1
|
Ali ML, Noushin F, Azme E, Hasan MM, Hoque N, Metu AF. Marine natural compounds as potential CBP bromodomain inhibitors for treating cancer: an in-silico approach using molecular docking, ADMET, molecular dynamics simulations and MM-PBSA binding free energy calculations. In Silico Pharmacol 2024; 12:85. [PMID: 39310674 PMCID: PMC11411048 DOI: 10.1007/s40203-024-00258-5] [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: 02/07/2024] [Accepted: 09/08/2024] [Indexed: 09/25/2024] Open
Abstract
The cAMP-responsive element binding protein (CREB) binding protein (CBP), a bromodomain-containing protein, engages with multiple transcription factors and enhances the activation of many genes. CBP bromodomain acts as an epigenetic reader and plays an important role in the CBP-chromatin interaction which makes it an important drug target for treating many diseases. Though inhibiting CBP bromodomain was reported to have great potential in cancer therapeutics, approved CBP bromodomain inhibitor is yet to come. We utilized various in silico approaches like molecular docking, ADMET, molecular dynamics (MD) simulations, MM-PBSA calculations, and in silico PASS predictions to identify potential CBP bromodomain inhibitors from marine natural compounds as they have been identified as having distinctive chemical structures and greater anticancer activities. To develop a marine natural compound library for this investigation, Lipinski's rule of five was used. Sequential investigations utilizing molecular docking, ADMET studies, 100 ns MD simulations, and MM-PBSA calculations revealed that three marine compounds-ascididemin, neoamphimedine, and stelletin A-demonstrated superior binding affinity compared to the standard inhibitor, 69 A. These compounds also exhibited suitable drug-like properties, a favorable safety profile, and formed stable protein-ligand complexes. The in-silico PASS tool predicted that these compounds have significant potential for anticancer activity. Among them, ascididemin demonstrated the highest binding affinity in both molecular docking and MM-PBSA calculations, as well as a better stability profile in MD simulations. Hence, ascididemin can be a potential inhibitor of CBP bromodomain. However, in vitro and in vivo validation is required for further confirmation of these findings. Graphical abstract Supplementary Information The online version contains supplementary material available at 10.1007/s40203-024-00258-5.
Collapse
Affiliation(s)
- Md. Liakot Ali
- Department of Pharmacy, Faculty of Biological Science, University of Chittagong, Chittagong, 4331 Bangladesh
| | - Fabiha Noushin
- Department of Pharmacy, Faculty of Biological Science, University of Chittagong, Chittagong, 4331 Bangladesh
| | - Eva Azme
- Department of Pharmacy, Faculty of Biological Science, University of Chittagong, Chittagong, 4331 Bangladesh
| | - Md. Mahmudul Hasan
- Department of Pharmacy, Faculty of Biological Science, University of Chittagong, Chittagong, 4331 Bangladesh
| | - Neamul Hoque
- Department of Pharmacy, Faculty of Biological Science, University of Chittagong, Chittagong, 4331 Bangladesh
| | - Afroz Fathema Metu
- Department of Pharmacy, Faculty of Biological Science, University of Chittagong, Chittagong, 4331 Bangladesh
| |
Collapse
|
2
|
Islam M, Hossain A, Yamari I, Abchir O, Chtita S, Ali F, Kawsar SMA. Synthesis, Antimicrobial, Molecular Docking Against Bacterial and Fungal Proteins and In Silico Studies of Glucopyranoside Derivatives as Potent Antimicrobial Agents. Chem Biodivers 2024; 21:e202400932. [PMID: 38949892 DOI: 10.1002/cbdv.202400932] [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: 04/19/2024] [Revised: 06/28/2024] [Accepted: 07/01/2024] [Indexed: 07/03/2024]
Abstract
Carbohydrate derivatives play a crucial roles in biochemical and medicinal research, especially in the fields of chemistry and biochemistry. From this perspective, the present study was designed to explore the synthesis of methyl α-D-glucopyranoside derivatives (1-8), focusing on their efficacy against bacterial and fungal inhibition. The structure of the synthesized compounds was ascertained using FTIR, 1H-NMR, 13C-NMR, mass and elemental analyses. Antimicrobial screening revealed strong antifungal properties, with compound 7 exhibiting minimum inhibitory concentrations (MICs) ranging from 16-32 μg/L and minimum bactericidal concentrations (MBCs) ranging from 64-128 μg/L. Incorporating decanoyl acyl groups at C-2 and C-3 of (7) significantly improved the efficacy against bacteria and fungi. Structure-activity relationship (SAR) analysis indicated that adding nonanoyl and decanoyl groups to the ribose moiety enhanced potency against both bacterial and fungal strains. Computational methods, including molecular docking, density functional theory (DFT), Petra, Osiris, Molinspiration (POM) evaluation, and molecular dynamics (MD) simulations, were used to assess the efficacy of these derivatives. Compounds 6 and 7, which presented nonanoyl and decanoyl substituents, demonstrated greater efficacy. In addition, DFT studies identified compound 8 as possessing ideal electronic properties. Molecular docking revealed that compound 8 exhibits exceptional binding affinities to bacterial proteins, conferring potent antibacterial and antifungal activities. In addition, pharmacokinetic optimization via POM analysis highlighted compounds 1 and 2 as promising bioavailable drugs with minimal toxicity. Molecular dynamics simulations confirmed the stability of the 2-S. aureus complex, revealing the therapeutic potential of compounds 2 and 8. Future experiments are required to validate their efficacy for pharmaceutical development. The integration of in vitro and in silico methods, including DFT anchoring dynamics and molecular dynamics simulations, provides a solid framework for the advancement of effective anti-infective drugs.
Collapse
Affiliation(s)
- Mazherul Islam
- Laboratory of Carbohydrate and Nucleoside Chemistry (LCNC), Department of Chemistry, Faculty of Science, University of Chittagong, Chittagong, 4331, Bangladesh
| | - Ahad Hossain
- Laboratory of Carbohydrate and Nucleoside Chemistry (LCNC), Department of Chemistry, Faculty of Science, University of Chittagong, Chittagong, 4331, Bangladesh
| | - Imane Yamari
- Laboratory of Analytical and Molecular Chemistry, Faculty of Sciences Ben M'Sik, Hassan II University of Casablanca, Casablanca, Morocco
| | - Oussama Abchir
- Laboratory of Analytical and Molecular Chemistry, Faculty of Sciences Ben M'Sik, Hassan II University of Casablanca, Casablanca, Morocco
| | - Samir Chtita
- Laboratory of Analytical and Molecular Chemistry, Faculty of Sciences Ben M'Sik, Hassan II University of Casablanca, Casablanca, Morocco
| | - Ferdausi Ali
- Department of Microbiology, Faculty of Biological Science, University of Chittagong, Chittagong, 4331, Bangladesh
| | - Sarkar M A Kawsar
- Laboratory of Carbohydrate and Nucleoside Chemistry (LCNC), Department of Chemistry, Faculty of Science, University of Chittagong, Chittagong, 4331, Bangladesh
| |
Collapse
|
3
|
Gaiya DD, Muhammad A, Musa JS, Auta R, Dadah AJ, Bello RO, Hassan M, Eke SS, Odihi RI, Sankey M. In silico analysis of balsaminol as anti-viral agents targeting SARS-CoV-2 main protease, spike receptor binding domain and papain-like protease receptors. In Silico Pharmacol 2024; 12:75. [PMID: 39155972 PMCID: PMC11329488 DOI: 10.1007/s40203-024-00241-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 07/13/2024] [Indexed: 08/20/2024] Open
Abstract
Plant-derived phytochemicals from medicinal plants are becoming increasingly attractive natural sources of antimicrobial and antiviral agents due to their therapeutic value, mechanism of action, level of toxicity and bioavailability. The continued emergence of more immune-evasive strains and the rate of resistance to current antiviral drugs have created a need to identify new antiviral agents against SARS-CoV-2. This study investigated the antiviral potential of balsaminol, a bioactive compound from Momordica balsamina, and its inhibitory activities against SARS-CoV-2 receptor proteins. In this study, three Food and Drug Administration (FDA) COVID-19 approved drugs namely; nirmatrelvir, ritonavir and remdesivir were used as positive control. Molecular docking was performed to determine the predominant binding mode (most negative Gibbs free energy of binding/ΔG) and inhibitory activity of balsaminol against SARS-CoV-2 receptor proteins. The pharmacokinetics, toxicity, physicochemical and drug-like properties of balsaminol were evaluated to determine its potential as an active oral drug candidate as well as its non-toxicity in humans. The results show that balsaminol E has the highest binding affinity to the SARS CoV-2 papain-like protease (7CMD) with a free binding energy of - 8.7 kcal/mol, followed by balsaminol A interacting with the spike receptor binding domain (6VW1) with - 8.5 kcal/mol and balsaminol C had a binding energy of - 8.1 kcal/mol with the main protease (6LU7) comparable to the standard drugs namely ritonavir, nirmatrelvir and remdesivir. However, the ADMET and drug-like profile of balsaminol F favours it as a better potential drug candidate and inhibitor of the docked SARS-CoV-2 receptor proteins. Further preclinical studies are therefore recommended. Supplementary Information The online version contains supplementary material available at 10.1007/s40203-024-00241-0.
Collapse
Affiliation(s)
- Daniel Danladi Gaiya
- Biology Unit, Air Force Institute of Technology, Nigerian Air Force Base, P.M.B 2104, Kaduna, Nigeria
| | - Aliyu Muhammad
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, P.M.B. 1045, Samaru Zaria, Nigeria
| | - Joy Sim Musa
- Department of Chemistry, Faculty of Physical Sciences, Ahmadu Bello University, P.M.B. 1045, Samaru Zaria, Nigeria
| | - Richard Auta
- Department of Biochemistry, Faculty of Life Sciences, Kaduna State University, Tafawa Balewa Way, P.M.B. 2339, Kaduna, Nigeria
| | - Anthony John Dadah
- Department of Microbiology, Faculty of Life Sciences, Kaduna State University, Tafawa Balewa Way, P.M.B. 2339, Kaduna, Nigeria
| | | | - Madinat Hassan
- Biology Unit, Air Force Institute of Technology, Nigerian Air Force Base, P.M.B 2104, Kaduna, Nigeria
| | - Samuel Sunday Eke
- Biology Unit, Air Force Institute of Technology, Nigerian Air Force Base, P.M.B 2104, Kaduna, Nigeria
| | - Rebecca Imoo Odihi
- Department of Biological Science, Nigerian Defence Academy, Kaduna, Nigeria
| | - Musa Sankey
- Department of Chemistry, Kaduna State College of Education, Gidan Waya, Kaduna, Nigeria
| |
Collapse
|
4
|
Boukandou Mounanga MM, Mezui A, Mewono L, Mogangué JB, Aboughe Angone S. Medicinal plants used in Gabon for prophylaxis and treatment against COVID-19-related symptoms: an ethnobotanical survey. Front Pharmacol 2024; 15:1393636. [PMID: 39035990 PMCID: PMC11258373 DOI: 10.3389/fphar.2024.1393636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 06/04/2024] [Indexed: 07/23/2024] Open
Abstract
Background: Gabon faced COVID-19 with more than 49,000 individuals tested positive and 307 recorded fatalities since the first reported case in 2020. A popular hypothesis is that the low rate of cases and deaths in the country was attributed to the use of medicinal plants in prevention and treatment. This study aimed to document the plants used for remedial and preventive therapies by the Gabonese population during the COVID-19 pandemic and to pinpoint specific potential plant species that merit further investigation. Methods: An ethnobotanical survey involving 97 participants was conducted in Libreville. Traditional healers and medicinal plant vendors were interviewed orally using a semi-structured questionnaire sheet, while the general population responded to an online questionnaire format. Various quantitative indexes were calculated from the collected data and included the relative frequency of citation (RFC), use value (UV), informant consensus factor (ICF), relative importance (RI), and popular therapeutic use value (POPUT). One-way ANOVA and independent samples t-test were used for statistical analyses. p-values ≤0.05 were considered significant. Results: The survey identified 63 plant species belonging to 35 families. Prevalent symptoms treated included fever (18%), cough (16%), fatigue (13%), and cold (12%). The demographic data highlighted that 52.58% of male subjects (p > 0.94) aged 31-44 years were enrolled in the survey, of which 48.45% (p < 0.0001) and 74.73% (p < 0.99) of informants had university-level education. In addition, the results indicated that a total of 66% of the informants used medicinal plants for prophylaxis (34%), for both prevention and treatment (26%), exclusively for treatment (3%), and only for prevention (3%) while suffering from COVID-19, against 34% of the participants who did not use plants for prevention or treatment. Annickia chlorantha, Citrus sp., Alstonia congensis, Zingiber officinale, and Carica papaya emerged as the most commonly cited plants with the highest RFC (0.15-0.26), UV (0.47-0.75), and RI (35.72-45.46) values. Most of these plants were used either individually or in combination with others. Conclusion: The survey reinforces the use of traditional medicine as a method to alleviate COVID-19 symptoms, thereby advocating for the utilization of medicinal plants in managing coronavirus infections.
Collapse
Affiliation(s)
- Marlaine Michel Boukandou Mounanga
- Institut de Pharmacopée et de Médecine Traditionnelle (IPHAMETRA), Centre National de la Recherche Scientifique et Technologique (CENAREST), Libreville, Gabon
| | - Annais Mezui
- Centre Hospitalier Universitaire Mère- Enfant, Fondation Jeanne EBORI, Libreville, Gabon
| | - Ludovic Mewono
- Groupe de Recherche en Immunologie 2, Microbiologie appliquée, Hygiène et Physiologie, Département des Sciences de la Vie et de la Terre-Ecole Normale Supérieure, Libreville, Gabon
| | - Jean Bertrand Mogangué
- Institut de Pharmacopée et de Médecine Traditionnelle (IPHAMETRA), Centre National de la Recherche Scientifique et Technologique (CENAREST), Libreville, Gabon
| | - Sophie Aboughe Angone
- Institut de Pharmacopée et de Médecine Traditionnelle (IPHAMETRA), Centre National de la Recherche Scientifique et Technologique (CENAREST), Libreville, Gabon
| |
Collapse
|
5
|
Ja'afaru SC, Uzairu A, Chandra A, Sallau MS, Ndukwe GI, Ibrahim MT, Qamar I. Ligand based-design of potential schistosomiasis inhibitors through QSAR, homology modeling, molecular dynamics, pharmacokinetics, and DFT studies. J Taibah Univ Med Sci 2024; 19:429-446. [PMID: 38440085 PMCID: PMC10909894 DOI: 10.1016/j.jtumed.2024.02.003] [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: 11/08/2023] [Revised: 01/03/2024] [Accepted: 02/19/2024] [Indexed: 03/06/2024] Open
Abstract
Objectives Schistosomiasis, a neglected tropical disease, is a leading cause of mortality in affected geographic areas. Currently, because no vaccine for schistosomiasis is available, control measures rely on widespread administration of the drug praziquantel (PZQ). The mass administration of PZQ has prompted concerns regarding the emergence of drug resistance. Therefore, new therapeutic targets and potential compounds are necessary to combat schistosomiasis. Methods Twenty-four potent derivatives of PZQ were optimized via density functional theory (DFT) at the B3LYP/6-31G∗ level. Quantitative structureactivity relationship (QSAR) models were generated and statistically validated, and a lead candidate was selected to develop therapeutic options with improved efficacy against schistosomiasis. The biological and binding energies of the designed compounds were evaluated. In addition, molecular dynamics; drug-likeness; absorption, distribution, metabolism, excretion, and toxicity (ADMET); and DFT studies were performed on the newly designed compounds. Results Five QSAR models were generated, among which model 1 had favorable validation parameters (R2train: 0.957, R2adj: 0.941, LOF: 0.101, Q2cv: 0.906, and R2test: 0.783) and was chosen to identify a lead candidate. Other statistical parameters for the chosen model included variance inflation factor values ranging from 1.242 to 1.678, and a Y-scrambling coefficient (cRp2) of 0.747. Five new compounds were designed with improved predicted activity (ranging from 5.081 to 7.022) surpassing those of both the lead compound and PZQ (predicted pEC50 of 5.545). Molecular dynamics simulation revealed high binding affinity of the proposed compounds toward the target receptor. ADMET and drug-likeness assessments indicated adherence to Lipinski's rule of five criteria, thereby suggesting pharmacological and oral safety. In addition, DFT analysis indicated resistance to electronic alteration during chemical reactions. Conclusion The proposed compounds exhibited potential drug characteristics, thus indicating their suitability for further investigation to enhance schistosomiasis treatment options.
Collapse
Affiliation(s)
- Saudatu C. Ja'afaru
- Department of Chemistry, Ahmadu Bello University Zaria, Nigeria
- Department of Chemistry, Aliko Dangote University of Science and Technology, Wudil, Kano, Nigeria
| | - Adamu Uzairu
- Department of Chemistry, Ahmadu Bello University Zaria, Nigeria
| | - Anshuman Chandra
- School of Physical Sciences, JawaharLal Nehru University, New Delhi, India
| | | | | | | | - Imteyaz Qamar
- School of Biotechnology, Gautam Buddha University, Greater Noida, India
| |
Collapse
|
6
|
Hassan MA, Abd El-Aziz S, Nabil-Adam A, Tamer TM. Formulation of novel bioactive gelatin inspired by cinnamaldehyde for combating multi-drug resistant bacteria: Characterization, molecular docking, pharmacokinetic analyses, and in vitro assessments. Int J Pharm 2024; 652:123827. [PMID: 38253268 DOI: 10.1016/j.ijpharm.2024.123827] [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: 10/24/2023] [Revised: 01/13/2024] [Accepted: 01/19/2024] [Indexed: 01/24/2024]
Abstract
This study set out to formulate antibacterial and antioxidant gelatin boosted by cinnamaldehyde for combating multi-drug resistant bacteria previously obtained from chronic wounds. Towards this end, gelatin amine groups were conjugated with carbonyl groups of cinnamaldehyde, producing cinnamyl-gelatin Schiff bases. The physicochemical attributes of cinnamyl-gelatin Schiff bases were probed concerning alterations in chemical structures and microstructures compared to native gelatin. Besides, cinnamyl-gelatin Schiff bases exhibited higher thermal stability than gelatin, with a diminishing in solubility due to increases in hydrophobicity features. Interestingly, cinnamyl-gelatin derivatives exerted antibacterial activities versus multi-drug resistant Gram-negative and Gram-positive bacteria, showing maximum growth inhibition at the highest concentration of cinnamaldehyde incorporated into gelatin. The scavenging activities of gelatin against DPPH and ABTS•+ were promoted in cinnamyl-gelatin derivatives from 11.93 ± 0.6 % to 49.9 ± 2.5 % and 12.54 ± 0.63 % to 49.9 ± 3.12 %, respectively. Remarkably, cinnamyl-gelatin derivatives induced the proliferation of fibroblast cells, implying their prospective applications in tissue engineering. Molecular docking and pharmacokinetic investigations disclosed the potential antibacterial mechanisms of cinnamyl-gelatin derivatives alongside their biopharmaceutical applications. Altogether, these findings suggest that cinnamyl-gelatin derivatives could be utilized to tailor antibacterial-free antibiotics and antioxidant wound dressings against virulent bacteria to promote chronic wound recovery.
Collapse
Affiliation(s)
- Mohamed A Hassan
- Protein Research Department, Genetic Engineering and Biotechnology Research Institute (GEBRI), City of Scientific Research and Technological Applications (SRTA-City), New Borg El-Arab City, 21934 Alexandria, Egypt.
| | - Sarah Abd El-Aziz
- Polymer Materials Research Department, Advanced Technologies, and New Materials Research Institute (ATNMRI), City of Scientific Research and Technological Applications (SRTA-City), New Borg El-Arab City, 21934 Alexandria, Egypt
| | - Asmaa Nabil-Adam
- Marine Biotechnology and Natural Products Laboratory, National Institute of Oceanography & Fisheries, Cairo 11516, Egypt
| | - Tamer M Tamer
- Polymer Materials Research Department, Advanced Technologies, and New Materials Research Institute (ATNMRI), City of Scientific Research and Technological Applications (SRTA-City), New Borg El-Arab City, 21934 Alexandria, Egypt.
| |
Collapse
|
7
|
Zaki MEA, AL-Hussain SA, Al-Mutairi AA, Samad A, Masand VH, Ingle RG, Rathod VD, Gaikwad NM, Rashid S, Khatale PN, Burakale PV, Jawarkar RD. Application of in-silico drug discovery techniques to discover a novel hit for target-specific inhibition of SARS-CoV-2 Mpro's revealed allosteric binding with MAO-B receptor: A theoretical study to find a cure for post-covid neurological disorder. PLoS One 2024; 19:e0286848. [PMID: 38227609 PMCID: PMC10790994 DOI: 10.1371/journal.pone.0286848] [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: 03/22/2023] [Accepted: 05/24/2023] [Indexed: 01/18/2024] Open
Abstract
Several studies have revealed that SARS-CoV-2 damages brain function and produces significant neurological disability. The SARS-CoV-2 coronavirus, which causes COVID-19, may infect the heart, kidneys, and brain. Recent research suggests that monoamine oxidase B (MAO-B) may be involved in metabolomics variations in delirium-prone individuals and severe SARS-CoV-2 infection. In light of this situation, we have employed a variety of computational to develop suitable QSAR model using PyDescriptor and genetic algorithm-multilinear regression (GA-MLR) models (R2 = 0.800-793, Q2LOO = 0.734-0.727, and so on) on the data set of 106 molecules whose anti-SARS-CoV-2 activity was empirically determined. QSAR models generated follow OECD standards and are predictive. QSAR model descriptors were also observed in x-ray-resolved structures. After developing a QSAR model, we did a QSAR-based virtual screening on an in-house database of 200 compounds and found a potential hit molecule. The new hit's docking score (-8.208 kcal/mol) and PIC50 (7.85 M) demonstrated a significant affinity for SARS-CoV-2's main protease. Based on post-covid neurodegenerative episodes in Alzheimer's and Parkinson's-like disorders and MAO-B's role in neurodegeneration, the initially disclosed hit for the SARS-CoV-2 main protease was repurposed against the MAO-B receptor using receptor-based molecular docking, which yielded a docking score of -12.0 kcal/mol. This shows that the compound that inhibits SARS-CoV-2's primary protease may bind allosterically to the MAO-B receptor. We then did molecular dynamic simulations and MMGBSA tests to confirm molecular docking analyses and quantify binding free energy. The drug-receptor complex was stable during the 150-ns MD simulation. The first computational effort to show in-silico inhibition of SARS-CoV-2 Mpro and allosteric interaction of novel inhibitors with MAO-B in post-covid neurodegenerative symptoms and other disorders. The current study seeks a novel compound that inhibits SAR's COV-2 Mpro and perhaps binds MAO-B allosterically. Thus, this study will enable scientists design a new SARS-CoV-2 Mpro that inhibits the MAO-B receptor to treat post-covid neurological illness.
Collapse
Affiliation(s)
- Magdi E. A. Zaki
- Faculty of Science, Department of Chemistry, Imam Mohammad Ibn Saud Islamic University, Riyadh, Saudi Arabia
| | - Sami A. AL-Hussain
- Faculty of Science, Department of Chemistry, Imam Mohammad Ibn Saud Islamic University, Riyadh, Saudi Arabia
| | - Aamal A. Al-Mutairi
- Faculty of Science, Department of Chemistry, Imam Mohammad Ibn Saud Islamic University, Riyadh, Saudi Arabia
| | - Abdul Samad
- Faculty of Pharmacy, Department of Pharmaceutical Chemistry, Tishk International University, Erbil, Kurdistan Region, Iraq
| | - Vijay H. Masand
- Department of Chemistry, Vidya Bharti Mahavidyalaya, Amravati, Maharashtra, India
| | - Rahul G. Ingle
- Datta Meghe College of Pharmacy, DMIHER Deemed University, Wardha, India
| | - Vivek Digamber Rathod
- Department of Chemical Technology, Dr Babasaheb Ambedkar Marathwada University, Aurangabad, India
| | | | - Summya Rashid
- Department of Pharmacology & Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Pravin N. Khatale
- Department of Medicinal Chemistry and Drug Discovery, Dr Rajendra Gode Institute of Pharmacy, University Mardi Road, Amravati, Maharashtra, India
| | - Pramod V. Burakale
- Department of Medicinal Chemistry and Drug Discovery, Dr Rajendra Gode Institute of Pharmacy, University Mardi Road, Amravati, Maharashtra, India
| | - Rahul D. Jawarkar
- Department of Medicinal Chemistry and Drug Discovery, Dr Rajendra Gode Institute of Pharmacy, University Mardi Road, Amravati, Maharashtra, India
| |
Collapse
|
8
|
Singh D, Mittal N, Verma S, Singh A, Siddiqui MH. Applications of some advanced sequencing, analytical, and computational approaches in medicinal plant research: a review. Mol Biol Rep 2023; 51:23. [PMID: 38117315 DOI: 10.1007/s11033-023-09057-1] [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: 05/18/2023] [Accepted: 11/27/2023] [Indexed: 12/21/2023]
Abstract
The potential active chemicals found in medicinal plants, which have long been employed as natural medicines, are abundant. Exploring the genes responsible for producing these compounds has given new insights into medicinal plant research. Previously, the authentication of medicinal plants was done via DNA marker sequencing. With the advancement of sequencing technology, several new techniques like next-generation sequencing, single molecule sequencing, and fourth-generation sequencing have emerged. These techniques enshrined the role of molecular approaches for medicinal plants because all the genes involved in the biosynthesis of medicinal compound(s) could be identified through RNA-seq analysis. In several research insights, transcriptome data have also been used for the identification of biosynthesis pathways. miRNAs in several medicinal plants and their role in the biosynthesis pathway as well as regulation of the disease-causing genes were also identified. In several research articles, an in silico study was also found to be effective in identifying the inhibitory effect of medicinal plant-based compounds against virus' gene(s). The use of advanced analytical methods like spectroscopy and chromatography in metabolite proofing of secondary metabolites has also been reported in several recent research findings. Furthermore, advancement in molecular and analytic methods will give new insight into studying the traditionally important medicinal plants that are still unexplored.
Collapse
Affiliation(s)
- Dhananjay Singh
- Department of Biosciences, Integral University, Lucknow, Uttar Pradesh, 226026, India
| | - Nishu Mittal
- Institute of Biosciences and Technology, Shri Ramswaroop Memorial University, Barabanki, Uttar Pradesh, 225003, India
| | - Swati Verma
- College of Horticulture and Forestry Thunag, Dr. Y. S. Parmar University of Horticulture and Forestry, Nauni, Solan, Himachal Pradesh, 173230, India
| | - Anjali Singh
- Institute of Biosciences and Technology, Shri Ramswaroop Memorial University, Barabanki, Uttar Pradesh, 225003, India
| | | |
Collapse
|
9
|
Qais FA, Parveen N, Ahmad I, Husain FM, Khan A, Adil M. Multi-targeting of virulence factors of P. aeruginosa by β-lactam antibiotics to combat antimicrobial resistance. J Biomol Struct Dyn 2023:1-19. [PMID: 37904338 DOI: 10.1080/07391102.2023.2275181] [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: 09/11/2023] [Accepted: 10/20/2023] [Indexed: 11/01/2023]
Abstract
Antimicrobial resistance poses a significant challenge to public health, especially in developing countries, due to a substantial rise in bacterial resistance. This situation has become so concerning that we are now at risk of losing the effectiveness of antibiotics altogether. Recent research has firmly established that bacteria engage in a process called quorum sensing (QS). QS regulates various functions, including nutrient scavenging, immune response suppression, increased virulence, biofilm formation and mobility. Pseudomonas aeruginosa, an opportunistic bacterial pathogen, plays a significant role in various medical conditions such as chronic wounds, corneal infections, burn wounds and cystic fibrosis. While antibiotics are effective in killing bacteria, only a few antibiotics, particularly those from the β-lactam group, have been studied for their impact on the quorum sensing of P. aeruginosa. Given the lack of concentrated efforts in this area, we have investigated the role of β-lactam antibiotics on various potential targets of P. aeruginosa. Based on their toxicological profiles and the average binding energy obtained through molecular docking, azlocillin and moxalactam have emerged as lead antibiotics. The binding energy for the docking of azlocillin and moxalactam with LasA was determined to be -8.2 and -8.6 kcal/mol, respectively. Molecular simulation analysis has confirmed the stable interaction of both these ligands with all three target proteins (LasI, LasA and PqsR) under physiological conditions. The results of this research underscore the effectiveness of azlocillin and moxalactam. These two antibiotics may be repurposed to target the quorum sensing of P. aeruginosa.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Faizan Abul Qais
- Department of Agricultural Microbiology, Faculty of Agricultural Sciences, Aligarh Muslim University, Aligarh, UP, India
| | - Nagma Parveen
- Department of Zoology, Saifia College, Barkatullah University, Bhopal, India
| | - Iqbal Ahmad
- Department of Agricultural Microbiology, Faculty of Agricultural Sciences, Aligarh Muslim University, Aligarh, UP, India
| | | | - Altaf Khan
- Department of Pharmacology, College of Pharmacy, King Saud University, Riyadh, KSA
| | - Mohd Adil
- Department of Environmental Sciences, Dalhousie University, Truro, NS, Canada
| |
Collapse
|
10
|
G F N, V V, M G, S M, M P. Surface enhanced Raman scattering investigation of tecovirimat on silver, gold and platinum loaded silica nanocomposites: Theoretical analysis (DFT) and molecular modeling. Heliyon 2023; 9:e21122. [PMID: 37916120 PMCID: PMC10616345 DOI: 10.1016/j.heliyon.2023.e21122] [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: 08/04/2023] [Revised: 10/16/2023] [Accepted: 10/16/2023] [Indexed: 11/03/2023] Open
Abstract
As of today, there have been 612 million confirmed cases of coronavirus disease (COVID-19) around the world, with over 6 million fatalities. Tecovirimat (TPOXX) is an anti-viral drug, and it was the first drug approved for the treatment of anti-pox virus in the US. However, the effectiveness of this drug against COVID-19 has not yet been explored. Since TPOXX is an anti-viral drug, an attempt has been made to determine its ability to act as a COVID inhibitor. Recent medical advances have resulted in the development of nano cage-based drug delivery. Drug delivery clusters based on nano cages have recently been used in the medical industry. As such, we used DFT coupled to the B3LYP/LANL2DZ basis set to study the adsorption behavior of the anti-viral drug TPOXX on Au/Ag/Pt⋯SiO2loaded silica nanocomposites. In order to identify the active site of the molecule, we have used the frontier molecular orbital (FMO) theory of molecular electrostatic potential (MEP). The compound and its complexes obey Lipinski's rule of five and have good drug-likeness properties based on the bioactivity evaluation. The biological properties of organic molecules and nano metal clusters were compared. The TPOXX with its nanocomposites was also studied in terms of Electron Localization Function (ELF) and Localized Orbital Locator (LOL). Molecular docking was performed for both pure molecule and its silica nanocomposites-doped derivatives with the chosen proteins to discuss the protein-ligand binding properties. These results could be more helpful in designing the drug and exploring its application for the inhibition of SARS-CoV-2.
Collapse
Affiliation(s)
- Nivetha G F
- Department of Physics, Periyar University Centre for Post Graduate and Research Studies, Dharmapuri, 635205, India
| | - Vetrivelan V
- Department of Physics, Government College of Engineering, Srirangam, Tiruchirappalli, 620012, Tamilnadu, India
| | - Govindammal M
- Department of Physics, Government Arts College, Dharmapuri, 636705, India
| | - Muthu S
- Department of Physics, Arignar Anna Govt. Arts College, Cheyyar, 604407, Tamilnadu, India
| | - Prasath M
- Department of Physics, Periyar University Centre for Post Graduate and Research Studies, Dharmapuri, 635205, India
| |
Collapse
|
11
|
Rafiq A, Jabeen T, Aslam S, Ahmad M, Ashfaq UA, Mohsin NUA, Zaki MEA, Al-Hussain SA. A Comprehensive Update of Various Attempts by Medicinal Chemists to Combat COVID-19 through Natural Products. Molecules 2023; 28:4860. [PMID: 37375415 PMCID: PMC10305344 DOI: 10.3390/molecules28124860] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 06/05/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
The ongoing COVID-19 pandemic has resulted in a global panic because of its continual evolution and recurring spikes. This serious malignancy is caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Since the outbreak, millions of people have been affected from December 2019 till now, which has led to a great surge in finding treatments. Despite trying to handle the pandemic with the repurposing of some drugs, such as chloroquine, hydroxychloroquine, remdesivir, lopinavir, ivermectin, etc., against COVID-19, the SARS-CoV-2 virus continues its out-of-control spread. There is a dire need to identify a new regimen of natural products to combat the deadly viral disease. This article deals with the literature reports to date of natural products showing inhibitory activity towards SARS-CoV-2 through different approaches, such as in vivo, in vitro, and in silico studies. Natural compounds targeting the proteins of SARS-CoV-2-the main protease (Mpro), papain-like protease (PLpro), spike proteins, RNA-dependent RNA polymerase (RdRp), endoribonuclease, exoribonuclease, helicase, nucleocapsid, methyltransferase, adeno diphosphate (ADP) phosphatase, other nonstructural proteins, and envelope proteins-were extracted mainly from plants, and some were isolated from bacteria, algae, fungi, and a few marine organisms.
Collapse
Affiliation(s)
- Ayesha Rafiq
- Department of Chemistry, Government College University, Faisalabad 38000, Pakistan
| | - Tooba Jabeen
- Department of Chemistry, Government College University, Faisalabad 38000, Pakistan
| | - Sana Aslam
- Department of Chemistry, Government College Women University, Faisalabad 38000, Pakistan
| | - Matloob Ahmad
- Department of Chemistry, Government College University, Faisalabad 38000, Pakistan
| | - Usman Ali Ashfaq
- Department of Bioinformatics and Biotechnology, Government College University, Faisalabad 38000, Pakistan
| | - Noor ul Amin Mohsin
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Government College University, Faisalabad 38000, Pakistan
| | - Magdi E. A. Zaki
- Department of Chemistry, Faculty of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia
| | - Sami A. Al-Hussain
- Department of Chemistry, Faculty of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia
| |
Collapse
|
12
|
Nag A, Dasgupta A, Sengupta S, Lai TK, Acharya K. An in-silico pharmacophore-based molecular docking study to evaluate the inhibitory potentials of novel fungal triterpenoid Astrakurkurone analogues against a hypothetical mutated main protease of SARS-CoV-2 virus. Comput Biol Med 2023; 152:106433. [PMID: 36565483 PMCID: PMC9767885 DOI: 10.1016/j.compbiomed.2022.106433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 11/21/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND The main protease is an important structural protein of SARS-CoV-2, essential for its survivability inside a human host. Considering current vaccines' limitations and the absence of approved therapeutic targets, Mpro may be regarded as the potential candidate drug target. Novel fungal phytocompound Astrakurkurone may be studied as the potential Mpro inhibitor, considering its medicinal properties reported elsewhere. METHODS In silico molecular docking was performed with Astrakurkurone and its twenty pharmacophore-based analogues against the native Mpro protein. A hypothetical Mpro was also constructed with seven mutations and targeted by Astrakurkurone and its analogues. Furthermore, multiple parameters such as statistical analysis (Principal Component Analysis), pharmacophore alignment, and drug likeness evaluation were performed to understand the mechanism of protein-ligand molecular interaction. Finally, molecular dynamic simulation was done for the top-ranking ligands to validate the result. RESULT We identified twenty Astrakurkurone analogues through pharmacophore screening methodology. Among these twenty compounds, two analogues namely, ZINC89341287 and ZINC12128321 showed the highest inhibitory potentials against native and our hypothetical mutant Mpro, respectively (-7.7 and -7.3 kcal mol-1) when compared with the control drug Telaprevir (-5.9 and -6.0 kcal mol-1). Finally, we observed that functional groups of ligands namely two aromatic and one acceptor groups were responsible for the residual interaction with the target proteins. The molecular dynamic simulation further revealed that these compounds could make a stable complex with their respective protein targets in the near-native physiological condition. CONCLUSION To conclude, Astrakurkurone analogues ZINC89341287 and ZINC12128321 can be potential therapeutic agents against the highly infectious SARS-CoV-2 virus.
Collapse
Affiliation(s)
- Anish Nag
- Department of Life Sciences, CHRIST (Deemed to be University), Bangalore, Karnataka, India
| | - Adhiraj Dasgupta
- Department of Botany, University of Calcutta, Kolkata, West Bengal, India
| | - Sutirtha Sengupta
- Department of Life Sciences, CHRIST (Deemed to be University), Bangalore, Karnataka, India
| | - Tapan Kumar Lai
- Department of Chemistry, Vidyasagar Metropolitan College, Kolkata, West Bengal, India
| | - Krishnendu Acharya
- Department of Botany, University of Calcutta, Kolkata, West Bengal, India.
| |
Collapse
|
13
|
Daoui O, Elkhattabi S, Chtita S. Rational identification of small molecules derived from 9,10-dihydrophenanthrene as potential inhibitors of 3CL pro enzyme for COVID-19 therapy: a computer-aided drug design approach. Struct Chem 2022; 33:1667-1690. [PMID: 35818588 PMCID: PMC9261181 DOI: 10.1007/s11224-022-02004-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 06/23/2022] [Indexed: 01/11/2023]
Abstract
Small molecules such as 9,10-dihydrophenanthrene derivatives have remarkable activity toward inhibition of SARS-CoV-2 3CLpro and COVID-19 proliferation, which show a strong correlation between their structures and bioactivity. Therefore, these small compounds could be suitable for clinical pharmaceutical use against COVID-19. The objective of this study was to remodel the structures of 9,10-dihydrophenanthrene derivatives to achieve a powerful biological activity against 3CLpro and favorable pharmacokinetic properties for drug design and discovery. Therefore, by the use of bioinformatics techniques, we developed robust 3D-QSAR models that are capable of describing the structure-activity relationship for 46 molecules based on 9,10-dihydrophenanthrene derivatives using CoMFA/SE (R 2 = 0.97, Q 2 = 0.81, R 2 pred = 0.95, c R 2 p = 0.71) and CoMSIA/SEHDA (R 2 = 0.94, Q 2 = 0.76, R 2 pred = 0.91, c R 2 p = 0.65) techniques. Accordingly, 96 lead compounds were generated based on a template molecule that showed the highest observed activity in vitro (T40, pIC50 = 5.81) and predicted their activities and bioavailability in silico. The rational screening outputs of 3D-QSAR, Molecular docking, ADMET, and MM-GBSA led to the identification of 9 novel modeled molecules as potent noncovalent drugs against SARS-CoV-2-3CLpro. Finally, by molecular dynamics simulations, the stability and structural dynamics of 3CLpro free and complex (PDB code: 6LU7) were discussed in the presence of samples of 9,10-dihydrophenanthrene derivative in an aqueous environment. Overall, the retrosynthesis of the proposed drug compounds in this study and the evaluation of their bioactivity in vitro and in vivo may be interesting for designing and discovering a new drug effective against COVID-19. Supplementary Information The online version contains supplementary material available at 10.1007/s11224-022-02004-z.
Collapse
Affiliation(s)
- Ossama Daoui
- Laboratory of Engineering, Systems and Applications, National School of Applied Sciences, Sidi Mohamed Ben Abdellah-Fez University, BP Box 72, Fez, Morocco
| | - Souad Elkhattabi
- Laboratory of Engineering, Systems and Applications, National School of Applied Sciences, Sidi Mohamed Ben Abdellah-Fez University, BP Box 72, Fez, Morocco
| | - Samir Chtita
- Laboratory of Analytical and Molecular Chemistry, Faculty of Sciences Ben M’Sik, Hassan II University of Casablanca, B.P 7955 Casablanca, Morocco
| |
Collapse
|