(amidoamine) dendron, against a panel of Gram-negative bacteria, including multidrug-resistant Escherichia coli and Acinetobacter baumannii. AD1b exhibited effective activity against drug-resistant bacterial infections in vivo. Mechanistic studies revealed that AD1b targeted the membrane phospholipids phosphatidylglycerol (PG) and cardiolipin (CL), leading to the disruption of the bacterial membrane and proton motive force, metabolic disturbance, leakage of cellular components, and, ultimately, cell death. Together, AD1b that specifically interacts with PG/CL in bacterial membranes supports the use of small amphiphilic dendrimers as a promising strategy to target drug-resistant bacterial pathogens and addresses the global antibiotic crisis.
Collapse
Affiliation(s)
- Nian Zhang
- Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Dinesh Dhumal
- Aix-Marseille Universite, CNRS, Centre Interdisciplinaire de Nanoscience de Marseille, UMR 7325, "Equipe Labellisee Ligue Contre le Cancer," 13288 Marseille, France
| | - Shanny Hsuan Kuo
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Shi Qian Lew
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Pankaj D Patil
- Antimicrobial Discovery Center, Department of Biology, Northeastern University, Boston, MA, USA
| | - Raleb Taher
- Antimicrobial Discovery Center, Department of Biology, Northeastern University, Boston, MA, USA
| | - Sanika Vaidya
- Antimicrobial Discovery Center, Department of Biology, Northeastern University, Boston, MA, USA
| | - Christina Galanakou
- Aix-Marseille Universite, CNRS, Centre Interdisciplinaire de Nanoscience de Marseille, UMR 7325, "Equipe Labellisee Ligue Contre le Cancer," 13288 Marseille, France
| | - Abdechakour Elkihel
- Aix-Marseille Universite, CNRS, Centre Interdisciplinaire de Nanoscience de Marseille, UMR 7325, "Equipe Labellisee Ligue Contre le Cancer," 13288 Marseille, France
| | - Myung Whan Oh
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Sook Yin Chong
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Domenico Marson
- Molecular Biology and Nanotechnology Laboratory (MolBNL@UniTS), DEA, University of Trieste, Trieste, Italy
| | - Jun Zheng
- Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Oleg Rouvinski
- Antimicrobial Discovery Center, Department of Biology, Northeastern University, Boston, MA, USA
| | - Williams O Abolarin
- Antimicrobial Discovery Center, Department of Biology, Northeastern University, Boston, MA, USA
| | - Sabrina Pricl
- Molecular Biology and Nanotechnology Laboratory (MolBNL@UniTS), DEA, University of Trieste, Trieste, Italy
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Gee W Lau
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Leo Tsz On Lee
- Faculty of Health Sciences, University of Macau, Taipa, Macau, China
- Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
- Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau, China
| | - Ling Peng
- Aix-Marseille Universite, CNRS, Centre Interdisciplinaire de Nanoscience de Marseille, UMR 7325, "Equipe Labellisee Ligue Contre le Cancer," 13288 Marseille, France
| |
Collapse
|
94
|
Liu S, Hao X, Miao D, Zhang Y. A Study on the Binding Mechanism and the Impact of Key Residue Mutations between SND1 and MTDH Peptide through Molecular Dynamics Simulations. J Phys Chem B 2024; 128:9074-9085. [PMID: 39276108 DOI: 10.1021/acs.jpcb.4c02325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/16/2024]
Abstract
Metastasis of breast cancer is the main cause of death for patients with breast cancer. The interaction between metadherin (MTDH) and staphylococcal nuclease domain 1 (SND1) plays a pivotal role in promoting breast cancer development. However, the binding details between MTDH and SND1 remain unclear. In this study, we employed all-atom molecular dynamics simulations (MDs) and conducted binding energy calculations to investigate the binding details and the impact of key residue mutations on binding. The mutations in key residues have not significantly affected the overall stability of the structure and the fluctuation of residues near the binding site; they have exerted a substantial impact on the binding of SND1 and MTDH peptide. The electrostatic interactions and van der Waals interactions play an important role in the binding of SND1 and the MTDH peptide. The mutations in the key residues have a significant impact on electrostatic and van der Waals interactions, resulting in weakened binding. The energy contributions of key residues mainly come from the electrostatic energy and van der Waals interactions of the side chain. In addition, the key residues form an intricate and stable network of hydrogen bonds and salt-bridge interactions with the MTDH peptide. The mutations in key residues have directly disrupt the interactions formed between SND1 and MTDH peptide, consequently leading to changes in the binding mode of the MTDH peptide. These analyses unveil the detailed atomic-level interaction mechanism between SND1 and the MTDH peptide, providing a molecular foundation for the development of antibreast cancer drugs.
Collapse
Affiliation(s)
- Senchen Liu
- School of Mathematics & Physics, Hebei University of Engineering, Handan 056038, China
| | - Xiafei Hao
- Medical College, Hebei University of Engineering, Handan 056038, China
| | - Dongqiang Miao
- School of Mathematics & Physics, Hebei University of Engineering, Handan 056038, China
| | - Yanjun Zhang
- School of Mathematics & Physics, Hebei University of Engineering, Handan 056038, China
| |
Collapse
|
95
|
Trujillo P, Garavaglia P, Alvarez G, Aduviri S, Domene C, Cannata J, Asciutto EK, García GA, Pickholz M. Insight from atomistic molecular dynamics simulations into the supramolecular assembly of the aldo-keto reductase from Trypanosoma cruzi. J Mol Model 2024; 30:346. [PMID: 39316137 DOI: 10.1007/s00894-024-06153-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 09/19/2024] [Indexed: 09/25/2024]
Abstract
CONTEXT Currently, Chagas disease represents an important public health problem affecting more than 8 million people worldwide. The vector of this disease is the Trypanosoma cruzi (Tc) parasite. Our research specifically focuses on the structure and aggregation states of the enzyme aldo-keto reductase of Tc (TcAKR) reported in this parasite. TcAKR belongs to the aldo-keto reductase (AKR) superfamily, enzymes that catalyze redox reactions involved in crucial biological processes. While most AKRs are found in monomeric forms, some have been reported to form dimeric and tetrameric structures. This is the case for some TcAKR. To better understand how TcAKR multimers form and remain stable, we conducted a comprehensive computational analysis using molecular dynamics (MD) simulations. Our approach to elucidating the aggregation states of TcAKR involved two strategies. Initially, we explored the dynamic behaviour of pre-assembled TcAKR dimers. Subsequently, we examined the self-aggregation of eight monomers. This investigation led to the identification of crucial residues that contribute to the stabilization of protein-protein interactions. It was also found that TcAKRs can form stable supramolecular assemblies, with each monomer typically surrounded by three first neighbours. These findings align with experimental reports of tetrameric or more complex supramolecular structures. Our computational studies could guide further experimental investigations aiming at drug development and assist in designing strategies to modulate aggregation. METHOD Atomistic molecular dynamics simulations were carried out. The TcAKR 3D model structure was obtained by homology modelling using the Swiss Model for the TcAKR sequence (GenBank accession no. EU558869). Further, we checked the model with Alphafold2 and found a high degree of similarity between models. Several tools were used to build the dimers including CLUSPRO, GRAMM-Docking, Hdock, and Py-dock. Protein superstructures were built using the PACKMOL package. CHARMM-GUI was used to set up the simulation systems. GROMACS version 2020.5 was used to perform the simulations with the CHARMM36 force field for the protein and ions and the TIP3P model for water. Further analyses were performed using VMD, GROMACS, AMBER tools, MDLovoFit, bio3d, and in-house programs.
Collapse
Affiliation(s)
- Pablo Trujillo
- Department of Physics, Faculty of Exact and Natural Sciences, University of Buenos Aires, Buenos Aires, Argentina
- CONICET-University of Buenos Aires, Physics Institute of Buenos Aires (IFIBA), Buenos Aires, Argentina
| | - Patricia Garavaglia
- National Institute of Parasitology "Dr. Mario Fatala Chaben" ANLIS "Dr. Carlos G. Malbrán", Buenos Aires, Argentina
| | - Guadalupe Alvarez
- School of Science and Technology, National University of San Martín (UNSAM), ICIFI, CONICET, San Martín, Argentina
| | - Sebastian Aduviri
- Department of Physics, Faculty of Exact and Natural Sciences, University of Buenos Aires, Buenos Aires, Argentina
- CONICET-University of Buenos Aires, Physics Institute of Buenos Aires (IFIBA), Buenos Aires, Argentina
| | - Carmen Domene
- Department of Chemistry, University of Bath, 1 South Bldg, Claverton Down, Bath, BA27AY, UK
| | - Joaquín Cannata
- Institute for Biotechnological Research (IIB-INTECH) "Dr. Rodolfo A. Ugalde", National University of General San Martín-CONICET, San Martín, Argentina
| | - Eliana K Asciutto
- School of Science and Technology, National University of San Martín (UNSAM), ICIFI, CONICET, San Martín, Argentina
| | - Gabriela A García
- National Institute of Parasitology "Dr. Mario Fatala Chaben" ANLIS "Dr. Carlos G. Malbrán", Buenos Aires, Argentina
| | - Mónica Pickholz
- Department of Physics, Faculty of Exact and Natural Sciences, University of Buenos Aires, Buenos Aires, Argentina.
- CONICET-University of Buenos Aires, Physics Institute of Buenos Aires (IFIBA), Buenos Aires, Argentina.
| |
Collapse
|
96
|
Ling J, Ruan Y, Xu C, Liu D, Shi B, Yang Y, Jia Z, Zhang T, Guo M, Li R, Li X. Analysis of factors affecting microbial degradation of cyanobacterial toxins based on theoretical calculations. ENVIRONMENTAL GEOCHEMISTRY AND HEALTH 2024; 46:430. [PMID: 39316189 DOI: 10.1007/s10653-024-02192-z] [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: 05/27/2024] [Accepted: 08/24/2024] [Indexed: 09/25/2024]
Abstract
Cyanobacterial toxins are the most common algal toxins, which are highly toxic and can persist in the aquatic environment without easy degradation, posing risks to the ecosystem and human health that cannot be ignored. Although microbiological methods for the removal of cyanobacterial toxins from aqueous environments are highly efficient, their degradation efficiency is susceptible to many abiotic environmental factors. In this paper, Microcystin-LR (MC-LR) and its microbial degrading enzymes were selected to study the effects of common environmental factors (temperature (T), NO3-, NH4+, Cu2+, Zn2+) and their levels during microbial degradation of cyanobacterial toxins in aqueous environments by using molecular docking, molecular dynamics simulation, analytical factor design, and the combined toxicokinetics of TOPKAT (toxicity prediction). It was found that the addition of T, NO3- and Cu2+ to the aqueous environment promoted the microbial degradation of MC-LR, while the addition of NH4+ and Zn2+ inhibited the degradation; The level effect study showed that the microbial degradation of MC-LR was promoted by increasing levels of added T and NO3- in the aqueous environment, whereas it was inhibited and then promoted by increasing levels of NH4+, Cu2+ and Zn2+. In addition, the predicted toxicity of common Microcystins (MCs) showed that MC-LR, Microcystin-RR (MC-RR) and Microcystin-YR (MC-YR) were not carcinogenic, developmentally toxic, mutagenic or ocular irritants in humans. MC-LR and MC-RR are mild skin irritants and MC-YR is not a skin irritant. MC-YR has a higher chronic and acute toxicity in humans than MC-LR and MC-RR. Acute/chronic toxicity intensity for aquatic animals: MC-YR > MC-LR > MC-RR and for aquatic plants: MC-LR > MC-YR > MC-RR. This suggests that MC-YR also has a high environmental health risk. This paper provides theoretical support for optimizing the environmental conditions for microbial degradation of cyanobacterial toxins by studying the effects of common environmental factors and their level effects in the aquatic environment.
Collapse
Affiliation(s)
- Jianglong Ling
- Center for Environmental Health Risk Assessment and Research, Chinese Research Academy of Environmental Sciences, Beijing, 100012, China
- School of Public Health, Lanzhou University, Lanzhou, 730000, People's Republic of China
- Medical Center for Neck and Low Back Pain, Xijing Hospital, Fourth Military Medical University, Xi'an, 710000, China
| | - Ye Ruan
- School of Public Health, Lanzhou University, Lanzhou, 730000, People's Republic of China
| | - Congchao Xu
- Center for Environmental Health Risk Assessment and Research, Chinese Research Academy of Environmental Sciences, Beijing, 100012, China
| | - Di Liu
- Center for Environmental Health Risk Assessment and Research, Chinese Research Academy of Environmental Sciences, Beijing, 100012, China
| | - Bowen Shi
- Center for Environmental Health Risk Assessment and Research, Chinese Research Academy of Environmental Sciences, Beijing, 100012, China
| | - Yihong Yang
- Center for Environmental Health Risk Assessment and Research, Chinese Research Academy of Environmental Sciences, Beijing, 100012, China
| | - Zihao Jia
- Center for Environmental Health Risk Assessment and Research, Chinese Research Academy of Environmental Sciences, Beijing, 100012, China
| | - Tianyu Zhang
- Center for Environmental Health Risk Assessment and Research, Chinese Research Academy of Environmental Sciences, Beijing, 100012, China
| | - Mingxuan Guo
- Center for Environmental Health Risk Assessment and Research, Chinese Research Academy of Environmental Sciences, Beijing, 100012, China
| | - Rui Li
- Center for Environmental Health Risk Assessment and Research, Chinese Research Academy of Environmental Sciences, Beijing, 100012, China.
| | - Xixi Li
- National Engineering Laboratory for Lake Pollution Control and Ecological Restoration, State Environmental Protection Key Laboratory for Lake Pollution Control, Chinese Research Academy of Environmental Sciences, Beijing, 100012, China.
| |
Collapse
|
97
|
Wang Z, Bao W, Wujieti B, Liu M, Li X, Ma Z, Cui W, Tian Z. Molecular Photoswitching Unlocks Glucose Oxidase for Synergistically Reinforcing Fenton Reactions for Antitumor Chemodynamic Therapy. Angew Chem Int Ed Engl 2024:e202413633. [PMID: 39312192 DOI: 10.1002/anie.202413633] [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: 07/19/2024] [Indexed: 11/08/2024]
Abstract
We have developed a new type of nanoparticles with potent antitumor activity photoactivatable via the combination of molecular photoswitching of spiropyran (SP) and enzymatic reaction of glucose oxidase (GOx). As two key processes involved therein, Fe(III)-to-Fe(II) photoreduction in Fe(III) metal-organic frameworks (MOFs) brings about the release of free Fe2+/Fe3+ while the photoswitching of SP to merocyanine (MC) unlocks the enzymatic activity of GOx that was pre-passivated by SP. The release of free Fe3+ boosts its hydrolysis and therefore enables the acidification of microenvironment, which is further reinforced by one of the products of the GOx-mediated glucose oxidation reaction, gluconic acid (GlcA). Based on the generation of Fe2+ and acidic milieu together with another product of the oxidation reaction, hydrogen peroxide (H2O2), these two processes jointly present triple enabling factors for generating lethal hydroxyl radicals (⋅OH) species via Fenton reactions and therefore oxidative stress capable of inhibiting tumor. The antitumor potency of such nanoparticle is verified in tumor-bearing model mice in vivo, proclaiming its potential as a potent and safe agent based on the unique mechanism of optically manipulating enzyme activity for synergistic antitumor therapeutics with high spatial precision, enhanced efficacy and minimized side effects.
Collapse
Affiliation(s)
- Zicheng Wang
- School of Chemical Sciences, University of Chinese Academy of Sciences (UCAS), 100049, Beijing, P. R. China
| | - Weier Bao
- School of Chemical Sciences, University of Chinese Academy of Sciences (UCAS), 100049, Beijing, P. R. China
| | - Baerlike Wujieti
- School of Chemical Sciences, University of Chinese Academy of Sciences (UCAS), 100049, Beijing, P. R. China
| | - Ming Liu
- School of Chemical Sciences, University of Chinese Academy of Sciences (UCAS), 100049, Beijing, P. R. China
| | | | - Zhecheng Ma
- School of Chemical Sciences, University of Chinese Academy of Sciences (UCAS), 100049, Beijing, P. R. China
| | - Wei Cui
- School of Chemical Sciences, University of Chinese Academy of Sciences (UCAS), 100049, Beijing, P. R. China
| | - Zhiyuan Tian
- School of Chemical Sciences, University of Chinese Academy of Sciences (UCAS), 100049, Beijing, P. R. China
| |
Collapse
|
98
|
Zhang Y, Dou W, Zhao Z, Li G, Li C, Chen X, Mou L. Stereo-selectivity of enantiomeric inhibitors to ubiquitin-specific protease 7 (USP7) dissected by molecular docking, molecular dynamics simulations, and binding free energy calculations. Mol Divers 2024:10.1007/s11030-024-10948-z. [PMID: 39298085 DOI: 10.1007/s11030-024-10948-z] [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: 06/20/2024] [Accepted: 07/25/2024] [Indexed: 09/21/2024]
Abstract
The ubiquitin-specific protease 7 (USP7), as a member of deubiquitination enzymes, represents an attractive therapeutic target for various cancers, including prostate cancer and liver cancer. The change of the inhibitor stereocenter from the S to R stereochemistry (S-ALM → R-ALM34) markedly improved USP7 inhibitory activity. However, the molecular mechanism for the stereo-selectivity of enantiomeric inhibitors to USP7 is still unclear. In this work, molecular docking, molecular dynamics (MD) simulations, molecular mechanics/Generalized-Born surface area (MM/GBSA) calculations, and free energy landscapes were performed to address this mystery. MD simulations revealed that S-ALM34 showed a high degree of conformational flexibility compared to the R-ALM34 counterpart, and S-ALM34 binding led to the enhanced intradomain motions of USP7, especially the BL1 and BL2 loops and the two helices α4 and α5. MM/GBSA calculations showed that the binding strength of R-ALM34 to USP7 was stronger than that of S-ALM34 by - 4.99 kcal/mol, a similar trend observed by experimental data. MM/GBSA free energy decomposition was further performed to differentiate the ligand-residue spectrum. These analyses not only identified the hotspot residues interacting with R-ALM34, but also revealed that the hydrophobic interactions from F409, K420, H456, and Y514 play the major determinants in the binding of R-ALM34 to USP7. This result is anticipated to shed light on energetic basis and conformational dynamics information to aid in the design of more potent and selective inhibitors targeting USP7.
Collapse
Affiliation(s)
- Yusheng Zhang
- Department of Urology, Affiliated Hospital of Shandong Second Medical University, Shandong Second Medical University, Weifang, 261031, Shandong, China
| | - Wenwen Dou
- Department of Infectious Diseases, Affiliated Hospital of Shandong Second Medical University, Shandong Second Medical University, Weifang, 261031, Shandong, China
| | - Ziqi Zhao
- Department of Urology, Affiliated Hospital of Shandong Second Medical University, Shandong Second Medical University, Weifang, 261031, Shandong, China
| | - Guozhen Li
- Department of Urology, Affiliated Hospital of Shandong Second Medical University, Shandong Second Medical University, Weifang, 261031, Shandong, China
| | - Chunlong Li
- Department of Urology, Affiliated Hospital of Shandong Second Medical University, Shandong Second Medical University, Weifang, 261031, Shandong, China.
| | - Xiangyu Chen
- School of Medical Laboratory, Shandong Second Medical University, Weifang, 261031, Shandong, China.
| | - Linkai Mou
- Department of Urology, Affiliated Hospital of Shandong Second Medical University, Shandong Second Medical University, Weifang, 261031, Shandong, China.
| |
Collapse
|
99
|
Arief I, Sunnardianto GK, Khairi S, Saputri WD. The potential of Mitragyna speciosa leaves as a natural source of antioxidants for disease prevention. J Integr Bioinform 2024:jib-2023-0030. [PMID: 39286883 DOI: 10.1515/jib-2023-0030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 07/09/2024] [Indexed: 09/19/2024] Open
Abstract
Mitragyna speciosa is famous for its addictive effect. On the other hand, this plant has good potential as an antioxidant agent, and so far, it was not explicitly explained what the most contributing compound in the leaves to that activity is. This study has been conducted using several computational methods to determine which compounds are the most active in interacting with cytochrome P450, myeloperoxidase, and NADPH oxidase proteins. First, virtual screening was carried out based on molecular docking, followed by profiling the properties of adsorption, distribution, metabolism, excretion, and toxicity (ADMET); the second one is the molecular dynamics (MD) simulations for 100 ns. The virtual screening results showed that three compounds acted as inhibitors for each protein: (-)-epicatechin, sitogluside, and corynoxeine. The ADMET profiles of the three compounds exhibit good drug ability and toxicity. The trajectories study from MD simulations predicts that the complexes of these three compounds with their respective target proteins are stable. Furthermore, these compounds identified in this computational study can be a potential guide for future experiments aimed at assessing the antioxidant properties through in vitro testing.
Collapse
Affiliation(s)
- Ihsanul Arief
- Research Center for Quantum Physics, 599846 National Research and Innovation Agency (BRIN) , South Tangerang 15314, Indonesia
- Akademi Farmasi Yarsi Pontianak, Pontianak 78232, Indonesia
| | - Gagus Ketut Sunnardianto
- Research Center for Quantum Physics, 599846 National Research and Innovation Agency (BRIN) , South Tangerang 15314, Indonesia
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
| | - Syahrul Khairi
- Department of Chemical Engineering, Faculty of Engineering, Universitas Tanjungpura, Pontianak 78124, Indonesia
| | - Wahyu Dita Saputri
- Research Center for Quantum Physics, 599846 National Research and Innovation Agency (BRIN) , South Tangerang 15314, Indonesia
| |
Collapse
|
100
|
Goel KK, Chahal S, Kumar D, Jaiswal S, Nainwal N, Singh R, Mahajan S, Rawat P, Yadav S, Fartyal P, Ahmad G, Jha V, Dwivedi AR. Repurposing of USFDA-approved drugs to identify leads for inhibition of acetylcholinesterase enzyme: a plausible utility as an anti-Alzheimer agent. RSC Med Chem 2024:d4md00461b. [PMID: 39371435 PMCID: PMC11447705 DOI: 10.1039/d4md00461b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 09/10/2024] [Indexed: 10/08/2024] Open
Abstract
In the quest to identify new anti-Alzheimer agents, we employed drug repositioning or drug repositioning techniques on approved USFDA small molecules. Herein, we report the structure-based virtual screening (SBVS) of 1880 USFDA-approved drugs. The in silico-based identification was followed by calculating Prime MMGB-SA binding energy and molecular dynamics simulation studies. The cumulative analysis led to identifying domperidone as an identified hit. Domperidone was further corroborated in vitro using anticholinesterase-based assessment, keeping donepezil as a positive control. The analysis revealed that the identified lead (domperidone) could induce an inhibitory effect on AChE in a dose-dependent manner with an IC50 of 3.67 μM as compared to donepezil, which exhibited an IC50 of 1.37 μM. However, as domperidone is known to have poor BBB permeability, we rationally proposed new analogues utilizing the principles of bioisosterism. The bioisostere-clubbed analogues were found to have better BBB permeability, affinity, and stability within the catalytic domain of AChE via molecular docking and dynamics studies. The proposed bioisosteres may be synthesized in the future. They may plausibly be explored for their implication in the developmental progress of new anti-Alzheimer agent achieved via repurposing techniques in future.
Collapse
Affiliation(s)
- Kapil Kumar Goel
- Department of Pharmaceutical Sciences, Gurukul Kangri (Deemed to Be University) Haridwar 249404 Uttarakhand India
| | - Sandhya Chahal
- Department of Chemistry, Chaudhary Ranbir Singh University Jind India-126102
| | - Devendra Kumar
- School of Pharmacy, Narsee Monjee Institute of Management Studies (NMIMS) Dist. Dhule Maharashtra 424006 India
| | - Shivani Jaiswal
- Institute of Pharmaceutical Research, GLA University Mathura, 17 Km Stone, National Highway, Delhi-Mathura Road, P.O. Chaumuha Mathura-281406 Uttar Pradesh India
| | - Nidhi Nainwal
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University Premanagar Dehradun 248007 Uttarakhand India
| | - Rahul Singh
- University Centre for Research and Development, Chandigarh University Mohali Punjab India
| | - Shriya Mahajan
- Centre of Research Impact and Outcome, Chitkara University Rajpura 140401 Punjab India
| | - Pramod Rawat
- Graphic Era (Deemed to be University) Clement Town Dehradun-248002 India
- Graphic Era Hill University Clement Town Dehradun-248002 India
| | - Savita Yadav
- IES Institute of Pharmacy, IES University Bhopal Madhya Pradesh 462044 India
| | - Prachi Fartyal
- Department of Mathematics, Govt PG college Bajpur (US Nagar) Uttarakhand India
| | - Gazanfar Ahmad
- Prabha Harjilal College of Pharmacy and Paraclinical Sciences, Madre Meharban Campus of Health Sciences (Affiliated to University of Jammu) Jammu J&K 181122 India
| | - Vibhu Jha
- Institute of Cancer Therapeutics School of Pharmacy and Medical Sciences, University of Bradford UK
| | | |
Collapse
|