1
|
Rustagi V, Rameshwari R, Kumar Singh I. Identification of potential inhibitors for MAP4K4 in glaucoma using meta-dynamics-based dissociation free energy calculation. Brain Res 2025; 1847:149300. [PMID: 39500479 DOI: 10.1016/j.brainres.2024.149300] [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: 05/23/2024] [Revised: 10/16/2024] [Accepted: 10/26/2024] [Indexed: 11/12/2024]
Abstract
Glaucoma, a prevalent eye ailment causing irreversible vision loss, affects over 295 million individuals globally, necessitating the exploration of novel therapeutic avenues. Despite extensive research on targets like the phosphodiesterase enzyme and rho kinase, the potential of MAP4K4 in glaucoma remains untapped. This study aims to identify potent MAP4K4 inhibitors to counteract retinal cell apoptosis and oxidative stress associated with glaucoma. Using HTVS and XP docking, 911,059 compounds were screened. The MMGBSA calculation and pharmacokinetics analysis were used to shortlist the compounds. After performing 75 molecular dynamics simulations, further meta-dynamics were employed to calculate dissociation-free energy and find potential MAP4K4 inhibitors. Findings indicated that ZINC06717217 and ZINC38836256 exhibited remarkable promise, with docking scores of -9.57 and -11.12 and MMGBSA binding energies of -91.07 kcal/mol and -87.52 kcal/mol, respectively. Comparative analysis with the reference compound Q27453723 underscored their superior stability, requiring dissociation-free energies of -15.11 kcal/mol and -12.46 kcal/mol to disengage from the docked complex. This underscored their robust binding affinity. ZINC06717217 and ZINC38836256 show promising stability and strong binding to the MAP4K4 protein. Hence, these findings are promising in inhibiting MAP4K4 for glaucoma treatment, potentially leading to more effective treatment and curing blindness. KEY MESSAGES: First to incorporate the dissociation-free energy for identifying compounds for glaucoma treatment. In-silico analysis showed that ZINC06717217 and ZINC38836256 are promising compounds for targeting MAP4K4.
Collapse
Affiliation(s)
- Vanshika Rustagi
- Department of Biotechnology, School of Engineering and Technology, Manav Rachna International Institute of Research and Studies, Faridabad, Haryana 121004, India
| | - Rashmi Rameshwari
- Department of Biotechnology, School of Engineering and Technology, Manav Rachna International Institute of Research and Studies, Faridabad, Haryana 121004, India.
| | - Indrakant Kumar Singh
- Molecular Biology Research Lab, Department of Zoology, Deshbandhu College, University of Delhi, Kalkaji, New Delhi 110019, India; Delhi School of Public Health, Institute of Eminence, University of Delhi, Delhi 110007, India.
| |
Collapse
|
2
|
Balta O, Yilmaz E, Tatar Yilmaz G. Exploring Inhibition Mechanisms in Wildtype and T315I BCR-ABL1: An In Silico Approach Integrating Virtual Screening, MD Simulations, and MM-GBSA Analysis. J Comput Chem 2025; 46:e27545. [PMID: 39636243 DOI: 10.1002/jcc.27545] [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: 07/19/2024] [Revised: 09/17/2024] [Accepted: 11/15/2024] [Indexed: 12/07/2024]
Abstract
The BCR-ABL tyrosine kinase which is responsible for the pathogenesis of chronic myeloid leukemia (CML), has emerged as a promising therapeutic target. To address this issue, we employed a comprehensive computational approach integrating virtual screening, molecular dynamics (MD) simulations, and MM-GBSA (Molecular Mechanics/Generalized Born Surface Area) analysis to identify potential inhibitors and elucidate their binding mechanisms. Initially, virtual screening was conducted on 994 compounds from the ZINC database and, these compounds were docked against wildtype and T315I mutant ABL1 for the Type I and Type II ABL1 kinase inhibition mechanisms. In our molecular docking analysis for Type I inhibition, compound 911 demonstrated notable affinity towards the wildtype ABL1, with a binding energy of -14.91 kcal/mol, while compound 972 showed significant binding affinity towards the mutant ABL1, with a binding energy of -14.27 kcal/mol. In the Type II inhibition mechanism, the compounds with the highest binding affinity were compound 261 in wildtype ABL1 with -17.05 kcal/mol binding energy and compound 966 to the mutant ABL1 with a binding energy of -16.29 kcal/mol. Furthermore, analyses of MD simulations and MM/GBSA binding free energy (ΔG) were performed for target proteins with compounds, that exhibited the most favorable binding affinities with target proteins. The selected hit compounds showed ΔG scores ranging from -118.09 to -74.85 kJ/mol in both wildtype and mutant ABL1. Considering all in silico studies performed, it can be inferred that the identified molecules hold promise as potential candidates for drug design aimed at targeting CML.
Collapse
Affiliation(s)
- Ozlen Balta
- Department of Hematology, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey
- Department of Bioinformatics, Institute of Health Sciences, Karadeniz Technical University, Trabzon, Turkey
| | - Ercument Yilmaz
- Department of Computer Technologies, Karadeniz Technical University, Trabzon, Turkey
- Yılmaz Bilişim R&D Consulting Software Engineering and Services Trade Limited Company, Trabzon, Turkey
| | - Gizem Tatar Yilmaz
- Department of Bioinformatics, Institute of Health Sciences, Karadeniz Technical University, Trabzon, Turkey
- Yılmaz Bilişim R&D Consulting Software Engineering and Services Trade Limited Company, Trabzon, Turkey
- Department of Biostatistics and Medical Informatics, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey
| |
Collapse
|
3
|
Scott CE, Juechter LA, Rocha J, Jones LD, Outten B, Aishman TD, Ivers AR, Shields GC. Impact of Intracellular Proteins on μ-Opioid Receptor Structure and Ligand Binding. J Phys Chem B 2024. [PMID: 39699881 DOI: 10.1021/acs.jpcb.4c05214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2024]
Abstract
Chronic pain is a prevalent problem affecting approximately one out of every five adults in the U.S. The most effective way to treat chronic pain is with opioids, but they cause dangerous side effects such as tolerance, addiction, and respiratory depression, which makes them quite deadly. Opioids, such as fentanyl, target the μ-opioid receptor (MOR), which can then bind to the intracellular Gi protein or the β-arrestin protein. The Gi pathway is primarily responsible for pain relief and potential side effects, but the β-arrestin pathway is chiefly responsible for the unwanted side effects. Ideally, an effective pain medication without side effects would bind to MOR, which would bias signaling solely through the Gi pathway. We used the Bio3D library to conduct principal component analysis to compare the cryo-electron microscopy MOR structures in complex with the Gi versus an X-ray crystallography MOR structure with a nanobody acting as a Gi mimic. Our results agree with a previous study by Munro, which concluded that nanobody-bound MOR is structurally different than Gi-bound MOR. Furthermore, we investigated the structural diversity of opioids that can bind to MOR. Quantum mechanical calculations show that the low energy solution structures of fentanyl differ from the one bound to MOR in the experimental structure, and pKa calculations reveal that fentanyl is protonated in aqueous solution. Glide docking studies show that higher energy structures of fentanyl in solution form favorable docking complexes with MOR. Our calculations show the relative abundance of each fentanyl conformation in solution as well as the energetic barriers that need to be overcome to bind to MOR. Docking studies confirm that multiple fentanyl conformations can bind to the receptor. Perhaps a variety of conformations of fentanyl can stabilize multiple conformations of the MOR, which can explain why fentanyl can induce different intracellular signaling and multiple physiological effects.
Collapse
Affiliation(s)
- Caitlin E Scott
- Department of Chemistry and Biochemistry, California State University, Los Angeles, California, 90032, United States
- Department of Chemistry, Hendrix College, Conway, Arkansas 72032, United States
| | - Leah A Juechter
- Department of Chemistry, Furman University, Greenville, South Carolina 29613, United States
| | - Josephine Rocha
- Department of Chemistry and Biochemistry, California State University, Los Angeles, California, 90032, United States
| | - Lauren D Jones
- Department of Chemistry, Furman University, Greenville, South Carolina 29613, United States
| | - Brenna Outten
- Department of Chemistry, Furman University, Greenville, South Carolina 29613, United States
| | - Taylor D Aishman
- Department of Chemistry, Hendrix College, Conway, Arkansas 72032, United States
| | - Alaina R Ivers
- Department of Chemistry, Hendrix College, Conway, Arkansas 72032, United States
| | - George C Shields
- Department of Chemistry, Furman University, Greenville, South Carolina 29613, United States
| |
Collapse
|
4
|
Ranade SD, Alegaon SG, Khatib NA, Gharge S, Kavalapure RS, Kumar BRP. Reversal of insulin resistance to combat type 2 diabetes mellitus by newer thiazolidinedione's in fructose induced insulin resistant rats. Eur J Med Chem 2024; 280:116939. [PMID: 39396421 DOI: 10.1016/j.ejmech.2024.116939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 09/30/2024] [Accepted: 10/02/2024] [Indexed: 10/15/2024]
Abstract
In our pursuit of discovering new antidiabetic agents to manage type 2 diabetes mellitus (T2DM), our approach aimed to identify the bioactive feature/pharmacophore responsible for PPAR-γ expression, as it is accountable for the glucose homeostasis and lipid metabolism. This was achieved by pharmacophore model generation, screening of rationally designed newer thiazolidinedione's library, identifying synthesizing and characterizing the top ten molecules (5a-5j) for their (Invitro & invivo) antidiabetic activity. Preliminary screening of all the ligands by Invitro glucose uptake assay in L6 myotubes (skeletal muscle cell line of rats) revealed compound 5b and 5f stimulated the glucose uptake with 79.29 ± 1.02 % and 74.58 ± 1.02 % respectively compared to pioglitazone with 82.36 ± 0.98 %. This was validated by PPAR-γ TF expression assay, which highlighted a dose dependent increase in transactivation of PPAR-γ. These compounds 5b and 5f were evaluated in fructose induced insulin resistance rat model. Where the treatment with 5b and 5f markedly increased the exogenous clearance of glucose and exogenous insulin via OGTT and ITT respectively, also improved the glucose utilization by significantly increasing content of glycogen and uptake of glucose in rat hemidiaphragm and reversed insulin resistance. Likewise a significant decreased in the VLDL and triglyceride levels was seen in 5b and 5f treated groups compared to insulin resistant (IR) group. It improved glycogenesis by catabolism of glucose and maintained glycaemic control. Similarly it had marked action on enzymatic oxidative biomarkers. Compound 5b displayed better, improved T1/2 (half-life) of 4.21 h and Kel (elimination constant) of 0.381 was noticed in comparison to compound 5f indicating the pharmacokinetic profile. Insilico studies like DFT calculations refined the geometry of 5b and 5f ligands, docking and molecular simulation provided the insights in binding affinity, dynamic behaviour and stability of ligands in PPAR-γ ligand binding domain. MM/GBSA provided the energetics of 5b and 5f in binding pocket. Finally network pharmacology identified ADIPOQ (adiponectin), NR1C3 (PPAR-γ), SLC2A4 (GLUT4), and LEP (leptin) proteins associate with compound 5b and 5f and enriched in Adipocytokine pathway, and PPAR-γ signaling pathway.
Collapse
Affiliation(s)
- Shriram D Ranade
- Department of Pharmaceutical Chemistry, KLE College of Pharmacy, Belagavi, KLE Academy of Higher Education and Research, Belagavi, 590 010, Karnataka, India
| | - Shankar G Alegaon
- Department of Pharmaceutical Chemistry, KLE College of Pharmacy, Belagavi, KLE Academy of Higher Education and Research, Belagavi, 590 010, Karnataka, India.
| | - Nayeem A Khatib
- Department of Pharmacology, KLE College of Pharmacy, Belagavi, KLE Academy of Higher Education and Research, Belagavi, 590 010, Karnataka, India
| | - Shankar Gharge
- Department of Pharmaceutical Chemistry, KLE College of Pharmacy, Belagavi, KLE Academy of Higher Education and Research, Belagavi, 590 010, Karnataka, India
| | - Rohini S Kavalapure
- Department of Pharmaceutical Chemistry, KLE College of Pharmacy, Belagavi, KLE Academy of Higher Education and Research, Belagavi, 590 010, Karnataka, India
| | - B R Prashantha Kumar
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, Mysuru, JSS Academy of Higher Education and Research, Mysuru, 570015, Karnataka, India
| |
Collapse
|
5
|
Machado TMMM, Aquino IG, Franchin M, Zarraga MO, Bustos D, Spada FP, Napimoga MH, Clemente-Napimoga JT, Alencar SM, Benso B, Abdalla HB. Novel apocynin regulates TRPV1 activity in the trigeminal system and controls pain in a temporomandibular joint neurogenic model. Eur J Pharmacol 2024; 985:177093. [PMID: 39489280 DOI: 10.1016/j.ejphar.2024.177093] [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: 06/24/2024] [Revised: 09/20/2024] [Accepted: 10/31/2024] [Indexed: 11/05/2024]
Abstract
OBJECTIVE Herein, we investigate the potential analgesic effect of a newly synthesized chalcone-derived apocynin in a neurogenic pain model. METHODS Molecular docking was used to foretell the apocynin binding features and dynamics with the TRPV1 channel, and the activity was tested in vitro, using transfected HEK 293T cells with the rat TRPV1 receptor. The analgesic effect of apocynin was investigated using a capsaicin-induced pain model. The expression of TRPV1, TRPA1, TRPM8, and MAPKs was assessed by electrophoresis, and immunosorbent assays were performed to quantify the neurotransmitters Substance P, Glutamate, and CGRP. A survival assay using Galleria mellonella was carried out to determine the toxicity. RESULTS We observed that apocynin exhibits greater thermodynamic stability. Upon apocynin ligand binding, it changes the electrostatic potential for a predominantly electronegative state in the interior and neutral in its external vanilloid pocket. Treatment of apocynin induces antinociceptive effects against the noxious challenge of capsaicin. Histologically, apocynin decreased the number of TRPV1+ immunopositive cells. Electrophoresis showed reduced phosphorylation of p44/42 (ERK1/2) and decreased protein levels of substance P, and CGRP. In the survival assay, apocynin showed low toxicity. CONCLUSIONS In conclusion, we provide proof-of-principles that the newly synthesized apocynin compound effectively prevented nociception in a neurogenic model of orofacial pain.
Collapse
Affiliation(s)
| | | | - Marcelo Franchin
- School of Dentistry, Federal University of Alfenas (Unifal-MG), Alfenas, MG, Brazil; Bioactivity and Applications Lab, Department of Biological Sciences, Faculty of Science and Engineering, School of Natural Sciences, University of Limerick, Limerick, Ireland
| | - Miguel O Zarraga
- Department of Organic Chemistry, Faculty of Chemical Sciences, Universidad de Concepcion, Concepcion, Chile
| | - Daniel Bustos
- Centro de Investigación de Estudios Avanzados del Maule (CIEAM), Vicerrectoría de Investigación y Postgrado, Universidad Católica del Maule, Talca, Chile; Laboratorio de Bioinformática y Química Computacional (LBQC), Escuela de Bioingeniería Médica, Facultad de Medicina, Universidad Católica del Maule, Talca, Chile
| | - Fernanda Papa Spada
- Department of Agri-Food Industry, Food, and Nutrition, Luiz de Queiroz College of Agriculture, University of São Paulo (USP), Piracicaba, SP, Brazil
| | | | | | - Severino Matias Alencar
- Department of Agri-Food Industry, Food, and Nutrition, Luiz de Queiroz College of Agriculture, University of São Paulo (USP), Piracicaba, SP, Brazil
| | - Bruna Benso
- School of Dentistry, Faculty of Medicine, Pontificia Universidad Catolica de Chile, Santiago, Chile.
| | | |
Collapse
|
6
|
Sadamori K, Kubo T, Yoshida T, Yamamoto M, Shibata Y, Fukasawa K, Tokumura K, Horie T, Kadota T, Yamakawa R, Hojo H, Tanaka N, Kitao T, Shirahase H, Hinoi E. CDK8 inhibitor KY-065 rescues skeletal abnormalities in achondroplasia model mice. Biochim Biophys Acta Mol Basis Dis 2024; 1871:167626. [PMID: 39674288 DOI: 10.1016/j.bbadis.2024.167626] [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: 06/01/2024] [Revised: 12/10/2024] [Accepted: 12/10/2024] [Indexed: 12/16/2024]
Abstract
Cyclin-dependent kinase 8 (CDK8) is a transcription-related CDK family member implicated in the regulation of bone homeostasis, and we recently demonstrated that our internally developed CDK8 inhibitor KY-065 can prevent postmenopausal osteoporosis in a mouse model. Achondroplasia (ACH), the most common form of genetic dwarfism in humans, is caused by a gain-of-function mutation in fibroblast growth factor receptor 3 (FGFR3), a receptor tyrosine kinase that activates downstream mitogen-activated protein kinase (MAPK) and signal transducer and activator of transcription (STAT) signaling pathways. The first precision drug approved for the treatment of ACH in children, the C-type natriuretic peptide analog vosoritide, antagonizes the MAPK pathway, while there are currently no effective and safe medications targeting the STAT1 pathway. Here, we demonstrate that KY-065 rescues impaired chondrogenesis and stunted long bone growth in the Fgfr3Ach mouse model of ACH. KY-065 inhibited CDK8 with high affinity in vitro by competing with ATP. The CDK8 expression and STAT1Ser727 phosphorylation were upregulated in chondrocytes isolated from ACH model mice, and KY-065 repressed its phosphorylation and restored normal chondrogenic differentiation without affecting MAPK activation. Moreover, daily administration of 10 mg/kg KY-065 to Fgfr3Ach mice (yielding a peak concentration of 22.0 ± 1.47 μM in plasma) resulted in significant elongation of long bone and improved growth plate cytoarchitecture. Collectively, these findings identify the CDK8 in chondrocytes as a potential therapeutic target for ACH and KY-065 as a promising candidate drug treatment for this debilitating skeletal disease.
Collapse
Affiliation(s)
- Koki Sadamori
- Department of Bioactive Molecules, Pharmacology, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Takuya Kubo
- Department of Bioactive Molecules, Pharmacology, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Tomoki Yoshida
- School of Pharmacy, Kitasato University, Tokyo 108-8641, Japan
| | - Megumi Yamamoto
- Drug Discovery Research Department, Kyoto Pharmaceutical Industries, Ltd., Kyoto, Japan
| | - Yui Shibata
- Drug Discovery Research Department, Kyoto Pharmaceutical Industries, Ltd., Kyoto, Japan
| | - Kazuya Fukasawa
- Department of Bioactive Molecules, Pharmacology, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Kazuya Tokumura
- Department of Bioactive Molecules, Pharmacology, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Tetsuhiro Horie
- Department of Bioactive Molecules, Pharmacology, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Takuya Kadota
- Department of Bioactive Molecules, Pharmacology, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Ryotaro Yamakawa
- Department of Bioactive Molecules, Pharmacology, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Hironori Hojo
- Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan; Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Tokyo 113-8656, Japan
| | - Nobutada Tanaka
- School of Pharmacy, Kitasato University, Tokyo 108-8641, Japan
| | - Tatsuya Kitao
- Drug Discovery Research Department, Kyoto Pharmaceutical Industries, Ltd., Kyoto, Japan
| | - Hiroaki Shirahase
- Drug Discovery Research Department, Kyoto Pharmaceutical Industries, Ltd., Kyoto, Japan
| | - Eiichi Hinoi
- Department of Bioactive Molecules, Pharmacology, Gifu Pharmaceutical University, Gifu 501-1196, Japan; United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, Gifu 501-1196, Japan; Center for One Medicine Innovative Translational Research (COMIT), Division of Innovative Modality Development, Gifu University, Gifu 501-1196, Japan.
| |
Collapse
|
7
|
Nael MA, Ghoneim MM, Almuqbil M, Al-Serwi RH, El-Sherbiny M, Mostafa AE, Elokely KM. An evaluation of the precision of computational methods used in drug development initiatives. J Biomol Struct Dyn 2024:1-15. [PMID: 39659185 DOI: 10.1080/07391102.2024.2435633] [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: 01/04/2024] [Accepted: 03/29/2024] [Indexed: 12/12/2024]
Abstract
Computational approaches are commonly employed to expedite and provide decision-making for the drug development process. Drug development programs that involve targets without known crystal structures can be quite challenging. In many cases, a viable approach is to generate reliable homology models using the amino acid sequence of the target. This is followed by a series of validation steps, druggable pocket detection, and then moving forward with lead identification and validation. This study commenced by conducting an initial benchmark exercise using a series of computationally designed sequences for steroid-binding proteins. By conducting an unbiased comparison with the released X-ray crystal structures, the homology models that were generated demonstrated reliable outcomes. The aligned homology models showed a root mean square deviation (RMSD) of less than 0.6 Å when compared to the corresponding X-ray structures. Three different methods were used to detect the druggable cavities for comparison, and the identified pockets closely resembled those of the crystal structures. The achievement of near-native pose prediction was made possible by utilizing the comprehensive binding energy function that characterizes the interaction between each pose and the neighboring residues. In order to address the issue of limited correlation between entropy and internal energy in docking, an alternative was devised by incorporating entropy as a post-docking optimization step to enhance the accuracy of ligand binding affinity predictions and improve the overall quality of the results.
Collapse
Affiliation(s)
- Manal A Nael
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta, Egypt
- Department of Chemistry, Institute for Computational Molecular Science, Temple University, Philadelphia, Pennsylvania, USA
| | - Mohammed M Ghoneim
- Department of Pharmacy Practice, College of Pharmacy, AlMaarefa University, Riyadh, Saudi Arabia
| | - Mansour Almuqbil
- Clinical Pharmacy Department, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Rasha Hamed Al-Serwi
- Department of Basic Dental Sciences, College of Dentistry, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Mohamed El-Sherbiny
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, Riyadh, Saudi Arabia
| | - Ahmad E Mostafa
- Pharmacognosy and Medicinal Plants Department, Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Khaled M Elokely
- Department of Chemistry, Institute for Computational Molecular Science, Temple University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
8
|
Ohadi D, Kumar K, Ravula S, DesJarlais RL, Seierstad MJ, Shih AY, Hack MD, Schiffer JM. Input Pose is Key to Performance of Free Energy Perturbation: Benchmarking with Monoacylglycerol Lipase. J Chem Inf Model 2024; 64:8859-8869. [PMID: 39560439 DOI: 10.1021/acs.jcim.4c01223] [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: 11/20/2024]
Abstract
Free energy perturbation (FEP) methodologies have become commonplace methods for modeling potency in hit-to-lead and lead optimization stages of drug discovery. The conformational states of the initial poses of compounds for FEP+ calculations are often set up by alignment to a cocrystal structure ligand, but it is not clear if this method provides the best result for all proteins or all ligands. Not only are ligand conformational states potential variables in modeling compound potency in FEP but also the selection of crystallographic water molecules for inclusion in the FEP input structures can impact FEP models. Here, we report the results of FEP calculations using FEP+ from Schrödinger and starting from maximum common substructure alignment and docked poses generated with an array of docking methodologies. As a benchmark data set, we use monoacylglycerol lipase (MAGL), an important clinical drug target in cancer malignancy, neurological diseases, and metabolic disorders, and a set of 17 MAGL inhibitors. We found a large variation among FEP+ correlations to experimental IC50 values depending on the method used to generate the input pose and that the inclusion of ligand-based information in the docking process, with some methods, increases the correlation between FEP+ free energies and IC50 values. Upon analysis of the initial poses, we found that the differences in FEP+ correlations stemmed from rotation around a tertiary amide bond as well as translation of the compound toward the more hydrophobic side of the MAGL pocket. FEP+ estimation improved across all pose modeling methods when hydrogen bond constraint information was added. However, simple maximum common substructure alignment in the presence of all crystallographic water molecules outperformed all other methods in correlation between estimated and experimental IC50 values. Taken together, these findings suggest that pose selection and crystallographic water inclusion greatly impact how well FEP+ estimated IC50 values align with experimental IC50 values and that modelers should benchmark a few different pose generation methodologies and different water inclusion strategies for their hit-to-lead and lead optimization drug discovery projects.
Collapse
Affiliation(s)
- Donya Ohadi
- Johnson & Johnson Innovative Medicine, 1400 McKean Road, Spring House, Pennsylvania 19477, United States
| | - Kiran Kumar
- Johnson & Johnson Innovative Medicine, 1400 McKean Road, Spring House, Pennsylvania 19477, United States
| | - Suchitra Ravula
- Johnson & Johnson Innovative Medicine, 3210 Merryfield Row, San Diego, California 92121, United States
| | - Renee L DesJarlais
- Johnson & Johnson Innovative Medicine, 1400 McKean Road, Spring House, Pennsylvania 19477, United States
| | - Mark J Seierstad
- Johnson & Johnson Innovative Medicine, 3210 Merryfield Row, San Diego, California 92121, United States
| | - Amy Y Shih
- Johnson & Johnson Innovative Medicine, 3210 Merryfield Row, San Diego, California 92121, United States
| | - Michael D Hack
- Johnson & Johnson Innovative Medicine, 3210 Merryfield Row, San Diego, California 92121, United States
| | - Jamie M Schiffer
- Johnson & Johnson Innovative Medicine, 3210 Merryfield Row, San Diego, California 92121, United States
| |
Collapse
|
9
|
Bruno P, Pala D, Micoli A, Corsi M, Accetta A, Carzaniga L, Ronchi P, Fiorelli C, Formica M, Pizzirani D, Mazzucato R, Guariento S, Bertolini S, Martucci C, Allen AD, Mileo V, Capacchi S, Gallo PM, Fioni A, Xanxo Fernandez S, Villetti G, Puccini P, Civelli M, Guala M, Retini M, Martinelli P, Visentini F, Pavoni V, Daldosso M, Fontana S, Biagetti M, Capelli AM. Discovery of CHF-6523, an Inhaled Selective PI3Kδ Inhibitor for the Treatment of Chronic Obstructive Pulmonary Disease. J Med Chem 2024. [PMID: 39635891 DOI: 10.1021/acs.jmedchem.4c02062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
The design of inhaled selective phosphatidylinositol 3-kinase delta (PI3Kδ) inhibitors for the treatment of inflammatory lung diseases was pursued. Knowledge-based design of a novel isocoumarin scaffold that was able to adopt a propeller-shape topology ensured the desired PI3Kδ selectivity. Achievement of low nanomolar cellular potencies through hinge binder group optimization, reduction of intrinsic permeability through head group optimization to extend lung retention, and screening of crystalline forms suitable for administration as dry powders culminated in the identification of compound 18. This novel inhaled selective PI3Kδ inhibitor displayed durable anti-inflammatory activity in a disease-relevant rat model of Th-2-driven acute lung inflammation and safe in vitro and in vivo preclinical profiles. Therefore, compound 18 showed the appropriate discovery profile and was progressed to clinical trials in healthy volunteers and chronic obstructive pulmonary disease (COPD) patients as CHF-6523.
Collapse
Affiliation(s)
- Paolo Bruno
- Chiesi Farmaceutici S.p.A, Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| | - Daniele Pala
- Chiesi Farmaceutici S.p.A, Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| | - Alessandra Micoli
- Aptuit, an Evotec Company, Via Alessandro Fleming, 4, 37135 Verona, Italy
| | - Mauro Corsi
- Aptuit, an Evotec Company, Via Alessandro Fleming, 4, 37135 Verona, Italy
| | - Alessandro Accetta
- Chiesi Farmaceutici S.p.A, Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| | - Laura Carzaniga
- Chiesi Farmaceutici S.p.A, Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| | - Paolo Ronchi
- Chiesi Farmaceutici S.p.A, Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| | - Claudio Fiorelli
- Chiesi Farmaceutici S.p.A, Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| | - Michele Formica
- Chiesi Farmaceutici S.p.A, Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| | - Daniela Pizzirani
- Chiesi Farmaceutici S.p.A, Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| | - Roberta Mazzucato
- Chiesi Farmaceutici S.p.A, Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| | - Sara Guariento
- Chiesi Farmaceutici S.p.A, Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| | - Serena Bertolini
- Chiesi Farmaceutici S.p.A, Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| | - Cataldo Martucci
- Chiesi Farmaceutici S.p.A, Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| | - Andrew Dennis Allen
- Chiesi Farmaceutici S.p.A, Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| | - Valentina Mileo
- Chiesi Farmaceutici S.p.A, Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| | - Silvia Capacchi
- Chiesi Farmaceutici S.p.A, Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| | - Paola Maria Gallo
- Chiesi Farmaceutici S.p.A, Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| | - Alessandro Fioni
- Chiesi Farmaceutici S.p.A, Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| | | | - Gino Villetti
- Chiesi Farmaceutici S.p.A, Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| | - Paola Puccini
- Chiesi Farmaceutici S.p.A, Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| | - Maurizio Civelli
- Chiesi Farmaceutici S.p.A, Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| | - Matilde Guala
- Aptuit, an Evotec Company, Via Alessandro Fleming, 4, 37135 Verona, Italy
| | - Michele Retini
- Aptuit, an Evotec Company, Via Alessandro Fleming, 4, 37135 Verona, Italy
| | - Prisca Martinelli
- Aptuit, an Evotec Company, Via Alessandro Fleming, 4, 37135 Verona, Italy
| | - Filippo Visentini
- Aptuit, an Evotec Company, Via Alessandro Fleming, 4, 37135 Verona, Italy
| | - Valentina Pavoni
- Aptuit, an Evotec Company, Via Alessandro Fleming, 4, 37135 Verona, Italy
| | - Matteo Daldosso
- Aptuit, an Evotec Company, Via Alessandro Fleming, 4, 37135 Verona, Italy
| | - Stefano Fontana
- Aptuit, an Evotec Company, Via Alessandro Fleming, 4, 37135 Verona, Italy
| | - Matteo Biagetti
- Chiesi Farmaceutici S.p.A, Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| | - Anna Maria Capelli
- Chiesi Farmaceutici S.p.A, Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| |
Collapse
|
10
|
Camci M, Şenol H, Kose A, Karaman Mayack B, Alayoubi MM, Karali N, Gezginci MH. Bioisosteric replacement of the carboxylic acid group in Hepatitis-C virus NS5B thumb site II inhibitors: phenylalanine derivatives. Eur J Med Chem 2024; 279:116832. [PMID: 39288595 DOI: 10.1016/j.ejmech.2024.116832] [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: 07/11/2023] [Revised: 11/03/2023] [Accepted: 09/01/2024] [Indexed: 09/19/2024]
Abstract
Hepatitis C virus (HCV) is a global health concern and the NS5B RNA-dependent RNA polymerase (RdRp) of HCV is an attractive target for drug discovery due to its role in viral replication. This study focuses on NS5B thumb site II inhibitors, specifically phenylalanine derivatives, and explores bioisosteric replacement and prodrug strategies to overcome limitations associated with carboxylic acid functionality. The synthesized compounds demonstrated antiviral activity, with compound 6d showing the most potent activity with an EC50 value of 3.717 μM. The hydroxamidine derivatives 7a-d showed EC50 values ranging from 3.9 μM to 11.3 μM. However, the acidic heterocyclic derivatives containing the oxadiazolone (8a-d) and oxadiazolethione (9a-d) rings did not exhibit measurable activity. A methylated heterocycle 10b showed a hint of activity at 8.09 μM. The pivaloyloxymethyl derivatives 11a and 11b did not show antiviral activity. Further studies are warranted to fully understand the effects of these modifications and to explore additional strategies for developing novel therapeutic options for HCV.
Collapse
Affiliation(s)
- Merve Camci
- Istanbul University, Faculty of Pharmacy, Department of Pharmaceutical Chemistry, 34116, Istanbul, Turkey; Graduate School of Health Sciences, Istanbul University, 34126, Istanbul, Turkey.
| | - Halil Şenol
- Bezmialem Vakif University, Faculty of Pharmacy, Department of Pharmaceutical Chemistry, 34093, Istanbul, Turkey.
| | - Aytekin Kose
- Aksaray University, Faculty of Science and Letters, Department of Chemistry, 68100, Aksaray, Turkey.
| | - Berin Karaman Mayack
- Istanbul University, Faculty of Pharmacy, Department of Pharmaceutical Chemistry, 34116, Istanbul, Turkey; Department of Pharmacology, School of Medicine, University of California Davis, Davis, CA, 95616, USA.
| | | | - Nilgun Karali
- Istanbul University, Faculty of Pharmacy, Department of Pharmaceutical Chemistry, 34116, Istanbul, Turkey.
| | - Mikail Hakan Gezginci
- Istanbul University, Faculty of Pharmacy, Department of Pharmaceutical Chemistry, 34116, Istanbul, Turkey.
| |
Collapse
|
11
|
Shakir M, Ali A, Lakshmi S, Garg M, Abdulhameed Almuqdadi HT, Irfan I, Kamthan M, Joshi MC, Javed S, Rawat DS, Abid M. Synthesis and mechanistic studies of 4-aminoquinoline-Isatin molecular hybrids and Schiff's bases as promising antimicrobial agents. Eur J Med Chem 2024; 283:117127. [PMID: 39673862 DOI: 10.1016/j.ejmech.2024.117127] [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: 09/13/2024] [Revised: 11/09/2024] [Accepted: 11/22/2024] [Indexed: 12/16/2024]
Abstract
In this investigation, to determine their potential as specific antibacterial agents, Schiff's bases (LT-SB1-23 and SB1-SB12) and novel quinoline-isatin hybrids were subjected to microbiological testing. The in-vitro screening against bacterial strains (Escherichia coli, Enterococcus faecalis, Bacillus subtilis, Staphylococcus aureus, Pseudomonas aeruginosa, and Salmonella typhi) exhibited their antibacterial potential with many of the compounds showing inhibition range of 90-100 % at 200 μg/mL, against most of the tested strains. The MIC values of some of the compounds showed good antibacterial efficacy with values ranging from 32 to 128 μg/mL. Their bacterial growth inhibitory potential was further supported by disk diffusion and growth curve assays. Interestingly, one of the Schiff's bases (LT-SB7) displayed strong synergistic activity against E. coli and S. typhi with 16-64 folds reduction in MIC values. Additionally, it exhibited up to 85 % suppression of biofilm at ½MIC against AA209 environmental bacterial isolate and reduced the development of multidrug-resistant bacterial isolates. Promising compound LT-SB7 underwent 100 ns molecular dynamics simulations with biofilm-causing protein (PDB ID: 7C7U) to assess conformational changes and complex stability. Overall, this study identified compounds as effective antibacterial alternatives for the future.
Collapse
Affiliation(s)
- Mohd Shakir
- Medicinal Chemistry Laboratory, Department of Biosciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India; Department of Chemistry, University of Delhi, Delhi, 110007, India
| | - Asghar Ali
- Medicinal Chemistry Laboratory, Department of Biosciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India; Clinical Biochemistry Lab, Department of Biochemistry, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, 110062, India
| | - Swati Lakshmi
- Medicinal Chemistry Laboratory, Department of Biosciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India
| | - Manika Garg
- Clinical Biochemistry Lab, Department of Biochemistry, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, 110062, India
| | - Haider Thaer Abdulhameed Almuqdadi
- Medicinal Chemistry Laboratory, Department of Biosciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India; Department of Chemistry, College of Science, Al-Nahrain University, Baghdad, Iraq
| | - Iram Irfan
- Medicinal Chemistry Laboratory, Department of Biosciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India
| | - Mohan Kamthan
- Clinical Biochemistry Lab, Department of Biochemistry, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, 110062, India
| | - Mukesh C Joshi
- Department of Chemistry, Kirori Mal College, University of Delhi, Delhi, 110007, India
| | - Saleem Javed
- Department of Chemistry, Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India
| | - Diwan S Rawat
- Department of Chemistry, University of Delhi, Delhi, 110007, India.
| | - Mohammad Abid
- Medicinal Chemistry Laboratory, Department of Biosciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India.
| |
Collapse
|
12
|
Kotzampasi DM, Papadourakis M, Burke JE, Cournia Z. Free energy landscape of the PI3Kα C-terminal activation. Comput Struct Biotechnol J 2024; 23:3118-3131. [PMID: 39229338 PMCID: PMC11369385 DOI: 10.1016/j.csbj.2024.07.010] [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: 02/27/2024] [Revised: 07/05/2024] [Accepted: 07/05/2024] [Indexed: 09/05/2024] Open
Abstract
The gene PIK3CA, encoding the catalytic subunit p110α of PI3Kα, is the second most frequently mutated gene in cancer, with the highest frequency oncogenic mutants occurring in the C-terminus of the kinase domain. The C-terminus has a dual function in regulating the kinase, playing a putative auto-inhibitory role for kinase activity and being absolutely essential for binding to the cell membrane. However, the molecular mechanisms by which these C-terminal oncogenic mutations cause PI3Kα overactivation remain unclear. To understand how a spectrum of C-terminal mutations of PI3Kα alter kinase activity compared to the WT, we perform unbiased and biased Molecular Dynamics simulations of several C-terminal mutants and report the free energy landscapes for the C-terminal "closed-to-open" transition in the WT, H1047R, G1049R, M1043L and N1068KLKR mutants. Results are consistent with HDX-MS experimental data and provide a molecular explanation why H1047R and G1049R reorient the C-terminus with a different mechanism compared to the WT and M1043L and N1068KLKR mutants. Moreover, we show that in the H1047R mutant, the cavity, where the allosteric ligands STX-478 and RLY-2608 bind, is more accessible contrary to the WT. This study provides insights into the molecular mechanisms underlying activation of oncogenic PI3Kα by C-terminal mutations and represents a valuable resource for continued efforts in the development of mutant selective inhibitors as therapeutics.
Collapse
Affiliation(s)
- Danai Maria Kotzampasi
- Biomedical Research Foundation, Academy of Athens, Athens 11527, Greece
- Department of Biology, University of Crete, Heraklion 71500, Greece
| | | | - John E. Burke
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC V8W 2Y2, Canada
- Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Zoe Cournia
- Biomedical Research Foundation, Academy of Athens, Athens 11527, Greece
| |
Collapse
|
13
|
Del Rio Flores A, Zhai R, Kastner DW, Seshadri K, Yang S, De Matias K, Shen Y, Cai W, Narayanamoorthy M, Do NB, Xue Z, Marzooqi DA, Kulik HJ, Zhang W. Enzymatic synthesis of azide by a promiscuous N-nitrosylase. Nat Chem 2024; 16:2066-2075. [PMID: 39333393 PMCID: PMC11611683 DOI: 10.1038/s41557-024-01646-2] [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: 07/18/2023] [Accepted: 08/29/2024] [Indexed: 09/29/2024]
Abstract
Azides are energy-rich compounds with diverse representation in a broad range of scientific disciplines, including material science, synthetic chemistry, pharmaceutical science and chemical biology. Despite ubiquitous usage of the azido group, the underlying biosynthetic pathways for its formation remain largely unknown. Here we report the characterization of an enzymatic route for de novo azide construction. We demonstrate that Tri17, a promiscuous ATP- and nitrite-dependent enzyme, catalyses organic azide synthesis through sequential N-nitrosation and dehydration of aryl hydrazines. Through biochemical, structural and computational analyses, we further propose a plausible molecular mechanism for azide synthesis that sets the stage for future biocatalytic applications and biosynthetic pathway engineering.
Collapse
Affiliation(s)
- Antonio Del Rio Flores
- Department of Chemical and Biomolecular Engineering, University of California Berkeley, Berkeley, CA, USA
| | - Rui Zhai
- Department of Chemical and Biomolecular Engineering, University of California Berkeley, Berkeley, CA, USA
| | - David W Kastner
- Department of Bioengineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Kaushik Seshadri
- Department of Chemical and Biomolecular Engineering, University of California Berkeley, Berkeley, CA, USA
| | - Siyue Yang
- Department of Chemistry, University of California Berkeley, Berkeley, CA, USA
| | - Kyle De Matias
- Department of Chemical and Biomolecular Engineering, University of California Berkeley, Berkeley, CA, USA
| | - Yuanbo Shen
- Department of Chemistry, University of California Berkeley, Berkeley, CA, USA
| | - Wenlong Cai
- Department of Chemical and Biomolecular Engineering, University of California Berkeley, Berkeley, CA, USA
| | | | - Nicholas B Do
- Department of Chemical and Biomolecular Engineering, University of California Berkeley, Berkeley, CA, USA
| | - Zhaoqiang Xue
- Department of Chemical and Biomolecular Engineering, University of California Berkeley, Berkeley, CA, USA
| | - Dunya Al Marzooqi
- Department of Chemical and Biomolecular Engineering, University of California Berkeley, Berkeley, CA, USA
| | - Heather J Kulik
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - Wenjun Zhang
- Department of Chemical and Biomolecular Engineering, University of California Berkeley, Berkeley, CA, USA.
| |
Collapse
|
14
|
Kifayat S, Almuqdadi HTA, Singh RP, Singh M, Abid M, Sanapalli BKR. An in silico approach for identification of lead compound as FtsZ inhibitor. Mol Divers 2024; 28:3937-3948. [PMID: 38775995 DOI: 10.1007/s11030-023-10787-4] [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: 10/28/2023] [Accepted: 11/30/2023] [Indexed: 12/21/2024]
Abstract
The remarkable conservation of the FtsZ among Gram-positive and Gram-negative bacteria, a crucial GTPase in bacterial cell division, has emerged as a promising antibacterial drug target to combat antibacterial resistance. There have been several coordinated efforts to develop inhibitors against FtsZ which can also serve as potential candidates for future antibiotics. In the present study, a natural product-like library (≈50,000 compounds) was employed to conduct HTVS against Staphylococcus aureus FtsZ protein (PDB Id: 6KVP). Additionally, molecular docking was carried out in two modes, SP and XP docking, using the Schrödinger suite. The glide scores of ligands obtained by XP docking were further summarized and compared with the control ligands (ZI1- co-crystal and PC190723-a compound undergoing clinical trial). Using the Prime-MM-GBSA approach, BFE calculations were performed on the top XP-scored ligands (≈598 compounds). These hits were also evaluated for ADMET parameters using the Qikprop algorithm, SwissADME, and in silico carcinogenicity testing using Carcinopred-El. Based on the results, ligand 4-FtsZ complex was considered for the 300 ns MDS analysis to get insights into its binding modes within the catalytic pocket of FtsZ protein. The analysis revealed that the amide linkage sandwiched between the triazole and 1-oxa-8-azaspirodecan-8-ium moiety (Val203) as well as the aminoethyl group present at 1st position on the triazole moiety (Leu209, Leu200, Asp210, and Ala202) were responsible for the FtsZ inhibitory activity, owing to their crucial interactions with key amino acid residues. Further, the complex also displayed good protein-ligand stability, ultimately predicting ligand 4 as a potent lead compound for the inhibition of FtsZ. Thus, our in silico findings will serve as a framework for in-depth in-vitro and in-vivo investigations encouraging the development of FtsZ inhibitors as a new generation of antibacterial agents.
Collapse
Affiliation(s)
- Sumaiya Kifayat
- Department of Pharmacology, NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, 303121, India
| | | | - Ravindra Pal Singh
- Department of Pharmaceutics, NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, 303121, India
| | - Mithilesh Singh
- Department of Pharmaceutical Chemistry, NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, 303121, India
| | - Mohammad Abid
- Medicinal Chemistry Laboratory, Department of Biosciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India.
| | - Bharat Kumar Reddy Sanapalli
- Department of Pharmacology, NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, 303121, India.
- Department of Pharmacology, School of Pharmacy and Technology Management, SVKM's Narsee Monjee Institute of Management Studies (NMIMS) Deemed-to-Be-University, Jadcherla, Hyderabad, 509301, India.
| |
Collapse
|
15
|
Fernandes Silva S, Hollunder Klippel A, Sigurdardóttir S, Mahdizadeh SJ, Tiukova I, Bourgard C, Salazar-Alvarez LC, do Amaral Prado HM, de Araujo RV, Costa FTM, Bilsland E, King RD, Brauer Massirer K, Eriksson LA, Bengtson MH, Zanelli CF, Sunnerhagen P. An experimental target-based platform in yeast for screening Plasmodium vivax deoxyhypusine synthase inhibitors. PLoS Negl Trop Dis 2024; 18:e0012690. [PMID: 39621767 PMCID: PMC11637365 DOI: 10.1371/journal.pntd.0012690] [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: 06/10/2024] [Revised: 12/12/2024] [Accepted: 11/12/2024] [Indexed: 12/14/2024] Open
Abstract
The enzyme deoxyhypusine synthase (DHS) catalyzes the first step in the post-translational modification of the eukaryotic translation factor 5A (eIF5A). This is the only protein known to contain the amino acid hypusine, which results from this modification. Both eIF5A and DHS are essential for cell viability in eukaryotes, and inhibiting DHS is a promising strategy to develop new therapeutic alternatives. DHS proteins from many are sufficiently different from their human orthologs for selective targeting against infectious diseases; however, no DHS inhibitor selective for parasite orthologs has previously been reported. Here, we established a yeast surrogate genetics platform to identify inhibitors of DHS from Plasmodium vivax, one of the major causative agents of malaria. We constructed genetically modified Saccharomyces cerevisiae strains expressing DHS genes from Homo sapiens (HsDHS) or P. vivax (PvDHS) in place of the endogenous DHS gene from S. cerevisiae. Compared with a HsDHS complemented strain with a different genetic background that we previously generated, this new strain background was ~60-fold more sensitive to an inhibitor of human DHS. Initially, a virtual screen using the ChEMBL-NTD database was performed. Candidate ligands were tested in growth assays using the newly generated yeast strains expressing heterologous DHS genes. Among these, two showed promise by preferentially reducing the growth of the PvDHS-expressing strain. Further, in a robotized assay, we screened 400 compounds from the Pathogen Box library using the same S. cerevisiae strains, and one compound preferentially reduced the growth of the PvDHS-expressing yeast strain. Western blot revealed that these compounds significantly reduced eIF5A hypusination in yeast. The compounds showed antiplasmodial activity in the asexual erythrocyte stage; EC50 in high nM to low μM range, and low cytotoxicity. Our study demonstrates that this yeast-based platform is suitable for identifying and verifying candidate small molecule DHS inhibitors, selective for the parasite over the human ortholog.
Collapse
Affiliation(s)
- Suélen Fernandes Silva
- Department of Chemistry and Molecular Biology, University of Gothenburg, Göteborg, Sweden
- Chemistry Institute, São Paulo State University - UNESP, Araraquara, São Paulo, Brazil
- Center for Medicinal Chemistry - CQMED, Center for Molecular Biology and Genetic Engineering - CBMEG, Universidade Estadual de Campinas, Campinas, São Paulo, Brazil
| | - Angélica Hollunder Klippel
- Center for Medicinal Chemistry - CQMED, Center for Molecular Biology and Genetic Engineering - CBMEG, Universidade Estadual de Campinas, Campinas, São Paulo, Brazil
- School of Pharmaceutical Sciences, São Paulo State University—UNESP, Araraquara, São Paulo, Brazil
| | - Sunniva Sigurdardóttir
- Department of Chemistry and Molecular Biology, University of Gothenburg, Göteborg, Sweden
| | | | | | - Catarina Bourgard
- Department of Chemistry and Molecular Biology, University of Gothenburg, Göteborg, Sweden
- Laboratory of Tropical Diseases, Institute of Biology, Universidade Estadual de Campinas - UNICAMP, Campinas, São Paulo, Brazil
| | - Luis Carlos Salazar-Alvarez
- Laboratory of Tropical Diseases, Institute of Biology, Universidade Estadual de Campinas - UNICAMP, Campinas, São Paulo, Brazil
| | - Heloísa Monteiro do Amaral Prado
- Center for Medicinal Chemistry - CQMED, Center for Molecular Biology and Genetic Engineering - CBMEG, Universidade Estadual de Campinas, Campinas, São Paulo, Brazil
| | - Renan Vinicius de Araujo
- Center for Medicinal Chemistry - CQMED, Center for Molecular Biology and Genetic Engineering - CBMEG, Universidade Estadual de Campinas, Campinas, São Paulo, Brazil
| | - Fabio Trindade Maranhão Costa
- Laboratory of Tropical Diseases, Institute of Biology, Universidade Estadual de Campinas - UNICAMP, Campinas, São Paulo, Brazil
| | - Elizabeth Bilsland
- Department of Structural and Functional Biology, Institute of Biology, Universidade Estadual de Campinas - UNICAMP, Campinas, São Paulo, Brazil
| | - Ross D. King
- Department of Life Sciences, Chalmers, Göteborg, Sweden
| | - Katlin Brauer Massirer
- Center for Medicinal Chemistry - CQMED, Center for Molecular Biology and Genetic Engineering - CBMEG, Universidade Estadual de Campinas, Campinas, São Paulo, Brazil
| | - Leif A. Eriksson
- Department of Chemistry and Molecular Biology, University of Gothenburg, Göteborg, Sweden
| | - Mário Henrique Bengtson
- Center for Medicinal Chemistry - CQMED, Center for Molecular Biology and Genetic Engineering - CBMEG, Universidade Estadual de Campinas, Campinas, São Paulo, Brazil
- Department of Biochemistry and Tissue Biology, Institute of Biology, Universidade Estadual de Campinas, Campinas, São Paulo, Brazil
| | - Cleslei Fernando Zanelli
- Chemistry Institute, São Paulo State University - UNESP, Araraquara, São Paulo, Brazil
- School of Pharmaceutical Sciences, São Paulo State University—UNESP, Araraquara, São Paulo, Brazil
| | - Per Sunnerhagen
- Department of Chemistry and Molecular Biology, University of Gothenburg, Göteborg, Sweden
| |
Collapse
|
16
|
Hunt HL, Goncalves BG, Biggs MA, Rico MI, Murray ME, Lebedenko CG, Banerjee IA. Design and investigation of interactions of novel peptide conjugates of purine and pyrimidine derivatives with EGFR and its mutant T790M/L858R: an in silico and laboratory study. Mol Divers 2024; 28:3683-3711. [PMID: 38240950 DOI: 10.1007/s11030-023-10772-x] [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: 07/17/2023] [Accepted: 11/17/2023] [Indexed: 12/21/2024]
Abstract
Peptide-based therapeutics have been gaining attention due to their ability to actively target tumor cells. Additionally, several varieties of nucleotide derivatives have been developed to reduce cell proliferation and induce apoptosis of tumor cells. In this work, we have developed novel peptide conjugates with newly designed purine analogs and pyrimidine derivatives and explored the binding interactions with the kinase domain of wild-type EGFR and its mutant EGFR [L858R/ T790M] which are known to be over-expressed in tumor cells. The peptides explored included WNWKV (derived from sea cucumber) and LARFFS, which in previous work was predicted to bind to Domain I of EGFR. Computational studies conducted to explore binding interactions include molecular docking studies, molecular dynamics simulations and MMGBSA to investigate the binding abilities and stability of the complexes. The results indicate that conjugation enhanced binding capabilities, particularly for the WNWKV conjugates. MMGBSA analysis revealed nearly twofold higher binding toward the T790M/L858R double mutant receptor. Several conjugates were shown to have strong and stable binding with both wild-type and mutant EGFR. As a proof of concept, we synthesized pyrimidine conjugates with both peptides and determined the KD values using SPR analysis. The results corroborated with the computational analyses. Additionally, cell viability and apoptosis studies with lung cancer cells expressing the wild-type and double mutant proteins revealed that the WNWKV conjugate showed greater potency than the LARFFS conjugate, while LARFFS peptide alone showed poor binding to the kinase domain. Thus, we have designed peptide conjugates that show potential for further laboratory studies for developing therapeutics for targeting the EGFR receptor and its mutant T790M/L858R.
Collapse
Affiliation(s)
- Hannah L Hunt
- Department of Chemistry, Fordham University, 441 East Fordham Road, Bronx, NY, 10458, USA
| | - Beatriz G Goncalves
- Department of Chemistry, Fordham University, 441 East Fordham Road, Bronx, NY, 10458, USA
| | - Mary A Biggs
- Department of Chemistry, Fordham University, 441 East Fordham Road, Bronx, NY, 10458, USA
| | - Mia I Rico
- Department of Chemistry, Fordham University, 441 East Fordham Road, Bronx, NY, 10458, USA
| | - Molly E Murray
- Department of Chemistry, Fordham University, 441 East Fordham Road, Bronx, NY, 10458, USA
| | - Charlotta G Lebedenko
- Department of Chemistry, Fordham University, 441 East Fordham Road, Bronx, NY, 10458, USA
| | - Ipsita A Banerjee
- Department of Chemistry, Fordham University, 441 East Fordham Road, Bronx, NY, 10458, USA.
| |
Collapse
|
17
|
Kumar S, Ali I, Abbas F, Shafiq F, Yadav AK, Ghate MD, Kumar D. In-silico identification and exploration of small molecule coumarin-1,2,3-triazole hybrids as potential EGFR inhibitors for targeting lung cancer. Mol Divers 2024; 28:4301-4324. [PMID: 38470555 DOI: 10.1007/s11030-024-10817-9] [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: 08/14/2023] [Accepted: 01/25/2024] [Indexed: 03/14/2024]
Abstract
Globally, lung cancer is a significant public health concern due to its role as the leading cause of cancer-related mortalities. The promising target of EGFR for lung cancer treatment has been identified, providing a potential avenue for more effective therapies. The purpose of the study was to design a library of 1843 coumarin-1,2,3-triazole hybrids and screen them based on a designed pharmacophore to identify potential inhibitors targeting EGFR in lung cancer with minimum or no side effects. Pharmacophore-based screening was carried out and 60 hits were obtained. To gain a better understanding of the binding interactions between the compounds and the targeted receptor, molecular docking was conducted on the 60 screened compounds. In-silico ADME and toxicity studies were also conducted to assess the drug-likeness and safety of the identified compounds. The results indicated that coumarin-1,2,3-triazole hybrids COUM-0849, COUM-0935, COUM-0414, COUM-1335, COUM-0276, and COUM-0484 exhibit dock score of - 10.2, - 10.2, - 10.1, - 10.1, - 10, - 10 while reference molecule - 7.9 kcal/mol for EGFR (PDB ID: 4HJO) respectively. The molecular docking and molecular dynamics simulations revealed that the identified compounds formed stable interactions with the active site of EGFR, indicating their potential as inhibitors. The in-silico ADME and toxicity studies showed that the compounds had favorable drug-likeness properties and low toxicity, further supporting their potential as therapeutic agents. Finally, we performed DFT studies on the best-selected ligands to gain further insights into their electronic properties. The findings of this study provide important insights into the potential of coumarin-1,2,3-triazole hybrids as promising EGFR inhibitors for the management of lung cancer.
Collapse
Affiliation(s)
- Sunil Kumar
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Shoolini University, Solan, Himachal Pradesh, 173229, India
| | - Iqra Ali
- Department of Biosciences, COMSATS University Islamabad, Islamabad Campus, Islamabad, 45550, Pakistan
| | - Faheem Abbas
- Key Lab of Organic Optoelectronics and Molecular Engineering of Ministry of Education, Department of Chemistry, Tsinghua University, Beijing, 100084, People's Republic of China
| | - Faiza Shafiq
- Department of Chemistry, University of Agriculture, Faisalabad, 38040, Pakistan
| | - Ashok Kumar Yadav
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, 160014, India
| | - Manjunath D Ghate
- School of Pharmacy, National Forensic Sciences University, Gandhinagar, Gujarat, 382007, India
| | - Deepak Kumar
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Shoolini University, Solan, Himachal Pradesh, 173229, India.
| |
Collapse
|
18
|
Eldehna WM, Fares M, Bonardi A, Avgenikos M, Baselious F, Schmidt M, Al-Warhi T, Abdel-Aziz HA, Rennert R, Peat TS, Supuran CT, Wessjohann LA, Ibrahim HS. 4-(Pyrazolyl)benzenesulfonamide Ureas as Carbonic Anhydrases Inhibitors and Hypoxia-Mediated Chemo-Sensitizing Agents in Colorectal Cancer Cells. J Med Chem 2024; 67:20438-20454. [PMID: 39550697 PMCID: PMC11613454 DOI: 10.1021/acs.jmedchem.4c01894] [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: 08/12/2024] [Revised: 11/03/2024] [Accepted: 11/06/2024] [Indexed: 11/18/2024]
Abstract
Hypoxia in tumors contributes to chemotherapy resistance, worsened by acidosis driven by carbonic anhydrases (hCA IX and XII). Targeting these enzymes can mitigate acidosis, thus enhancing tumor sensitivity to cytotoxic drugs. Herein, novel 4-(pyrazolyl)benzenesulfonamide ureas (SH7a-t) were developed and evaluated for their inhibitory activity against hCA IX and XII. They showed promising results (hCA IX: KI = 15.9-67.6 nM, hCA XII: KI = 16.7-65.7 nM). Particularly, SH7s demonstrated outstanding activity (KIs = 15.9 nM for hCA IX and 55.2 nM for hCA XII) and minimal off-target kinase inhibition over a panel of 258 kinases. In NCI anticancer screening, SH7s exhibited broad-spectrum activity with an effective growth inhibition full panel GI50 (MG-MID) value of 3.5 μM and a subpanel GI50 (MG-MID) range of 2.4-6.3 μM. Furthermore, SH7s enhanced the efficacy of Taxol and 5-fluorouracil in cotreatment regimens under hypoxic conditions in HCT-116 colorectal cancer cells, indicating its potential as a promising anticancer agent.
Collapse
Affiliation(s)
- Wagdy M. Eldehna
- Department
of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh 33516, Egypt
- Department
of Pharmaceutical Chemistry, Faculty of Pharmacy, Pharos University in Alexandria, Canal El Mahmoudia Street, Alexandria 21648, Egypt
| | - Mohamed Fares
- Department
of Pharmaceutical Chemistry, Faculty of Pharmacy, Egyptian Russian University, Badr
City, Cairo 11829, Egypt
- Sydney
Pharmacy School, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Alessandro Bonardi
- Department
NEUROFARBA—Pharmaceutical and Nutraceutical Section, University of Firenze, via Ugo Schiff 6, Sesto
Fiorentino I-50019, Firenze, Italy
| | - Moscos Avgenikos
- Department
of Bioorganic Chemistry, Leibniz Institute
of Plant Biochemistry, Halle
(Saale) D-06120, Germany
| | - Fady Baselious
- Department
of Medicinal Chemistry, Institute of Pharmacy, Martin-Luther-University of Halle-Wittenberg, Halle (Saale) D-06120, Germany
| | - Matthias Schmidt
- Department
of Medicinal Chemistry, Institute of Pharmacy, Martin-Luther-University of Halle-Wittenberg, Halle (Saale) D-06120, Germany
| | - Tarfah Al-Warhi
- Department
of Chemistry, College of Science, Princess
Nourah Bint Abdulrahman University, Riyadh 11564, Saudi Arabia
| | - Hatem A. Abdel-Aziz
- Applied
Organic Chemistry Department, National Research
Center, Dokki, Giza 12622, Cairo, Egypt
| | - Robert Rennert
- Department
of Bioorganic Chemistry, Leibniz Institute
of Plant Biochemistry, Halle
(Saale) D-06120, Germany
| | - Thomas S. Peat
- School
of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Claudiu T. Supuran
- Department
NEUROFARBA—Pharmaceutical and Nutraceutical Section, University of Firenze, via Ugo Schiff 6, Sesto
Fiorentino I-50019, Firenze, Italy
| | - Ludger A. Wessjohann
- Department
of Bioorganic Chemistry, Leibniz Institute
of Plant Biochemistry, Halle
(Saale) D-06120, Germany
| | - Hany S. Ibrahim
- Department
of Pharmaceutical Chemistry, Faculty of Pharmacy, Egyptian Russian University, Badr
City, Cairo 11829, Egypt
- Department
of Bioorganic Chemistry, Leibniz Institute
of Plant Biochemistry, Halle
(Saale) D-06120, Germany
- Department
of Medicinal Chemistry, Institute of Pharmacy, Martin-Luther-University of Halle-Wittenberg, Halle (Saale) D-06120, Germany
| |
Collapse
|
19
|
Kalath H, Vishwakarma R, Banjan B, Ramakrishnan K, Koshy AJ, Raju R, Rehman N, Revikumar A. In-silico studies on evaluating the liver-protective effectiveness of a polyherbal formulation in preventing hepatocellular carcinoma progression. In Silico Pharmacol 2024; 12:109. [PMID: 39569037 PMCID: PMC11574239 DOI: 10.1007/s40203-024-00285-2] [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: 09/02/2024] [Accepted: 11/01/2024] [Indexed: 11/22/2024] Open
Abstract
Liv-52, an herbal formulation consisting of seven distinct plants and Mandur Bhasma, is recognized for its hepatoprotective, anti-inflammatory, and antioxidant properties. To investigate the pharmacological potential of each phytochemical from these plants, we conducted ADMET analysis, molecular docking, and molecular dynamic simulations to identify potent molecules capable of inhibiting the interaction between Alpha-fetoprotein (AFP) and Cysteine aspartyl protease 3 (Caspase-3/CASP3). In our study, we have used molecular docking of all the compounds against AFP and filtered them on the basis of ADME properties. Among the compounds analyzed, (-) Syringaresinol from Solanum nigrum, exhibited good binding interactions with AFP, the highest binding free energy, and maintained stability throughout the simulation along with favorable drug likeness properties based on ADME and Toxicity analysis. These findings have strongly indicated that (-) Syringaresinol is a potential inhibitor of AFP, providing a promising therapeutic avenue for hepatocellular carcinoma (HCC) treatment by inhibiting the interaction between AFP and CASP3, thereby reinstating normal CASP3 activity. Further in vitro studies are imperative to validate the therapeutic efficacy of (-) Syringaresinol as an AFP inhibitor, potentially impeding the progression of HCC. Supplementary Information The online version contains supplementary material available at 10.1007/s40203-024-00285-2.
Collapse
Affiliation(s)
- Haritha Kalath
- Centre for Integrative Omics Data Science (CIODS), Yenepoya (Deemed to be University), Mangalore, 575018 Karnataka India
| | - Riya Vishwakarma
- Centre for Integrative Omics Data Science (CIODS), Yenepoya (Deemed to be University), Mangalore, 575018 Karnataka India
| | - Bhavya Banjan
- Centre for Integrative Omics Data Science (CIODS), Yenepoya (Deemed to be University), Mangalore, 575018 Karnataka India
| | - Krishnapriya Ramakrishnan
- Centre for Integrative Omics Data Science (CIODS), Yenepoya (Deemed to be University), Mangalore, 575018 Karnataka India
| | - Abel John Koshy
- Centre for Integrative Omics Data Science (CIODS), Yenepoya (Deemed to be University), Mangalore, 575018 Karnataka India
| | - Rajesh Raju
- Centre for Integrative Omics Data Science (CIODS), Yenepoya (Deemed to be University), Mangalore, 575018 Karnataka India
| | - Niyas Rehman
- Centre for Integrative Omics Data Science (CIODS), Yenepoya (Deemed to be University), Mangalore, 575018 Karnataka India
| | - Amjesh Revikumar
- Centre for Integrative Omics Data Science (CIODS), Yenepoya (Deemed to be University), Mangalore, 575018 Karnataka India
- Kerala Genome Data Centre, Kerala Development and Innovation Strategic Council, Vazhuthacaud, Thiruvananthapuram, 695014 Kerala India
| |
Collapse
|
20
|
Palol VV, Waidha K, Moovarkumudalvan B, Valavath Baburajan N, Saravanan SK, Lakshmanan D, Subramanyam V, Chinnadurai RK. β-1,3-glucan from Euglena gracilis: a promising epidrug targeting epigenetic regulators PRMTs and SIRTs for therapeutic applications in ovarian cancer. J Biomol Struct Dyn 2024:1-16. [PMID: 39535161 DOI: 10.1080/07391102.2024.2425832] [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: 09/20/2023] [Accepted: 04/23/2024] [Indexed: 11/16/2024]
Abstract
Natural products serve as a valuable resource in drug discovery and the identification of bioactive molecules in the field of epimedicine, which targets epigenetic regulator enzymes through epidrugs. In this study, β-1,3-glucan (BG), a natural storage polysaccharide in Euglena gracilis, a well-known immunostimulatory agent, is propounded as a promising epidrug. To elucidate the therapeutic efficacy of BG against ovarian cancer, the molecular interactions between BG and epigenetic regulators, Protein Arginine Methyltransferases (PRMTs) and Sirtuins (SIRTs) were investigated using computational methods followed by in vitro gene expression studies in SKOV-3 ovarian cancer cell line. The binding energies of PRMT5 and SIRT5 against BG were observed as -65.5 and -68.2 kcal/mol, respectively. The in vitro cytotoxic effects of BG against human ovarian cancer cell line, SKOV-3 showed an IC50 of 150 µg/mL at 48 h. Significant epigenetic modifications were observed to be influenced by BG which increased the gene expression of PRMT5, SIRT5 and Nrf2 to 0.3, 0.5, and 0.7 fold-change respectively, while the Nrf1/2 plasmid showed reduced reporter activity by 29%. Collectively, both in silico and in vitro studies provided valuable insights into the epigenetic regulation of PRMT5 and SIRT5 by BG via Nrf1/2. Nonetheless, further preclinical and clinical investigations are essential to validate the therapeutic properties of BG as an epidrug against ovarian cancer.
Collapse
Affiliation(s)
- Varsha Virendra Palol
- Mahatma Gandhi Medical Advanced Research Institute (MGMARI), Sri Balaji Vidyapeeth (Deemed-to-be University), Pillayarkuppam, Puducherry, India
| | - Kamran Waidha
- The Shraga Segal Department of Microbiology, Immunology & Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Balasubramanian Moovarkumudalvan
- Mahatma Gandhi Medical Advanced Research Institute (MGMARI), Sri Balaji Vidyapeeth (Deemed-to-be University), Pillayarkuppam, Puducherry, India
| | | | - Suresh Kumar Saravanan
- Mahatma Gandhi Medical Preclinical Research Centre (MGMPRC), Sri Balaji Vidyapeeth (Deemed-to-be University), Pillayarkuppam, Puducherry, India
| | - Divya Lakshmanan
- Mahatma Gandhi Medical Advanced Research Institute (MGMARI), Sri Balaji Vidyapeeth (Deemed-to-be University), Pillayarkuppam, Puducherry, India
| | - Veni Subramanyam
- Mahatma Gandhi Medical Advanced Research Institute (MGMARI), Sri Balaji Vidyapeeth (Deemed-to-be University), Pillayarkuppam, Puducherry, India
| | - Raj Kumar Chinnadurai
- Mahatma Gandhi Medical Advanced Research Institute (MGMARI), Sri Balaji Vidyapeeth (Deemed-to-be University), Pillayarkuppam, Puducherry, India
| |
Collapse
|
21
|
Oluwafemi KA, Jimoh RB, Omoboyowa DA, Olonisakin A, Adeforiti AF, Iqbal N. Investigating the effect of 1, 2-Dibenzoylhydrazine on Staphylococcus aureus using integrated computational approaches. In Silico Pharmacol 2024; 12:102. [PMID: 39524456 PMCID: PMC11549268 DOI: 10.1007/s40203-024-00278-1] [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: 08/14/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Staphylococcus aureus, a notorious member of the ESKAPE pathogens, poses significant public health challenges due to its virulence and multidrug-resistant nature, particularly in methicillin-resistant S. aureus (MRSA) strains. With the increasing threat of antibiotic resistance, there is an urgent need to develop novel antibiotic agents. This study therefore aims to explore the antibacterial potential of 1,2-dibenzoylhydrazine (DBH) as a scaffold against S. aureus drug target enzymes, using integrated computational approaches. The study utilized molecular docking, lead optimization, and structure-based virtual screening techniques to evaluate the binding affinities of DBH and its derivatives against various S. aureus enzymes. Prime/MM-GBSA calculations were performed to validate the binding affinities obtained, and molecular dynamics (MD) simulations were conducted to assess the stability of the DBHs-enzyme complexes. Results indicated that, out of twenty enzymes from S. aureus examined against DBH, carotenoid dehydrosqualene synthase was predicted as a suitable target enzyme for DBH, showing a binding affinity of -8.027 kcal/mol. A lead optimization operation of the compound generated 27 DBH derivatives out of which four exhibited enhanced binding affinities compared to both DBH and a standard antibiotic, ofloxacin. The QSAR model predicted that, DBH and molecule_D_1 have higher PIC50 of 4.779 µM compared with the standard drug (ofloxacin = 4.678 µM). MD simulations confirmed the stability of the top-scoring derivatives within the enzyme's binding pocket, with RMSD and RMSF analyses supporting their potential as inhibitors of the enzyme. In conclusion, this study has predicted the effect of DBH derivatives on S. aureus based on their in silico inhibitory capacity against the carotenoid dehydrosqualene synthase from the organism. Future work will seek to experimentally validate these findings against the suggested enzyme. Supplementary Information The online version contains supplementary material available at 10.1007/s40203-024-00278-1.
Collapse
Affiliation(s)
- Kola A. Oluwafemi
- Department of Chemical Sciences, Adekunle Ajasin University, Akungba-Akoko, Nigeria
| | - Rashidat B. Jimoh
- Department of Chemical Sciences, Adekunle Ajasin University, Akungba-Akoko, Nigeria
| | - Damilola A. Omoboyowa
- Phyto-medicine and Computational Biology Laboratory, Adekunle Ajasin University, Akungba-Akoko, Nigeria
- Department of Biochemistry, Adekunle Ajasin University, Akungba-Akoko, Nigeria
| | - Adebisi Olonisakin
- Department of Chemical Sciences, Adekunle Ajasin University, Akungba-Akoko, Nigeria
| | - Anthony F. Adeforiti
- Department of Chemical Sciences, Adekunle Ajasin University, Akungba-Akoko, Nigeria
| | - Naveed Iqbal
- Department of BioinformaticsInstitute of Biochemistry, Biotechnology and Bioinformatics, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| |
Collapse
|
22
|
Sanches VHDS, Lobato CC, Silva LB, dos Santos IVF, Barros EDS, Maciel ADA, Ferreira EFB, da Costa KS, Espejo-Román JM, Rosa JMC, Kimani NM, Santos CBR. Rational Approach to New Chemical Entities with Antiproliferative Activity on Ab1 Tyrosine Kinase Encoded by the BCR-ABL Gene: An Hierarchical Biochemoinformatics Analysis. Pharmaceuticals (Basel) 2024; 17:1491. [PMID: 39598402 PMCID: PMC11597596 DOI: 10.3390/ph17111491] [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: 08/30/2024] [Revised: 10/28/2024] [Accepted: 10/29/2024] [Indexed: 11/29/2024] Open
Abstract
Background: This study began with a search in three databases, totaling six libraries (ChemBridge-DIVERSet, ChemBridge-DIVERSet-EXP, Zinc_Drug Database, Zinc_Natural_Stock, Zinc_FDA_BindingDB, Maybridge) with approximately 2.5 million compounds with the aim of selecting potential inhibitors with antiproliferative activity on the chimeric tyrosine kinase encoded by the BCR-ABL gene. Methods: Through hierarchical biochemoinformatics, ADME/Tox analyses, biological activity prediction, molecular docking simulations, synthetic accessibility and theoretical synthetic routes of promising compounds and their lipophilicity and water solubility were realized. Results: Predictions of toxicological and pharmacokinetic properties (ADME/Tox) using the top100/base (600 structures), in comparison with the commercial drug imatinib, showed that only nine exhibited the desired properties. In the prediction of biological activity, the results of the nine selected structures ranged from 13.7% < Pa < 65.8%, showing them to be potential protein kinase inhibitors. In the molecular docking simulations, the promising molecules LMQC01 and LMQC04 showed significant values in molecular targeting (PDB 1IEP-resolution 2.10 Å). LMQC04 presented better binding affinity (∆G = -12.2 kcal mol-1 with a variation of ±3.6 kcal mol-1) in relation to LMQC01. The LMQC01 and LMQC04 molecules were advanced for molecular dynamics (MD) simulation followed by Molecular Mechanics with generalized Born and Surface Area solvation (MM-GBSA); the comparable, low and stable RMSD and ΔE values for the protein and ligand in each complex suggest that the selected compounds form a stable complex with the Abl kinase domain. This stability is a positive indicator that LMQC01 and LMQC04 can potentially inhibit enzyme function. Synthetic accessibility (SA) analysis performed on the AMBIT and SwissADME webservers showed that LMQC01 and LMQC04 can be considered easy to synthesize. Our in silico results show that these molecules could be potent protein kinase inhibitors with potential antiproliferative activity on tyrosine kinase encoded by the BCR-ABL gene. Conclusions: In conclusion, the results suggest that these ligands, particularly LMQC04, may bind strongly to the studied target and may have appropriate ADME/Tox properties in experimental studies. Considering future in vitro or in vivo assays, we elaborated the theoretical synthetic routes of the promising compounds identified in the present study. Based on our in silico findings, the selected ligands show promise for future studies in developing chronic myeloid leukemia treatments.
Collapse
Affiliation(s)
- Vitor H. da S. Sanches
- Biodiversity and Biotechnology Network of the Legal Amazon, Biotechnology Department, Federal University of Amapá, Macapá 68903-419, AP, Brazil; (V.H.d.S.S.); (C.C.L.); (L.B.S.); (I.V.F.d.S.)
- Graduate Program in Medicinal Chemistry and Molecular Modeling, Federal University of Pará, Belém 66075-110, PA, Brazil
- Laboratory of Modeling and Computational Chemistry, Department of Biological and Health Sciences, Federal University of Amapá, Macapá 68903-419, AP, Brazil; (E.d.S.B.); (A.d.A.M.); (E.F.B.F.)
| | - Cleison C. Lobato
- Biodiversity and Biotechnology Network of the Legal Amazon, Biotechnology Department, Federal University of Amapá, Macapá 68903-419, AP, Brazil; (V.H.d.S.S.); (C.C.L.); (L.B.S.); (I.V.F.d.S.)
- Graduate Program in Medicinal Chemistry and Molecular Modeling, Federal University of Pará, Belém 66075-110, PA, Brazil
- Laboratory of Modeling and Computational Chemistry, Department of Biological and Health Sciences, Federal University of Amapá, Macapá 68903-419, AP, Brazil; (E.d.S.B.); (A.d.A.M.); (E.F.B.F.)
| | - Luciane B. Silva
- Biodiversity and Biotechnology Network of the Legal Amazon, Biotechnology Department, Federal University of Amapá, Macapá 68903-419, AP, Brazil; (V.H.d.S.S.); (C.C.L.); (L.B.S.); (I.V.F.d.S.)
- Graduate Program in Medicinal Chemistry and Molecular Modeling, Federal University of Pará, Belém 66075-110, PA, Brazil
- Laboratory of Modeling and Computational Chemistry, Department of Biological and Health Sciences, Federal University of Amapá, Macapá 68903-419, AP, Brazil; (E.d.S.B.); (A.d.A.M.); (E.F.B.F.)
| | - Igor V. F. dos Santos
- Biodiversity and Biotechnology Network of the Legal Amazon, Biotechnology Department, Federal University of Amapá, Macapá 68903-419, AP, Brazil; (V.H.d.S.S.); (C.C.L.); (L.B.S.); (I.V.F.d.S.)
- Laboratory of Modeling and Computational Chemistry, Department of Biological and Health Sciences, Federal University of Amapá, Macapá 68903-419, AP, Brazil; (E.d.S.B.); (A.d.A.M.); (E.F.B.F.)
| | - Elcimar de S. Barros
- Laboratory of Modeling and Computational Chemistry, Department of Biological and Health Sciences, Federal University of Amapá, Macapá 68903-419, AP, Brazil; (E.d.S.B.); (A.d.A.M.); (E.F.B.F.)
- Graduate Program of Pharmaceutical Innovation, Federal University of Amapá, Macapá 68902-280, AP, Brazil
| | - Alexandre de A. Maciel
- Laboratory of Modeling and Computational Chemistry, Department of Biological and Health Sciences, Federal University of Amapá, Macapá 68903-419, AP, Brazil; (E.d.S.B.); (A.d.A.M.); (E.F.B.F.)
- Graduate Program of Pharmaceutical Innovation, Federal University of Amapá, Macapá 68902-280, AP, Brazil
| | - Elenilze F. B. Ferreira
- Laboratory of Modeling and Computational Chemistry, Department of Biological and Health Sciences, Federal University of Amapá, Macapá 68903-419, AP, Brazil; (E.d.S.B.); (A.d.A.M.); (E.F.B.F.)
- Graduate Program of Pharmaceutical Innovation, Federal University of Amapá, Macapá 68902-280, AP, Brazil
- Laboratory of Organic Chemistry and Biochemistry, University of the State of Amapá, Macapá 68900-070, AP, Brazil
| | - Kauê S. da Costa
- Computational Simulation Laboratory, Institute of Biodiversity, Federal University of Western Pará, Vera Paz Street, w/n Salé, Santarém 68040-255, PA, Brazil;
| | - José M. Espejo-Román
- Department of Pharmaceutical and Organic Chemistry, Faculty of Pharmacy, Campus of Cartuja, University of Granada, 18071 Granada, Spain; (J.M.E.-R.); (J.M.C.R.)
| | - Joaquín M. C. Rosa
- Department of Pharmaceutical and Organic Chemistry, Faculty of Pharmacy, Campus of Cartuja, University of Granada, 18071 Granada, Spain; (J.M.E.-R.); (J.M.C.R.)
| | - Njogu M. Kimani
- Natural Product Chemistry and Computational Drug Discovery Laboratory, Embu P.O. Box 6-60100, Kenya;
| | - Cleydson B. R. Santos
- Biodiversity and Biotechnology Network of the Legal Amazon, Biotechnology Department, Federal University of Amapá, Macapá 68903-419, AP, Brazil; (V.H.d.S.S.); (C.C.L.); (L.B.S.); (I.V.F.d.S.)
- Graduate Program in Medicinal Chemistry and Molecular Modeling, Federal University of Pará, Belém 66075-110, PA, Brazil
- Laboratory of Modeling and Computational Chemistry, Department of Biological and Health Sciences, Federal University of Amapá, Macapá 68903-419, AP, Brazil; (E.d.S.B.); (A.d.A.M.); (E.F.B.F.)
- Graduate Program of Pharmaceutical Innovation, Federal University of Amapá, Macapá 68902-280, AP, Brazil
| |
Collapse
|
23
|
Xu L, Xu Y, Wang G, Tu X, Xu J, Zheng H, Wang D, Su Y, Zhang XK, Zeng Z. Halogenated retinoid derivatives as dual RARα and RXRα modulators for treating acute promyelocytic leukemia cells. Eur J Med Chem 2024; 277:116779. [PMID: 39163777 DOI: 10.1016/j.ejmech.2024.116779] [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: 05/23/2024] [Revised: 08/06/2024] [Accepted: 08/12/2024] [Indexed: 08/22/2024]
Abstract
Acute promyelocytic leukemia (APL), a distinctive subtype of acute myeloid leukemia (AML), is characterized by the t(15; 17) translocation forming the PML-RARα fusion protein. Recent studies have revealed a crucial role of retinoid X receptor α (RXRα) in PML-RARα's tumorigenesis. This necessitates the development of dual RARα and RXRα targeting compounds for treating APL. Here, we developed a pair of brominated retinoid isomers, 5a and 5b, exhibiting RARα agonistic selectivity among the RAR subtypes and RXRα partial agonistic activities. In the treatment of APL cells, low doses (RARα activation range) of 5a and 5b degrade PML-RARα and strongly induce differentiation, while higher doses (RXRα activation range) induce G2/M arrest and apoptosis in both all-trans retinoic acid (ATRA)-sensitive and resistant cells. We replaced the bromine in 5a with chlorine or iodine to obtain compounds 7 or 8a. Interestingly, the chlorinated compound 7 tends to activate RXRα and induce G2/M arrest and apoptosis, while the iodinated compound 8a tends to activate RARα and induce differentiation. Together, our work underscores several advantages and characteristics of halogens in the rational design of RARα and RXRα ligands, offering three promising drug candidates for treating both ATRA-sensitive and resistant APL.
Collapse
Affiliation(s)
- Lin Xu
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, 361102, Fujian, China
| | - Yunqing Xu
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, 361102, Fujian, China
| | - Guijiang Wang
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, 361102, Fujian, China
| | - Xuhuang Tu
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, 361102, Fujian, China
| | - Jiale Xu
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, 361102, Fujian, China
| | - Hongzhi Zheng
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, 361102, Fujian, China
| | - Daohu Wang
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, 361102, Fujian, China
| | - Ying Su
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, 361102, Fujian, China; NucMito Pharmaceuticals Co., Ltd., Xiamen, 361000, China
| | - Xiao-Kun Zhang
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, 361102, Fujian, China.
| | - Zhiping Zeng
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, 361102, Fujian, China.
| |
Collapse
|
24
|
Aydın S, Ergün Y, Ghazy S, Çelebi A, Kilic T, Avşar T, Durdağı S. Synthesis of Tryptamine-Thiazolidin-4-one Derivatives and the Combined In Silico and In Vitro Evaluation of their Biological Activity and Cytotoxicity. ACS OMEGA 2024; 9:44262-44281. [PMID: 39524642 PMCID: PMC11541526 DOI: 10.1021/acsomega.4c04456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 08/22/2024] [Accepted: 09/03/2024] [Indexed: 11/16/2024]
Abstract
Tryptamine, a monoamine alkaloid with an indole ring structure, is derived from the decarboxylation of the amino acid tryptophan, which is present in fungi, plants, and animals. Tryptamine analogues hold significant therapeutic potential due to their broad pharmacological activities, including roles as neurotransmitters and potential therapeutic agents for various diseases. Structural modifications of tryptamine enhance receptor selectivity and metabolic stability, improving therapeutic efficacy. These modifications are crucial for optimizing pharmacokinetic and pharmacodynamic properties, making the analogues more effective and safer for clinical use. In this study, novel tryptamine-thiazolidin-4-one (YS1-12) derivatives were synthesized via a one-pot three-component condensation reaction. The synthesized compounds are characterized by different spectroscopy techniques such as FT-IR, 1H NMR, 13C NMR, and HR-NMS. The synthesized compounds were subjected to binary QSAR disease models for bioactivity prediction and a target prediction model for target analysis. Potential targets were identified, and physics-based molecular simulations were conducted. Additionally, MM/GBSA binding free energy analysis was performed to calculate the average binding free energies of YS1-12 compared to reference molecules. Our computational results indicated promising biological activities for these new compounds. To further investigate these activities, the compounds were tested in vitro using two different cancer cell lines: YKG-1 glioblastoma and SH-SY5Y neuroblastoma cells. The results confirmed the potential activities of these novel compounds. Notably, compounds YS4 and YS10 exhibited favorable activities compared to the control compounds 5-FU and Temozolomide. YS4 demonstrated an IC50 value of 20 nM against YKG-1 cells, while YS10 exhibited an IC50 value of 0.44 nM against SH-SY5Y cells.
Collapse
Affiliation(s)
- Seher Aydın
- Dokuz
Eylul University, The Graduate School
of Natural and Applied Sciences, Kaynaklar Campus, Buca, Izmir 35160, Türkiye
| | - Yavuz Ergün
- Dokuz
Eylul University, Faculty of Sciences,
Department of Chemistry, Kaynaklar Campus, Buca, Izmir 35160, Türkiye
| | - Salma Ghazy
- Computational
Biology and Molecular Simulations Laboratory, Department of Biophysics, School of Medicine, Bahçeşehir University, Istanbul 34353, Türkiye
- Lab
for Innovative Drugs (Lab4IND), Computational Drug Design Center (HİTMER), Bahçeşehir University, İstanbul 34353, Türkiye
| | - Asuman Çelebi
- Department
of Medical Biology, School of Medicine,
Bahcesehir University, Istanbul 34353, Türkiye
| | - Turker Kilic
- Department
of Neurosurgery, School of Medicine, Bahcesehir
University, Istanbul 34353, Türkiye
| | - Timuçin Avşar
- Department
of Medical Biology, School of Medicine,
Bahcesehir University, Istanbul 34353, Türkiye
| | - Serdar Durdağı
- Computational
Biology and Molecular Simulations Laboratory, Department of Biophysics, School of Medicine, Bahçeşehir University, Istanbul 34353, Türkiye
- Lab
for Innovative Drugs (Lab4IND), Computational Drug Design Center (HİTMER), Bahçeşehir University, İstanbul 34353, Türkiye
- Molecular
Therapy Lab, Department of Pharmaceutical Chemistry, School of Pharmacy, Bahçeşehir University, Istanbul 34353, Türkiye
| |
Collapse
|
25
|
Sharma S, Rana P, Chadha VD, Dhingra N, Kaur T. Exploring characteristic features for effective HCN1 channel inhibition using integrated analytical approaches: 3D QSAR, molecular docking, homology modelling, ADME and molecular dynamics. EUROPEAN BIOPHYSICS JOURNAL : EBJ 2024; 53:447-464. [PMID: 39488633 DOI: 10.1007/s00249-024-01726-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 09/14/2024] [Accepted: 10/03/2024] [Indexed: 11/04/2024]
Abstract
Neuropathic pain (NP) is characterized by hyperalgesia, allodynia, and spontaneous pain. Hyperpolarization-activated cyclic nucleotide-gated (HCN) channel involved in neuronal hyperexcitability, has emerged as an important target for the drug development of NP. HCN channels exist in four different isoforms, where HCN1 is majorly expressed in dorsal root ganglion having an imperative role in NP pathophysiology. A specific HCN1 channel inhibitor will hold the better potential to treat NP without disturbing the physiological roles of other HCN isoforms. The main objective is to identify and analyze the chemical properties of scaffolds with higher HCN1 channel specificity. The 3D-QSAR studies highlight the hydrophobic & hydrogen bond donor groups enhance specificity towards the HCN1 channel. Further, the molecular interaction of the scaffolds with the HCN1 pore was studied by generating an open-pore model of the HCN1 channel using homology modelling and then docking the molecules with it. In addition, the important residues involved in the interaction between HCN1 pore and scaffolds were also identified. Moreover, ADME predictions revealed that compounds had good oral bioavailability and solubility characteristics. Subsequently, molecular dynamics simulation studies revealed the better stability of the lead molecules A7 and A9 during interactions and ascertained them as potential drug candidates. Cumulative studies provided the important structural features for enhancing HCN1 channel-specific inhibition, paving the way to design and develop novel specific HCN1 channel inhibitors.
Collapse
Affiliation(s)
- Shiwani Sharma
- Department of Biophysics, Panjab University, Chandigarh, 160014, India
| | - Priyanka Rana
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, 160014, India
| | | | - Neelima Dhingra
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, 160014, India.
| | - Tanzeer Kaur
- Department of Biophysics, Panjab University, Chandigarh, 160014, India.
| |
Collapse
|
26
|
Setlur AS, Karunakaran C, Anusha V, Shendre AA, Uttarkar A, Niranjan V, Ashok Kumar HG, Kusanur R. Investigating the Molecular Interactions of Quinoline Derivatives for Antibacterial Activity Against Bacillus subtilis: Computational Biology and In Vitro Study Interpretations. Mol Biotechnol 2024; 66:3252-3273. [PMID: 37930509 DOI: 10.1007/s12033-023-00933-6] [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: 06/02/2023] [Accepted: 10/05/2023] [Indexed: 11/07/2023]
Abstract
Bacterial infections are evolving and one of the chief problems is emergence and prevalence of antibacterial resistance. Moreover, certain strains of Bacillus subtilis have become resistant to several antibiotics. To counteract this menace, the present work aimed to comprehend the antibacterial activity of synthesized two quinoline derivatives against Bacillus subtilis. Toxicity predictions via Protox II, SwissADME and T.E.S.T (Toxicity Estimation Software Tool) revealed that these derivatives were non-toxic and had little to no adverse effects. Molecular docking studies carried out in Schrodinger with two quinoline derivatives (referred Q1 and Q2) docked against selected target proteins (PDB IDs: 2VAM and1FSE) of B. subtilis demonstrated ideal binding energies (2VAM-Q1: - 4.63 kcal/mol and 2VAM-Q2: - 4.46 kcal/mol, and 1FSE-Q1: - 3.51 kcal/mol, 1FSE-Q2: - 6.34 kcal/mol). These complexes were simulated at 100 ns and the outcomes revealed their stability with slight conformational changes. Anti-microbial assay via disc diffusion method revealed zones of inhibition showing that B. subtilis was inhibited by both Q1 and Q2, with Q2 performing slightly better than Q1, pointing towards its effectiveness against this organism and necessitating further study on other bacteria in prospective studies. Thus, this study demonstrates that our novel quinoline derivatives exhibit antibacterial properties against Bacillus subtilis and can act as potent anti-bacterials.
Collapse
Affiliation(s)
- Anagha S Setlur
- Department of Biotechnology, R.V College of Engineering, Bangalore, 560059, India
| | | | - V Anusha
- Department of Biotechnology, R.V College of Engineering, Bangalore, 560059, India
| | - Aditya A Shendre
- Department of Biotechnology, R.V College of Engineering, Bangalore, 560059, India
| | - Akshay Uttarkar
- Department of Biotechnology, R.V College of Engineering, Bangalore, 560059, India
| | - Vidya Niranjan
- Department of Biotechnology, R.V College of Engineering, Bangalore, 560059, India
| | - H G Ashok Kumar
- Department of Biotechnology, R.V College of Engineering, Bangalore, 560059, India
| | - Raviraj Kusanur
- Department of Chemistry, R.V. College of Engineering, Bangalore, 560059, India.
| |
Collapse
|
27
|
Cartereau A, Bouchouireb Z, Kaaki S, Héricourt F, Taillebois E, Le Questel JY, Thany SH. Pharmacology and molecular modeling studies of sulfoxaflor, flupyradifurone and neonicotinoids on the human neuronal α7 nicotinic acetylcholine receptor. Toxicol Appl Pharmacol 2024; 492:117123. [PMID: 39393466 DOI: 10.1016/j.taap.2024.117123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 10/06/2024] [Accepted: 10/06/2024] [Indexed: 10/13/2024]
Abstract
We conducted electrophysiological and molecular docking studies using a heterologous expression system (Xenopus oocytes) to compare the effects of four neonicotinoids (acetamiprid, imidacloprid, clothianidin and thiamethoxam), one sulfoximine, (sulfoxaflor), and one butenolide (flupyradifurone), on human α7 neuronal nicotinic acetylcholine receptors (nAChRs). All neonicotinoids (except thiamethoxam), as well as the recently introduced nAChR competitive modulators, flupyradifurone and sulfoxaflor, appear to be weaker agonists than acetylcholine. Two mutations in loop C (E211N and E211P) and one mutation in loop D (Q79K), known to be involved in the binding properties of neonicotinoids were introduced to the α7 wild type. Interestingly, the acetylcholine and nicotine-evoked activation was not modified in human α7 mutated receptors, but the net charge was enhanced for clothianidin and imidacloprid, respectively. Flupyradifurone responses strongly increased under the Q79K mutation. The molecular docking investigations demonstrated that the orientations and interactions of the ligands considered were in accordance with those observed experimentally. Specifically, the charged fragments of acetylcholine and nicotine, used as reference ligands, and their neonicotinoid homologs were found to be surrounded by aromatic residues, with key interactions with Trp171 and Y210. Furthermore, the molecular docking investigations predicted the water-mediated interaction between the carbonyl oxygen of acetylcholine and the Nsp2 nitrogen of the pyridine ring for nicotine (as well as for the majority of the corresponding neonicotinoid fragments) and main chain NH of L141. The docking scores, extending over a significant range of 6 kcal/mol, showed that most neonicotinoids were poorly stabilized in the α7 nAChR compared to acetylcholine, except sulfoxaflor.
Collapse
Affiliation(s)
- Alison Cartereau
- Université d'Orléans, USC INRAE 1328, Laboratoire Physiologie, Ecologie et Environnement (P2E), 1 rue de Chartres, 45067 Orléans, France
| | | | - Sara Kaaki
- Université d'Orléans, USC INRAE 1328, Laboratoire Physiologie, Ecologie et Environnement (P2E), 1 rue de Chartres, 45067 Orléans, France
| | - François Héricourt
- Université d'Orléans, USC INRAE 1328, Laboratoire Physiologie, Ecologie et Environnement (P2E), 1 rue de Chartres, 45067 Orléans, France
| | - Emiliane Taillebois
- Université d'Orléans, USC INRAE 1328, Laboratoire Physiologie, Ecologie et Environnement (P2E), 1 rue de Chartres, 45067 Orléans, France
| | | | - Steeve H Thany
- Université d'Orléans, USC INRAE 1328, Laboratoire Physiologie, Ecologie et Environnement (P2E), 1 rue de Chartres, 45067 Orléans, France; Institut Universitaire de France (IUF), 1 rue Descartes, 75005 Paris, France.
| |
Collapse
|
28
|
Hu Y, Yang H, Li M, Zhong Z, Zhou Y, Bai F, Wang Q. Exploring Protein Conformational Changes Using a Large-Scale Biophysical Sampling Augmented Deep Learning Strategy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400884. [PMID: 39387316 PMCID: PMC11600214 DOI: 10.1002/advs.202400884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 07/22/2024] [Indexed: 10/15/2024]
Abstract
Inspired by the success of deep learning in predicting static protein structures, researchers are now actively exploring other deep learning algorithms aimed at predicting the conformational changes of proteins. Currently, a major challenge in the development of such models lies in the limited training data characterizing different conformational transitions. To address this issue, molecular dynamics simulations is combined with enhanced sampling methods to create a large-scale database. To this end, the study simulates the conformational changes of 2635 proteins featuring two known stable states, and collects the structural information along each transition pathway. Utilizing this database, a general deep learning model capable of predicting the transition pathway for a given protein is developed. The model exhibits general robustness across proteins with varying sequence lengths (ranging from 44 to 704 amino acids) and accommodates different types of conformational changes. Great agreement is shown between predictions and experimental data in several systems and successfully apply this model to identify a novel allosteric regulation in an important biological system, the human β-cardiac myosin. These results demonstrate the effectiveness of the model in revealing the nature of protein conformational changes.
Collapse
Affiliation(s)
- Yao Hu
- Department of PhysicsUniversity of Science and Technology of ChinaHefeiAnhui230026China
| | - Hao Yang
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and TechnologyShanghaiTech University393 Middle Huaxia RoadShanghai201210China
| | - Mingwei Li
- Department of PhysicsUniversity of Science and Technology of ChinaHefeiAnhui230026China
| | - Zhicheng Zhong
- Department of PhysicsUniversity of Science and Technology of ChinaHefeiAnhui230026China
| | - Yongqi Zhou
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and TechnologyShanghaiTech University393 Middle Huaxia RoadShanghai201210China
| | - Fang Bai
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and TechnologyShanghaiTech University393 Middle Huaxia RoadShanghai201210China
- School of Information Science and TechnologyShanghaiTech University393 Middle Huaxia RoadShanghai201210China
- Shanghai Clinical Research and Trial CenterShanghai201210China
| | - Qian Wang
- Department of PhysicsUniversity of Science and Technology of ChinaHefeiAnhui230026China
| |
Collapse
|
29
|
Berluti F, Baselious F, Hagemann S, Hilscher S, Schmidt M, Hüttelmaier S, Schutkowski M, Sippl W, Ibrahim HS. Development of new pyrazoles as class I HDAC inhibitors: Synthesis, molecular modeling, and biological characterization in leukemia cells. Arch Pharm (Weinheim) 2024; 357:e2400437. [PMID: 39291901 DOI: 10.1002/ardp.202400437] [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: 05/31/2024] [Revised: 08/26/2024] [Accepted: 08/27/2024] [Indexed: 09/19/2024]
Abstract
Class I histone deacetylases (HDACs) are considered promising targets in current cancer research. To obtain subtype-selective and potent HDAC inhibitors, we used the aminobenzamide scaffold as the zinc-binding group and prepared new derivatives with a pyrazole ring as the linking group. The synthesized compounds were analyzed in vitro using an enzymatic assay against HDAC1, -2, and -3. Compounds 12b, 15b, and 15i were found to be potent HDAC1 inhibitors, also in comparison to the reference compounds entinostat and tacedinaline, with IC50 values of 0.93, 0.22, and 0.68 μM, respectively. The best compounds were measured for their cellular effect and target engagement in acute myeloid leukemia (AML) cells. In addition, we studied the interaction of the compounds with HDAC subtypes using docking and molecular dynamic simulations. In summary, we have developed a new chemotype of HDAC1 inhibitors that can be used for further structure-based optimization.
Collapse
Affiliation(s)
- Francesco Berluti
- Department of Medicinal Chemistry, Institute of Pharmacy, Martin-Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Fady Baselious
- Department of Medicinal Chemistry, Institute of Pharmacy, Martin-Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Sven Hagemann
- Department of Molecular Medicine, Faculty of Medicine, Martin-Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Sebastian Hilscher
- Department of Enzymology, Institute of Biochemistry, Martin-Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Matthias Schmidt
- Department of Medicinal Chemistry, Institute of Pharmacy, Martin-Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Stefan Hüttelmaier
- Department of Molecular Medicine, Faculty of Medicine, Martin-Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Mike Schutkowski
- Department of Enzymology, Institute of Biochemistry, Martin-Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Wolfgang Sippl
- Department of Medicinal Chemistry, Institute of Pharmacy, Martin-Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Hany S Ibrahim
- Department of Medicinal Chemistry, Institute of Pharmacy, Martin-Luther University Halle-Wittenberg, Halle (Saale), Germany
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Egyptian Russian University, Badr City, Cairo, Egypt
| |
Collapse
|
30
|
Ataker Y, Öncü Ö, Gülmez D, Sabuncuoğlu S, Arikan-Akdagli S, Sari S. New Ester-Containing Azole Derivatives With Potent Anti-Candida Effects: Synthesis, Antifungal Susceptibility, Cytotoxicity, and Molecular Modeling Studies. Drug Dev Res 2024; 85:e70021. [PMID: 39551958 DOI: 10.1002/ddr.70021] [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: 08/13/2024] [Revised: 09/27/2024] [Accepted: 11/02/2024] [Indexed: 11/19/2024]
Abstract
Mortalities due to mycoses have dramatically increased with the emergence of drug-resistant strains and growing immune-compromised populations globally. Azole antifungals have been the first choice against fungal infections of a wide spectrum and several azole derivatives with ester function were reported for their potentially promising and favorable activity against Candida spp. In this study, we designed and synthesized a series of 1-(aryl)-2-(1H-imidazol-1-yl/1H-1,2,4-triazol-1-yl)ethyl esters, and tested them against seven reference Candida strains using EUCAST reference microdilution method. Among the series, 6a, 6d, and 6g proved highly potent in vitro compared to fluconazole; especially against Candida albicans and Candida tropicalis with minimum inhibitor concentration (MIC) values as low as 0.125 and 0.06 mg/L, respectively, although their activities against Candida krusei and Candida glabrata remained limited. The compounds also showed minimal toxicity to murine fibroblasts according to the in vitro cytotoxicity tests. Molecular modeling predicted 6g as an orally available druglike compound according to all parameters and CYP51 inhibition as the likely mechanism for their antifungal effects. The study underpins the promise of azoles with ester functionality as a potential scaffold for small-molecule antifungal drug design.
Collapse
Affiliation(s)
- Yusuf Ataker
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Başkent University, Ankara, Turkey
| | - Özge Öncü
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Dolunay Gülmez
- Department of Medical Microbiology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Suna Sabuncuoğlu
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Sevtap Arikan-Akdagli
- Department of Medical Microbiology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Suat Sari
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| |
Collapse
|
31
|
Kumar S, Ali I, Abbas F, Rana A, Pandey S, Garg M, Kumar D. In-silico design, pharmacophore-based screening, and molecular docking studies reveal that benzimidazole-1,2,3-triazole hybrids as novel EGFR inhibitors targeting lung cancer. J Biomol Struct Dyn 2024; 42:9416-9438. [PMID: 37646177 DOI: 10.1080/07391102.2023.2252496] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 08/18/2023] [Indexed: 09/01/2023]
Abstract
Lung cancer is a complex and heterogeneous disease, which has been associated with various molecular alterations, including the overexpression and mutations of the epidermal growth factor receptor (EGFR). In this study, designed a library of 1843 benzimidazole-1,2,3-triazole hybrids and carried out pharmacophore-based screening to identify potential EGFR inhibitors. The 164 compounds were further evaluated using molecular docking and molecular dynamics simulations to understand the binding interactions between the compounds and the receptor. In-si-lico ADME and toxicity studies were also conducted to assess the drug-likeness and safety of the identified compounds. The results of this study indicate that benzimidazole-1,2,3-triazole hybrids BENZI-0660, BENZI-0125, BENZI-0279, BENZI-0415, BENZI-0437, and BENZI-1110 exhibit dock scores of -9.7, -9.6, -9.6, -9.6, -9.6, -9.6 while referencing molecule -7.9 kcal/mol for EGFR (PDB ID: 4HJO), respectively. The molecular docking and molecular dynamics simulations revealed that the identified compounds formed stable interactions with the active site of EGFR, indicating their potential as inhibitors. The in-silico ADME and toxicity studies showed that the compounds had favorable drug-likeness properties and low toxicity, further supporting their potential as therapeutic agents. Finally, performed DFT studies on the best-selected ligands to gain further insights into their electronic properties. The findings of this study provide important insights into the potential of benzimidazole-1,2,3-triazole hybrids as promising EGFR inhibitors for the treatment of lung cancer. This research opens up a new avenue for the discovery and development of potent and selective EGFR inhibitors for the treatment of lung cancer.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Sunil Kumar
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Shoolini University, Solan, India
| | - Iqra Ali
- Department of Biosciences, COMSATS University Islamabad, Islamabad, Pakistan
| | - Faheem Abbas
- Key Lab of Organic Optoelectronics and Molecular Engineering of Ministry of Education, Department of Chemistry, Tsinghua University, Beijing, P. R. China
| | - Anurag Rana
- Yogananda School of Artificial Intelligence, Computers, and Data Sciences, Shoolini University, Solan, India
| | - Sadanand Pandey
- Department of Chemistry, College of Natural Science, Yeungnam University, Gyeongsan, Korea
| | - Manoj Garg
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University, Noida, India
| | - Deepak Kumar
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Shoolini University, Solan, India
| |
Collapse
|
32
|
Yousuf M, Zafar H, Atia-Tul-Wahab, Yousuf S, Rahman N, Ghoran SH, Ahmed A, Choudhary MI. Identification of new potential inhibitors of pteridine reductase-1 (PTR1) via biophysical and biochemical mechanism-based approaches: Step towards the treatment of Leishmaniasis. Int J Biol Macromol 2024; 282:137198. [PMID: 39489241 DOI: 10.1016/j.ijbiomac.2024.137198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 09/30/2024] [Accepted: 10/31/2024] [Indexed: 11/05/2024]
Abstract
Leishmaniasis is a parasitic disease, which spreads from the bite of an infected Phlebotomine fly to human hosts. The disease is characterized by a number of clinical manifestations, such as ulcerative lesions at the site of sandfly bite (cutaneous form), inflammation of mucosal membranes (mucosal leishmaniasis) or the deadly visceral form. This study was aimed to target pteridine reductase-1 (PTR1), a member of short chain dehydrogenases, which accounts for the reduction of conjugated and unconjugated pterins in Leishmania parasite. The ptr1-pET28a+-tev construct was expressed using BL21 (DE3) cells, followed by two tandem purification steps including affinity and gel permeation chromatography. In the next phase, functional studies of PTR1 were performed via screening of an in-house library of 500 compounds. The biochemical-mechanism based assay of PTR1 identified 11 hits that were also found to be non-cytotoxic against human fibroblast cell line (BJ) (except compound 6), and thus further studied via computational technique and saturation transfer difference-nuclear magnetic resonance (STD-NMR) spectroscopy. These high throughput techniques identified six compounds 2, 4, 5, 7, 9, and 11 as active, which were then assessed via in-vitro assay. Among them, compounds 2, 4, and 7 showed substantial leishmanicidal activity, comparable to the standard drug, miltefosine (IC50 value: 31.8 ± 0.2 μM). These results narrowed down the search to 3 compounds as potential leads, with prominent protein-ligand interaction profiles. Hence, the respective compounds can be further assessed for their therapeutic potential against leishmaniasis.
Collapse
Affiliation(s)
- Muhammad Yousuf
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Humaira Zafar
- Dr. Panjwani Center for Molecular and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan.
| | - Atia-Tul-Wahab
- Dr. Panjwani Center for Molecular and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan.
| | - Sammer Yousuf
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Noor Rahman
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Salar Hafez Ghoran
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Aftab Ahmed
- School of Pharmacy, Chapman University, 9401 Jeronimo Road, Irvine, CA 92618, USA.
| | - M Iqbal Choudhary
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan; Dr. Panjwani Center for Molecular and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan.
| |
Collapse
|
33
|
Almufarriji FM, Alotaibi BS, Alamri AS, Alkhalil SS, Alkhorayef N, Hakami MA. Unveiling the multitargeted potential of deprodone and control comparison with linezolid against hydrolase and transferase enzymes of methicillin-resistant Staphylococcus aureus. Int J Biol Macromol 2024; 279:135459. [PMID: 39250989 DOI: 10.1016/j.ijbiomac.2024.135459] [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: 04/27/2024] [Revised: 08/27/2024] [Accepted: 09/06/2024] [Indexed: 09/11/2024]
Abstract
Staphylococcus aureus (S. aureus), commonly found on the skin and nose, causes minor skin conditions to life-threatening diseases, including boils or impetigo, pneumonia, and bloodstream infections. MRSA (Methicillin-Resistant S. aureus) is a strain resistant to many antibiotics and poses a significant challenge in clinical settings. Nowadays, the alternative drug Linezolid is used, and it is not clear when MRSA starts resistance to it, necessitating the need for more alternative drugs with the least chance of developing resistance. This study aims to identify a multitargeted drug candidate with better efficacy than Linezolid. We have taken three hydrolase and transferase proteins from S. aureus, performed the multitargeted docking studies with human-approved drugs, and compared them with the control drug Linezolid. The docking and MM\GBSA scores ranging from -6.79 to -5.78 Kcal/mol and - 37.47 to 30.16 Kcal/mol, respectively, that revealed Deprodone (used for inflammatory skin disorders, bowel disease, and fatty acid metabolism disorders) can be a far better and multitargeted drug candidate than Linezolid. We extended our studies to include extensive pharmacokinetics and molecular interaction fingerprints for interaction pattern studies. Also, the DFT computations optimised the drug, and we extended our studies for MD Simulation in water for 100 ns, which showed the complexes among the identified drug with proteins are entirely stable with acceptable deviation, fluctuations and many intermolecular interactions that make them stable. We also performed the MM\GBSA studies on MD simulation's all 1000 frames to understand the complex energy level. All the results reveal promising interactions between Deprodone and the targeted enzymes, suggesting its potential as a multitargeted therapeutic agent-however, experimental studies need to validate Deprodone against MRSA.
Collapse
Affiliation(s)
- Fawaz M Almufarriji
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Shaqra University, Al-Quwayiyah 19257, Riyadh, Saudi Arabia.
| | - Bader S Alotaibi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Shaqra University, Al-Quwayiyah 19257, Riyadh, Saudi Arabia.
| | - Ahlam Saleh Alamri
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Shaqra University, Al-Quwayiyah 19257, Riyadh, Saudi Arabia.
| | - Samia S Alkhalil
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Shaqra University, Al-Quwayiyah 19257, Riyadh, Saudi Arabia.
| | - Nada Alkhorayef
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Shaqra University, Al-Quwayiyah 19257, Riyadh, Saudi Arabia.
| | - Mohammed Ageeli Hakami
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Shaqra University, Al-Quwayiyah 19257, Riyadh, Saudi Arabia.
| |
Collapse
|
34
|
Tse AL, Acreman CM, Ricardo-Lax I, Berrigan J, Lasso G, Balogun T, Kearns FL, Casalino L, McClain GL, Chandran AM, Lemeunier C, Amaro RE, Rice CM, Jangra RK, McLellan JS, Chandran K, Miller EH. Distinct pathways for evolution of enhanced receptor binding and cell entry in SARS-like bat coronaviruses. PLoS Pathog 2024; 20:e1012704. [PMID: 39546542 PMCID: PMC11602109 DOI: 10.1371/journal.ppat.1012704] [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: 07/05/2024] [Revised: 11/27/2024] [Accepted: 10/29/2024] [Indexed: 11/17/2024] Open
Abstract
Understanding the zoonotic risks posed by bat coronaviruses (CoVs) is critical for pandemic preparedness. Herein, we generated recombinant vesicular stomatitis viruses (rVSVs) bearing spikes from divergent bat CoVs to investigate their cell entry mechanisms. Unexpectedly, the successful recovery of rVSVs bearing the spike from SHC014-CoV, a SARS-like bat CoV, was associated with the acquisition of a novel substitution in the S2 fusion peptide-proximal region (FPPR). This substitution enhanced viral entry in both VSV and coronavirus contexts by increasing the availability of the spike receptor-binding domain to recognize its cellular receptor, ACE2. A second substitution in the S1 N-terminal domain, uncovered through the rescue and serial passage of a virus bearing the FPPR substitution, further enhanced spike:ACE2 interaction and viral entry. Our findings identify genetic pathways for adaptation by bat CoVs during spillover and host-to-host transmission, fitness trade-offs inherent to these pathways, and potential Achilles' heels that could be targeted with countermeasures.
Collapse
Affiliation(s)
- Alexandra L. Tse
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, New York, New York, United States of America
| | - Cory M. Acreman
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas, United States of America
| | - Inna Ricardo-Lax
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, New York, United States of America
| | - Jacob Berrigan
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, New York, New York, United States of America
| | - Gorka Lasso
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, New York, New York, United States of America
| | - Toheeb Balogun
- Department of Molecular Biology, University of California San Diego, La Jolla, California, United States of America
| | - Fiona L. Kearns
- Department of Molecular Biology, University of California San Diego, La Jolla, California, United States of America
| | - Lorenzo Casalino
- Department of Molecular Biology, University of California San Diego, La Jolla, California, United States of America
| | - Georgia L. McClain
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, New York, United States of America
| | - Amartya Mudry Chandran
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, New York, New York, United States of America
| | - Charlotte Lemeunier
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, New York, New York, United States of America
| | - Rommie E. Amaro
- Department of Molecular Biology, University of California San Diego, La Jolla, California, United States of America
| | - Charles M. Rice
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, New York, United States of America
| | - Rohit K. Jangra
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, New York, New York, United States of America
- Present address: Department of Microbiology and Immunology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana, United States of America
| | - Jason S. McLellan
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas, United States of America
| | - Kartik Chandran
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, New York, New York, United States of America
| | - Emily Happy Miller
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, New York, New York, United States of America
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, New York, United States of America
| |
Collapse
|
35
|
Modafferi C, Tabolacci E, Lo Vecchio F, Cassano I, Bertozzi R, Fargnoli A, Cafiero C, Lo Cascio E, Arcovito A, Grippaudo C, Chiurazzi P. New Insight into the genotype-phenotype correlation of PTH1R variants and primary failure of tooth eruption on an Italian Cohort. Eur J Hum Genet 2024; 32:1402-1411. [PMID: 39327493 DOI: 10.1038/s41431-024-01691-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 08/01/2024] [Accepted: 09/05/2024] [Indexed: 09/28/2024] Open
Abstract
Primary failure of tooth eruption (PFE) is an autosomal dominant disease with penetrance defect. While the clinical phenotype is relatively well-defined since the 70 s of the last centuries, much more need to be clarified about the genetic causes of this condition. In our previous paper we established clinical criteria to better identify PFE patients carrying PTH1R gene variants. We examined a new cohort of 32 patients, including one or more relatives for 7 patients (43 cases in total), referred to have PFE and recruited from our Hospital and from external outpatients. Sequencing analysis of the PTH1R coding sequence in this cohort of patients revealed 9 different variants, 4 exonic and 5 intronic. Through in silico prediction tools and databases, 3 of them (2 exonic and 1 in a splicing site) had been considered potentially involved in the PFE phenotype. Sequencing of cDNA was unsuccessfully attempted due to the low levels of PTH1R expression in the analysed tissues. The yield of the genetic test increases when the clinical selection of the patients with dental eruption failure is well-characterized. Dental eruption failure with pure clinical findings of PFE associated with familial history revealed variants in PTH1R gene, offering a diagnostic test for the family. Characterization of novel variants in the most relevant responsible gene of the PFE could bring to a more accurate diagnosis and therapeutic approach in the future and to a deeper comprehension of the disease.
Collapse
Affiliation(s)
- Clarissa Modafferi
- UOC Genetica Medica, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
| | - Elisabetta Tabolacci
- Dipartimento di Scienze della Vita e Sanità Pubblica, Sezione di Medicina Genomica, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
| | - Filomena Lo Vecchio
- UOC Genetica Medica, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
| | - Ilaria Cassano
- UOC Genetica Medica, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
| | - Roberto Bertozzi
- UOC Genetica Medica, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
| | - Arcangelo Fargnoli
- UOC Genetica Medica, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
| | - Concetta Cafiero
- Medical Oncology, SG Moscati Hospital, Statte, Ta, Italy
- Anatomic Pathology Unit, Fabrizio Spaziani Hospital, Frosinone, Italy
| | - Ettore Lo Cascio
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Sezione di Biochimica, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Alessandro Arcovito
- Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Sezione di Biochimica, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Cristina Grippaudo
- UOC di Clinica Odontoiatrica, Dipartimento di Neuroscienze, Organi di senso e Torace, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy.
- Dipartimento Universitario Testa Collo ed Organi di Senso, Università Cattolica del Sacro Cuore, Rome, Italy.
| | - Pietro Chiurazzi
- UOC Genetica Medica, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
- Dipartimento di Scienze della Vita e Sanità Pubblica, Sezione di Medicina Genomica, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
| |
Collapse
|
36
|
Alexopoulos S, McGawley M, Mathews R, Papakostopoulou S, Koulas S, Leonidas DD, Zwain T, Hayes JM, Skamnaki V. Evidence for the Quercetin Binding Site of Glycogen Phosphorylase as a Target for Liver-Isoform-Selective Inhibitors against Glioblastoma: Investigation of Flavanols Epigallocatechin Gallate and Epigallocatechin. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:24070-24081. [PMID: 39433280 PMCID: PMC11528470 DOI: 10.1021/acs.jafc.4c06920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 10/07/2024] [Accepted: 10/08/2024] [Indexed: 10/23/2024]
Abstract
Glycogen phosphorylase (GP) is the rate-determining enzyme in glycogenolysis, and its druggability has been extensively studied over the years for the development of therapeutics against type 2 diabetes (T2D) and, more recently, cancer. However, the conservation of binding sites between the liver and muscle isoforms makes the inhibitor selectivity challenging. Using a combination of kinetic, crystallographic, modeling, and cellular studies, we have probed the binding of dietary flavonoids epigallocatechin gallate (EGCG) and epigallocatechin (EGC) to GP isoforms. The structures of rmGPb-EGCG and rmGPb-EGC complexes were determined by X-ray crystallography, showing binding at the quercetin binding site (QBS) in agreement with kinetic studies that revealed both compounds as noncompetitive inhibitors of GP, with EGCG also causing a significant reduction in cell viability and migration of U87-MG glioblastoma cells. Interestingly, EGCG exhibits different binding modes to GP isoforms, revealing QBS as a promising site for GP targeting, offering new opportunities for the design of liver-selective GP inhibitors.
Collapse
Affiliation(s)
- Serafeim Alexopoulos
- Department
of Biochemistry and Biotechnology, University
of Thessaly, Biopolis, Larisa 41500, Greece
| | - Megan McGawley
- School
of Pharmacy & Biomedical Sciences, University
of Central Lancashire, Preston PR1 2HE, U.K.
| | - Roshini Mathews
- School
of Pharmacy & Biomedical Sciences, University
of Central Lancashire, Preston PR1 2HE, U.K.
| | - Souzana Papakostopoulou
- Department
of Biochemistry and Biotechnology, University
of Thessaly, Biopolis, Larisa 41500, Greece
| | - Symeon Koulas
- Department
of Biochemistry and Biotechnology, University
of Thessaly, Biopolis, Larisa 41500, Greece
| | - Demetres D. Leonidas
- Department
of Biochemistry and Biotechnology, University
of Thessaly, Biopolis, Larisa 41500, Greece
| | - Tamara Zwain
- School
of Pharmacy & Biomedical Sciences, University
of Central Lancashire, Preston PR1 2HE, U.K.
| | - Joseph M. Hayes
- School
of Pharmacy & Biomedical Sciences, University
of Central Lancashire, Preston PR1 2HE, U.K.
| | - Vasiliki Skamnaki
- Department
of Biochemistry and Biotechnology, University
of Thessaly, Biopolis, Larisa 41500, Greece
| |
Collapse
|
37
|
Pfannenstiel JJ, Duong MTH, Cluff D, Sherrill LM, Colquhoun I, Cadoux G, Thorne D, Pääkkönen J, Schemmel NF, O'Connor J, Saenjamsai P, Feng M, Hageman MJ, Johnson DK, Roy A, Lehtiö L, Ferraris DV, Fehr AR. Identification of a series of pyrrolo-pyrimidine based SARS-CoV-2 Mac1 inhibitors that repress coronavirus replication. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.28.620664. [PMID: 39554145 PMCID: PMC11565749 DOI: 10.1101/2024.10.28.620664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Coronaviruses (CoVs) can emerge from zoonotic sources and cause severe diseases in humans and animals. All CoVs encode for a macrodomain (Mac1) that binds to and removes ADP-ribose from target proteins. SARS-CoV-2 Mac1 promotes virus replication in the presence of interferon (IFN) and blocks the production of IFN, though the mechanisms by which it mediates these functions remain unknown. Mac1 inhibitors could help elucidate these mechanisms and serve as therapeutic agents against CoV-induced diseases. We previously identified compound 4a (a.k.a. MCD-628), a pyrrolo-pyrimidine that inhibited Mac1 activity in vitro at low micromolar levels. Here, we determined the binding mode of 4a by crystallography, further defining its interaction with Mac1. However, 4a did not reduce CoV replication, which we hypothesized was due to its acidic side chain limiting permeability. To test this hypothesis, we developed several hydrophobic derivatives of 4a . We identified four compounds that both inhibited Mac1 in vitro and inhibited murine hepatitis virus (MHV) replication: 5a , 5c , 6d , and 6e . Furthermore, 5c and 6e inhibited SARS-CoV-2 replication only in the presence of IFN γ , similar to a Mac1 deletion virus. To confirm their specificity, we passaged MHV in the presence of 5a to identify drug-resistant mutations and identified an alanine-to-threonine and glycine-to-valine double mutation in Mac1. Recombinant virus with these mutations had enhanced replication compared to WT virus when treated with 5a , demonstrating the specificity of these compounds during infection. However, this virus is highly attenuated in vivo , indicating that drug-resistance emerged at the expense of viral fitness. IMPORTANCE Coronaviruses (CoVs) present significant threats to human and animal health, as evidenced by recent outbreaks of MERS-CoV and SARS-CoV-2. All CoVs encode for a highly conserved macrodomain protein (Mac1) that binds to and removes ADP-ribose from proteins, which promotes virus replication and blocks IFN production, though the exact mechanisms remain unclear. Inhibiting Mac1 could provide valuable insights into these mechanisms and offer new therapeutic avenues for CoV-induced diseases. We have identified several unique pyrrolo-pyrimidine-based compounds as Mac1 inhibitors. Notably, at least two of these compounds inhibited both murine hepatitis virus (MHV) and SARS-CoV-2 replication. Furthermore, we identified a drug-resistant mutation in Mac1, confirming target specificity during infection. However, this mutant is highly attenuated in mice, indicating that drug-resistance appears to come at a fitness cost. These results emphasize the potential of Mac1 as a drug target and the promise of structure-based inhibitor design in combating coronavirus infections.
Collapse
|
38
|
Mukherjee MM, Biesbrock D, Abramowitz LK, Pavan M, Kumar B, Walter PJ, Azadi P, Jacobson KA, Hanover JA. Selective bioorthogonal probe for N-glycan hybrid structures. Nat Chem Biol 2024:10.1038/s41589-024-01756-5. [PMID: 39468349 DOI: 10.1038/s41589-024-01756-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 09/17/2024] [Indexed: 10/30/2024]
Abstract
Metabolic incorporation of chemically tagged monosaccharides is a facile means of tagging cellular glycoproteins and glycolipids. However, since the monosaccharide precursors are often shared by several pathways, selectivity has been difficult to attain. For example, N-linked glycosylation is a chemically complex and ubiquitous posttranslational modification, with three distinct classes of GlcNAc-containing N-glycan structures: oligomannose, hybrid and complex. Here we describe the synthesis of 1,3-Pr2-6-OTs GlcNAlk (MM-JH-1) as a next-generation metabolic chemical reporter for the selective labeling of hybrid N-glycan structures. We first developed a general strategy for defining the selectivity of labeling with chemically tagged monosaccharides. We then applied this approach to establish that MM-JH-1 is selectively incorporated into hybrid N-glycans. Using this metabolic chemical reporter as a detection tool, we performed imaging and fractionation to define features of the intracellular localization and trafficking of target proteins bearing hybrid N-glycan structures.
Collapse
Affiliation(s)
- Mana Mohan Mukherjee
- Laboratory of Cell and Molecular Biology, NIDDK, National Institutes of Health, Bethesda, MD, USA
| | - Devin Biesbrock
- Laboratory of Cell and Molecular Biology, NIDDK, National Institutes of Health, Bethesda, MD, USA
| | - Lara K Abramowitz
- Laboratory of Cell and Molecular Biology, NIDDK, National Institutes of Health, Bethesda, MD, USA
| | - Matteo Pavan
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, NIDDK, NIH, Bethesda, MD, USA
| | - Bhoj Kumar
- Complex Carbohydrate Research Center, The University of Georgia, Athens, GA, USA
| | - Peter J Walter
- Clinical Mass Spectrometry Core, NIDDK, National Institutes of Health, Bethesda, MD, USA
| | - Parastoo Azadi
- Complex Carbohydrate Research Center, The University of Georgia, Athens, GA, USA
| | - Kenneth A Jacobson
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, NIDDK, NIH, Bethesda, MD, USA
| | - John A Hanover
- Laboratory of Cell and Molecular Biology, NIDDK, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
39
|
Kwon JJ, Dilly J, Liu S, Kim E, Bian Y, Dharmaiah S, Tran TH, Kapner KS, Ly SH, Yang X, Rabara D, Waybright TJ, Giacomelli AO, Hong AL, Misek S, Wang B, Ravi A, Doench JG, Beroukhim R, Lemke CT, Haigis KM, Esposito D, Root DE, Nissley DV, Stephen AG, McCormick F, Simanshu DK, Hahn WC, Aguirre AJ. Comprehensive structure-function analysis reveals gain- and loss-of-function mechanisms impacting oncogenic KRAS activity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.22.618529. [PMID: 39484452 PMCID: PMC11526993 DOI: 10.1101/2024.10.22.618529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
To dissect variant-function relationships in the KRAS oncoprotein, we performed deep mutational scanning (DMS) screens for both wild-type and KRASG12D mutant alleles. We defined the spectrum of oncogenic potential for nearly all possible KRAS variants, identifying several novel transforming alleles and elucidating a model to describe the frequency of KRAS mutations in human cancer as a function of transforming potential, mutational probability, and tissue-specific mutational signatures. Biochemical and structural analyses of variants identified in a KRASG12D second-site suppressor DMS screen revealed that attenuation of oncogenic KRAS can be mediated by protein instability and conformational rigidity, resulting in reduced binding affinity to effector proteins, such as RAF and PI3-kinases, or reduced SOS-mediated nucleotide exchange activity. These studies define the landscape of single amino acid alterations that modulate the function of KRAS, providing a resource for the clinical interpretation of KRAS variants and elucidating mechanisms of oncogenic KRAS inactivation for therapeutic exploitation.
Collapse
Affiliation(s)
- Jason J. Kwon
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Julien Dilly
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Shengwu Liu
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - Eejung Kim
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Yuemin Bian
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Srisathiyanarayanan Dharmaiah
- NCI RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Timothy H. Tran
- NCI RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Kevin S. Kapner
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - Seav Huong Ly
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Xiaoping Yang
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Dana Rabara
- NCI RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Timothy J. Waybright
- NCI RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | | | - Andrew L. Hong
- Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, Atlanta, GA, USA
| | - Sean Misek
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Belinda Wang
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Arvind Ravi
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - John G. Doench
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Rameen Beroukhim
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | | | - Kevin M. Haigis
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Dominic Esposito
- NCI RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - David E. Root
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Dwight V. Nissley
- NCI RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Andrew G. Stephen
- NCI RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Frank McCormick
- NCI RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
- University of California, San Francisco Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Dhirendra K. Simanshu
- NCI RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - William C. Hahn
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Andrew J. Aguirre
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| |
Collapse
|
40
|
Yilmaz YB, Güngör T, Dönmez S, Atalay HN, Siyah P, Durdağı S, Ay M, Boyunegmez Tumer T. Synthesis, in silico and bio-evaluation studies of new isothiocyanate derivatives with respect to COX inhibition and H 2S release profiles. RSC Med Chem 2024:d4md00495g. [PMID: 39507615 PMCID: PMC11536671 DOI: 10.1039/d4md00495g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 10/21/2024] [Indexed: 11/08/2024] Open
Abstract
The development of H2S-donating derivatives of non-steroidal anti-inflammatory drugs (NSAIDs) is considered important to reduce or overcome their gastrointestinal side effects. Sulforaphane, one of the most extensively studied isothiocyanates (ITCs), effectively releases H2S at a slow rate. Thus, we rationally designed, synthesized, and characterized new ITC derivatives (I1-3 and I1a-e) inspired by the natural compound sulforaphane. The anti-inflammatory properties of these compounds were evaluated by their inhibitory activities against cyclooxygenase targets COX-1 and COX-2. Additionally, the cytotoxicity of the compounds was tested using the MTT assay on LPS-induced RAW 264.7 cells, revealing no cytotoxic effects at low doses. Notably, compounds I1 and fluorine-containing ester derivative I1c emerged as the most potent and selective COX-2 inhibitors, with selectivity indexes of 2611.5 and 2582.4, respectively. The H2S-releasing capacities of ITC derivatives were investigated and compared with that of sulforaphane, showing that while compounds I1-3 exhibit slow and similar H2S release to sulforaphane, the release from compounds I1a-e was not as pronounced as that of the standard. Physics-based molecular modeling studies including molecular docking and molecular dynamics (MD) simulations, binding free energy calculations and absorption, distribution, metabolism, and excretion (ADME) analyses were also conducted. MD simulations analysis underscored the crucial amino acids such as Tyr385, Trp387, Phe518, Val523, and Ser530 in the interactions between I1c hit compound and COX-2. The combined in silico and in vitro findings suggest that compounds I1 and I1c are promising NSAID candidates against selective COX-2 inhibition.
Collapse
Affiliation(s)
- Yakup Berkay Yilmaz
- Graduate Program of Molecular Biology and Genetics, School of Graduate Studies, Canakkale Onsekiz Mart University Canakkale Türkiye
| | - Tuğba Güngör
- Natural Products and Drug Research Laboratory, Department of Chemistry, Faculty of Science, Canakkale Onsekiz Mart University Canakkale Türkiye
| | - Serhat Dönmez
- Graduate Program of Molecular Biology and Genetics, School of Graduate Studies, Canakkale Onsekiz Mart University Canakkale Türkiye
| | - Hazal Nazlıcan Atalay
- Graduate Program of Molecular Biology and Genetics, School of Graduate Studies, Canakkale Onsekiz Mart University Canakkale Türkiye
| | - Pınar Siyah
- Department of Biochemistry, School of Pharmacy, Bahçeşehir University Istanbul Türkiye
| | - Serdar Durdağı
- Computational Biology and Molecular Simulations Laboratory, Department of Biophysics, School of Medicine, Bahçeşehir University Istanbul Türkiye
- Molecular Therapy Lab, Department of Pharmaceutical Chemistry, School of Pharmacy, Bahçeşehir University Istanbul Türkiye
- Lab for Innovative Drugs (Lab4IND), Computational Drug Design Center (HITMER), Bahçeşehir University Istanbul Türkiye
| | - Mehmet Ay
- Natural Products and Drug Research Laboratory, Department of Chemistry, Faculty of Science, Canakkale Onsekiz Mart University Canakkale Türkiye
| | - Tugba Boyunegmez Tumer
- Department of Molecular Biology and Genetics, Faculty of Science, Canakkale Onsekiz Mart University Canakkale Türkiye
| |
Collapse
|
41
|
Das A, Biggs MA, Hunt HL, Mahabadi V, Goncalves BG, Phan CAN, Banerjee IA. Design and investigation of novel iridoid-based peptide conjugates for targeting EGFR and its mutants L858R and T790M/L858R/C797S: an in silico study. Mol Divers 2024:10.1007/s11030-024-11007-3. [PMID: 39424745 DOI: 10.1007/s11030-024-11007-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Accepted: 09/29/2024] [Indexed: 10/21/2024]
Abstract
In this work, we designed novel peptide conjugates with plant-based iridoid and lichen-derived depside derivatives to target the wild-type EGFR (WT) and its mutants, L858R and T790M/L858R/C797S triple mutant. These mutations are often expressed in multiple cancers, particularly lung cancer. Specifically, the iridoids included 7-deoxyloganetic acid (7-DGA) and loganic acid (LG), while the depside derivative was sekikaic acid (SK). These compounds are known for their innate anticancer properties and were conjugated with two separate peptide sequences KLPGWSG (K) and YSIPKSS (Y). These sequences have been shown to target EGFR in previous phage display library screening, although the mechanism is unknown. Thus, we created the di-conjugates for dual targeting and investigated their interactions of the di-conjugates and that of the neat peptides with the kinase domain of EGFR (WT) and the two mutants using molecular docking, molecular dynamics (MD) simulations, and MM-GBSA analysis. Docking studies revealed that the (7-DGA)2-K showed the highest binding affinity at - 9.3 kcal/mol with the L858R mutant, while (LG)2-Y displayed the highest binding affinity at - 9.0 kcal/mol for the triple mutant receptor. Our results indicated that several of the conjugates interacted with crucial residues of the kinase domain, including ASP855 and THR854 (activation loop), MET793 and PRO794 (hinge region), ARG841 (catalytic loop), and LYS728 and LEU718 of the glycine-rich P-loop. Interestingly, strong hydrophobic interactions were also observed with the C-terminal tail residues, such as PHE997 and ALA1000 as well as with ARG999 for the YSIPKSS peptide and most of the conjugates. The hydroxyl group of the cyclopentane ring and the oxygen of the pyran ring of the (7-DGA)2-peptide conjugates contributed to binding particularly in the hinge region, while the peptide components formed an extended structure that bound well into the C-lobe. The (SK)2-Y di-conjugate and KLPGWSG peptide formed hydrogen bonds with the SER797 residue of the triple mutant. Overall, our results show that the (7-DGA)2-K, di-conjugate, the (7-DGA)2-Y di-conjugate, and the neat YSIPKSS demonstrated strong and stable binding with the L858R mutant and the highly resistant triple mutant EGFR, respectively. The novel designed conjugates demonstrate potential for further optimization for laboratory studies aimed at developing new therapeutics for targeting specific EGFR mutant expressing cells.
Collapse
Affiliation(s)
- Amrita Das
- Department of Chemistry and Biochemistry, Fordham University, 441 East Fordham Road, Bronx, NY, 10458, USA
| | - Mary A Biggs
- Department of Chemistry and Biochemistry, Fordham University, 441 East Fordham Road, Bronx, NY, 10458, USA
| | - Hannah L Hunt
- Department of Chemistry and Biochemistry, Fordham University, 441 East Fordham Road, Bronx, NY, 10458, USA
| | - Vida Mahabadi
- Department of Chemistry and Biochemistry, Fordham University, 441 East Fordham Road, Bronx, NY, 10458, USA
| | - Beatriz G Goncalves
- Department of Chemistry and Biochemistry, Fordham University, 441 East Fordham Road, Bronx, NY, 10458, USA
| | - Chau Anh N Phan
- Department of Chemistry and Biochemistry, Fordham University, 441 East Fordham Road, Bronx, NY, 10458, USA
| | - Ipsita A Banerjee
- Department of Chemistry and Biochemistry, Fordham University, 441 East Fordham Road, Bronx, NY, 10458, USA.
| |
Collapse
|
42
|
Facen E, Assoni G, Donati G, Paladino D, Carreira A, Bonomo I, Pietra VL, Lotti R, Houser J, Fava LL, Seneci P, Marinelli L, Arosio D, Provenzani A. Novel, soluble 3-heteroaryl-substituted tanshinone mimics attenuate the inflammatory response in murine macrophages. Sci Rep 2024; 14:24501. [PMID: 39424621 PMCID: PMC11489580 DOI: 10.1038/s41598-024-73309-8] [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: 03/11/2024] [Accepted: 09/16/2024] [Indexed: 10/21/2024] Open
Abstract
The RNA binding protein Human Antigen R (HuR) has been identified as a main regulator of the innate immune response and its inhibition can lead to beneficial anti-inflammatory effects. To this aim, we previously synthesized a novel class of small molecules named Tanshinone Mimics (TMs) able to interfere with HuR-RNA binding, and that dampen the LPS-induced immune response. Herein, we present a novel series of TMs, encompassing thiophene 3/TM9 and 4/TM10, furan 5/TM11 and 6/TM12, pyrrole 7b/TM13, and pyrazole 8. The furan-containing 5(TM11) showed the greatest inhibitory effect of the series on HuR-RNA complex formation, as suggested by RNA Electromobility Shift Assay and Time-Resolved FRET. Molecular Dynamics Calculation of HuR - 5/TM11 interaction, quantum mechanics approaches and Surface Plasmon Resonance data, all indicates that, within the novel heteroaryl substituents, the furan ring better recapitulates the chemical features of the RNA bound to HuR. Compound 5/TM11 also showed improved aqueous solubility compared to previously reported TMs. Real-time monitoring of cell growth and flow cytometry analyses showed that 5/TM11 preferentially reduced cell proliferation rather than apoptosis in murine macrophages at immunomodulatory doses. We observed its effects on the innate immune response triggered by lipopolysaccharide (LPS) in macrophages, showing that 5/TM11 significantly reduced the expression of proinflammatory cytokines as Cxcl10 and Il1b.
Collapse
Affiliation(s)
- Elisa Facen
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, Trento, 38123, Italy
| | - Giulia Assoni
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, Trento, 38123, Italy
- Department of Chemistry, University of Milan, Via Golgi 19, Milan, 20133, Italy
- Department of Chemistry and Applied Biosciences, ETH Hoenggerberg, HCI H498, Zurich, 8093, Switzerland
| | - Greta Donati
- Department of Pharmacy, University of Napoli Federico II, Via D. Montesano 49, Napoli, 80131, Italy
| | - Dalila Paladino
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, Trento, 38123, Italy
| | - Agata Carreira
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, Trento, 38123, Italy
| | - Isabelle Bonomo
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, Trento, 38123, Italy
| | - Valeria La Pietra
- Department of Pharmacy, University of Napoli Federico II, Via D. Montesano 49, Napoli, 80131, Italy
| | - Roberta Lotti
- DERMOLAB, University of Modena and Reggio Emilia, via del Pozzo, 71, Modena, 41124, Italy
| | - Josef Houser
- Central European Institute of Technology, Masaryk University, Kamenice 753/5, Brno, 625 00, Czech Republic
| | - Luca L Fava
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, Trento, 38123, Italy
| | - Pierfausto Seneci
- Department of Chemistry, University of Milan, Via Golgi 19, Milan, 20133, Italy
| | - Luciana Marinelli
- Department of Pharmacy, University of Napoli Federico II, Via D. Montesano 49, Napoli, 80131, Italy.
| | - Daniela Arosio
- Istituto di Scienze e Tecnologie Chimiche (SCITEC) 'Giulio Natta', Consiglio Nazionale delle Ricerche (CNR), Via C. Golgi 19, Milan, 20133, Italy.
| | - Alessandro Provenzani
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, Trento, 38123, Italy.
| |
Collapse
|
43
|
Dhanabalan AK, Devadasan V, Haribabu J, Krishnasamy G. Machine learning models to identify lead compound and substitution optimization to have derived energetics and conformational stability through docking and MD simulations for sphingosine kinase 1. Mol Divers 2024:10.1007/s11030-024-10997-4. [PMID: 39417979 DOI: 10.1007/s11030-024-10997-4] [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: 08/14/2024] [Accepted: 09/16/2024] [Indexed: 10/19/2024]
Abstract
Sphingosine kinases (SphKs) are a group of important enzymes that circulate at low micromolar concentrations in mammals and have received considerable attention due to the roles they play in a broad array of biological processes including apoptosis, mutagenesis, lymphocyte migration, radio- and chemo-sensitization, and angiogenesis. In the present study, we constructed three classification models by four machine learning (ML) algorithms including naive bayes (NB), support vector machine (SVM), logistic regression, and random forest from 395 compounds. The generated ML models were validated by fivefold cross validation. Five different scaffold hit fragments resulted from SVM model-based virtual screening and docking results indicate that all the five fragments exhibit common hydrogen bond interaction a catalytic residue of SphK1. Further, molecular dynamics (MD) simulations and binding free energy calculation had been carried out with the identified five fragment leads and three cocrystal inhibitors. The best 15 fragments were selected. Molecular dynamics (MD) simulations showed that among these compounds, 7 compounds have favorable binding energy compared with cocrystal inhibitors. Hence, the study showed that the present lead fragments could act as potential inhibitors against therapeutic target of cancers and neurodegenerative disorders.
Collapse
Affiliation(s)
- Anantha Krishnan Dhanabalan
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, 603203, India
| | - Velmurugan Devadasan
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, 603203, India.
| | - Jebiti Haribabu
- Facultad de Medicina, Universidad de Atacama, Los Carreras 1579, 1532502, Copiapó, Chile
- Chennai Institute of Technology (CIT), Chennai, Tamil Nadu, 600069, India
| | - Gunasekaran Krishnasamy
- Centre of Advanced Study in Crystallography and Biophysics, University of Madras, Guindy Campus, Chennai, Tamil Nadu, 600025, India.
| |
Collapse
|
44
|
Alsaiari AA, Almufarriji FM, Hazazi A, Almarghalani DA, Bakhuraysah MM, Alrehaili AA, Algethami SM, Almehmadi KA, Bahwerth FS, Hakami MA. Multitargeted docking approach reveals droxidopa against DNA replication and repair-related protein of cervical cancer. Sci Rep 2024; 14:24301. [PMID: 39414839 PMCID: PMC11484978 DOI: 10.1038/s41598-024-72770-9] [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: 04/27/2024] [Accepted: 09/10/2024] [Indexed: 10/18/2024] Open
Abstract
Cervical cancer begins in the cells lining the cervix and is caused by persistent infection with certain types of human papillomavirus (HPV). Initially, it has no symptoms, and later it causes pelvic pain, abnormal vaginal bleeding, and pain during intercourse. It is the fourth-ranked cancer among women, and many women die from cervical cancer every year, particularly in low-income countries and the majority could be prevented with early detection and treatment. In this study, we have taken Cervical Cancer DNA Replication and Repair-Related Protein with the PDBID- 3H15, 5VBN, and 6NT9 and performed the multitargeted molecular docking with the FDA-approved drug library using HTVS, SP and XP docking. Then, the poses were filtered with MM\GBSA for proper computations of free energy, identified a multitargeted inhibitor Droxidopa with docking and MM\GBSA scores ranging from - 5.559 to - 6.835 Kcal/mol and - 26.04 to - 37.33 Kcal/mol, respectively. We also performed interaction fingerprints revealing 2VAL, 2LYS, 1ALA, 1ARG, 1ASN, 1CYS, 1GLN, 1GLU, 1ILE, 1MET, 1PHE, 1PRO, 1SER, and 1THR were most interacted residues and computed the ADMET properties with QikProp and DFT with Jaguar, which supported the study and compounds' suitability. Moreover, we performed the 100ns MD simulation in water, showing the controlled deviation and fluctuations of the residues with many interactions, and MM\GBSA was performed with the same trajectories, showing a better understanding of each frame's total complex and binding-free energy. The whole study favours droxidopa as an inhibitor of cervical cancer DNA Replication and Repair-Related Proteins-however, experimental studies are needed before use.
Collapse
Affiliation(s)
- Ahad Amer Alsaiari
- Department of Clinical Laboratory Science, College of Applied Medical Science, Taif University, Taif, 21944, Saudi Arabia
| | - Fawaz M Almufarriji
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Shaqra University, Al-Quwayiyah, 19257, Saudi Arabia
| | - Ali Hazazi
- Department of Pathology and Laboratory Medicine, Security Forces Hospital Program, Riyadh, Saudi Arabia
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Daniyah A Almarghalani
- Department of Pharmacology and Toxicology, College of Pharmacy, Taif University, P.O. Box 11099, Taif, 21944, Saudi Arabia
- Stroke Research Unit, Taif University, P.O. Box 11099, Taif, 21944, Saudi Arabia
| | - Maha Mahfouz Bakhuraysah
- Department of Clinical Laboratory Science, College of Applied Medical Science, Taif University, Taif, 21944, Saudi Arabia
| | - Amani A Alrehaili
- Department of Clinical Laboratory Science, College of Applied Medical Science, Taif University, Taif, 21944, Saudi Arabia
| | - Shatha M Algethami
- Department of Clinical Laboratory Science, College of Applied Medical Science, Taif University, Taif, 21944, Saudi Arabia
| | - Khulood A Almehmadi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | | | - Mohammed Ageeli Hakami
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Shaqra University, Al-Quwayiyah, 19257, Saudi Arabia.
| |
Collapse
|
45
|
Kroupova A, Spiteri VA, Rutter ZJ, Furihata H, Darren D, Ramachandran S, Chakraborti S, Haubrich K, Pethe J, Gonzales D, Wijaya AJ, Rodriguez-Rios M, Sturbaut M, Lynch DM, Farnaby W, Nakasone MA, Zollman D, Ciulli A. Design of a Cereblon construct for crystallographic and biophysical studies of protein degraders. Nat Commun 2024; 15:8885. [PMID: 39406745 PMCID: PMC11480361 DOI: 10.1038/s41467-024-52871-9] [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: 03/14/2024] [Accepted: 09/19/2024] [Indexed: 10/19/2024] Open
Abstract
The ubiquitin E3 ligase cereblon (CRBN) is the target of therapeutic drugs thalidomide and lenalidomide and is recruited by most targeted protein degraders (PROTACs and molecular glues) in clinical development. Biophysical and structural investigation of CRBN has been limited by current constructs that either require co-expression with the adaptor DDB1 or inadequately represent full-length protein, with high-resolution structures of degrader ternary complexes remaining rare. We present the design of CRBNmidi, a construct that readily expresses from E. coli with high yields as soluble, stable protein without DDB1. We benchmark CRBNmidi for wild-type functionality through a suite of biophysical techniques and solve high-resolution co-crystal structures of its binary and ternary complexes with degraders. We qualify CRBNmidi as an enabling tool to accelerate structure-based discovery of the next generation of CRBN based therapeutics.
Collapse
Grants
- Almirall, Protac Programme, 35480b_CRT (118945), 03.01.2021-31.12.2024 Boehringer Ingelheim, Building a Protac, 8144e_CRT (115737), 01.12.16-31.12.2025 EUbOPEN (CEC), Enabling and Unlocking Biology in the Open, 35733_GR (118810), 01.05.2020-30.04.2025 Eisai, Research Collaboration, 34788_CRT (118489), 01.07.19-30.09-2025 JSPS Fellowship, 03.04.2023 but no separate funding for consumables Tocris, Development of a Covalent BromoTag System, 39186_CRT (119776), 10.01.2023-10.01.2025
Collapse
Affiliation(s)
- Alena Kroupova
- Centre for Targeted Protein Degradation, School of Life Sciences, University of Dundee, Dundee, DD1 5JJ, UK
| | - Valentina A Spiteri
- Centre for Targeted Protein Degradation, School of Life Sciences, University of Dundee, Dundee, DD1 5JJ, UK
| | - Zoe J Rutter
- Centre for Targeted Protein Degradation, School of Life Sciences, University of Dundee, Dundee, DD1 5JJ, UK
| | - Hirotake Furihata
- Centre for Targeted Protein Degradation, School of Life Sciences, University of Dundee, Dundee, DD1 5JJ, UK
| | - Darren Darren
- Centre for Targeted Protein Degradation, School of Life Sciences, University of Dundee, Dundee, DD1 5JJ, UK
- Cancer Science Institute Singapore, National University of Singapore, Singapore, 117599, Singapore
| | - Sarath Ramachandran
- Centre for Targeted Protein Degradation, School of Life Sciences, University of Dundee, Dundee, DD1 5JJ, UK
- Biocon BMS R&D Center, Bommasandra Industrial Area, Bommasandra, Karnataka, 560099, India
| | - Sohini Chakraborti
- Centre for Targeted Protein Degradation, School of Life Sciences, University of Dundee, Dundee, DD1 5JJ, UK
| | - Kevin Haubrich
- Centre for Targeted Protein Degradation, School of Life Sciences, University of Dundee, Dundee, DD1 5JJ, UK
| | - Julie Pethe
- Centre for Targeted Protein Degradation, School of Life Sciences, University of Dundee, Dundee, DD1 5JJ, UK
- National Heart and Lung Institute, Imperial College London, London, SW3 6LY, UK
| | - Denzel Gonzales
- Centre for Targeted Protein Degradation, School of Life Sciences, University of Dundee, Dundee, DD1 5JJ, UK
- Institute of Cell Biology, University of Edinburgh, Edinburgh, EH9 3BF, UK
| | - Andre J Wijaya
- Centre for Targeted Protein Degradation, School of Life Sciences, University of Dundee, Dundee, DD1 5JJ, UK
- PT Kalbe Farma, Jl. Let. Jend Suprapto Kav 4, Kalbe Farma, Jakarta, 10510, Indonesia
| | - Maria Rodriguez-Rios
- Centre for Targeted Protein Degradation, School of Life Sciences, University of Dundee, Dundee, DD1 5JJ, UK
| | - Manon Sturbaut
- Centre for Targeted Protein Degradation, School of Life Sciences, University of Dundee, Dundee, DD1 5JJ, UK
| | - Dylan M Lynch
- Centre for Targeted Protein Degradation, School of Life Sciences, University of Dundee, Dundee, DD1 5JJ, UK
| | - William Farnaby
- Centre for Targeted Protein Degradation, School of Life Sciences, University of Dundee, Dundee, DD1 5JJ, UK
| | - Mark A Nakasone
- Centre for Targeted Protein Degradation, School of Life Sciences, University of Dundee, Dundee, DD1 5JJ, UK
| | - David Zollman
- Centre for Targeted Protein Degradation, School of Life Sciences, University of Dundee, Dundee, DD1 5JJ, UK.
| | - Alessio Ciulli
- Centre for Targeted Protein Degradation, School of Life Sciences, University of Dundee, Dundee, DD1 5JJ, UK.
| |
Collapse
|
46
|
Fan L, Zhuang Y, Wu H, Li H, Xu Y, Wang Y, He L, Wang S, Chen Z, Cheng J, Xu HE, Wang S. Structural basis of psychedelic LSD recognition at dopamine D 1 receptor. Neuron 2024; 112:3295-3310.e8. [PMID: 39094559 DOI: 10.1016/j.neuron.2024.07.003] [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: 04/03/2024] [Revised: 06/11/2024] [Accepted: 07/05/2024] [Indexed: 08/04/2024]
Abstract
Understanding the kinetics of LSD in receptors and subsequent induced signaling is crucial for comprehending both the psychoactive and therapeutic effects of LSD. Despite extensive research on LSD's interactions with serotonin 2A and 2B receptors, its behavior on other targets, including dopamine receptors, has remained elusive. Here, we present cryo-EM structures of LSD/PF6142-bound dopamine D1 receptor (DRD1)-legobody complexes, accompanied by a β-arrestin-mimicking nanobody, NBA3, shedding light on the determinants of G protein coupling versus β-arrestin coupling. Structural analysis unveils a distinctive binding mode of LSD in DRD1, particularly with the ergoline moiety oriented toward TM4. Kinetic investigations uncover an exceptionally rapid dissociation rate of LSD in DRD1, attributed to the flexibility of extracellular loop 2 (ECL2). Moreover, G protein can stabilize ECL2 conformation, leading to a significant slowdown in ligand's dissociation rate. These findings establish a solid foundation for further exploration of G protein-coupled receptor (GPCR) dynamics and their relevance to signal transduction.
Collapse
Affiliation(s)
- Luyu Fan
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China.
| | - Youwen Zhuang
- State Key Laboratory of Drug Research, Center for Structure and Function of Drug Targets, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Hongyu Wu
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Huiqiong Li
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Youwei Xu
- State Key Laboratory of Drug Research, Center for Structure and Function of Drug Targets, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yue Wang
- State Key Laboratory of Drug Research, Center for Structure and Function of Drug Targets, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Licong He
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Shishan Wang
- Laboratory of Anesthesia and Critical Care Medicine in Colleges and Universities of Shandong Province, School of Anesthesiology, Shandong Second Medical University, Weifang 261021, China
| | - Zhangcheng Chen
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Jianjun Cheng
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - H Eric Xu
- State Key Laboratory of Drug Research, Center for Structure and Function of Drug Targets, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China; Lingang Laboratory, Shanghai 200031, China.
| | - Sheng Wang
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China; Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China.
| |
Collapse
|
47
|
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
|
48
|
Jagtap P, Meena VK, Sambhare S, Basu A, Abraham P, Cherian S. Exploring Niclosamide as a Multi-target Drug Against SARS-CoV-2: Molecular Dynamics Simulation Studies on Host and Viral Proteins. Mol Biotechnol 2024:10.1007/s12033-024-01296-2. [PMID: 39373955 DOI: 10.1007/s12033-024-01296-2] [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: 08/29/2024] [Accepted: 09/23/2024] [Indexed: 10/08/2024]
Abstract
Niclosamide has emerged as a promising repurposed drug against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). In vitro studies suggested that niclosamide inhibits the host transmembrane protein 16F (hTMEM16F), crucial for lipid scramblase activity, which consequently reduces syncytia formation that aids viral spread. Based on other in vitro reports, niclosamide may also target viral proteases such as papain-like protease (PLpro) and main protease (Mpro), essential for viral replication and maturation. However, the precise interactions by which niclosamide interacts with these multiple targets remain largely unclear. Docking and molecular dynamics (MD) simulation studies were undertaken based on a homology model of the hTMEM16F and available crystal structures of SARS-CoV-2 PLpro and Mpro. Niclosamide was observed to bind stably throughout a 400 ns MD simulation at the extracellular exit gate of the hTMEM16F tunnel, forming crucial interactions with residues spanning the TM1-TM2 loop (Gln350), TM3 (Phe481), and TM5-TM6 loop (Lys573, Glu594, and Asp596). Among the SARS-CoV-2 proteases, niclosamide was found to interact effectively with conserved active site residues of PLpro (Tyr268), exhibiting better stability in comparison to the control inhibitor, GRL0617. In conclusion, our in silico analyses support niclosamide as a multi-targeted drug inhibiting viral and host proteins involved in SARS-CoV-2 infections.
Collapse
Affiliation(s)
- Prachi Jagtap
- Bioinformatics & Data Management Group, ICMR National Institute of Virology, 20A Dr. Ambedkar Road, Pune, Maharashtra, 411 001, India
| | - Virendra Kumar Meena
- ICMR National Institute of Virology, 20A Dr. Ambedkar Road, Pune, Maharashtra, 411 001, India
| | - Susmit Sambhare
- ICMR National Institute of Virology, 20A Dr. Ambedkar Road, Pune, Maharashtra, 411 001, India
| | - Atanu Basu
- ICMR National Institute of Virology, 20A Dr. Ambedkar Road, Pune, Maharashtra, 411 001, India
| | - Priya Abraham
- Christian Medical College, Vellore, Tamil Nadu, India
| | - Sarah Cherian
- Bioinformatics & Data Management Group, ICMR National Institute of Virology, 20A Dr. Ambedkar Road, Pune, Maharashtra, 411 001, India.
| |
Collapse
|
49
|
Mohamed MA, Elsaman T, Elderdery AY, Alsrhani A, Ghanem HB, Alruwaili MM, Hamza SMA, Mekki SEI, Alotaibi HA, Mills J. Unveiling the Anticancer Potential: Computational Exploration of Nitrogenated Derivatives of (+)-Pancratistatin as Topoisomerase I Inhibitors. Int J Mol Sci 2024; 25:10779. [PMID: 39409108 PMCID: PMC11476810 DOI: 10.3390/ijms251910779] [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: 09/22/2024] [Revised: 10/03/2024] [Accepted: 10/05/2024] [Indexed: 10/20/2024] Open
Abstract
Cancer poses a substantial global health challenge, driving the need for innovative therapeutic solutions that offer improved effectiveness and fewer side effects. Topoisomerase I (Topo I) has emerged as a validated molecular target in the pursuit of developing anticancer drugs due to its critical role in DNA replication and transcription. (+)-Pancratistatin (PST), a naturally occurring compound found in various Amaryllidaceae plants, exhibits promising anticancer properties by inhibiting Topo I activity. However, its clinical utility is hindered by issues related to limited chemical availability and aqueous solubility. To address these challenges, molecular modelling techniques, including virtual screening, molecular docking, molecular mechanics with generalised born and surface area solvation (MM-GBSA) calculations, and molecular dynamics simulations were utilised to evaluate the binding interactions and energetics of PST analogues with Topo I, comparing them with the well-known Topo I inhibitor, Camptothecin. Among the compounds screened for this study, nitrogenated analogues emerged as the most encouraging drug candidates, exhibiting improved binding affinities, favourable interactions with the active site of Topo I, and stability of the protein-ligand complex. Structural analysis pinpointed key molecular determinants responsible for the heightened potency of nitrogenated analogues, shedding light on essential structural modifications for increased activity. Moreover, in silico absorption, distribution, metabolism, excretion, and toxicity (ADMET) predictions highlighted favourable drug-like properties and reduced toxicity profiles for the most prominent nitrogenated analogues, further supporting their potential as effective anticancer agents. In summary, this screening study underscores the significance of nitrogenation in augmenting the anticancer efficacy of PST analogues targeting Topo I. The identified lead compounds exhibit significant potential for subsequent experimental validation and optimisation, thus facilitating the development of novel and efficacious anticancer therapeutics with enhanced pharmacological profiles.
Collapse
Affiliation(s)
- Magdi Awadalla Mohamed
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University, Sakaka 72388, Saudi Arabia
| | - Tilal Elsaman
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University, Sakaka 72388, Saudi Arabia
| | - Abozer Y. Elderdery
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka 42421, Saudi Arabia; (A.Y.E.); (A.A.); (H.B.G.)
| | - Abdullah Alsrhani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka 42421, Saudi Arabia; (A.Y.E.); (A.A.); (H.B.G.)
| | - Heba Bassiony Ghanem
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka 42421, Saudi Arabia; (A.Y.E.); (A.A.); (H.B.G.)
| | - Majed Mowanes Alruwaili
- Nursing Administration & Education Department, College of Nursing, Jouf University, Sakaka 72388, Saudi Arabia;
| | - Siddiqa M. A. Hamza
- Department of Pathology, College of Medicine, Umm Alqura University, Algunfudah 21912, Saudi Arabia;
| | | | | | - Jeremy Mills
- School of Medicine, Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth PO1 2DT, UK;
| |
Collapse
|
50
|
Sentürk NB, Kasapoglu B, Sahin E, Ozcan O, Ozansoy M, Ozansoy MB, Siyah P, Sezerman U, Sahin F. The Potential Role of Boron in the Modulation of Gut Microbiota Composition: An In Vivo Pilot Study. Pharmaceuticals (Basel) 2024; 17:1334. [PMID: 39458975 PMCID: PMC11510266 DOI: 10.3390/ph17101334] [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: 07/09/2024] [Revised: 09/13/2024] [Accepted: 09/24/2024] [Indexed: 10/28/2024] Open
Abstract
Background/Objectives: The role of the gut microbiome in the development and progression of many diseases has received increased attention in recent years. Boron, a trace mineral found in dietary sources, has attracted interest due to its unique electron depletion and coordination characteristics in chemistry, as well as its potential role in modulating the gut microbiota. This study investigates the effects of inorganic boron derivatives on the gut microbiota of mice. Methods: For three weeks, boric acid (BA), sodium pentaborate pentahydrate (NaB), and sodium perborate tetrahydrate (SPT) were dissolved (200 mg/kg each) in drinking water and administered to wild-type BALB/c mice. The composition of the gut microbiota was analyzed to determine the impact of these treatments. Results: The administration of BA significantly altered the composition of the gut microbiota, resulting in a rise in advantageous species such as Barnesiella and Alistipes. Additionally, there was a decrease in some taxa associated with inflammation and illness, such as Clostridium XIVb and Bilophila. Notable increases in genera like Treponema and Catellicoccus were observed, suggesting the potential of boron compounds to enrich microbial communities with unique metabolic functions. Conclusions: These findings indicate that boron compounds may have the potential to influence gut microbiota composition positively, offering potential prebiotic effects. Further research with additional analyses is necessary to fully understand the interaction between boron and microbiota and to explore the possibility of their use as prebiotic agents in clinical settings.
Collapse
Affiliation(s)
- Nermin Basak Sentürk
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, 34755 Istanbul, Turkey; (N.B.S.); (B.K.)
| | - Burcu Kasapoglu
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, 34755 Istanbul, Turkey; (N.B.S.); (B.K.)
- Abdi Ibrahim Pharmaceuticals, Biotechnological Products Production Facility (AbdiBio), 34538 Istanbul, Turkey
| | - Eray Sahin
- Biostatistics and Bioinformatics PhD Program, Institute of Health Sciences, Acibadem Mehmet Ali Aydinlar University, 34752 Istanbul, Turkey;
| | | | - Mehmet Ozansoy
- Department of Physiology, International School of Medicine, Istanbul Medipol University, 34810 Istanbul, Turkey;
| | - Muzaffer Beyza Ozansoy
- Department of Physical Therapy and Rehabilitation, Faculty of Health Sciences, Fenerbahçe University, 34758 Istanbul, Turkey;
| | - Pinar Siyah
- Department of Biochemistry, School of Pharmacy, Bahçeşehir University, 34353 Istanbul, Turkey;
| | - Ugur Sezerman
- Biostatistics and Bioinformatics PhD Program, Institute of Health Sciences, Acibadem Mehmet Ali Aydinlar University, 34752 Istanbul, Turkey;
- Department of Biostatistics and Medical Informatics, Faculty of Medicine, Acibadem Mehmet Ali Aydinlar University, 34752 Istanbul, Turkey
| | - Fikrettin Sahin
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, 34755 Istanbul, Turkey; (N.B.S.); (B.K.)
| |
Collapse
|