1
|
Mishra S, Rout M, Singh MK, Dehury B, Pati S. Classical molecular dynamics simulation identifies catechingallate as a promising antiviral polyphenol against MPOX palmitoylated surface protein. Comput Biol Chem 2024; 110:108070. [PMID: 38678726 DOI: 10.1016/j.compbiolchem.2024.108070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 04/04/2024] [Accepted: 04/06/2024] [Indexed: 05/01/2024]
Abstract
Cumulative global prevalence of the emergent monkeypox (MPX) infection in the non-endemic countries has been professed as a global public health predicament. Lack of effective MPX-specific treatments sets the baseline for designing the current study. This research work uncovers the effective use of known antiviral polyphenols against MPX viral infection, and recognises their mode of interaction with the target F13 protein, that plays crucial role in formation of enveloped virions. Herein, we have employed state-of-the-art machine learning based AlphaFold2 to predict the three-dimensional structure of F13 followed by molecular docking and all-atoms molecular dynamics (MD) simulations to investigate the differential mode of F13-polyphenol interactions. Our extensive computational approach identifies six potent polyphenols Rutin, Epicatechingallate, Catechingallate, Quercitrin, Isoquecitrin and Hyperoside exhibiting higher binding affinity towards F13, buried inside a positively charged binding groove. Intermolecular contact analysis of the docked and MD simulated complexes divulges three important residues Asp134, Ser137 and Ser321 that are observed to be involved in ligand binding through hydrogen bonds. Our findings suggest that ligand binding induces minor conformational changes in F13 to affect the conformation of the binding site. Concomitantly, essential dynamics of the six-MD simulated complexes reveals Catechin gallate, a known antiviral agent as a promising polyphenol targeting F13 protein, dominated with a dense network of hydrophobic contacts. However, assessment of biological activities of these polyphenols need to be confirmed through in vitro and in vivo assays, which may pave the way for development of new novel antiviral drugs.
Collapse
Affiliation(s)
- Sarbani Mishra
- Bioinformatics Division, ICMR-Regional Medical Research Centre, Nalco Square, Chandrasekharpur, Bhubaneswar, Odisha 751023, India
| | - Madhusmita Rout
- Bioinformatics Division, ICMR-Regional Medical Research Centre, Nalco Square, Chandrasekharpur, Bhubaneswar, Odisha 751023, India
| | - Mahender Kumar Singh
- Data Science Laboratory, National Brain Research Centre, Gurgaon, Haryana 122052, India
| | - Budheswar Dehury
- Bioinformatics Division, ICMR-Regional Medical Research Centre, Nalco Square, Chandrasekharpur, Bhubaneswar, Odisha 751023, India; Department of Bioinformatics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, India.
| | - Sanghamitra Pati
- Bioinformatics Division, ICMR-Regional Medical Research Centre, Nalco Square, Chandrasekharpur, Bhubaneswar, Odisha 751023, India.
| |
Collapse
|
2
|
Paymal SB, Barale SS, Supanekar SV, Sonawane KD. Structure based virtual screening, molecular dynamic simulation to identify the oxadiazole derivatives as inhibitors of Enterococcus D-Ala-D-Ser ligase for combating vancomycin resistance. Comput Biol Med 2023; 159:106965. [PMID: 37119552 DOI: 10.1016/j.compbiomed.2023.106965] [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: 12/01/2022] [Revised: 04/03/2023] [Accepted: 04/19/2023] [Indexed: 05/01/2023]
Abstract
Vancomycin resistance in enterococci mainly arises due to alteration in terminal peptidoglycan dipeptide. A comprehensive structural analysis for substrate specificity of dipeptide modifying d-Alanine: d-Serine ligase (Ddls) is essential to screen its inhibitors for combating vancomycin resistance. In this study modeled 3D structure of EgDdls from E. gallinarum was used for structure based virtual screening (SBVS) of oxadiazole derivatives. Initially, fifteen oxadiazole derivatives were identified as inhibitors at the active site of EgDdls from PubChem database. Further, four EgDdls inhibitors were evaluated using pharmacokinetic profile and molecular docking. The results of molecular docking showed that oxadiazole inhibitors could bind preferentially at ATP binding pocket with the lowest binding energy. Further, molecular dynamics simulation results showed stable behavior of EgDdls in complex with screened inhibitors. The residues Phe172, Lys174, Glu217, Phe292, and Asn302 of EgDdls were mainly involved in interactions with screened inhibitors. Furthermore, MM-PBSA calculation showed electrostatic and van der Waals interactions mainly contribute to overall binding energy. The PCA analysis showed motion of central domain and omega loop of EgDdls. This is involved in the formation of native dipeptide and stabilized after binding of 2-(1-(Ethylsulfonyl) piperidin-4-yl)-5-(furan-2-yl)-1,3,4-oxadiazole, which could be reason for the inhibition of EgDdls. Hence, in this study we have screened inhibitors of EgDdls which could be useful to alleviate the vancomycin resistance problem in enterococci, involved in hospital-acquired infections, especially urinary tract infections (UTI).
Collapse
Affiliation(s)
- Sneha B Paymal
- Department of Microbiology, Shivaji University, Vidyanagar, Kolhapur, 416004, Maharashtra, India; Rayat Institute of Research and Development (RIRD), Satara, 415001, Maharashtra, India
| | - Sagar S Barale
- Structural Bioinformatics Unit, Department of Biochemistry, Shivaji University, Vidyanagar, Kolhapur, 416004, Maharashtra, India
| | | | - Kailas D Sonawane
- Structural Bioinformatics Unit, Department of Biochemistry, Shivaji University, Vidyanagar, Kolhapur, 416004, Maharashtra, India; Department of Microbiology, Shivaji University, Vidyanagar, Kolhapur, 416004, Maharashtra, India; Department of Chemistry, Shivaji University, Vidyanagar, Kolhapur, 416004, Maharashtra, India.
| |
Collapse
|
3
|
Al-Sehemi AG, Parulekar RS, Pannipara M, P P MA, Zubaidha PK, Bhatia MS, Mohanta TK, Al-Harrasi A. In silico evaluation of NO donor heterocyclic vasodilators as SARS-CoV-2 M pro protein inhibitor. J Biomol Struct Dyn 2023; 41:280-297. [PMID: 34809523 DOI: 10.1080/07391102.2021.2005682] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) which causes COVID-19 disease has been exponentially increasing throughout the world. The mortality rate is increasing gradually as effective treatment is unavailable to date. In silico based screening for novel testable hypotheses on SARS-CoV-2 Mpro protein to discover the potential lead drug candidate is an emerging area along with the discovery of a vaccine. Administration of NO-releasing agents, NO inducers or the NO gas itself may be useful as therapeutics in the treatment of SARS-CoV-2. In the present study, a 3D structure of SARS-CoV-2 Mpro protein was used for the rational setting of inhibitors to the binding pocket of enzyme which proposed that phenyl furoxan derivative gets efficiently dock in the target pocket. Molecular docking and molecular dynamics simulations helped to investigate possible effective inhibitor candidates bound to SARS-CoV-2 Mpro substrate binding pocket. Molecular mechanics Poisson-Boltzmann surface area (MM/PBSA) calculations revealed energetic contributions of active site residues of Mpro in binding with most stable proposed NO donor heterocyclic vasodilator inhibitor molecules. Furthermore, principal component analysis (PCA) showed that the NO donor heterocyclic inhibitor molecules 14, 16, 18 and 19 was strongly bound to catalytic core of SARS-CoV-2 Mpro protein, limiting its movement to form stable complex as like control. Thus, overall in silico investigations revealed that 5-oxopiperazine-2-carboxylic acid coupled furoxan derivatives was found to be key pharmacophore in drug design for the treatment of SARS-CoV-2, a global pandemic disease with a dual mechanism of action as NO donor and a worthwhile ligand to act as SARS-CoV-2 Mpro protein inhibitor.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Abdullah G Al-Sehemi
- Research center for Advanced Materials Science, King Khalid University, Abha, Saudi Arabia.,Department of Chemistry, King Khalid University, Abha, Saudi Arabia
| | - Rishikesh S Parulekar
- Department of Pharmaceutical Chemistry, Bharati Vidyapeeth College of Pharmacy, Kolhapur, Maharashtra, India
| | - Mehboobali Pannipara
- Research center for Advanced Materials Science, King Khalid University, Abha, Saudi Arabia.,Department of Chemistry, King Khalid University, Abha, Saudi Arabia
| | - Manzur Ali P P
- Department of Biotechnology, MES College, Marampally, Kerala, India
| | | | - Manish S Bhatia
- Department of Pharmaceutical Chemistry, Bharati Vidyapeeth College of Pharmacy, Kolhapur, Maharashtra, India
| | - Tapan Kumar Mohanta
- Natural and Medical Sciences Research Center, University of Nizwa, Nizwa, Oman
| | - Ahmed Al-Harrasi
- Natural and Medical Sciences Research Center, University of Nizwa, Nizwa, Oman
| |
Collapse
|
4
|
Basha GM, Parulekar RS, Al-Sehemi AG, Pannipara M, Siddaiah V, Kumari S, Choudhari PB, Tamboli Y. Design and in silico investigation of novel Maraviroc analogues as dual inhibition of CCR-5/SARS-CoV-2 M pro. J Biomol Struct Dyn 2022; 40:11095-11110. [PMID: 34308790 DOI: 10.1080/07391102.2021.1955742] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
A sudden increase in life-threatening COVID-19 infections around the world inflicts global crisis and emotional trauma. In current study two druggable targets, namely SARS-COV-2 Mpro and CCR-5 were selected due to their significant nature in the viral life cycle and cytokine molecular storm respectively. The systematic drug repurposing strategy has been utilized to recognize inhibitory mechanism through extensive in silico investigation of novel Maraviroc analogues as promising inhibitors against SARS-CoV-2 Mpro and CCR-5. The dual inhibition specificity approach implemented in present study using molecular docking, molecular dynamics (MD), principal component analysis (PCA), free energy landscape (FEL) and MM/PBSA binding energy studies. The proposed Maraviroc analogues obtained from in silico investigation could be easily synthesized and constructive in developing significant drug against COVID-19 pandemic, with essentiality of their in vivo/in vitro evaluation to affirm the conclusions of this study. This will further fortify the concept of single drug targeting dual inhibition mechanism for treatment of COVID-19 infection and complications.
Collapse
Affiliation(s)
- G Mahaboob Basha
- Department of Organic Chemistry, Foods, Drugs and Water, College of Science and Technology, Andhra University, Visakhapatnam, India
| | - Rishikesh S Parulekar
- Department of Pharmaceutical Chemistry, BharatiVidyapeeth College of Pharmacy, Kolhapur, India
| | - Abdullah G Al-Sehemi
- Research Center for Advanced Materials Science, King Khalid University, Abha, Saudi Arabia.,Department of Chemistry, King Khalid University, Abha, Saudi Arabia
| | - Mehboobali Pannipara
- Research Center for Advanced Materials Science, King Khalid University, Abha, Saudi Arabia.,Department of Chemistry, King Khalid University, Abha, Saudi Arabia
| | - Vidavalur Siddaiah
- Department of Organic Chemistry, Foods, Drugs and Water, College of Science and Technology, Andhra University, Visakhapatnam, India
| | - Sunanda Kumari
- Department of Microbiology, Andhra University, Visakhapatnam, India
| | - Prafulla B Choudhari
- Department of Pharmaceutical Chemistry, BharatiVidyapeeth College of Pharmacy, Kolhapur, India
| | | |
Collapse
|
5
|
Ilimaquinone (Marine Sponge Metabolite) Induces Apoptosis in HCT-116 Human Colorectal Carcinoma Cells via Mitochondrial-Mediated Apoptosis Pathway. Mar Drugs 2022; 20:md20090582. [PMID: 36135771 PMCID: PMC9503335 DOI: 10.3390/md20090582] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/03/2022] [Accepted: 09/15/2022] [Indexed: 12/05/2022] Open
Abstract
Ilimaquinone (IQ), a metabolite found in marine sponges, has been reported to have a number of biological properties, including potential anticancer activity against colon cancer. However, no clear understanding of the precise mechanism involved is known. The aim of this study was to examine the molecular mechanism by which IQ acts on HCT-116 cells. The anticancer activity of IQ was investigated by means of a cell viability assay followed by the determination of induction of apoptosis by means of the use of acridine orange–ethidium bromide (AO/EB) staining, Annexin V/PI double staining, DNA fragmentation assays, and TUNEL assays. The mitochondrial membrane potential (ΔΨm) was detected using the JC-1 staining technique, and the apoptosis-associated proteins were analyzed using real-time qRT-PCR. A molecular docking study of IQ with apoptosis-associated proteins was also conducted in order to assess the interaction between IQ and them. Our results suggest that IQ significantly suppressed the viability of HCT-116 cells in a dose-dependent manner. Fluorescent microscopy, flow cytometry, DNA fragmentation and the TUNEL assay in treated cells demonstrated apoptotic death mode. As an additional confirmation of apoptosis, the increased level of caspase-3 and caspase-9 expression and the downregulation of Bcl-2 and mitochondrial dysfunction were observed in HCT-116 cells after treatment with IQ, which was accompanied by a decrease in mitochondrial membrane potential (ΔΨm). Overall, the results of our studies demonstrate that IQ could trigger mitochondria-mediated apoptosis as demonstrated by a decrease in ΔΨm, activation of caspase-9/-3, damage of DNA and a decrease in the proportion of Bcl-2 through the mitochondrial-mediated apoptosis pathway.
Collapse
|
6
|
Parulekar RS, Sonawane KD. Structure elucidation study of aminoglycoside phosphotransferase from B. cereus sensu lato: a comprehensive outlook for drug discovery. Struct Chem 2022. [DOI: 10.1007/s11224-022-02040-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
|
7
|
Thakur M, Parulekar RS, Barale SS, Sonawane KD, Muniyappa K. Interrogating the substrate specificity landscape of UvrC reveals novel insights into its non-canonical function. Biophys J 2022; 121:3103-3125. [PMID: 35810330 PMCID: PMC9463653 DOI: 10.1016/j.bpj.2022.07.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 05/23/2022] [Accepted: 07/07/2022] [Indexed: 11/29/2022] Open
Abstract
Although it is relatively unexplored, accumulating data highlight the importance of tripartite crosstalk between nucleotide excision repair (NER), DNA replication, and recombination in the maintenance of genome stability; however, elucidating the underlying mechanisms remains challenging. While Escherichia coli uvrA and uvrB can fully complement polAΔ cells in DNA replication, uvrC attenuates this alternative DNA replication pathway, but the exact mechanism by which uvrC suppresses DNA replication is unknown. Furthermore, the identity of bona fide canonical and non-canonical substrates for UvrCs are undefined. Here, we reveal that Mycobacterium tuberculosis UvrC (MtUvrC) strongly binds to, and robustly cleaves, key intermediates of DNA replication/recombination as compared with the model NER substrates. Notably, inactivation of MtUvrC ATPase activity significantly attenuated its endonuclease activity, thus suggesting a causal link between these two functions. We built an in silico model of the interaction of MtUvrC with the Holliday junction (HJ), using a combination of homology modeling, molecular docking, and molecular dynamic simulations. The model predicted residues that were potentially involved in HJ binding. Six of these residues were mutated either singly or in pairs, and the resulting MtUvrC variants were purified and characterized. Among them, residues Glu595 and Arg597 in the helix-hairpin-helix motif were found to be crucial for the interaction between MtUvrC and HJ; consequently, mutations in these residues, or inhibition of ATP hydrolysis, strongly abrogated its DNA-binding and endonuclease activities. Viewed together, these findings expand the substrate specificity landscape of UvrCs and provide crucial mechanistic insights into the interplay between NER and DNA replication/recombination.
Collapse
Affiliation(s)
- Manoj Thakur
- Department of Biochemistry, Indian Institute of Science, Bengaluru, India.
| | | | - Sagar S Barale
- Structural Bioinformatics Unit, Shivaji University, Kolhapur, India
| | - Kailas D Sonawane
- Department of Microbiology, Shivaji University, Kolhapur, India; Structural Bioinformatics Unit, Shivaji University, Kolhapur, India
| | - Kalappa Muniyappa
- Department of Biochemistry, Indian Institute of Science, Bengaluru, India.
| |
Collapse
|
8
|
In Silico Exploration of Binding Potentials of Anti SARS-CoV-1 Phytochemicals against Main Protease of SARS-CoV-2. JOURNAL OF SAUDI CHEMICAL SOCIETY 2022. [PMCID: PMC8915462 DOI: 10.1016/j.jscs.2022.101453] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
9
|
Barale SS, Ghane SG, Sonawane KD. Purification and characterization of antibacterial surfactin isoforms produced by Bacillus velezensis SK. AMB Express 2022; 12:7. [PMID: 35084596 PMCID: PMC8795249 DOI: 10.1186/s13568-022-01348-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 01/16/2022] [Indexed: 11/10/2022] Open
Abstract
Bacillus velezensis SK having broad-spectrum antimicrobial activity has been isolated from soil. The efficient extraction of antimicrobial compounds produced in various mediums has been done using Diaion HP-20 resin. Further, characterization of an antimicrobial compound by TLC, FTIR, in-situ bioautography analysis revealed the presence of cyclic lipopeptides, which is then purified by the combination of silica gel, size exclusion, dual gradient, and RP-HPLC chromatography techniques. Growth kinetic studies showed that Bacillus velezensis SK produces a mixture of lipopeptides (1.33 gL-1). The lipopeptide exhibits good pH (2-10) and temperature stability up to 80 °C. LC-ESI-MS analysis of partially purified lipopeptide identified variant of surfactin, further analysis of purified chromatographic fractions revealed the occurrence of most abundant C15-surfactin homologues (m/z 1036.72 Da). The isolated surfactin exhibits good antimicrobial activity (1600 AU/ml) against drug-resistant food-born B. cereus and human pathogen Staphylococcus aureus. Hence, identified strain B. velezensis SK and its potent antibacterial surfactin lipopeptide could be used in various food and biomedical applications.
Collapse
|
10
|
Haddaji F, Papetti A, Noumi E, Colombo R, Deshpande S, Aouadi K, Adnan M, Kadri A, Selmi B, Snoussi M. Bioactivities and in silico study of Pergularia tomentosa L. phytochemicals as potent antimicrobial agents targeting type IIA topoisomerase, TyrRS, and Sap1 virulence proteins. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:25349-25367. [PMID: 33454827 DOI: 10.1007/s11356-020-11946-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 12/02/2020] [Indexed: 06/12/2023]
Abstract
Pergularia tomentosa L. (P. tomentosa) has been largely used in Tunisian folk medicine as remedies against skin diseases, asthma, and bronchitis. The main objectives of this study were to identify phytochemical compounds that have antioxidant and antimicrobial properties from the stem, leaves, and fruit crude methanolic extracts of P. tomentosa, and to search for tyrosyl-tRNA synthetase (TyrRS), topoisomerase type IIA, and Candidapepsin-1 (SAP1) enzyme inhibitors through molecular docking study. Phytochemical quantification revealed that fruit and leaves extracts displayed the highest total flavonoids (582 mg QE/g Ex; 219 mg QE/g Ex) and tannins content (375 mg TAE/g Ex; 216 mg TAE/g Ex), also exhibiting significant scavenging activity to decrease free radicals for ABTS, DPPH, β-carotene, and FRAP assay with IC50 values (> 1 mg/mL). Additionally, promising antimicrobial activities towards different organs have been observed against several bacteria and Candida strains. From the liquid chromatography-mass spectrometry (LC-MS) analysis, five polyphenolic compounds, namely digitoxigenin, digitonin glycoside and calactina in the leaves, kaempferol in the fruit, and calotropagenin in the stems, were identified. They were also analyzed for their drug likeliness, based on computational methods. Molecular docking study affirmed that the binding affinity of calactin and actodigin to the active site of TyrRS, topoisomerase type IIA, and SAP1 target virulence proteins was the highest among the examined dominant compounds. Therefore, this study indicated that P. tomentosa methanolic extracts displayed great potential to become a potent antimicrobial agent and might be a promising source for therapeutic and nutritional functions. These phytocompounds could be further promoted as a candidate for drug discovery and development.
Collapse
Affiliation(s)
- Fatma Haddaji
- Laboratory of Genetics, Biodiversity and Valorization of Bio-resources (LR11ES41), University of Monastir, Higher Institute of Biotechnology of Monastir, Avenue Tahar Haddad, BP74, 5000, Monastir, Tunisia
| | - Adele Papetti
- Nutraceutical & Food Chemical-Toxicological Analysis Laboratory, Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100, Pavia, Italy
| | - Emira Noumi
- Department of Biology, College of Science, University of Ha'il, P.O. 2440, Hail, Saudi Arabia
- Laboratory of Bioressources: Integrative Biology and Valorization, (LR14-ES06), University of Monastir, Higher Institute of Biotechnology of Monastir, Avenue Tahar Haddad, BP 74, 5000, Monastir, Tunisia
| | - Raffaella Colombo
- Nutraceutical & Food Chemical-Toxicological Analysis Laboratory, Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100, Pavia, Italy
| | - Sumukh Deshpande
- Central Biotechnology Services, College of Biomedical and Life Sciences, Cardiff University, Cardiff, Wales, CF14 4XN, UK
| | - Kaïss Aouadi
- Faculty of Science and Arts in Baljurashi, Albaha University, P.O. Box 1988, Albaha, Saudi Arabia
- Faculty of Science of Monastir, Laboratory of Hetrocyclic Chemistry, Natural Products and Reactivity, Avenue of the Environment, University of Monastir, 5019, Monastir, Tunisia
| | - Mohd Adnan
- Department of Biology, College of Science, University of Ha'il, P.O. 2440, Hail, Saudi Arabia
| | - Adel Kadri
- Faculty of Science of Sfax, Department of Chemistry, University of Sfax, B.P. 1171, 3000, Sfax, Tunisia
- Department of Chemistry, College of Science, Qassim University, Buraidah, 51452, Saudi Arabia
| | - Boulbaba Selmi
- Laboratory of Bioressources: Integrative Biology and Valorization, (LR14-ES06), University of Monastir, Higher Institute of Biotechnology of Monastir, Avenue Tahar Haddad, BP 74, 5000, Monastir, Tunisia
| | - Mejdi Snoussi
- Department of Biology, College of Science, University of Ha'il, P.O. 2440, Hail, Saudi Arabia.
| |
Collapse
|
11
|
Thymus musilii Velen. as a promising source of potent bioactive compounds with its pharmacological properties: In vitro and in silico analysis. ARAB J CHEM 2020. [DOI: 10.1016/j.arabjc.2020.06.032] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
12
|
Al-Sehemi AG, Pannipara M, Parulekar RS, Patil O, Choudhari PB, Bhatia MS, Zubaidha PK, Tamboli Y. Potential of NO donor furoxan as SARS-CoV-2 main protease (M pro) inhibitors: in silico analysis. J Biomol Struct Dyn 2020; 39:5804-5818. [PMID: 32643550 PMCID: PMC7441807 DOI: 10.1080/07391102.2020.1790038] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The sharp spurt in positive cases of novel coronavirus-19 (SARS-CoV-2) worldwide has created a big threat to human. In view to expedite new drug leads for COVID-19, Main Proteases (Mpro) of novel Coronavirus (SARS‐CoV‐2) has emerged as a crucial target for this virus. Nitric oxide (NO) inhibits the replication cycle of SARS-CoV. Inhalation of nitric oxide is used in the treatment of severe acute respiratory syndrome. Herein, we evaluated the phenyl furoxan, a well-known exogenous NO donor to identify the possible potent inhibitors through in silico studies such as molecular docking as per target analysis for candidates bound to substrate binding pocket of SARS-COV-2 Mpro. Molecular dynamics (MD) simulations of most stable docked complexes (Mpro-22 and Mpro-26) helped to confirm the notable conformational stability of these docked complexes under dynamic state. Furthermore, Molecular mechanics Poisson-Boltzmann surface area (MM-PBSA) calculations revealed energetic contributions of key residues of Mpro in binding with potent furoxan derivatives 22, 26. In the present study to validate the molecular docking, MD simulation and MM-PBSA results, crystal structure of Mpro bound to experimentally known inhibitor X77 was used as control and the obtained results are presented herein. We envisaged that spiro-isoquinolino-piperidine-furoxan moieties can be used as effective ligand for SARS-CoV-2 Mpro inhibition due to the presence of key isoquinolino-piperidine skeleton with additional NO effect. Communicated by Ramaswamy H. Sarma
Collapse
Affiliation(s)
- Abdullah G Al-Sehemi
- Research center for Advanced Materials Science, King Khalid University, Abha, Saudi Arabia.,Department of Chemistry, King Khalid University, Abha, Saudi Arabia
| | - Mehboobali Pannipara
- Research center for Advanced Materials Science, King Khalid University, Abha, Saudi Arabia.,Department of Chemistry, King Khalid University, Abha, Saudi Arabia
| | - Rishikesh S Parulekar
- Department of Pharmaceutical Chemistry, Bharati Vidyapeeth College of Pharmacy, Kolhapur, Maharashtra, India
| | - Omkar Patil
- Department of Pharmaceutical Chemistry, Bharati Vidyapeeth College of Pharmacy, Kolhapur, Maharashtra, India
| | - Prafulla B Choudhari
- Department of Pharmaceutical Chemistry, Bharati Vidyapeeth College of Pharmacy, Kolhapur, Maharashtra, India
| | - M S Bhatia
- Department of Pharmaceutical Chemistry, Bharati Vidyapeeth College of Pharmacy, Kolhapur, Maharashtra, India
| | - P K Zubaidha
- School of Chemical Sciences, SRTM University, Nanded, Maharashtra, India
| | - Yasinalli Tamboli
- School of Chemical Sciences, SRTM University, Nanded, Maharashtra, India
| |
Collapse
|
13
|
Adnan M, Patel M, Deshpande S, Alreshidi M, Siddiqui AJ, Reddy MN, Emira N, De Feo V. Effect of Adiantum philippense Extract on Biofilm Formation, Adhesion With Its Antibacterial Activities Against Foodborne Pathogens, and Characterization of Bioactive Metabolites: An in vitro-in silico Approach. Front Microbiol 2020; 11:823. [PMID: 32477292 PMCID: PMC7237743 DOI: 10.3389/fmicb.2020.00823] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Accepted: 04/06/2020] [Indexed: 12/13/2022] Open
Abstract
Adiantum philippense (A. philippense), an ethnomedicinally important fern, has become an interesting herb in the search for novel bioactive metabolites, which can also be used as therapeutic agents. Primarily, in this study, A. philippense crude extract was screened for its phytochemical constituents, antagonistic potential, and effect on bacterial adhesion and biofilm formation against common food pathogens. Phytochemical profiling of A. philippense was carried out by using High Resolution-Liquid Chromatography and Mass Spectroscopy (HR-LCMS) followed by antibacterial activity via agar cup/well diffusion, broth microdilution susceptibility methods, and growth curve analysis. Antibiofilm potency and efficacy were assessed on the development, formation, and texture of biofilms through light microscopy, fluorescent microscopy, scanning electron microscopy, and the assessment of exopolysaccharide production. Correspondingly, a checkerboard test was performed to evaluate the combinatorial effect of A. philippense and chloramphenicol. Lastly, molecular docking studies of identified phytochemicals with adhesin proteins of tested food pathogens, which helps the bacteria in surface attachment and leads to biofilm formation, were assessed. A. philippense crude extract was found to be active against all tested food pathogens, displaying the rapid time-dependent kinetics of bacterial killing. A. philippense crude extract also impedes the biofilm matrix by reducing the total content of exopolysaccharide, and, likewise, the microscopic images revealed a great extent of disruption in the architecture of biofilms. A synergy was observed between A. philippense crude extract and chloramphenicol for E. coli, S. aureus, and P. aeruginosa, whereas an additive effect was observed for S. flexneri. Various bioactive phytochemicals were categorized from A. philippense crude extract using HR-LCMS. The molecular docking of these identified phytochemicals was interrelated with the active site residues of adhesin proteins, IcsA, Sortase A, OprD, EspA, and FimH from S. flexneri, S. aureus, P. aeruginosa, and E. coli, respectively. Thus, our findings represent the bioactivity and potency of A. philippense crude extract against food pathogens not only in their planktonic forms but also against/in biofilms for the first time. We have also correlated these findings with the possible mechanism of biofilm inhibition via targeting adhesin proteins, which could be explored further to design new bioactive compounds against biofilm producing foodborne bacterial pathogens.
Collapse
Affiliation(s)
- Mohd Adnan
- Department of Biology, College of Science, University of Ha'il, Ha'il, Saudi Arabia
| | - Mitesh Patel
- Department of Biosciences, Bapalal Vaidya Botanical Research Centre, Veer Narmad South Gujarat University, Surat, India
| | - Sumukh Deshpande
- Central Biotechnology Services, College of Biomedical and Life Sciences, Cardiff University, Cardiff, United Kingdom
| | - Mousa Alreshidi
- Department of Biology, College of Science, University of Ha'il, Ha'il, Saudi Arabia
| | - Arif Jamal Siddiqui
- Department of Biology, College of Science, University of Ha'il, Ha'il, Saudi Arabia
| | - Mandadi Narsimha Reddy
- Department of Biosciences, Bapalal Vaidya Botanical Research Centre, Veer Narmad South Gujarat University, Surat, India
| | - Noumi Emira
- Department of Biology, College of Science, University of Ha'il, Ha'il, Saudi Arabia
| | - Vincenzo De Feo
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, Fisciano, Italy
| |
Collapse
|
14
|
Antibiotic resistance and inhibition mechanism of novel aminoglycoside phosphotransferase APH(5) from B. subtilis subsp. subtilis strain RK. Braz J Microbiol 2019; 50:887-898. [PMID: 31401782 DOI: 10.1007/s42770-019-00132-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 08/02/2019] [Indexed: 01/05/2023] Open
Abstract
Bacterial resistance towards aminoglycoside antibiotics mainly occurs because of aminoglycoside phosphotransferases (APHs). It is thus necessary to provide a rationale for focusing inhibitor development against APHs. The nucleotide triphosphate (NTP) binding site of eukaryotic protein kinases (ePKs) is structurally conserved with APHs. However, ePK inhibitors cannot be used against APHs due to cross reactivity. Thus, understanding bacterial resistance at the atomic level could be useful to design new inhibitors against such resistant pathogens. Hence, we carried out in vitro studies of APH from newly deposited multidrug-resistant organism Bacillus subtilis subsp. subtilis strain RK. Enzymatic modification studies of different aminoglycoside antibiotics along with purification and characterization revealed a novel class of APH, i.e., APH(5), with molecular weight 27 kDa approximately. Biochemical analysis of virtually screened inhibitor ZINC71575479 by coupled spectrophotometric assay showed complete enzymatic inhibition of purified APH(5). In silico toxicity study comparison of ZINC71575479 with known inhibitor of APH, i.e., tyrphostin AG1478, predicted its acceptable values for 96 h fathead minnow LC50, 48 h Tetrahymena pyriformis IGC50, oral rat LD50, and developmental toxicity using different QSAR methodologies. Thus, the present study gives novel insight into the aminoglycoside resistance and inhibition mechanism of APH(5) by applying experimental and computational techniques synergistically.
Collapse
|
15
|
Swain SS, Paidesetty SK, Dehury B, Sahoo J, Vedithi SC, Mahapatra N, Hussain T, Padhy RN. Molecular docking and simulation study for synthesis of alternative dapsone derivative as a newer antileprosy drug in multidrug therapy. J Cell Biochem 2018; 119:9838-9852. [DOI: 10.1002/jcb.27304] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 06/28/2018] [Indexed: 12/24/2022]
Affiliation(s)
- Shasank S. Swain
- Central Research Laboratory, Institute of Medical Sciences and Sum Hospital, Siksha “O” Anusandhan (Deemed to be University) Bhubaneswar Odisha India
- NCDs Division ICMR‐Regional Medical Research Centre Bhubaneswar Odisha India
| | - Sudhir K. Paidesetty
- Department of Pharmaceutical Chemistry School of Pharmaceutical Sciences, Siksha “O” Anusandhan (Deemed to be University) Bhubaneswar Odisha India
| | - Budheswar Dehury
- Biomedical Informatics Centre, ICMR‐Regional Medical Research Centre Bhubaneswar Odisha India
| | - Jyotirmaya Sahoo
- Department of Pharmaceutical Chemistry School of Pharmaceutical Sciences, Siksha “O” Anusandhan (Deemed to be University) Bhubaneswar Odisha India
| | - Sundeep Chaitanya Vedithi
- Schieffelin Institute of Health‐Research and Leprosy Centre (SIH R & LC), Karigiri Vellore Tamil Nadu India
- Department of Biochemistry University of Cambridge Cambridge UK
| | - Namita Mahapatra
- Biomedical Informatics Centre, ICMR‐Regional Medical Research Centre Bhubaneswar Odisha India
| | - Tahziba Hussain
- NCDs Division ICMR‐Regional Medical Research Centre Bhubaneswar Odisha India
| | - Rabindra N. Padhy
- Central Research Laboratory, Institute of Medical Sciences and Sum Hospital, Siksha “O” Anusandhan (Deemed to be University) Bhubaneswar Odisha India
| |
Collapse
|
16
|
Nadaf NH, Parulekar RS, Patil RS, Gade TK, Momin AA, Waghmare SR, Dhanavade MJ, Arvindekar AU, Sonawane KD. Biofilm inhibition mechanism from extract of Hymenocallis littoralis leaves. JOURNAL OF ETHNOPHARMACOLOGY 2018; 222:121-132. [PMID: 29698774 DOI: 10.1016/j.jep.2018.04.031] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 02/17/2018] [Accepted: 04/21/2018] [Indexed: 06/08/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Hymenocallis littoralis (Jacq.) Salisb. has been referred as beach spider lily and commonly known for its rich phytochemical diversity. Phytochemicals such as alkaloids, volatile constituents, phenols, flavonoids, flavonols extracted from different parts of these plants like bulbs, flowers, leaf, stem and root had been used in folk medicines from ancient times because of their excellent antimicrobial and antioxidant properties. The leaf and bulb extract of H. littoralis plant was traditionally used for wound healing. Alkaloids extracted from bulb of this plant possess anti-viral, anti-neoplastic and cytotoxic properties. However, these phytochemicals have also shown antibiofilm activity, which is considered as one of the important factor accountable for the drug resistance in microorganisms. Thus, the investigation of medicinal properties of H. littoralis could be useful to control biofilm producing pathogens. AIM OF THE STUDY Explore antimicrobial, antibiofilm and antioxidant potentials of H. littoralis against pathogenic microorganisms using experimental and computational biology approach. MATERIALS AND METHODS Phytochemical extraction from dried powder of H. littoralis leaves was done by solvent extraction using methanol. Antimicrobial and antibiofilm activities of leaves extract were carried out using agar well diffusion method, growth curve, minimum inhibitory concentration (MIC) and Scanning Electron Microscopy (SEM). Liquid Chromatography and Mass Spectroscopy (LCMS) technique was used for the identification of phytochemicals. Molecular docking studies of antibiofilm agents with adhesin proteins were performed using Autodock 4.2. Antioxidant activity of extract was carried out by FRAP assay. The noxious effect of extract was investigated by histological studies on rat skin. RESULTS The preliminary phytochemical analysis of methanolic leaves extract revealed the presence of alkaloids, flavonoids, terpenoid, glycosides, terpene, terpenoids and phenolics. The various phytochemicals such as Apigenin 7-(4'', 6'' diacetylalloside)-4'- alloside, Catechin 7-O- apiofuranoside, Emodic acid, Epicatechin 3-O- β-D-glucopyranoside, 4 - Methylesculetin, Methylisoeugenol, Quercetin 5,7,3',4'-tetramethyl ether 3-rutinoside, 4 - Methylumbelliferyl β-D- glucuronide were extracted, characterized and recognized from the leaves extract of H. littoralis. The identification of these phytochemicals was performed using LC-MS. The antimicrobial property of H. littoralis leaf extract was investigated against different pathogenic microorganisms. Out of these tested microorganisms, promising antibiofilm and antimicrobial activities were confirmed against S. aureus NCIM 2654 and C. albicans NCIM 3466 by using growth curve and SEM analysis. MIC of this leaf extract was identified as 45 µg/ml and 70 µg/ml for S. aureus NCIM 2654 and C. albicans NCIM 3466 respectively. The leaves extract also showed good antioxidant activity due to presence of phenols and flavonoids. Molecular docking of these identified antibiofilm components interacts with the active site residues of adhesin proteins, Sortase A and Als3 from S. aureus and C. albicans respectively. Histological studies of extracted phytochemicals revealed non-noxious effects on rat skin. CONCLUSION Thus, the present study revealed that the leaves extract of H. littoralis contains various phytochemicals having good extent of antimicrobial, antibiofilm and antioxidant properties. The in-vitro and in-silico results would be useful to design new lead compounds against biofilm producing pathogenic microorganisms.
Collapse
Affiliation(s)
- Naiem H Nadaf
- Department of Microbiology, Shivaji University, Vidyanagar, Kolhapur, 416004 Maharashtra, India
| | - Rishikesh S Parulekar
- Department of Microbiology, Shivaji University, Vidyanagar, Kolhapur, 416004 Maharashtra, India
| | - Rahul S Patil
- Department of Biochemistry, Shivaji University, Vidyanagar, Kolhapur, 416004 Maharashtra, India
| | - Trupti K Gade
- Department of Microbiology, Shivaji University, Vidyanagar, Kolhapur, 416004 Maharashtra, India
| | - Anjum A Momin
- Department of Microbiology, Shivaji University, Vidyanagar, Kolhapur, 416004 Maharashtra, India
| | - Shailesh R Waghmare
- Department of Microbiology, Shivaji University, Vidyanagar, Kolhapur, 416004 Maharashtra, India
| | - Maruti J Dhanavade
- Department of Microbiology, Shivaji University, Vidyanagar, Kolhapur, 416004 Maharashtra, India
| | - Akalpita U Arvindekar
- Department of Biochemistry, Shivaji University, Vidyanagar, Kolhapur, 416004 Maharashtra, India
| | - Kailas D Sonawane
- Department of Microbiology, Shivaji University, Vidyanagar, Kolhapur, 416004 Maharashtra, India; Department of Biochemistry, Shivaji University, Vidyanagar, Kolhapur, 416004 Maharashtra, India.
| |
Collapse
|
17
|
Parulekar RS, Sonawane KD. Insights into the antibiotic resistance and inhibition mechanism of aminoglycoside phosphotransferase from
Bacillus cereus
: In silico and in vitro perspective. J Cell Biochem 2018; 119:9444-9461. [DOI: 10.1002/jcb.27261] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 06/22/2018] [Indexed: 01/13/2023]
Affiliation(s)
| | - Kailas Dashrath Sonawane
- Department of Microbiology Shivaji University Kolhapur Maharashtra India
- Structural Bioinformatics Unit, Department of Biochemistry Shivaji University Kolhapur Maharashtra India
| |
Collapse
|
18
|
Nimbalkar P, Khedkar MA, Parulekar RS, Chandgude VK, Sonawane KD, Chavan PV, Bankar SB. Role of Trace Elements as Cofactor: An Efficient Strategy toward Enhanced Biobutanol Production. ACS SUSTAINABLE CHEMISTRY & ENGINEERING 2018; 6:9304-9313. [PMID: 30271690 PMCID: PMC6156106 DOI: 10.1021/acssuschemeng.8b01611] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 06/02/2018] [Indexed: 05/07/2023]
Abstract
Metabolic engineering has the potential to steadily enhance product titers by inducing changes in metabolism. Especially, availability of cofactors plays a crucial role in improving efficacy of product conversion. Hence, the effect of certain trace elements was studied individually or in combinations, to enhance butanol flux during its biological production. Interestingly, nickel chloride (100 mg L-1) and sodium selenite (1 mg L-1) showed a nearly 2-fold increase in solvent titer, achieving 16.13 ± 0.24 and 12.88 ± 0.36 g L-1 total solvents with yields of 0.30 and 0.33 g g-1, respectively. Subsequently, the addition time (screened entities) was optimized (8 h) to further increase solvent production up to 18.17 ± 0.19 and 15.5 ± 0.13 g L-1 by using nickel and selenite, respectively. A significant upsurge in butanol dehydrogenase (BDH) levels was observed, which reflected in improved solvent productions. Additionally, a three-dimensional structure of BDH was also constructed using homology modeling and subsequently docked with substrate, cofactor, and metal ion to investigate proper orientation and molecular interactions.
Collapse
Affiliation(s)
- Pranhita
R. Nimbalkar
- Department
of Bioproducts and Biosystems, School of Chemical Engineering, Aalto University P.O.
Box 16100, FI-00076 Aalto, Finland
- Department
of Chemical Engineering, Bharati Vidyapeeth
Deemed University College of Engineering, Pune 411043, India
| | - Manisha A. Khedkar
- Department
of Chemical Engineering, Bharati Vidyapeeth
Deemed University College of Engineering, Pune 411043, India
| | | | - Vijaya K. Chandgude
- Department
of Bioproducts and Biosystems, School of Chemical Engineering, Aalto University P.O.
Box 16100, FI-00076 Aalto, Finland
| | - Kailas D. Sonawane
- Department
of Microbiology, Shivaji University, Kolhapur 416004, India
- Department
of Biochemistry, Structural Bioinformatics Unit, Shivaji University, Kolhapur 416004, India
| | - Prakash V. Chavan
- Department
of Chemical Engineering, Bharati Vidyapeeth
Deemed University College of Engineering, Pune 411043, India
| | - Sandip B. Bankar
- Department
of Bioproducts and Biosystems, School of Chemical Engineering, Aalto University P.O.
Box 16100, FI-00076 Aalto, Finland
- E-mail: ; . Tel.: +358 505777898
| |
Collapse
|
19
|
Peptide Similarity Search Based and Virtual Screening Based Strategies to Identify Small Molecules to Inhibit CarD–RNAP Interaction in M. tuberculosis. Int J Pept Res Ther 2018. [DOI: 10.1007/s10989-018-9716-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
|