1
|
Tiwari U, Akhtar S, Mir SS, Khan MKA. Evaluation of selected indigenous spices- and herbs-derived small molecules as potential inhibitors of SREBP and its implications for breast cancer using MD simulations and MMPBSA calculations. Mol Divers 2025:10.1007/s11030-025-11122-9. [PMID: 39899124 DOI: 10.1007/s11030-025-11122-9] [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: 11/05/2024] [Accepted: 01/23/2025] [Indexed: 02/04/2025]
Abstract
In this study, we conducted an extensive analysis of 252 bioactive compounds derived from native spices and herbs for their potential anti-breast cancer activity against sterol regulatory element-binding protein (SREBP), using in silico techniques. To evaluate the oral bioavailability, overall pharmacokinetics, and safety profiles of these compounds, we employed Lipinski's rule of five and ADME descriptors, which depicted 66 lead molecules. These molecules were then docked with the SREBP using molecular docking tools, which revealed that diosgenin and smilagenin were the most promising hits compared to the reference inhibitor betulin, with average binding affinities of - 7.42 and - 7.37 kcal/mol and - 6.27 kcal/mol, respectively. To further assess the stability of these complexes along with betulin, we conducted molecular dynamics simulations over a 100 ns simulation period. We employed various parameters, including the root-mean-square deviation, root-mean-square fluctuation, solvent-accessible surface area, free energy of solvation, and radius of gyration, followed by principal component analysis. Furthermore, we evaluated the toxicity of the selected compounds against various anticancer cell lines, as well as their metabolic activity related to CYP450 metabolism and biological activity spectrum. Based on these results, both molecules exhibited promising drug candidate potential and could be utilized for further experimental analysis to elucidate their anticancer potential.
Collapse
Affiliation(s)
- Urvashi Tiwari
- Department of Biosciences, Integral University, Lucknow, Uttar Pradesh, 226026, India
| | - Salman Akhtar
- Department of Bioengineering, Integral University, Lucknow, Uttar Pradesh, 226026, India
| | - Snober S Mir
- Department of Biosciences, Integral University, Lucknow, Uttar Pradesh, 226026, India
| | | |
Collapse
|
2
|
Gattan HS, Al-Ahmadi BM, Shater AF, Saeedi NH, Alruhaili MH. Structural exploration of the PfBLM Helicase-ATP Binding Domain and implications in the quest for antimalarial therapies. J Vector Borne Dis 2024; 61:389-399. [PMID: 38287768 DOI: 10.4103/jvbd.jvbd_176_23] [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: 11/13/2023] [Accepted: 01/23/2024] [Indexed: 01/31/2024] Open
Abstract
BACKGROUND OBJECTIVES The battle against malaria has witnessed remarkable progress in recent years, characterized by increased funding, development of life-saving tools, and a significant reduction in disease prevalence. Yet, the formidable challenge of drug resistance persists, threatening to undo these gains. METHODS To tackle this issue, it is imperative to identify new effective drug candidates against the malaria parasite that exhibit minimal toxicity. This study focuses on discovering such candidates by targeting PfRecQ1, also known as PfBLM, a vital protein within the malaria parasite Plasmodium falciparum . PfRecQ1 plays a crucial role in the parasite's life cycle and DNA repair processes, making it an attractive drug development target. The study employs advanced computational techniques, including molecular modeling, structure-based virtual screening (SBVS), ADMET profiling, molecular docking, and dynamic simulations. RESULTS The study sources ligand molecules from the extensive MCULE database and utilizes strict filters to ensure that the compounds meet essential criteria. Through these techniques, the research identifies MCULE-3763806507-0-9 as a promising antimalarial drug candidate, surpassing the binding affinity of potential antimalarial drugs. However, it is essential to underscore that drug-like properties are primarily based on in silico experiments, and wet lab experiments are necessary to validate these candidates' therapeutic potential. INTERPRETATION CONCLUSION This study represents a critical step in addressing the challenge of drug resistance in the fight against malaria.
Collapse
Affiliation(s)
- Hattan S Gattan
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Special Infectious Agents Unit, King Fahad Medical Research center, Jeddah, Saudi Arabia
| | - Bassam M Al-Ahmadi
- Department of Biology, Faculty of Science, Taibah University, Kingdom of Saudi Arabia
| | - Abdullah F Shater
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences. University of Tabuk, Tabuk 71491, Kingdom of Saudi Arabia
| | - Nizar H Saeedi
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk, Saudi Arabia
| | - Mohammed H Alruhaili
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, King AbdulAziz University, Jeddah, Saudi Arabia
- Special Infectious Agents Unit, King Fahad Medical Research center, King AbdulAziz University, Jeddah, Saudi Arabia
| |
Collapse
|
3
|
Barbosa F, Rocha BA, Souza MCO, Bocato MZ, Azevedo LF, Adeyemi JA, Santana A, Campiglia AD. Polycyclic aromatic hydrocarbons (PAHs): Updated aspects of their determination, kinetics in the human body, and toxicity. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART B, CRITICAL REVIEWS 2023; 26:28-65. [PMID: 36617662 DOI: 10.1080/10937404.2022.2164390] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Polycyclic aromatic hydrocarbons (PAHs) are legacy pollutants of considerable public health concern. Polycyclic aromatic hydrocarbons arise from natural and anthropogenic sources and are ubiquitously present in the environment. Several PAHs are highly toxic to humans with associated carcinogenic and mutagenic properties. Further, more severe harmful effects on human- and environmental health have been attributed to the presence of high molecular weight (HMW) PAHs, that is PAHs with molecular mass greater than 300 Da. However, more research has been conducted using low molecular weight (LMW) PAHs). In addition, no HMW PAHs are on the priority pollutants list of the United States Environmental Protection Agency (US EPA), which is limited to only 16 PAHs. However, limited analytical methodologies for separating and determining HMW PAHs and their potential isomers and lack of readily available commercial standards make research with these compounds challenging. Since most of the PAH kinetic data originate from animal studies, our understanding of the effects of PAHs on humans is still minimal. In addition, current knowledge of toxic effects after exposure to PAHs may be underrepresented since most investigations focused on exposure to a single PAH. Currently, information on PAH mixtures is limited. Thus, this review aims to critically assess the current knowledge of PAH chemical properties, their kinetic disposition, and toxicity to humans. Further, future research needs to improve and provide the missing information and minimize PAH exposure to humans.
Collapse
Affiliation(s)
- Fernando Barbosa
- Analytical and System Toxicology Laboratory, Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Bruno A Rocha
- Analytical and System Toxicology Laboratory, Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Marília C O Souza
- Analytical and System Toxicology Laboratory, Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Mariana Z Bocato
- Analytical and System Toxicology Laboratory, Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Lara F Azevedo
- Analytical and System Toxicology Laboratory, Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Joseph A Adeyemi
- Department of Biology, School of Sciences, Federal University of Technology, Akure, Nigeria
| | - Anthony Santana
- Department of Chemistry, University of Central Florida, Orlando, FL, USA
| | - Andres D Campiglia
- Department of Chemistry, University of Central Florida, Orlando, FL, USA
| |
Collapse
|
4
|
Kowalczyk K, Roszak J, Sobańska Z, Stępnik M. Review of mechanisms of genotoxic action of dibenzo[def,p]chrysene (formerly dibenzo[a,l]pyrene). TOXIN REV 2022. [DOI: 10.1080/15569543.2022.2124419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Affiliation(s)
| | - J. Roszak
- Department of Translational Research, Nofer Institute of Occupational Medicine, Łódź, Poland
| | - Z. Sobańska
- Department of Translational Research, Nofer Institute of Occupational Medicine, Łódź, Poland
| | | |
Collapse
|
5
|
Khan MKA, Ahmad S, Rabbani G, Shahab U, Khan MS. Target-based virtual screening, computational multiscoring docking and molecular dynamics simulation of small molecules as promising drug candidate affecting kinesin-like protein KIFC1. Cell Biochem Funct 2022; 40:451-472. [PMID: 35758564 DOI: 10.1002/cbf.3707] [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: 03/26/2022] [Revised: 04/28/2022] [Accepted: 05/04/2022] [Indexed: 11/07/2022]
Abstract
The kinesin family member C1 (KIFC1) is an essential protein that facilitates the bipolar division of neoplastic cells. Inhibiting KIFC1 by small molecules is a lucrative strategy to impede bipolar mitosis leading to the apoptosis of cancerous cells. The research aims to envisage small-molecule inhibitors targeting KIFC1. The Mcule database, a comprehensive online digital platform containing more than five million chemical compounds, was used for structure-based virtual screening (SBVS). Druglikeness filtration sifted 2,293,282 chemical hits that further narrowed down to 49 molecules after toxicity profiling. Finally, 39 compounds that comply with the BOILED-Egg permeation predictive model of the ADME rules were carried forward for multiscoring docking using the AutoDock Vina inbuilt to Mcule drug discovery platform, DockThor and SwissDock tools. The mean of ΔG terms produced by docking tools was computed to find consensus top ligand hits. AZ82 exhibited stronger binding (Consensus ΔG: -7.99 kcal mol-1 ) with KIFC1 among reference inhibitors, for example, CW069 (-7.57 kcal mol-1 ) and SR31527 (-7.01 kcal mol-1 ). Ten ligand hits namely, Mcule-4895338547 (Consensus ΔG: -8.69 kcal mol-1 ), Mcule-7035674888 (-8.42 kcal mol-1 ), Mcule-5531166845 (-8.53 kcal mol-1 ), Mcule-3248415882 (-8.55 kcal mol-1 ), Mcule-291881733 (-8.41 kcal mol-1 ), Mcule-5918624394 (-8.44), Mcule-3470115427 (-8.47), Mcule-3686193135 (-8.18 kcal mol-1 ), Mcule-3955355291 (8.09 kcal mol-1 ) and Mcule-9534899193 (-8.01 kcal mol-1 ) depicted strong binding interactions with KIFC1 in comparison to potential reference inhibitor AZ82. The top four ligands and AZ82 were considered for molecular dynamics simulation of 50 ns duration. Toxicity profiling, physicochemical properties, lipophilicity, solubility, pharmacokinetics, druglikeness, medicinal chemistry attributes, average potential energy, RMSD, RMSF, SASA, ΔGsolv and Rg analyses forecast the ligand mcule-4895338547 as a promising inhibitor of KIFC1.
Collapse
Affiliation(s)
| | - Saheem Ahmad
- Department of Biosciences, Integral University, Lucknow, India
| | - Gulam Rabbani
- Nano Diagnostics & Devices (NDD), IT Medical Fusion Center, Gumi-si, Gyeongbuk, Republic of Korea
| | - Uzma Shahab
- Department of Biotechnology, KMC Language University, Lucknow, India
| | - Mohd Shahnawaz Khan
- Department of Biochemistry, College of Sciences, King Saud University, Riyadh, Kingdom of Saudi Arabia
| |
Collapse
|
6
|
Revealing the Mechanism of EGCG, Genistein, Rutin, Quercetin, and Silibinin Against hIAPP Aggregation via Computational Simulations. Interdiscip Sci 2020; 12:59-68. [PMID: 31894470 DOI: 10.1007/s12539-019-00352-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 11/21/2019] [Accepted: 11/28/2019] [Indexed: 12/31/2022]
Abstract
To inhibit hIAPP aggregation and reduce toxicity of its oligomers are one of the potential strategies for the treatment of Type 2 diabetes (T2D). It has been reported that there is an effective inhibitory effect on hIAPP aggregation by five natural flavonoids, including Genistein, Rutin, Quercetin, Epigallocatechin gallate (EGCG), and Silibinin, which are widely found in our daily food. However, the detailed mechanisms to inhibit hIAPP aggregation remain unclear. Here, we explore the mechanisms of the five flavonoids against hIAPP aggregation by molecular docking and molecular dynamics simulations. We show that these flavonoids can disaggregate Chain A and Chain B of hIAPP to reduce the extended conformation by binding with two regions of hIAPP, Leu12-Ala13-Asn14 and Asn31-Val32-Gly33-Ser34-Asn35, with the inhibitory ability of Genistein > Rutin > Quercetin > EGCG > Silibinin. These five compounds exhibit a common mechanism for disaggregation of the hIAPP pentamer; that is, they loosen the two nearest peptide chains to potentially destroy the hIAPP oligomer. Mutations of eight key residues remarkably affected by the flavonoids indicate that the secondary structures of the hIAPP pentamer change from β-sheet to be random coil, thereby to destroy its structural stability; moreover, the 28th (Ser), 12th (Leu) and 32nd (Val) amino acids exhibit significant effects on structural stability of the hIAPP pentamer, providing an important hint that these amino acids can be considered as potential targets for design of new candidate inhibitors against hIAPP oligomers. This work is beneficial to understanding of mechanism of these inhibits against hIAPP aggregation and will facilitate screening, modification, and design of new inhibitors.
Collapse
|
7
|
Malik A, Afaq S, Gamal BE, Ellatif MA, Hassan WN, Dera A, Noor R, Tarique M. Molecular docking and pharmacokinetic evaluation of natural compounds as targeted inhibitors against Crz1 protein in Rhizoctonia solani. Bioinformation 2019; 15:277-286. [PMID: 31285645 PMCID: PMC6599437 DOI: 10.6026/97320630015277] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 03/27/2019] [Indexed: 11/29/2022] Open
Abstract
Crz1p regulates Calcineurin, a serine-threonine-specific protein phosphatase, in Rhizoctonia solani. It has attracted consideration as a novel target of antifungal therapy based on studies in numerous pathogenic fungi, including, Cryptococcus neoformans, Candida albicans and Aspergillus fumigatus. To investigate whether Calcineurin can be a useful target for the treatment of Crz1 protein in R. solani causing wet root rot in Chickpea. The work presented here reports the in-silico studies of Crz1 protein against natural compounds. This study Comprises of quantitative structure-toxicity relationship (QSTR) and quantitative structure-activity relationship (QSAR). All compounds showed high binding energy for Crz1 protein through molecular docking. Further, a pharmacokinetic study revealed that these compounds had minimal side effects. Biological activity spectrum prediction of these compounds showed potential antifungal properties by showing significant interaction with Crz1. Hence, these compounds can be used for the prevention and treatment of wet root rot in Chickpea.
Collapse
Affiliation(s)
- Ajit Malik
- Department of Clinical Biochemistry, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Sarah Afaq
- Department of Clinical Biochemistry, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Basiouny El Gamal
- Department of Clinical Biochemistry, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Mohamed Abd Ellatif
- Department of Clinical Biochemistry, College of Medicine, King Khalid University, Abha, Saudi Arabia
- Department of Medical Biochemistry,Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Waleed N Hassan
- Department of Clinical Biochemistry, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Ayed Dera
- Departments of Clinical Laboratory Science, College of Applied MedicalScience, King Khalid University, Abha, Saudi Arabia
| | - Rana Noor
- 5Department of Biochemistry, Faculty of Dentistry, Jamia Millia Islamia, New Delhi-110025, India
| | - Mohammed Tarique
- Center for InterdisciplinaryResearch in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi-110025, India
| |
Collapse
|
8
|
Khan MKA, Akhtar S, Arif JM. Development of In Silico Protocols to Predict Structural Insights into the Metabolic Activation Pathways of Xenobiotics. Interdiscip Sci 2017; 10:329-345. [PMID: 28527150 DOI: 10.1007/s12539-017-0237-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Revised: 05/05/2017] [Accepted: 05/09/2017] [Indexed: 11/26/2022]
Abstract
To establish in silico model to predict the structural insight into the metabolic bioactivation pathway of xenobiotics, we considered two specific and one non-specific mammary procarcinogen [e.g., dibenzo[a,l]pyrene (DBP), 7,12-dimethylbenz[a]anthracene (DMBA), and benzo[a]pyrene (BP)]. The CYP1A1, 1B1, 2C9, 1A2 and 2B6 reported in wet-lab studies to actively metabolize DBP also showed strong binding energies (kcal/mol) of -11.50, -10.67, -10.37, -9.76 and -9.72, respectively, with inhibition constants ranging between 0.01 and 0.08 µM. The CYP3A4 depicted minimum binding energy (-9.51 kcal/mol) which is in agreement with the wet-lab reports. Further, relatively better affinity of CYP1A1 and CYP1B1 with the dibenzo[a,l]pyrene-11,12-diol (DBPD) might be indicative of their involvement in carcinogenicity of parent compound. Like DBP, BP (-10.13 kcal/mol, Ki: 0.04 µM) and BP-diols (BPD) (-9.01 kcal/mol, Ki: 0.25 µM) observed plausible binding with CYP1A1 supporting to the reported data that emphasize the major contribution of CYP1A1 in the activation of similar procarcinogens and mutagens. Likewise, in silico results further highlighted the CYP1A1 as key player in bioactivation of DMBA to its carcinogenic metabolites. In case of PhIP metabolism, strong binding interaction predicted with CYP1A1 (-9.63 kcal/mol) rather than CYP1A2 (-8.84 kcal/mol). Dissimilarity in the binding affinity of PhIP might be due to its basic scaffold. Further, molecular dynamics (MD) simulation of 10 ns has been revealed that docked complexes of CYP1A1 with DBP, DMBA and BP are comparatively more stable than the complex of PhIP. Moreover, the current findings might be valuable as reference model in prediction and elucidation of the approximate metabolic pathway of xenobiotics.
Collapse
Affiliation(s)
- M Kalim A Khan
- Department of Bioengineering, Faculty of Engineering, Integral University, Lucknow, 226026, India
| | - Salman Akhtar
- Department of Bioengineering, Faculty of Engineering, Integral University, Lucknow, 226026, India
| | - Jamal M Arif
- Department of Biosciences, Faculty of Applied Sciences, Integral University, Kursi Road, P.O. Box Basha, Lucknow, 226026, India.
| |
Collapse
|