1
|
Putri IS, Shamsudin NF, Abdullah MA, Nurcholis M, Imran S, Yu CX, Tham CL, Mohd Aluwi MFF, Leong SW, Joko Raharjo S, Ibrahim Z, Islami D, Huq AM, Taher M, Rullah K. Theoretical investigation of selective inhibitory activity of chromone-based compounds against monoamine oxidase (MAO)-A and -B. J Biomol Struct Dyn 2024:1-18. [PMID: 39633610 DOI: 10.1080/07391102.2024.2436553] [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: 11/02/2023] [Accepted: 05/10/2024] [Indexed: 12/07/2024]
Abstract
Monoamine oxidase (MAO) is crucial for the breakdown of monoamine neurotransmitters, making it a promising target for treating neurodegenerative disorders, such as depression, Alzheimer's disease, and Parkinson's disease. In this study, we investigated the selective inhibitory activity of chromone-based compounds against MAO-A and MAO-B for neurodegenerative disease treatment. In literary sources, thirty chromone derivatives have been identified as potential ligands for MAO-A and MAO-B inhibitors. We utilized molecular docking to evaluate how the most active compound interacted with the targeted MAO-A and MAO-B. Compound 2 g, the most active for MAO-A, demonstrated a lower CDOCKER energy compared to the co-crystallized ligand. Meanwhile, compound 2f, the most active for MAO-B, showed a CDOCKER energy similar to the co-crystallized ligand and exhibited similar binding patterns. Furthermore, we constructed a quantitative structure-activity relationship (QSAR) model to predict the properties and estimate IC50 values for 30 chromone derivatives functioning as MAO-A and MAO-B inhibitors. The model predictions were validated against experimental measurements. Our 2D QSAR model demonstrated robustness, with a statistically significant non-cross-validated coefficient (r2 < 0.9), cross-validated correlation coefficient (q2 < 0.6), and predictive squared correlation coefficient (r2pred < 0.8). Additionally, MD simulations confirmed the stable binding of compounds 2 g and 2f with MAO-A and MAO-B, respectively, displaying substantial binding energy. The most effective pharmacophore model identified key features, such as hydrogen bond acceptors and hydrophobic interactions, that contribute significantly to inhibitory potency. This study offers valuable insight into the selection of compounds with improved selectivity for MAO inhibition.
Collapse
Affiliation(s)
- Intan Salsabila Putri
- Department of Food Science and Technology, Faculty of Agricultural Technology, Brawijaya University, Malang, Indonesia
- Drug Discovery and Synthetic Chemistry Research Group, Department of Pharmaceutical Chemistry, Kulliyyah of Pharmacy, International Islamic University Malaysia, Kuantan, Pahang, Malaysia
| | - Nur Farisya Shamsudin
- Drug Discovery and Synthetic Chemistry Research Group, Department of Pharmaceutical Chemistry, Kulliyyah of Pharmacy, International Islamic University Malaysia, Kuantan, Pahang, Malaysia
| | - Maryam Aisyah Abdullah
- Drug Discovery and Synthetic Chemistry Research Group, Department of Pharmaceutical Chemistry, Kulliyyah of Pharmacy, International Islamic University Malaysia, Kuantan, Pahang, Malaysia
| | - Mochamad Nurcholis
- Department of Food Science and Technology, Faculty of Agricultural Technology, Brawijaya University, Malang, Indonesia
| | - Syahrul Imran
- Atta-ur-Rahman Institute for Natural Products Discovery (AuRIns), Universiti Teknologi MARA, Bandar Puncak Alam, Selangor Darul Ehsan, Malaysia
| | - Chai Xin Yu
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Chau Ling Tham
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | | | - Sze-Wei Leong
- Department of Chemistry, Faculty of Science, University of Malaya, Kuala Lumpur, Malaysia
| | - Sentot Joko Raharjo
- Academic of Pharmacy and Food Analysis of Putra Indonesia Malang, East Java, Indonesia
| | - Zalikha Ibrahim
- Drug Discovery and Synthetic Chemistry Research Group, Department of Pharmaceutical Chemistry, Kulliyyah of Pharmacy, International Islamic University Malaysia, Kuantan, Pahang, Malaysia
| | - Deri Islami
- Faculty of Pharmacy and Health Sciences, Universitas Abdurrab, Pekanbaru, Riau, Indonesia
| | - Akm Moyeenul Huq
- Centre for Bio-Aromatic Research, Universiti Malaysia Pahang, Gambang, Pahang, Malaysia
| | - Muhammad Taher
- Department of Pharmaceutical Technology, Kulliyyah of Pharmacy, International Islamic University Malaysia, Kuantan, Pahang, Malaysia
| | - Kamal Rullah
- Drug Discovery and Synthetic Chemistry Research Group, Department of Pharmaceutical Chemistry, Kulliyyah of Pharmacy, International Islamic University Malaysia, Kuantan, Pahang, Malaysia
| |
Collapse
|
2
|
Prichard K, Chau N, Xue J, Krauss M, Sakoff JA, Gilbert J, Bahnik C, Muehlbauer M, Radetzki S, Robinson PJ, Haucke V, McCluskey A. Inhibition Clathrin Mediated Endocytosis: Pitstop 1 and Pitstop 2 Chimeras. ChemMedChem 2024; 19:e202400253. [PMID: 38894585 DOI: 10.1002/cmdc.202400253] [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: 04/08/2024] [Revised: 06/18/2024] [Accepted: 06/18/2024] [Indexed: 06/21/2024]
Abstract
Twenty-five chimera compounds of Pitstop 1 and 2 were synthesised and screened for their ability to block the clathrin terminal domain-amphiphysin protein-protein interaction (NTD-PPI using an ELISA) and clathrin mediated endocytosis (CME) in cells. Library 1 was based on Pitstop 2, but no notable clathrin PPI or in-cell activity was observed. With the Pitstop 1, 16 analogues were produced with 1,8-naphthalic imide core as a foundation. Analogues with methylene spaced linkers and simple amides showed a modest to good range of PPI inhibition (7.6-42.5 μM, naphthyl 39 and 4-nitrophenyl 40 respectively) activity. These data reveal the importance of the naphthalene sulfonate moiety, with no des-SO3 analogue displaying PPI inhibition. This was consistent with the observed analogue docked poses within the clathrin terminal domain Site 1 binding pocket. Further modifications targeted the naphthalene imide moiety, with the installation of 5-Br (45 a), 5-OH (45 c) and 5-propyl ether (45 d) moieties. Among them, the OH 45 c and propyl ether 45 d retained PPI inhibition, with propyl ether 45 d being the most active with a PPI inhibition IC50=7.3 μM. This is 2x more potent than Pitstop 2 and 3x more potent than Pitstop 1.
Collapse
Affiliation(s)
- Kate Prichard
- Chemistry, School of Environmental & Life Sciences, The University of Newcastle, University Drive, Callaghan, NSW, 2308, Australia
| | - Ngoc Chau
- Cell Signalling Unit, Children's Medical Research Institute, The University of Sydney, Hawkesbury Road, Westmead, Sydney, Australia
| | - Jing Xue
- Cell Signalling Unit, Children's Medical Research Institute, The University of Sydney, Hawkesbury Road, Westmead, Sydney, Australia
| | - Michael Krauss
- Leibniz Institute fur Molecular Pharmacologie, Department of Biology, Chemistry, Pharmacy, Robert-Roessle-Strasse 10, Berlin, 13125, Germany
| | - Jennette A Sakoff
- Experimental Therapeutics Group, Medical Oncology, Calvary Mater Newcastle Hospital, Edith Street, Waratah, NSW, 2298, Australia
| | - Jayne Gilbert
- Experimental Therapeutics Group, Medical Oncology, Calvary Mater Newcastle Hospital, Edith Street, Waratah, NSW, 2298, Australia
| | - Claudia Bahnik
- Leibniz Institute fur Molecular Pharmacologie, Department of Biology, Chemistry, Pharmacy, Robert-Roessle-Strasse 10, Berlin, 13125, Germany
| | - Maria Muehlbauer
- Leibniz Institute fur Molecular Pharmacologie, Department of Biology, Chemistry, Pharmacy, Robert-Roessle-Strasse 10, Berlin, 13125, Germany
| | - Silke Radetzki
- Leibniz Institute fur Molecular Pharmacologie, Department of Biology, Chemistry, Pharmacy, Robert-Roessle-Strasse 10, Berlin, 13125, Germany
| | - Phillip J Robinson
- Cell Signalling Unit, Children's Medical Research Institute, The University of Sydney, Hawkesbury Road, Westmead, Sydney, Australia
| | - Volker Haucke
- Leibniz Institute fur Molecular Pharmacologie, Department of Biology, Chemistry, Pharmacy, Robert-Roessle-Strasse 10, Berlin, 13125, Germany
| | - Adam McCluskey
- Chemistry, School of Environmental & Life Sciences, The University of Newcastle, University Drive, Callaghan, NSW, 2308, Australia
| |
Collapse
|
3
|
Jabeen Y, Yousaf N, Sarjadi MS, Gansau JA, Goh LPW. Bioactive compounds derived from marine source: a potential immunotherapy treatment. J Biomol Struct Dyn 2024; 42:5657-5668. [PMID: 37387587 DOI: 10.1080/07391102.2023.2227717] [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: 01/04/2023] [Accepted: 06/15/2023] [Indexed: 07/01/2023]
Abstract
Immunotherapy using checkpoint inhibitors blocks the checkpoint proteins (programmed cell death receptor-1; PD-1) from binding with their corresponding ligands (programmed cell death receptor ligand-1; PD-L1) to regulate cell signaling pathways. The marine environment holds a huge source of small molecules that are understudied which can be developed as an inhibitor. Hence, this study investigated the inhibitory effect of 19 algae-derived small molecules against PD-L1 by using molecular docking, absorption, distribution, metabolism, and elimination (ADME) properties and molecular dynamics simulations (MDS). The molecular docking revealed that the binding energy of the six best compounds ranges from -11.1 to -9.1 kcal/mol. Fucoxanthinol, in particular, has the strongest binding energy at -11.1 kcal/mol with three hydrogen bonds (ASN:63A, GLN:66A, and ASP:122A). Meanwhile, the MDS demonstrated that the ligands were strongly bound to the protein, indicating the stability of the complexes. In summary, the identified compounds are potential PD-L1 inhibitors in immunotherapy.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Yaruq Jabeen
- Department of Biosciences, COMSATS University Islamabad, Islamabad, Pakistan
| | - Numan Yousaf
- Department of Biosciences, COMSATS University Islamabad, Islamabad, Pakistan
| | - Mohd Sani Sarjadi
- Faculty of Science and Natural Resources, Universiti Malaysia Sabah, Kota Kinabalu, Sabah, Malaysia
| | - Jualang Azlan Gansau
- Faculty of Science and Natural Resources, Universiti Malaysia Sabah, Kota Kinabalu, Sabah, Malaysia
| | - Lucky Poh Wah Goh
- Faculty of Science and Natural Resources, Universiti Malaysia Sabah, Kota Kinabalu, Sabah, Malaysia
| |
Collapse
|
4
|
Żołek T, Dömötör O, Żabiński J. Binding mechanism of pentamidine derivatives with human serum acute phase protein α 1-acid glycoprotein. Int J Biol Macromol 2024; 266:131405. [PMID: 38582487 DOI: 10.1016/j.ijbiomac.2024.131405] [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: 02/18/2024] [Revised: 04/02/2024] [Accepted: 04/03/2024] [Indexed: 04/08/2024]
Abstract
Drug binding and interactions with plasma proteins play a crucial role in determining the efficacy of drug delivery, thus significantly impacting the overall pharmacological effect. AGP, the second most abundant plasma protein in blood circulation, has the unique capability to bind drugs and transport various compounds. In our present study, for the first time, we investigated whether AGP, a major component of the acute phase lipocalin in human plasma, can bind with pentamidine derivatives known for their high activity against the fungal pathogen Pneumocystis carinii. This investigation was conducted using integrated spectroscopic techniques and computer-based approaches. According to the results, it was concluded that compounds having heteroatoms (-NCH3) in the aliphatic linker and the addition of a Br atom and a methoxy substituent at the C-2 and C-6 positions on the benzene ring, exhibit strong interactions with the AGP binding site. These compounds are identified as potential candidates for recognition by this protein. MD studies indicated that the tested analogues complexed with AGPs reach an equilibrium state after 60 ns, suggesting the stability of the complexes. This observation was further corroborated by experimental results. Therefore, exploring the interaction mechanism of pentamidine derivatives with plasma proteins holds promise for the development of bis-benzamidine-designed pharmaceutically important drugs.
Collapse
Affiliation(s)
- Teresa Żołek
- Department of Organic and Physical Chemistry, Faculty of Pharmacy, Medical University of Warsaw, Banacha 1, 02-097 Warsaw, Poland.
| | - Orsolya Dömötör
- Department of Molecular and Analytical Chemistry, Interdisciplinary Excellence Centre, University of Szeged, Dóm tér 7-8, 6720 Szeged, Hungary
| | - Jerzy Żabiński
- Department of Organic and Physical Chemistry, Faculty of Pharmacy, Medical University of Warsaw, Banacha 1, 02-097 Warsaw, Poland
| |
Collapse
|
5
|
Sun J, Baker JR, Russell CC, Pham HNT, Goldsmith CD, Cossar PJ, Sakoff JA, Scarlett CJ, McCluskey A. Novel piperazine-1,2,3-triazole leads for the potential treatment of pancreatic cancer. RSC Med Chem 2023; 14:2246-2267. [PMID: 37974967 PMCID: PMC10650957 DOI: 10.1039/d2md00289b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 06/26/2023] [Indexed: 11/19/2023] Open
Abstract
From lead 1, (N-(4-((4-(3-(4-(3-methoxyphenyl)-1H-1,2,3-triazol-1-yl)propyl)piperazin-1-yl)sulfonyl)-phenyl)acetamide), a S100A2-p53 protein-protein interaction inhibitor based on an in silico modelling driven hypothesis, four focused libraries were designed and synthesised. Growth inhibition screening was performed against 16 human cancer cell lines including the pancreatic cell lines MiaPaCa2, BxPC3, AsPC-1, Capan-2, HPAC, PANC-1 and the drug resistant CFPAC1. Modification of 1's phenylacetamide moiety, gave Library 1 with only modest pancreatic cancer activity. Modification of the 3-OCH3Ph moiety (Library 2) gave 4-CH3 (26), 4-CH2CH3 (27), 4-CF3 (31) and 4-NO2 (32) with sterically bulky groups more active. A 4-CF3 acetamide replacement enhanced cytotoxicity (Library 3). The 4-C(CH3)336 resulted in a predicted steric clash in the S100A2-p53 binding groove, with a potency decrease. Alkyl moieties afforded more potent analogues, 34 (4-CH3) and 35 (CH2CH3), a trend evident against pancreatic cancer: GI50 3.7 (35; BxPC-3) to 18 (40; AsPC-1) μM. Library 4 analogues with a 2-CF3 and 3-CF3 benzenesulfonamide moiety were less active than the corresponding Library 3 analogues. Two additional analogues were designed: 51 (4-CF3; 4-OCH3) and 52 (4-CF3; 2-OCH3) revealed 52 to be 10-20 fold more active than 51, against the pancreatic cancer cell lines examined with sub-micromolar GI50 values 0.43 (HPAC) to 0.61 μM (PANC-1). MOE calculated binding scores for each pose are also consistent with the observed biological activity with 52. The obtained SAR data is consistent with the proposed interaction within the S100A2-p53 bonding groove.
Collapse
Affiliation(s)
- Jufeng Sun
- Chemistry, School of Environmental & Life Sciences, The University of Newcastle University Drive Callaghan NSW 2308 Australia
- Medicinal Chemistry, School of Pharmacy, Binzhou Medical University Yantai 264003 China
| | - Jennifer R Baker
- Chemistry, School of Environmental & Life Sciences, The University of Newcastle University Drive Callaghan NSW 2308 Australia
| | - Cecilia C Russell
- Chemistry, School of Environmental & Life Sciences, The University of Newcastle University Drive Callaghan NSW 2308 Australia
| | - Hong N T Pham
- Experimental Therapeutics Group, Department of Medical Oncology, Calvary Mater Newcastle Hospital Edith Street Waratah NSW 2298 Australia
| | - Chloe D Goldsmith
- Experimental Therapeutics Group, Department of Medical Oncology, Calvary Mater Newcastle Hospital Edith Street Waratah NSW 2298 Australia
| | - Peter J Cossar
- Chemistry, School of Environmental & Life Sciences, The University of Newcastle University Drive Callaghan NSW 2308 Australia
| | - Jennette A Sakoff
- Experimental Therapeutics Group, Department of Medical Oncology, Calvary Mater Newcastle Hospital Edith Street Waratah NSW 2298 Australia
| | - Christopher J Scarlett
- School of Environmental & Life Sciences, The University of Newcastle Ourimbah NSW 2258 Australia
| | - Adam McCluskey
- Chemistry, School of Environmental & Life Sciences, The University of Newcastle University Drive Callaghan NSW 2308 Australia
| |
Collapse
|
6
|
Poonia P, Sharma M, Jha P, Chopra M. Pharmacophore-based virtual screening of ZINC database, molecular modeling and designing new derivatives as potential HDAC6 inhibitors. Mol Divers 2023; 27:2053-2071. [PMID: 36214962 DOI: 10.1007/s11030-022-10540-3] [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: 09/10/2022] [Accepted: 09/30/2022] [Indexed: 11/25/2022]
Abstract
To date, many HDAC6 inhibitors have been identified and developed but none is clinically approved as of now. Through this study, we aim to obtain novel HDAC6 selective inhibitors and provide new insights into the detailed structural design of potential HDAC6 inhibitors. A HypoGen-based 3D QSAR HDAC6 pharmacophore was built and used as a query model to screen approximately 8 million ZINC database compounds. First, the ZINC Database was filtered using ADMET, followed by pharmacophore-based library screening. Using fit value and estimated activity cutoffs, a final set of 54 ZINC hits was obtained that were further investigated using molecular docking with the crystal structure of human histone deacetylase 6 catalytic domain 2 in complex with Trichostatin A (PDB ID: 5EDU). Through detailed in silico screening of the ZINC database, we shortlisted three hits as the lead molecules for designing novel HDAC6 inhibitors with better efficacy. Docking with 5EDU, followed by ADMET and TOPKAT analysis of modified ZINC hits provided 9 novel potential HDAC6 inhibitors that possess better docking scores and 2D interactions as compared to the control ZINC hit molecules. Finally, a 50 ns MD analysis run followed by Protein-Ligand Interaction Energy (PLIE) analysis of the top scored hits provided a novel molecule N1 that showed promisingly similar results to that of Ricolinostat (a known HDAC6 inhibitor). The comparable result of the designed hits to established HDAC6 inhibitors suggests that these compounds might prove to be successful HDAC6 inhibitors in future. Designed novel hits that might act as good HDAC6 inhibitors derived from ZINC database using combined molecular docking and modeling approaches.
Collapse
Affiliation(s)
- Priya Poonia
- Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, Delhi, 110036, India
| | - Monika Sharma
- Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, Delhi, 110036, India
| | - Prakash Jha
- Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, Delhi, 110036, India
| | - Madhu Chopra
- Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, Delhi, 110036, India.
| |
Collapse
|
7
|
Szewczyk-Roszczenko OK, Roszczenko P, Shmakova A, Finiuk N, Holota S, Lesyk R, Bielawska A, Vassetzky Y, Bielawski K. The Chemical Inhibitors of Endocytosis: From Mechanisms to Potential Clinical Applications. Cells 2023; 12:2312. [PMID: 37759535 PMCID: PMC10527932 DOI: 10.3390/cells12182312] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
Endocytosis is one of the major ways cells communicate with their environment. This process is frequently hijacked by pathogens. Endocytosis also participates in the oncogenic transformation. Here, we review the approaches to inhibit endocytosis, discuss chemical inhibitors of this process, and discuss potential clinical applications of the endocytosis inhibitors.
Collapse
Affiliation(s)
| | - Piotr Roszczenko
- Department of Biotechnology, Medical University of Bialystok, Kilinskiego 1, 15-089 Bialystok, Poland; (P.R.); (A.B.)
| | - Anna Shmakova
- CNRS, UMR 9018, Institut Gustave Roussy, Université Paris-Saclay, 94800 Villejuif, France;
| | - Nataliya Finiuk
- Department of Regulation of Cell Proliferation and Apoptosis, Institute of Cell Biology of National Academy of Sciences of Ukraine, Drahomanov 14/16, 79005 Lviv, Ukraine;
| | - Serhii Holota
- Department of Pharmaceutical, Organic and Bioorganic Chemistry, Danylo Halytsky Lviv National Medical University, Pekarska 69, 79010 Lviv, Ukraine; (S.H.); (R.L.)
| | - Roman Lesyk
- Department of Pharmaceutical, Organic and Bioorganic Chemistry, Danylo Halytsky Lviv National Medical University, Pekarska 69, 79010 Lviv, Ukraine; (S.H.); (R.L.)
| | - Anna Bielawska
- Department of Biotechnology, Medical University of Bialystok, Kilinskiego 1, 15-089 Bialystok, Poland; (P.R.); (A.B.)
| | - Yegor Vassetzky
- CNRS, UMR 9018, Institut Gustave Roussy, Université Paris-Saclay, 94800 Villejuif, France;
| | - Krzysztof Bielawski
- Department of Synthesis and Technology of Drugs, Medical University of Bialystok, Kilinskiego 1, 15-089 Bialystok, Poland;
| |
Collapse
|
8
|
Odell LR, Jones NC, Chau N, Robertson MJ, Ambrus JI, Deane FM, Young KA, Whiting A, Xue J, Prichard K, Daniel JA, Gorgani NN, O'Brien TJ, Robinson PJ, McCluskey A. The sulfonadyns: a class of aryl sulfonamides inhibiting dynamin I GTPase and clathrin mediated endocytosis are anti-seizure in animal models. RSC Med Chem 2023; 14:1492-1511. [PMID: 37593570 PMCID: PMC10429932 DOI: 10.1039/d2md00371f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 04/15/2023] [Indexed: 08/19/2023] Open
Abstract
We show that dansylcadaverine (1) a known in-cell inhibitor of clathrin mediated endocytosis (CME), moderately inhibits dynamin I (dynI) GTPase activity (IC50 45 μM) and transferrin (Tfn) endocytosis in U2OS cells (IC50 205 μM). Synthesis gave a new class of GTP-competitive dynamin inhibitors, the Sulfonadyns™. The introduction of a terminal cinnamyl moiety greatly enhanced dynI inhibition. Rigid diamine or amide links between the dansyl and cinnamyl moieties were detrimental to dynI inhibition. Compounds with in vitro inhibition of dynI activity <10 μM were tested in-cell for inhibition of CME. These data unveiled a number of compounds, e.g. analogues 33 ((E)-N-(6-{[(3-(4-bromophenyl)-2-propen-1-yl]amino}hexyl)-5-isoquinolinesulfonamide)) and 47 ((E)-N-(3-{[3-(4-bromophenyl)-2-propen-1-yl]amino}propyl)-1-naphthalenesulfonamide)isomers that showed dyn IC50 <4 μM, IC50(CME) <30 μM and IC50(SVE) from 12-265 μM. Both analogues (33 and 47) are at least 10 times more potent that the initial lead, dansylcadaverine (1). Enzyme kinetics revealed these sulfonamide analogues as being GTP competitive inhibitors of dynI. Sulfonadyn-47, the most potent SVE inhibitor observed (IC50(SVE) = 12.3 μM), significantly increased seizure threshold in a 6 Hz mouse psychomotor seizure test at 30 (p = 0.003) and 100 mg kg-1 ip (p < 0.0001), with similar anti-seizure efficacy to the established anti-seizure medication, sodium valproate (400 mg kg-1). The Sulfonadyn™ class of drugs target dynamin and show promise as novel leads for future anti-seizure medications.
Collapse
Affiliation(s)
- Luke R Odell
- Chemistry, Centre for Chemical Biology, School of Environmental & Life Science, The University of Newcastle University Drive Callaghan NSW 2308 Australia +612 4921 5472 +612 4921 6486
| | - Nigel C Jones
- Department of Neuroscience, Central Clinical School, Monash University Melbourne Victoria 3004 Australia
- Department of Neurology, The Alfred Hospital Commercial Road Melbourne Victoria 3004 Australia
- Department of Medicine (Royal Melbourne Hospital), University of Melbourne Parkville Victoria 3052 Australia
| | - Ngoc Chau
- Cell Signaling Unit, Children's Medical Research Institute, The University of Sydney 214 Hawkesbury Road Westmead NSW 2145 Australia +612 8865 2915
| | - Mark J Robertson
- Chemistry, Centre for Chemical Biology, School of Environmental & Life Science, The University of Newcastle University Drive Callaghan NSW 2308 Australia +612 4921 5472 +612 4921 6486
| | - Joseph I Ambrus
- Chemistry, Centre for Chemical Biology, School of Environmental & Life Science, The University of Newcastle University Drive Callaghan NSW 2308 Australia +612 4921 5472 +612 4921 6486
| | - Fiona M Deane
- Chemistry, Centre for Chemical Biology, School of Environmental & Life Science, The University of Newcastle University Drive Callaghan NSW 2308 Australia +612 4921 5472 +612 4921 6486
| | - Kelly A Young
- Chemistry, Centre for Chemical Biology, School of Environmental & Life Science, The University of Newcastle University Drive Callaghan NSW 2308 Australia +612 4921 5472 +612 4921 6486
| | - Ainslie Whiting
- Cell Signaling Unit, Children's Medical Research Institute, The University of Sydney 214 Hawkesbury Road Westmead NSW 2145 Australia +612 8865 2915
| | - Jing Xue
- Cell Signaling Unit, Children's Medical Research Institute, The University of Sydney 214 Hawkesbury Road Westmead NSW 2145 Australia +612 8865 2915
| | - Kate Prichard
- Chemistry, Centre for Chemical Biology, School of Environmental & Life Science, The University of Newcastle University Drive Callaghan NSW 2308 Australia +612 4921 5472 +612 4921 6486
| | - James A Daniel
- Cell Signaling Unit, Children's Medical Research Institute, The University of Sydney 214 Hawkesbury Road Westmead NSW 2145 Australia +612 8865 2915
| | - Nick N Gorgani
- Cell Signaling Unit, Children's Medical Research Institute, The University of Sydney 214 Hawkesbury Road Westmead NSW 2145 Australia +612 8865 2915
| | - Terence J O'Brien
- Department of Neurology, The Alfred Hospital Commercial Road Melbourne Victoria 3004 Australia
- Department of Medicine (Royal Melbourne Hospital), University of Melbourne Parkville Victoria 3052 Australia
| | - Phillip J Robinson
- Cell Signaling Unit, Children's Medical Research Institute, The University of Sydney 214 Hawkesbury Road Westmead NSW 2145 Australia +612 8865 2915
| | - Adam McCluskey
- Chemistry, Centre for Chemical Biology, School of Environmental & Life Science, The University of Newcastle University Drive Callaghan NSW 2308 Australia +612 4921 5472 +612 4921 6486
| |
Collapse
|
9
|
Seo J, Park M, Ko D, Kim S, Park JM, Park S, Nam KD, Farrand L, Yang J, Seok C, Jung E, Kim YJ, Kim JY, Seo JH. Ebastine impairs metastatic spread in triple-negative breast cancer by targeting focal adhesion kinase. Cell Mol Life Sci 2023; 80:132. [PMID: 37185776 PMCID: PMC10130003 DOI: 10.1007/s00018-023-04760-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 01/12/2023] [Accepted: 03/15/2023] [Indexed: 05/17/2023]
Abstract
We sought to investigate the utility of ebastine (EBA), a second-generation antihistamine with potent anti-metastatic properties, in the context of breast cancer stem cell (BCSC)-suppression in triple-negative breast cancer (TNBC). EBA binds to the tyrosine kinase domain of focal adhesion kinase (FAK), blocking phosphorylation at the Y397 and Y576/577 residues. FAK-mediated JAK2/STAT3 and MEK/ERK signaling was attenuated after EBA challenge in vitro and in vivo. EBA treatment induced apoptosis and a sharp decline in the expression of the BCSC markers ALDH1, CD44 and CD49f, suggesting that EBA targets BCSC-like cell populations while reducing tumor bulk. EBA administration significantly impeded BCSC-enriched tumor burden, angiogenesis and distant metastasis while reducing MMP-2/-9 levels in circulating blood in vivo. Our findings suggest that EBA may represent an effective therapeutic for the simultaneous targeting of JAK2/STAT3 and MEK/ERK for the treatment of molecularly heterogeneous TNBC with divergent profiles. Further investigation of EBA as an anti-metastatic agent for the treatment of TNBC is warranted.
Collapse
Affiliation(s)
- Juyeon Seo
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea
- Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea
- Department of Biomedical Research Center, Korea University Guro Hospital, Korea University, Seoul, 08308, Republic of Korea
| | - Minsu Park
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea
- Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea
- Department of Biomedical Research Center, Korea University Guro Hospital, Korea University, Seoul, 08308, Republic of Korea
| | - Dongmi Ko
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea
- Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea
- Department of Biomedical Research Center, Korea University Guro Hospital, Korea University, Seoul, 08308, Republic of Korea
| | - Seongjae Kim
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea
- Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea
- Department of Biomedical Research Center, Korea University Guro Hospital, Korea University, Seoul, 08308, Republic of Korea
| | - Jung Min Park
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea
- Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea
- Department of Biomedical Research Center, Korea University Guro Hospital, Korea University, Seoul, 08308, Republic of Korea
| | - Soeun Park
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea
- Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea
- Department of Biomedical Research Center, Korea University Guro Hospital, Korea University, Seoul, 08308, Republic of Korea
| | - Kee Dal Nam
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea
- Department of Biomedical Research Center, Korea University Guro Hospital, Korea University, Seoul, 08308, Republic of Korea
| | - Lee Farrand
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, 5000, Australia
| | - Jinsol Yang
- Galux Inc, Gwanak-Gu, Seoul, 08738, Republic of Korea
| | - Chaok Seok
- Galux Inc, Gwanak-Gu, Seoul, 08738, Republic of Korea
- Department of Chemistry, Seoul National University, Seoul, 08826, Republic of Korea
| | - Eunsun Jung
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea.
- Department of Biomedical Research Center, Korea University Guro Hospital, Korea University, Seoul, 08308, Republic of Korea.
- Guro Hospital Campus, Korea University, 97 Gurodong-Gil, Guro-Guu, Seoul, 08308, Republic of Korea.
| | - Yoon-Jae Kim
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea.
- Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea.
- Department of Biomedical Research Center, Korea University Guro Hospital, Korea University, Seoul, 08308, Republic of Korea.
- Guro Hospital Campus, Korea University, 97 Gurodong-Gil, Guro-Guu, Seoul, 08308, Republic of Korea.
| | - Ji Young Kim
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea.
- Department of Biomedical Research Center, Korea University Guro Hospital, Korea University, Seoul, 08308, Republic of Korea.
- Guro Hospital Campus, Korea University, 97 Gurodong-Gil, Guro-Guu, Seoul, 08308, Republic of Korea.
| | - Jae Hong Seo
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea.
- Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea.
- Department of Biomedical Research Center, Korea University Guro Hospital, Korea University, Seoul, 08308, Republic of Korea.
- Guro Hospital Campus, Korea University, 97 Gurodong-Gil, Guro-Guu, Seoul, 08308, Republic of Korea.
| |
Collapse
|
10
|
Asiedu SO, Gupta Y, Nicolaescu V, Gula H, Caulfield TR, Durvasula R, Kempaiah P, Kwofie SK, Wilson MD. Mycolactone: A Broad Spectrum Multitarget Antiviral Active in the Picomolar Range for COVID-19 Prevention and Cure. Int J Mol Sci 2023; 24:ijms24087151. [PMID: 37108313 PMCID: PMC10139166 DOI: 10.3390/ijms24087151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/25/2023] [Accepted: 03/29/2023] [Indexed: 04/29/2023] Open
Abstract
We have previously shown computationally that Mycolactone (MLN), a toxin produced by Mycobacterium ulcerans, strongly binds to Munc18b and other proteins, presumably blocking degranulation and exocytosis of blood platelets and mast cells. We investigated the effect of MLN on endocytosis using similar approaches, and it bound strongly to the N-terminal of the clathrin protein and a novel SARS-CoV-2 fusion protein. Experimentally, we found 100% inhibition up to 60 nM and 84% average inhibition at 30 nM in SARS-CoV-2 live viral assays. MLN was also 10× more potent than remdesivir and molnupiravir. MLN's toxicity against human alveolar cell line A549, immortalized human fetal renal cell line HEK293, and human hepatoma cell line Huh7.1 were 17.12%, 40.30%, and 36.25%, respectively. The cytotoxicity IC50 breakpoint ratio versus anti-SARS-CoV-2 activity was more than 65-fold. The IC50 values against the alpha, delta, and Omicron variants were all below 0.020 µM, and 134.6 nM of MLN had 100% inhibition in an entry and spread assays. MLN is eclectic in its actions through its binding to Sec61, AT2R, and the novel fusion protein, making it a good drug candidate for treating and preventing COVID-19 and other similarly transmitted enveloped viruses and pathogens.
Collapse
Affiliation(s)
- Seth Osei Asiedu
- Department of Parasitology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Accra P.O. Box GA 337, Ghana
| | - Yash Gupta
- Department of Medicine, Division of Infectious Diseases, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224, USA
| | - Vlad Nicolaescu
- Department of Microbiology, Ricketts Laboratory, University of Chicago, Chicago, IL 60637, USA
| | - Haley Gula
- Department of Microbiology, Ricketts Laboratory, University of Chicago, Chicago, IL 60637, USA
| | - Thomas R Caulfield
- Department of Neuroscience, Division of QHS Computational Biology, Mayo Clinic, Jacksonville, FL 32224, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| | - Ravi Durvasula
- Department of Medicine, Division of Infectious Diseases, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224, USA
| | - Prakasha Kempaiah
- Department of Medicine, Division of Infectious Diseases, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224, USA
| | - Samuel K Kwofie
- Department of Biomedical Engineering, School of Engineering, University of Ghana, Legon, Accra P.O. Box 77, Ghana
| | - Michael D Wilson
- Department of Parasitology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Accra P.O. Box GA 337, Ghana
| |
Collapse
|
11
|
Das B, Bhardwaj PK, Sharma N, Sarkar A, Haldar PK, Mukherjee PK. Evaluation of Mollugo oppositifolia Linn. as cholinesterase and β-secretase enzymes inhibitor. Front Pharmacol 2023; 13:990926. [PMID: 36686717 PMCID: PMC9846241 DOI: 10.3389/fphar.2022.990926] [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: 07/11/2022] [Accepted: 12/05/2022] [Indexed: 01/05/2023] Open
Abstract
Mollugo oppositifolia Linn. is traditionally used in neurological complications. The study aimed to investigate in-vitro neuroprotective effect of the plant extracts through testing against acetylcholinesterase (AChE), butyrylcholinesterase (BChE), and β-secretase linked to Alzheimer's disease (AD). To understand the safety aspects, the extracts were tested for CYP450 isozymes and human hepatocellular carcinoma cell (HepG2) inhibitory potential. The heavy metal contents were estimated using atomic absorption spectroscopy (AAS). Further, the antioxidant capacities as well as total phenolic content and total flavonoid content (TFC) were measured spectrophotometrically. UPLC-QTOF-MS/MS analysis was employed to identify phytometabolites present in the extract. The interactions of the ligands with the target proteins (AChE, BChE, and BACE-1) were studied using AutoDockTools 1.5.6. The results showed that M. oppositifolia extract has more selectivity towards BChE (IC50 = 278.23 ± 1.89 μg/ml) as compared to AChE (IC50 = 322.87 ± 2.05 μg/ml). The IC50 value against β-secretase was 173.93 μg/ml. The extract showed a CC50 value of 965.45 ± 3.07 μg/ml against HepG2 cells and the AAS analysis showed traces of lead 0.02 ± 0.001 which was found to be within the WHO prescribed limits. Moreover, the IC50 values against CYP3A4 (477.03 ± 2.01 μg/ml) and CYP2D6 (249.65 ± 2.46 μg/ml) isozymes justify the safety aspects of the extract. The in silico molecular docking analysis of the target enzymes showed that the compound menthoside was found to be the most stable and showed a good docking score among all the identified metabolites. Keeping in mind the multi-targeted drug approach, the present findings suggested that M. oppositifolia extract have anti-Alzheimer's potential.
Collapse
Affiliation(s)
- Bhaskar Das
- School of Natural Product Studies, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, India,Institute of Bioresources and Sustainable Development, Department of Biotechnology, Government of India, Imphal, India
| | - Pardeep K. Bhardwaj
- Institute of Bioresources and Sustainable Development, Department of Biotechnology, Government of India, Imphal, India,*Correspondence: Pardeep K. Bhardwaj,
| | - Nanaocha Sharma
- Institute of Bioresources and Sustainable Development, Department of Biotechnology, Government of India, Imphal, India
| | - Arnab Sarkar
- Department of Pharmaceutical Technology, Jadavpur University, Kolkata, India
| | - Pallab Kanti Haldar
- School of Natural Product Studies, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, India
| | - Pulok K. Mukherjee
- Institute of Bioresources and Sustainable Development, Department of Biotechnology, Government of India, Imphal, India
| |
Collapse
|
12
|
Alkafaas SS, Abdallah AM, Ghosh S, Loutfy SA, Elkafas SS, Abdel Fattah NF, Hessien M. Insight into the role of clathrin-mediated endocytosis inhibitors in SARS-CoV-2 infection. Rev Med Virol 2023; 33:e2403. [PMID: 36345157 PMCID: PMC9877911 DOI: 10.1002/rmv.2403] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 10/08/2022] [Accepted: 10/10/2022] [Indexed: 11/10/2022]
Abstract
Emergence of SARS-CoV-2 variants warrants sustainable efforts to upgrade both the diagnostic and therapeutic protocols. Understanding the details of cellular and molecular basis of the virus-host cell interaction is essential for developing variant-independent therapeutic options. The internalization of SARS-CoV-2, into lung epithelial cells, is mediated by endocytosis, especially clathrin-mediated endocytosis (CME). Although vaccination is the gold standard strategy against viral infection, selective inhibition of endocytic proteins, complexes, and associated adaptor proteins may present a variant-independent therapeutic strategy. Although clathrin and/or dynamins are the most important proteins involved in CME, other endocytic mechanisms are clathrin and/or dynamin independent and rely on other proteins. Moreover, endocytosis implicates some subcellular structures, like plasma membrane, actin and lysosomes. Also, physiological conditions, such as pH and ion concentrations, represent an additional factor that mediates these events. Accordingly, endocytosis related proteins are potential targets for small molecules that inhibit endocytosis-mediated viral entry. This review summarizes the potential of using small molecules, targeting key proteins, participating in clathrin-dependent and -independent endocytosis, as variant-independent antiviral drugs against SARS-CoV-2 infection. The review takes two approaches. The first outlines the potential role of endocytic inhibitors in preventing endocytosis-mediated viral entry and its mechanism of action, whereas in the second computational analysis was implemented to investigate the selectivity of common inhibitors against endocytic proteins in SARS-CoV-2 endocytosis. The analysis revealed that remdesivir, methyl-β-cyclodextrin, rottlerin, and Bis-T can effectively inhibit clathrin, HMG-CoA reductase, actin, and dynamin I GTPase and are more potent in inhibiting SARS-CoV-2 than chloroquine. CME inhibitors for SARS-CoV-2 infection remain understudied.
Collapse
Affiliation(s)
- Samar Sami Alkafaas
- Molecular Cell Biology UniteDivision of BiochemistryDepartment of ChemistryFaculty of ScienceTanta UniversityTantaEgypt
| | - Abanoub Mosaad Abdallah
- Narcotic Research DepartmentNational Center for Social and Criminological Research (NCSCR)GizaEgypt
| | - Soumya Ghosh
- Department of GeneticsFaculty of Natural and Agricultural SciencesUniversity of the Free StateBloemfonteinSouth Africa
| | - Samah A. Loutfy
- Virology and Immunology UnitCancer Biology DepartmentNational Cancer Institute (NCI)Cairo UniversityCairoEgypt
- Nanotechnology Research CenterBritish UniversityCairoEgypt
| | - Sara Samy Elkafas
- Production Engineering and Mechanical Design DepartmentFaculty of EngineeringMenofia UniversityMenofiaEgypt
| | - Nasra F. Abdel Fattah
- Virology and Immunology UnitCancer Biology DepartmentNational Cancer Institute (NCI)Cairo UniversityCairoEgypt
| | - Mohamed Hessien
- Molecular Cell Biology UniteDivision of BiochemistryDepartment of ChemistryFaculty of ScienceTanta UniversityTantaEgypt
| |
Collapse
|
13
|
Thirunavukkarasu MK, Karuppasamy R. Drug repurposing combined with MM/PBSA based validation strategies towards MEK inhibitors screening. J Biomol Struct Dyn 2022; 40:12392-12403. [PMID: 34459701 DOI: 10.1080/07391102.2021.1970629] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Emergence of oncogenic mutations in the MAPK pathway gaining more impact in the recent years. Importantly, MEK is a core element of this pathway as it is easy to inhibit and is a gatekeeper of multiple malignancies. Therefore, we performed in-silico strategy to screen repurposed candidate for MEK protein using a library of 11,808 compounds from different clusters in the DrugBank database. Glide docking, Prime-MM/GBSA and QikProp analysis were implemented to retrieve the hits with high precision. The stability of the binding mode and binding affinity of the resultant hit were explored using molecular dynamic simulations and MM/PBSA approach. The results highlight that Nebivolol (DB04861) not only achieved a stable conformation in the MEK binding pocket but also displayed highest binding affinity than the other molecules investigated in our study. Taken together, we hypothesized that Nebivolol is an excellent candidate for the inhibition of MEK in NSCLC patients in future.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Muthu Kumar Thirunavukkarasu
- Department of Biotechnology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - Ramanathan Karuppasamy
- Department of Biotechnology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| |
Collapse
|
14
|
Behera S, Behera A, Mekap SK, Behera CC, Kadam A, Mohanty PK. Periplaneta americana L . a potential source of traditional medicine: chemometric analysis, in vitro and in silico study. J Biomol Struct Dyn 2022; 40:9931-9947. [PMID: 34151747 DOI: 10.1080/07391102.2021.1938681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
'Mayurbhanj is the ethnic dominant tribal population district in Odisha, India. The triabl's of Mayurbhanj depends on traditional medicines since time immemorial for health-related issues. Due to the imperative ethnic claim of traditional healers, the financial stringency of the patient community and the necessity to produce a better therapeutic effect has led to investigate ethno zoological sources and to find out the biochemical moiety responsible for the healing process. Considering the ethnic communities' acceptability of the zoological source as traditional medicine, the current evidence-based research study is conducted to investigate the biochemical moiety present in Periplaneta americana, responsible for therapeutic activity. The whole powdered Periplaneta americana was extracted using maceration techniques with n-hexane and methanol as solvent. The obtained extracts were subjected to GC-MS analysis to identify the biochemical moiety. To check the potential biological activity, an in-vitro antimicrobial test was carried out in both turbidimetry and a viable count method against E. coli. Moreover, the obtained biochemical molecules were exposed to in silico studies for their binding modes and their affinity using Discovery studio software. The major compounds were found to be hexadecanoic acid, methyl ester, n-hexadecanoic acid, oleic acid, octadecanoic acid along with other minor constituents. The maximum inhibitory activity of n-hexane and methanol extract against S. aureus at a concentration of 400 µg/mL was found to be 89 and 87%, respectively. The binding models of almost all identified compounds confer very good binding affinities with some key and strong non-covalent interactions with various amino acid residues of receptor active site pocket, which predict the compounds to be potent inhibitors of various infectious bacteria. These findings suggested that the hexane extract of P. americana could be exploited as a potential natural source. Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Suchismeeta Behera
- Postgraduate Department of Zoology, Utkal University, Bhubaneswar, India.,State Forensic Science Laboratory, Bhubaneswar, India
| | - Amulyaratna Behera
- School of Pharmacy and Life Sciences, Centurion University of Technology and Management, Bhubaneswar, India
| | - Suman Kumar Mekap
- School of Pharmacy and Life Sciences, Centurion University of Technology and Management, Bhubaneswar, India
| | - Chinmaya Chidananda Behera
- School of Pharmacy and Life Sciences, Centurion University of Technology and Management, Bhubaneswar, India
| | - Atul Kadam
- Department of Pharmaceutics, Shree Santkrupa College of Pharmacy, Ghogaon, India
| | - Prafulla K Mohanty
- Postgraduate Department of Zoology, Utkal University, Bhubaneswar, India
| |
Collapse
|
15
|
Ali HSHM, Altayb HN, Firoz A, Bayoumi AAM, El Omri A, Chaieb K. Inhibitory activity of marine sponge metabolites on SARS-CoV-2 RNA dependent polymerase: virtual screening and molecular dynamics simulation. J Biomol Struct Dyn 2022; 40:10191-10202. [PMID: 34151745 DOI: 10.1080/07391102.2021.1940283] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Marine species are known as rich sources of metabolites involved mainly in the pharmaceutical industry. This study aimed to evaluate the effect of biologically active compounds in the marine sponge on the SARS-CoV-2 RNA-dependent-RNA polymerase protein (RdRp) using the in-silico method. A total of 51 marine compounds were checked for their possible interaction with SARS-CoV-2 RdRp using Maestro interface for molecular docking, molecular dynamic (MD) simulation, and MM/GBSA method to estimate compounds binding affinities. Among the 51 compounds screened in this study, two (mycalamide A, and nakinadine B) exhibited the lowest docking energy and best interaction. Among these compounds, mycalamide A was identified as a potent inhibitor of SARS-CoV-2 RdRp that showed the best and stable interaction during molecular dynamic simulation, with residues (Asp760 and Asp761) found in the catalytic domain of RdRp. The analysis through MM/GBSA for molecular dynamic simulation results revealed binding energy -59.7 ± 7.18 for Mycalamide A and -56 ± 10.55 for Nakinadine B. These results elucidate the possible use of mycalamide A for treating coronavirus disease.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Hani S H Mohammed Ali
- Faculty of Science, Department of Biological Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Hisham N Altayb
- Faculty of Science, Department of Biochemistry, King Abdulaziz University, Jeddah, Saudi Arabia.,Centre for Artificial Intelligence in Precision Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ahmad Firoz
- Faculty of Science, Department of Biological Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | | | - Abdelfatteh El Omri
- Center of Excellence in Bio-nanoscience Research, King Abdulaziz University, Jeddah, Saudi Arabia.,Faculty of Science, Genomics and Biotechnology Section and Research Group, Department of Biological Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Kamel Chaieb
- Faculty of Science, Department of Biochemistry, King Abdulaziz University, Jeddah, Saudi Arabia.,Laboratory of Analysis, Treatment, and valorization of Pollutants of the Environment and Products, Faculty of Pharmacy, Monastir University, Monastir, Tunisia
| |
Collapse
|
16
|
Azmatullah S, Khan AU, Qazi NG, Nadeem H, Irshad N. Pharmacological evaluation of newly synthesized organotin IV complex for antiulcer potential. BMC Pharmacol Toxicol 2022; 23:58. [PMID: 35906691 PMCID: PMC9335977 DOI: 10.1186/s40360-022-00596-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 07/12/2022] [Indexed: 11/29/2022] Open
Abstract
The present study aims to investigate the newly synthesized organotin (IV) complex (2E, 2′E) dibutylstannanediyl bis (4-(4-nitrophenyl) amino)-4-oxobut-2-enoate (DTN) for its anti-ulcer potential. Characterization performed by carbon nuclear magnetic resonance spectroscopy proved that all values are in the expected ranges of the new compound. Gastroprotective activity of DTN was evaluated through in-silico, anti-H. pylori, in-vitro, in-vivo, and ex-vivo proteomic analysis. In-silico analysis shows that DTN possess stable binding with protein targets involved in gastric ulcer pathophysiology. DTN exhibited an inhibitory effect against 2,2-diphenyl-1-picrylhydrazyl, H. pylori and hydrogen potassium ATPase (H+/K+-ATPase). The antiulcer activity was performed using an ethanol-induced gastric ulcer model in rats. Anti-oxidant profile of DTN showed a significant increase in glutathione-S-transferase, glutathione and catalase levels whereas lipid peroxidation levels were reduced. Histopathological findings confirmed that DTN protected the gastric mucosa of rats. Inflammatory markers tumor necrosis factor-alpha, nuclear factor kappa B, cyclooxygenase-2, interleukin 6 and interleukin-1β were reduced and prostaglandin-E2 restored expression of these cytokines in DTN pretreated animals when analyzed by using immunohistochemistry, enzyme-linked immunosorbent assay and western blot techniques. In real-time polymerase chain reaction technique, the expression of H+/K+-ATPase was downregulated in DTN pretreated group. DTN did not cause any mortality up to 400 mg/Kg. This study indicates that the newly synthesized compound DTN, possess stable binding against selected targets. DTN exhibits a gastro-protective effect, mediated via anti-H. pylori, H+/K+-ATPase inhibition, anti-oxidant and anti-inflammatory pathways, exploring its therapeutic potential in gastric ulcer management.
Collapse
Affiliation(s)
- Syed Azmatullah
- Department of Pharmacology, Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, Pakistan
| | - Arif-Ullah Khan
- Department of Pharmacology, Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, Pakistan.
| | - Neelam Gul Qazi
- Department of Pharmacology, Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, Pakistan
| | - Humaira Nadeem
- Department of Pharmaceutical Chemistry, Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, Pakistan
| | - Nadeem Irshad
- Department of Pharmacy, Quaid i Azam University, Islamabad, Pakistan
| |
Collapse
|
17
|
Kandi V, Vundecode A, Godalwar TR, Dasari S, Vadakedath S, Godishala V. The Current Perspectives in Clinical Research: Computer-Assisted Drug Designing, Ethics, and Good Clinical Practice. BORNEO JOURNAL OF PHARMACY 2022. [DOI: 10.33084/bjop.v5i2.3013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
In the era of emerging microbial and non-communicable diseases and re-emerging microbial infections, the medical fraternity and the public are plagued by under-preparedness. It is evident by the severity of the Coronavirus disease (COVID-19) pandemic that novel microbial diseases are a challenge and are challenging to control. This is mainly attributed to the lack of complete knowledge of the novel microbe’s biology and pathogenesis and the unavailability of therapeutic drugs and vaccines to treat and control the disease. Clinical research is the only answer utilizing which can handle most of these circumstances. In this review, we highlight the importance of computer-assisted drug designing (CADD) and the aspects of molecular docking, molecular superimposition, 3D-pharmacophore technology, ethics, and good clinical practice (GCP) for the development of therapeutic drugs, devices, and vaccines.
Collapse
|
18
|
Evaluation of Cyc 1 protein stability in Acidithiobacillus ferrooxidans bacterium after E121D mutation by molecular dynamics simulation to improve electron transfer. JOURNAL OF MICROBIOLOGY (SEOUL, KOREA) 2022; 60:526-532. [PMID: 35286603 DOI: 10.1007/s12275-022-1645-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/12/2022] [Accepted: 01/13/2022] [Indexed: 10/18/2022]
Abstract
Cyc1 (Cytochrome c552) is a protein in the electron transport chain of the Acidithiobacillus ferrooxidans (Af) bacteria which obtain their energy from oxidation Fe2+ to Fe3+. The electrons are directed through Cyc2, RCY (rusticyanin), Cyc1 and Cox aa3 proteins to O2. Cyc1 protein consists of two chains, A and B. In the present study, a novel mutation (E121D) in the A chain of Cyc1 protein was selected due to electron receiving from Histidine 143 of RCY. Then, the changes performed in the E121D mutant were evaluated by MD simulations analyzes. Cyc1 and RCY proteins were docked by a Patchdock server. By E121D mutation, the connection between Zn 1388 of chain B and aspartate 121 of chain A weaken. Asp 121 gets farther from Zn 1388. Therefore, the aspartate gets closer to Cu 1156 of the RCY leading to the higher stability of the RCY/Cyc1 complex. Further, an acidic residue (Glu121) becomes a more acidic residue (Asp121) and improves the electron transfer to Cyc1 protein. The results of RMSF analysis showed further ligand flexibility in mutation. This leads to fluctuation of the active site and increases redox potential at the mutation point and the speed of electron transfer. This study also predicts that in all respiratory chain proteins, electrons probably enter the first active site via glutamate and exit histidine in the second active site of each respiratory chain protein.
Collapse
|
19
|
Ghara A, Andhale GS, Matada GSP, Dhiwar PS. Design and Synthesis of Tri-substituted Imidazole Derivatives as CD73 Inhibitors for Their Anticancer Activity. LETT DRUG DES DISCOV 2022. [DOI: 10.2174/1570180818666210604113849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background:
Monoclonal antibodies licensed by the US Food and Drug Administration
(FDA) target diverse biological targets relevant to immuno-oncology, and small compounds in clinical
trials target various aspects of immuno-oncology. Several small compounds that target CD73 are
at various stages of clinical studies. Several imidazoles are currently being utilized to treat malignancies,
including Dacarbazine, Zoledronic acid, Mercaptopurine, and others. As a result, we evaluated
the cytotoxicity of modified tri-phenyl imidazoles against breast cancer cell lines, as well as conducted
virtual tests.
Methods:
We used Accelrys Drug Discovery Studio 3.5 software to undertake molecular docking,
ADMET, and molecular properties studies on 68 proposed imidazole derivatives. The synthesized
compounds' binding mechanisms were investigated against the CD73 protein (PDB Code: 4H1S).
To find the drugs with the best pharmacokinetics, researchers assessed ADMET solubility, BBB
penetration, hepatotoxicity, PPB binding, and polar surface area. The MDA-MB-231 breast cancer
cell line was treated with these produced compounds, and the MTT test method was used to determine
the IC50 values.
Results:
The selected 14 compounds showed good binding in the active site of CD73 by forming Hbonds
with amino acid residues, according to molecular docking studies. Breast cancer cell lines
were treated with substituted tri-phenyl imidazole derivatives, which displayed anticancer activity.
Compounds 3a and 3h, which had an electron-donating group at the 2nd and 3rd positions and
p-substitutions of the chloro and nitro groups, respectively, showed considerable anticancer action.
Conclusion:
Fourteen imidazole derivatives were produced and tested against breast cancer cell
lines based on in-silico research. The MDA-MB-231 cell line was strongly suppressed by compounds
3a and 3h. In-vitro enzyme inhibition experiments revealed that only 3h demonstrated considerable
inhibition.
Collapse
Affiliation(s)
- Abhishek Ghara
- Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru, Karnataka,
560107, India
| | - Ganesh Sakharam Andhale
- Department of Pharmaceutical Chemistry, Alard College of Pharmacy, Pune, Maharashtra, 411028, India
| | | | - Prasad Sanjay Dhiwar
- Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru, Karnataka,
560107, India
| |
Collapse
|
20
|
Prichard KL, O'Brien NS, Murcia SR, Baker JR, McCluskey A. Role of Clathrin and Dynamin in Clathrin Mediated Endocytosis/Synaptic Vesicle Recycling and Implications in Neurological Diseases. Front Cell Neurosci 2022; 15:754110. [PMID: 35115907 PMCID: PMC8805674 DOI: 10.3389/fncel.2021.754110] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 12/10/2021] [Indexed: 12/17/2022] Open
Abstract
Endocytosis is a process essential to the health and well-being of cell. It is required for the internalisation and sorting of “cargo”—the macromolecules, proteins, receptors and lipids of cell signalling. Clathrin mediated endocytosis (CME) is one of the key processes required for cellular well-being and signalling pathway activation. CME is key role to the recycling of synaptic vesicles [synaptic vesicle recycling (SVR)] in the brain, it is pivotal to signalling across synapses enabling intracellular communication in the sensory and nervous systems. In this review we provide an overview of the general process of CME with a particular focus on two key proteins: clathrin and dynamin that have a central role to play in ensuing successful completion of CME. We examine these two proteins as they are the two endocytotic proteins for which small molecule inhibitors, often of known mechanism of action, have been identified. Inhibition of CME offers the potential to develop therapeutic interventions into conditions involving defects in CME. This review will discuss the roles and the current scope of inhibitors of clathrin and dynamin, providing an insight into how further developments could affect neurological disease treatments.
Collapse
|
21
|
Thirunavukkarasu MK, Suriya U, Rungrotmongkol T, Karuppasamy R. In Silico Screening of Available Drugs Targeting Non-Small Cell Lung Cancer Targets: A Drug Repurposing Approach. Pharmaceutics 2021; 14:59. [PMID: 35056955 PMCID: PMC8778223 DOI: 10.3390/pharmaceutics14010059] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 12/16/2021] [Accepted: 12/20/2021] [Indexed: 01/03/2023] Open
Abstract
The RAS-RAF-MEK-ERK pathway plays a key role in malevolent cell progression in many tumors. The high structural complexity in the upstream kinases limits the treatment progress. Thus, MEK inhibition is a promising strategy since it is easy to inhibit and is a gatekeeper for the many malignant effects of its downstream effector. Even though MEK inhibitors are under investigation in many cancers, drug resistance continues to be the principal limiting factor to achieving cures in patients with cancer. Hence, we accomplished a high-throughput virtual screening to overcome this bottleneck by the discovery of dual-targeting therapy in cancer treatment. Here, a total of 11,808 DrugBank molecules were assessed through high-throughput virtual screening for their activity against MEK. Further, the Glide docking, MLSF and prime-MM/GBSA methods were implemented to extract the potential lead compounds from the database. Two compounds, DB012661 and DB07642, were outperformed in all the screening analyses. Further, the study results reveal that the lead compounds also have a significant binding capability with the co-target PIM1. Finally, the SIE-based free energy calculation reveals that the binding of compounds was majorly affected by the van der Waals interactions with MEK receptor. Overall, the in silico binding efficacy of these lead compounds against both MEK and PIM1 could be of significant therapeutic interest to overcome drug resistance in the near future.
Collapse
Affiliation(s)
- Muthu Kumar Thirunavukkarasu
- Department of Biotechnology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore 632014, India;
| | - Utid Suriya
- Program in Biotechnology, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand;
| | - Thanyada Rungrotmongkol
- Biocatalyst and Environmental Biotechnology Research Unit, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
- Program in Bioinformatics and Computational Biology, Graduate School, Chulalongkorn University, Bangkok 10330, Thailand
| | - Ramanathan Karuppasamy
- Department of Biotechnology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore 632014, India;
| |
Collapse
|
22
|
Rampogu S, Lee G, Kulkarni AM, Kim D, Yoon S, Kim MO, Lee KW. Computational Approaches to Discover Novel Natural Compounds for SARS-CoV-2 Therapeutics. ChemistryOpen 2021; 10:593-599. [PMID: 34010501 PMCID: PMC8133350 DOI: 10.1002/open.202000332] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 03/18/2021] [Indexed: 12/12/2022] Open
Abstract
Scientists all over the world are facing a challenging task of finding effective therapeutics for the coronavirus disease (COVID-19). One of the fastest ways of finding putative drug candidates is the use of computational drug discovery approaches. The purpose of the current study is to retrieve natural compounds that have obeyed to drug-like properties as potential inhibitors. Computational molecular modelling techniques were employed to discover compounds with potential SARS-CoV-2 inhibition properties. Accordingly, the InterBioScreen (IBS) database was obtained and was prepared by minimizing the compounds. To the resultant compounds, the absorption, distribution, metabolism, excretion and toxicity (ADMET) and Lipinski's Rule of Five was applied to yield drug-like compounds. The obtained compounds were subjected to molecular dynamics simulation studies to evaluate their stabilities. In the current article, we have employed the docking based virtual screening method using InterBioScreen (IBS) natural compound database yielding two compounds has potential hits. These compounds have demonstrated higher binding affinity scores than the reference compound together with good pharmacokinetic properties. Additionally, the identified hits have displayed stable interaction results inferred by molecular dynamics simulation results. Taken together, we advocate the use of two natural compounds, STOCK1N-71493 and STOCK1N-45683 as SARS-CoV-2 treatment regime.
Collapse
Affiliation(s)
- Shailima Rampogu
- Division of Life SciencesResearch Institute of Natural ScienceGyeongsang National University501 Jinju-daeroJinju52828South Korea
| | - Gihwan Lee
- Division of Life SciencesResearch Institute of Natural ScienceGyeongsang National University501 Jinju-daeroJinju52828South Korea
| | - Apoorva M. Kulkarni
- Division of Life SciencesResearch Institute of Natural ScienceGyeongsang National University501 Jinju-daeroJinju52828South Korea
| | - Donghwan Kim
- Division of Life SciencesResearch Institute of Natural ScienceGyeongsang National University501 Jinju-daeroJinju52828South Korea
| | - Sanghwa Yoon
- Division of Life SciencesResearch Institute of Natural ScienceGyeongsang National University501 Jinju-daeroJinju52828South Korea
| | - Myeong Ok Kim
- Division of Life Science and Applied Life ScienceCollege of Natural SciencesGyeongsang National UniversityJinjuSouth Korea
| | - Keun Woo Lee
- Division of Life SciencesResearch Institute of Natural ScienceGyeongsang National University501 Jinju-daeroJinju52828South Korea
| |
Collapse
|
23
|
Gogoi B, Chowdhury P, Goswami N, Gogoi N, Naiya T, Chetia P, Mahanta S, Chetia D, Tanti B, Borah P, Handique PJ. Identification of potential plant-based inhibitor against viral proteases of SARS-CoV-2 through molecular docking, MM-PBSA binding energy calculations and molecular dynamics simulation. Mol Divers 2021; 25:1963-1977. [PMID: 33856591 PMCID: PMC8047602 DOI: 10.1007/s11030-021-10211-9] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Accepted: 03/16/2021] [Indexed: 12/05/2022]
Abstract
The Coronavirus disease 2019 (COVID-19), caused by the novel coronavirus, SARS-CoV-2, has recently emerged as a pandemic. Here, an attempt has been made through in-silico high throughput screening to explore the antiviral compounds from traditionally used plants for antiviral treatments in India namely, Tea, Neem and Turmeric, as potential inhibitors of two widely studied viral proteases, main protease (Mpro) and papain-like protease (PLpro) of the SARS-CoV-2. Molecular docking study using BIOVIA Discovery Studio 2018 revealed, (−)-epicatechin-3-O-gallate (ECG), a tea polyphenol has a binding affinity toward both the selected receptors, with the lowest CDocker energy − 46.22 kcal mol−1 for SARS-CoV-2 Mpro and CDocker energy − 44.72 kcal mol−1 for SARS-CoV-2 PLpro, respectively. The SARS-CoV-2 Mpro complexed with (−)-epicatechin-3-O-gallate, which had shown the best binding affinity was subjected to molecular dynamics simulations to validate its binding affinity, during which, the root-mean-square-deviation values of SARS-CoV-2 Mpro–Co-crystal ligand (N3) and SARS-CoV-2 Mpro- (−)-epicatechin-3-O-gallate systems were found to be more stable than SARS-CoV-2 Mpro system. Further, (−)-epicatechin-3-O-gallate was subjected to QSAR analysis which predicted IC50 of 0.3281 nM against SARS-CoV-2 Mpro. Overall, (−)-epicatechin-3-O-gallate showed a potential binding affinity with SARS-CoV-2 Mpro and could be proposed as a potential natural compound for COVID-19 treatment.
Collapse
Affiliation(s)
- Bhaskarjyoti Gogoi
- Department of Biotechnology, Royal Global University, Guwahati, Assam, 781035, India
| | - Purvita Chowdhury
- Department of Health Research, Model Rural Health Research Unit, Tripura, 799035, India
| | - Nabajyoti Goswami
- Bioinformatics Infrastructure Facility, College of Veterinary Science, Assam Agricultural University, Khanapara, Guwahati, Assam, 781025, India
| | - Neelutpal Gogoi
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, 786004, India
| | - Tufan Naiya
- Department of Biotechnology, Maulana Abul Kalam Azad University of Technology, West Bengal, 700064, India
| | - Pankaj Chetia
- Department of Life Sciences, Dibrugarh University, Dibrugarh, 786004, Assam, India
| | - Saurov Mahanta
- National Institute of Electronics and Information Technology (NIELIT), Guwahati, Assam, 781008, India.
| | - Dipak Chetia
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, 786004, India
| | - Bhaben Tanti
- Department of Botany, Gauhati University, Guwahati, Assam, 781014, India
| | - Probodh Borah
- Bioinformatics Infrastructure Facility, College of Veterinary Science, Assam Agricultural University, Khanapara, Guwahati, Assam, 781025, India
| | | |
Collapse
|
24
|
Żołek T, Trzeciak A, Maciejewska D. Theoretical evaluation of EGFR kinase inhibition and toxicity of di-indol-3-yl disulphides with anti-cancer potency. J Biomol Struct Dyn 2020; 40:622-634. [PMID: 32880212 DOI: 10.1080/07391102.2020.1815576] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Research aimed at developing potent di-indol-3-yl disulphides for cancer diseases makes use of various theoretical techniques to evaluate the drug-likeness parameters and the mode of action. A drug-likeness filter helps evaluate the therapeutic potency of four bis-indole derivatives, structurally related to 3,3'-methanediyl-bis-indole (DIM) but having the S-S instead of the methylene linker and showing a high inhibitory impact on the variants of cancer cell lines (among them HL-60 and DU-145). Based on in vitro experimental results for their close analogues, a correlation was found between the epidermal growth factor receptor kinase (EGFR) inhibition and the theoretical energy of complexation. Docking studies of ligands followed by molecular dynamics were performed at the ATP-binding site of EGFR tyrosine kinase to scrutinize the inhibition of the di-indol-3-yl disulphides at a molecular level. Derivatives with bromine or iodine substituents at C-5 positions of the indole moieties made strong complexes by interaction with the most important hinge region residues Met-793 and Cys-733. The inhibition model for EGFR kinase and the proposed procedures can be very informative in the biological testing of selected bis-indoles and may be useful for future research on effective inhibitors for the treatment of EGFR-related cancer.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Teresa Żołek
- Department of Organic Chemistry, Faculty of Pharmacy, Medical University of Warsaw, Warsaw, Poland
| | - Aleksandra Trzeciak
- Department of Organic Chemistry, Faculty of Pharmacy, Medical University of Warsaw, Warsaw, Poland
| | - Dorota Maciejewska
- Department of Organic Chemistry, Faculty of Pharmacy, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
25
|
Mahanta S, Chowdhury P, Gogoi N, Goswami N, Borah D, Kumar R, Chetia D, Borah P, Buragohain AK, Gogoi B. Potential anti-viral activity of approved repurposed drug against main protease of SARS-CoV-2: an in silico based approach. J Biomol Struct Dyn 2020; 39:3802-3811. [PMID: 32406317 DOI: 10.1080/07391102.2020.1768902] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) which was first reported in Wuhan province of China, has become a deadly pandemic causing alarmingly high morbidity and mortality. In the absence of new targeted drugs and vaccines against SARS-CoV-2 at present, the choices for effective treatments are limited. Therefore, considering the exigency of the situation, we focused on identifying the available approved drugs as potential inhibitor against the promising Coronavirus drug target, the Main Protease, using computer-aided methods. We created a library of U. S. Food and Drug Administration approved anti-microbial drugs and virtually screened it against the available crystal structures of Main Protease of the virus. The study revealed that Viomycin showed the highest -CDocker energy after docking at the active site of SARS-CoV-2 Main Protease. It is noteworthy that Viomycin showed higher -CDocker energy as compared to the drugs currently under clinical trial for SARS-CoV-2 treatment viz. Ritonavir and Lopinavir. Additionally, Viomycin formed higher number of H-bonds with SARS-CoV-2 Main Protease than its co-crystallised inhibitor compound N3. Molecular dynamics simulation further showed that Viomycin embedded deeply inside the binding pocket and formed robust binding with SARS-CoV-2 Main Protease. Therefore, we propose that Viomycin may act as a potential inhibitor of the Main Protease of SARS-CoV-2. Further optimisations with the drug may support the much-needed rapid response to mitigate the pandemic.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Saurov Mahanta
- National Institute of Electronics and Information Technology (NIELIT), Guwahati, Guwahati, Assam, India
| | | | - Neelutpal Gogoi
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, India
| | - Nabajyoti Goswami
- Bioinformatics Infrastructure Facility, College of Veterinary Science, Assam Agricultural University, Guwahati, India
| | - Debajit Borah
- Department of Biotechnology, Royal Global University, Guwahati, India
| | - Rupesh Kumar
- Department of Biotechnology, Royal Global University, Guwahati, India
| | - Dipak Chetia
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, India
| | - Probodh Borah
- Bioinformatics Infrastructure Facility, College of Veterinary Science, Assam Agricultural University, Guwahati, India
| | - Alak K Buragohain
- Department of Biotechnology, Royal Global University, Guwahati, India
| | | |
Collapse
|
26
|
Zhang R, Tang L, Tian Y, Ji X, Hu Q, Zhou B, Zhenyu D, Heng X, Yang L. Cholesterol-modified DP7 enhances the effect of individualized cancer immunotherapy based on neoantigens. Biomaterials 2020; 241:119852. [PMID: 32120313 DOI: 10.1016/j.biomaterials.2020.119852] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 02/01/2020] [Accepted: 02/07/2020] [Indexed: 02/07/2023]
Abstract
Personalized cancer vaccines based on neoantigens have become an important research direction in cancer immunotherapy. However, their therapeutic effects are limited by the efficiency of antigen uptake and presentation by antigen presenting cells. Here, the low-toxicity cholesterol-modified antimicrobial peptide (AMP) DP7 (DP7-C), which has dual functions as a carrier and an immune adjuvant, improved the dendritic cell (DC)-based vaccine efficacy. As a delivery carrier, DP7-C can efficiently delivery various antigen peptides into 75-95% of DCs via caveolin- and clathrin-dependent pathways. As an immune adjuvant, DP7-C can induce DC maturation and proinflammatory cytokine release via the TLR2-MyD88-NF-κB pathway and effectively increase antigen presentation efficiency. In addition, DP7-C enhanced the efficacy of DC-based individualized cancer immunotherapy and achieved excellent antitumor effects on mouse tumor models using the OVA antigen peptides and LL2-neoantigens. Excitingly, after DP7-C stimulation, the antigen uptake efficiency of monocytes-derived DCs (MoDCs) in patients with advanced lung cancer increased from 14-40% to 88-98%, the presentation efficiency increased from approximately 15% to approximately 65%, and the proportion of mature MoDCs increased from approximately 20% to approximately 60%. These findings suggest that our approach may be a potentially alternative strategy to produce cancer vaccines designed for individual patients.
Collapse
Affiliation(s)
- Rui Zhang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lin Tang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yaomei Tian
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiao Ji
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qiuyue Hu
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Bailing Zhou
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ding Zhenyu
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xu Heng
- Precision Medicine Center, State Key Laboratory of Biotherapy, and Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, China
| | - Li Yang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
27
|
HRLC-ESI-MS based identification of active small molecules from Cissus quadrangularis and likelihood of their action towards the primary targets of osteoarthritis. J Mol Struct 2020. [DOI: 10.1016/j.molstruc.2019.127048] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
28
|
Mishra A, Cross M, Hofmann A, Coster MJ, Karim A, Sattar A. Identification of a Novel Scaffold for Inhibition of Dipeptidyl Peptidase-4. J Comput Biol 2019; 26:1470-1486. [PMID: 31390221 DOI: 10.1089/cmb.2019.0201] [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] [Indexed: 12/26/2022] Open
Abstract
Dipeptidyl peptidase-4 (DPP-4) is considered a major drug target for type 2 diabetes mellitus (T2DM). In addition to T2DM, a regulatory role of DPP-4 was also found in cardiovascular diseases. Existing DPP-4 inhibitors have been reported to have several adverse effects. In this study, a computer-aided drug design approach and its use to detect a novel class of inhibitor for DPP-4 are reported. Through structure and pharmacophore-based screening, we identified 13 hit compounds from an ∼4-million-compound library. Physical interactions of these hits with DPP-4 were studied using docking and explicit solvent molecular dynamics (MD) simulations. Later, MMPBSA binding energy was calculated for the ligand/protein simulation trajectories to determine the stability of compounds in the binding cavity. These compounds have a novel scaffold and exhibited a stable binding mode. "Best-in-screen" compounds (or their closest available analogs) were resourced and their inhibition of DPP-4 activity was experimentally validated using an in vitro enzyme activity assay in the presence of 100 and 10 μM compounds. These assays identified a compound with a spirochromanone center with 53% inhibition activity at a 100 μM concentration. A further five spirochromanone compounds were synthesized and examined in silico and in vitro; again, one compound showed 53% inhibitory activity action at 100 μM. Overall, this study identified two novel "spirochromanone" compounds that lowered DPP-4 activity by more than ∼50% at 100 μM. This study also showed the impact of fast in silico drug design techniques utilizing virtual screening and MD to identify novel scaffolds to bind and inhibit DPP-4. Spirochromanone motif identified here may be used to design molecules to achieve drug-like inhibitory action against DPP-4.
Collapse
Affiliation(s)
- Avinash Mishra
- Institute for Integrated and Intelligent Systems, Griffith University, Nathan, Australia.,Novo Informatics Pvt. Ltd., New Delhi, India
| | - Megan Cross
- Griffith Institute for Drug Discovery, Griffith University, Nathan, Australia
| | - Andreas Hofmann
- Griffith Institute for Drug Discovery, Griffith University, Nathan, Australia.,Faculty of Veterinary and Agricultural Sciences, Melbourne Veterinary School, The University of Melbourne, Parkville, Australia
| | - Mark J Coster
- Griffith Institute for Drug Discovery, Griffith University, Nathan, Australia
| | - Abdul Karim
- Institute for Integrated and Intelligent Systems, Griffith University, Nathan, Australia
| | - Abdul Sattar
- Institute for Integrated and Intelligent Systems, Griffith University, Nathan, Australia
| |
Collapse
|
29
|
Nnyigide OS, Lee SG, Hyun K. In Silico Characterization of the Binding Modes of Surfactants with Bovine Serum Albumin. Sci Rep 2019; 9:10643. [PMID: 31337814 PMCID: PMC6650617 DOI: 10.1038/s41598-019-47135-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 07/09/2019] [Indexed: 01/10/2023] Open
Abstract
The binding interactions of the surfactants: anionic sodium dodecyl sulphate (SDS), cationic cetyltrimethylammonium bromide (CTAB), non-ionic octyl glucoside (OG), and zwitterionic 3-[Hexadecyl(dimethyl)ammonio]-1-propanesulfonate (HPS), with bovine serum albumin (BSA) were investigated by computer simulation. The results disclosed that the surfactants bound stably between hydrophobic subdomain IIA and IIIA where tryptophan-213 residue, an important intrinsic fluorophore in BSA is housed. The interactions of the surfactants with the BSA were electrostatic and hydrophobic interactions. The head-groups of SDS, HPS and OG formed hydrogen bonds with the BSA, while that of CTAB was shielded from intermolecular hydrogen-bonding due to intervening methyl groups. Subsequently, molecular dynamics (MD) simulation of the protein-surfactant complexes revealed that hydrogen bonds formed by OG were stronger than those of SDS and HPS. However, the decomposed force-field energies showed that OG had the least interaction energy with the BSA. In addition to MD simulation, it was found by density functional theory (DFT) that the differences in the coulomb interaction energies can be attributed to charge distribution in the surfactants. Overall, free energies calculated by linear interaction energy (LIE) proved that the binding of each surfactant was dominated by differences between van der Waals interactions in bound and free states.
Collapse
Affiliation(s)
- Osita Sunday Nnyigide
- School of Chemical and Biomolecular Engineering, Pusan National University, Busan, 46241, Korea
| | - Sun-Gu Lee
- School of Chemical and Biomolecular Engineering, Pusan National University, Busan, 46241, Korea.
| | - Kyu Hyun
- School of Chemical and Biomolecular Engineering, Pusan National University, Busan, 46241, Korea.
| |
Collapse
|
30
|
Enkavi G, Javanainen M, Kulig W, Róg T, Vattulainen I. Multiscale Simulations of Biological Membranes: The Challenge To Understand Biological Phenomena in a Living Substance. Chem Rev 2019; 119:5607-5774. [PMID: 30859819 PMCID: PMC6727218 DOI: 10.1021/acs.chemrev.8b00538] [Citation(s) in RCA: 194] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Indexed: 12/23/2022]
Abstract
Biological membranes are tricky to investigate. They are complex in terms of molecular composition and structure, functional over a wide range of time scales, and characterized by nonequilibrium conditions. Because of all of these features, simulations are a great technique to study biomembrane behavior. A significant part of the functional processes in biological membranes takes place at the molecular level; thus computer simulations are the method of choice to explore how their properties emerge from specific molecular features and how the interplay among the numerous molecules gives rise to function over spatial and time scales larger than the molecular ones. In this review, we focus on this broad theme. We discuss the current state-of-the-art of biomembrane simulations that, until now, have largely focused on a rather narrow picture of the complexity of the membranes. Given this, we also discuss the challenges that we should unravel in the foreseeable future. Numerous features such as the actin-cytoskeleton network, the glycocalyx network, and nonequilibrium transport under ATP-driven conditions have so far received very little attention; however, the potential of simulations to solve them would be exceptionally high. A major milestone for this research would be that one day we could say that computer simulations genuinely research biological membranes, not just lipid bilayers.
Collapse
Affiliation(s)
- Giray Enkavi
- Department
of Physics, University of
Helsinki, P.O. Box 64, FI-00014 Helsinki, Finland
| | - Matti Javanainen
- Department
of Physics, University of
Helsinki, P.O. Box 64, FI-00014 Helsinki, Finland
- Institute
of Organic Chemistry and Biochemistry of the Czech Academy
of Sciences, Flemingovo naḿesti 542/2, 16610 Prague, Czech Republic
- Computational
Physics Laboratory, Tampere University, P.O. Box 692, FI-33014 Tampere, Finland
| | - Waldemar Kulig
- Department
of Physics, University of
Helsinki, P.O. Box 64, FI-00014 Helsinki, Finland
| | - Tomasz Róg
- Department
of Physics, University of
Helsinki, P.O. Box 64, FI-00014 Helsinki, Finland
- Computational
Physics Laboratory, Tampere University, P.O. Box 692, FI-33014 Tampere, Finland
| | - Ilpo Vattulainen
- Department
of Physics, University of
Helsinki, P.O. Box 64, FI-00014 Helsinki, Finland
- Computational
Physics Laboratory, Tampere University, P.O. Box 692, FI-33014 Tampere, Finland
- MEMPHYS-Center
for Biomembrane Physics
| |
Collapse
|
31
|
Ortiz CLD, Matel HD, Nellas RB. In Silico insights on enhancing thermostability and activity of a plant Fructosyltransferase from Pachysandra terminalis via introduction of disulfide bridges. J Mol Graph Model 2019; 89:250-260. [PMID: 30933883 DOI: 10.1016/j.jmgm.2019.03.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 03/18/2019] [Accepted: 03/18/2019] [Indexed: 12/18/2022]
Abstract
Drawbacks of industrially-used fructosyltransferases (FTs) such as low optimum temperature and low fructooligosaccharides (FOS) yield necessitates the search for engineered FTs that are highly thermostable and active. With the availability of the first plant FT crystal structure from Pachysandra terminalis (PDB ID: 3UGH), computer-aided protein engineering of plant FT is now feasible. To obtain insights on the effect of specific mutations i.e. disulfide bridge introduction, wild-type and mutant FTs were subjected to a 15 μs Martini Coarse-grained Molecular Dynamics (CGMD) simulations at 303 K and 334 K. We report here the five mutants, M31C-Q49C, L144C-S193C, P34C-W300C, S219C-L226C and V470C-S498C with enhanced thermostabilities and/or activities relative to the wild type. Interestingly, M31C-Q49C, which is located within the catalytic-carrying blade of the catalytic domain, has an activity enhancement at both temperatures. At 334 K, three mutations, L144C-S193C, P34C-W300C and V470C-S498C, achieved thermostability relative to the wild type. Intriguingly, both activity and stability enhancement exhibited only at 334 K can be achieved provided that the mutation is located either on the catalytic-carrying residue blade of the catalytic domain or on the non-catalytic domain. Our results suggest that V470C-S498C and L144C-S193C are promising mutants and that domain-specific approach may be exploited to customize enzyme properties.
Collapse
Affiliation(s)
| | - Hosea D Matel
- Research Center, Cavite State University, Don Severino De Las Alas Campus, Indang, Cavite, Philippines; Department of Physical Sciences, College of Arts and Sciences, Cavite State University, Don Severino De Las Alas Campus, Indang, Cavite, Philippines
| | - Ricky B Nellas
- Institute of Chemistry, College of Science, University of the Philippines Diliman, Diliman, Quezon City, Philippines.
| |
Collapse
|
32
|
Spirohydantoins and 1,2,4-triazole-3-carboxamide derivatives as inhibitors of histone deacetylase: Design, synthesis, and biological evaluation. Eur J Med Chem 2018; 146:79-92. [DOI: 10.1016/j.ejmech.2018.01.021] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 12/22/2017] [Accepted: 01/08/2018] [Indexed: 11/30/2022]
|
33
|
Computational Exploration for Lead Compounds That Can Reverse the Nuclear Morphology in Progeria. BIOMED RESEARCH INTERNATIONAL 2017; 2017:5270940. [PMID: 29226142 PMCID: PMC5684607 DOI: 10.1155/2017/5270940] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 09/24/2017] [Indexed: 01/01/2023]
Abstract
Progeria is a rare genetic disorder characterized by premature aging that eventually leads to death and is noticed globally. Despite alarming conditions, this disease lacks effective medications; however, the farnesyltransferase inhibitors (FTIs) are a hope in the dark. Therefore, the objective of the present article is to identify new compounds from the databases employing pharmacophore based virtual screening. Utilizing nine training set compounds along with lonafarnib, a common feature pharmacophore was constructed consisting of four features. The validated Hypo1 was subsequently allowed to screen Maybridge, Chembridge, and Asinex databases to retrieve the novel lead candidates, which were then subjected to Lipinski's rule of 5 and ADMET for drug-like assessment. The obtained 3,372 compounds were forwarded to docking simulations and were manually examined for the key interactions with the crucial residues. Two compounds that have demonstrated a higher dock score than the reference compounds and showed interactions with the crucial residues were subjected to MD simulations and binding free energy calculations to assess the stability of docked conformation and to investigate the binding interactions in detail. Furthermore, this study suggests that the Hits may be more effective against progeria and further the DFT studies were executed to understand their orbital energies.
Collapse
|
34
|
Rampogu S, Son M, Park C, Kim HH, Suh JK, Lee KW. Sulfonanilide Derivatives in Identifying Novel Aromatase Inhibitors by Applying Docking, Virtual Screening, and MD Simulations Studies. BIOMED RESEARCH INTERNATIONAL 2017; 2017:2105610. [PMID: 29312992 PMCID: PMC5664374 DOI: 10.1155/2017/2105610] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 07/31/2017] [Accepted: 08/27/2017] [Indexed: 01/04/2023]
Abstract
Breast cancer is one of the leading causes of death noticed in women across the world. Of late the most successful treatments rendered are the use of aromatase inhibitors (AIs). In the current study, a two-way approach for the identification of novel leads has been adapted. 81 chemical compounds were assessed to understand their potentiality against aromatase along with the four known drugs. Docking was performed employing the CDOCKER protocol available on the Discovery Studio (DS v4.5). Exemestane has displayed a higher dock score among the known drug candidates and is labeled as reference. Out of 81 ligands 14 have exhibited higher dock scores than the reference. In the second approach, these 14 compounds were utilized for the generation of the pharmacophore. The validated four-featured pharmacophore was then allowed to screen Chembridge database and the potential Hits were obtained after subjecting them to Lipinski's rule of five and the ADMET properties. Subsequently, the acquired 3,050 Hits were escalated to molecular docking utilizing GOLD v5.0. Finally, the obtained Hits were consequently represented to be ideal lead candidates that were escalated to the MD simulations and binding free energy calculations. Additionally, the gene-disease association was performed to delineate the associated disease caused by CYP19A1.
Collapse
Affiliation(s)
- Shailima Rampogu
- Division of Applied Life Science (BK21 Plus), Plant Molecular Biology and Biotechnology Research Center (PMBBRC), Systems and Synthetic Agrobiotech Center (SSAC), Research Institute of Natural Science (RINS), Gyeongsang National University (GNU), 501 Jinju-daero, Jinju 52828, Republic of Korea
| | - Minky Son
- Division of Applied Life Science (BK21 Plus), Plant Molecular Biology and Biotechnology Research Center (PMBBRC), Systems and Synthetic Agrobiotech Center (SSAC), Research Institute of Natural Science (RINS), Gyeongsang National University (GNU), 501 Jinju-daero, Jinju 52828, Republic of Korea
| | - Chanin Park
- Division of Applied Life Science (BK21 Plus), Plant Molecular Biology and Biotechnology Research Center (PMBBRC), Systems and Synthetic Agrobiotech Center (SSAC), Research Institute of Natural Science (RINS), Gyeongsang National University (GNU), 501 Jinju-daero, Jinju 52828, Republic of Korea
| | - Hyong-Ha Kim
- Division of Quality of Life, Korea Research Institute of Standards and Science, Daejeon 34113, Republic of Korea
| | - Jung-Keun Suh
- Bio-Computing Major, Korean German Institute of Technology, Seoul 07582, Republic of Korea
| | - Keun Woo Lee
- Division of Applied Life Science (BK21 Plus), Plant Molecular Biology and Biotechnology Research Center (PMBBRC), Systems and Synthetic Agrobiotech Center (SSAC), Research Institute of Natural Science (RINS), Gyeongsang National University (GNU), 501 Jinju-daero, Jinju 52828, Republic of Korea
| |
Collapse
|
35
|
Gogoi D, Baruah VJ, Chaliha AK, Kakoti BB, Sarma D, Buragohain AK. Identification of novel human renin inhibitors through a combined approach of pharmacophore modelling, molecular DFT analysis and in silico screening. Comput Biol Chem 2017; 69:28-40. [DOI: 10.1016/j.compbiolchem.2017.04.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2016] [Revised: 01/09/2017] [Accepted: 04/12/2017] [Indexed: 12/26/2022]
|
36
|
El Kfoury KA, Romond MB, Scuotto A, Alidjinou EK, Dabboussi F, Hamze M, Engelmann I, Sane F, Hober D. Bifidobacteria-derived lipoproteins inhibit infection with coxsackievirus B4 in vitro. Int J Antimicrob Agents 2017; 50:177-185. [PMID: 28595938 DOI: 10.1016/j.ijantimicag.2017.03.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 02/07/2017] [Accepted: 03/11/2017] [Indexed: 10/19/2022]
Abstract
The aim of the present study was to investigate the potential of bifidobacteria in protecting cells from coxsackievirus B4 (CV-B4) infection. Bifidobacterial screening identified two of five strains that protected human epithelial type 2 (HEp-2) cell viability when bifidobacteria were incubated with viral particles prior to inoculation. In contrast, no effect was shown by incubating HEp-2 cells with bifidobacteria prior to CV-B4 inoculation. Cell wall lipoprotein aggregates (LpAs) secreted by the selected strains were assayed for their antiviral activity. The two LpAs exhibited antiviral activity when they were incubated with viral particles prior to inoculation of HEp-2 cells. Recombinant LpA-derived protein exhibited identical antiviral activity. To identify the peptide sequences interacting with the virus particles, LpA proteins were aligned with the peptide sequences of the north canyon rim and puff footprint onto coxsackievirus and adenovirus receptor (CAR). The in silico molecular docking study using CV-B3 as template showed low-energy binding, indicating a stable system for the selected peptides and consequently a likely binding interaction with CV-B. Bifidobacterium longum and Bifidobacterium breve peptides homologous to the viral north rim footprint onto CAR sequence formed hydrogen bonds with several viral residues in the north rim of the canyon, which were already predicted as interacting with CAR. In conclusion, proteins from bifidobacterial LpAs can inhibit infection with CV-B4, likely through binding to the capsid amino acids that interact with CAR.
Collapse
Affiliation(s)
- Khalil Antoine El Kfoury
- Université de Lille, CHU Lille, Laboratoire de Virologie EA3610, Lille F-59000, France; Université Libanaise, Laboratoire de Microbiologie Santé et Environnement, Ecole Doctorale des Sciences et Technologie, Faculté de Santé Publique, Tripoli, Lebanon
| | | | - Angelo Scuotto
- Bifinove, 99 rue Jardin des Plantes, Lille 59000, France
| | | | - Fouad Dabboussi
- Université Libanaise, Laboratoire de Microbiologie Santé et Environnement, Ecole Doctorale des Sciences et Technologie, Faculté de Santé Publique, Tripoli, Lebanon
| | - Monzer Hamze
- Université Libanaise, Laboratoire de Microbiologie Santé et Environnement, Ecole Doctorale des Sciences et Technologie, Faculté de Santé Publique, Tripoli, Lebanon
| | - Ilka Engelmann
- Université de Lille, CHU Lille, Laboratoire de Virologie EA3610, Lille F-59000, France
| | - Famara Sane
- Université de Lille, CHU Lille, Laboratoire de Virologie EA3610, Lille F-59000, France
| | - Didier Hober
- Université de Lille, CHU Lille, Laboratoire de Virologie EA3610, Lille F-59000, France.
| |
Collapse
|
37
|
Odell LR, Abdel-Hamid MK, Hill TA, Chau N, Young KA, Deane FM, Sakoff JA, Andersson S, Daniel JA, Robinson PJ, McCluskey A. Pyrimidine-Based Inhibitors of Dynamin I GTPase Activity: Competitive Inhibition at the Pleckstrin Homology Domain. J Med Chem 2016; 60:349-361. [DOI: 10.1021/acs.jmedchem.6b01422] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Luke R. Odell
- Chemistry,
School of Environmental and Life Sciences, The University of Newcastle, Callaghan, New South Wales 2308, Australia
| | - Mohammed K. Abdel-Hamid
- Chemistry,
School of Environmental and Life Sciences, The University of Newcastle, Callaghan, New South Wales 2308, Australia
- Department
of Medicinal Chemistry, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt
| | - Timothy A. Hill
- Chemistry,
School of Environmental and Life Sciences, The University of Newcastle, Callaghan, New South Wales 2308, Australia
| | - Ngoc Chau
- Children’s
Medical Research Institute, The University of Sydney, 214 Hawkesbury
Road, Westmead New South
Wales 2145, Australia
| | - Kelly A. Young
- Chemistry,
School of Environmental and Life Sciences, The University of Newcastle, Callaghan, New South Wales 2308, Australia
| | - Fiona M. Deane
- Chemistry,
School of Environmental and Life Sciences, The University of Newcastle, Callaghan, New South Wales 2308, Australia
| | - Jennette A. Sakoff
- Experimental
Therapeutics Group, Department of Medical Oncology, Calvary Mater Newcastle Hospital, Edith Street, Waratah, 2298, New South Wales Australia
| | - Sofia Andersson
- Department
of Biology and Chemical Engineering, Mälardalens University, Box 325, S-631
05, Eskilstuna, Sweden
| | - James A. Daniel
- Children’s
Medical Research Institute, The University of Sydney, 214 Hawkesbury
Road, Westmead New South
Wales 2145, Australia
| | - Phillip J. Robinson
- Children’s
Medical Research Institute, The University of Sydney, 214 Hawkesbury
Road, Westmead New South
Wales 2145, Australia
| | - Adam McCluskey
- Chemistry,
School of Environmental and Life Sciences, The University of Newcastle, Callaghan, New South Wales 2308, Australia
| |
Collapse
|
38
|
Gogoi D, Baruah VJ, Chaliha AK, Kakoti BB, Sarma D, Buragohain AK. 3D pharmacophore-based virtual screening, docking and density functional theory approach towards the discovery of novel human epidermal growth factor receptor-2 (HER2) inhibitors. J Theor Biol 2016; 411:68-80. [PMID: 27693363 DOI: 10.1016/j.jtbi.2016.09.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 09/06/2016] [Accepted: 09/20/2016] [Indexed: 11/24/2022]
Abstract
Human epidermal growth factor receptor 2 (HER2) is one of the four members of the epidermal growth factor receptor (EGFR) family and is expressed to facilitate cellular proliferation across various tissue types. Therapies targeting HER2, which is a transmembrane glycoprotein with tyrosine kinase activity, offer promising prospects especially in breast and gastric/gastroesophageal cancer patients. Persistence of both primary and acquired resistance to various routine drugs/antibodies is a disappointing outcome in the treatment of many HER2 positive cancer patients and is a challenge that requires formulation of new and improved strategies to overcome the same. Identification of novel HER2 inhibitors with improved therapeutics index was performed with a highly correlating (r=0.975) ligand-based pharmacophore model (Hypo1) in this study. Hypo1 was generated from a training set of 22 compounds with HER2 inhibitory activity and this well-validated hypothesis was subsequently used as a 3D query to screen compounds in a total of four databases of which two were natural product databases. Further, these compounds were analyzed for compliance with Veber's drug-likeness rule and optimum ADMET parameters. The selected compounds were then subjected to molecular docking and Density Functional Theory (DFT) analysis to discern their molecular interactions at the active site of HER2. The findings thus presented would be an important starting point towards the development of novel HER2 inhibitors using well-validated computational techniques.
Collapse
Affiliation(s)
- Dhrubajyoti Gogoi
- DBT-Bioinformatics Infrastructure Facility, Centre for Biotechnology and Bioinformatics, School of Science and Engineering, Dibrugarh University, Dibrugarh, Assam, India
| | - Vishwa Jyoti Baruah
- DBT-Bioinformatics Infrastructure Facility, Centre for Biotechnology and Bioinformatics, School of Science and Engineering, Dibrugarh University, Dibrugarh, Assam, India
| | - Amrita Kashyap Chaliha
- DBT-Bioinformatics Infrastructure Facility, Centre for Biotechnology and Bioinformatics, School of Science and Engineering, Dibrugarh University, Dibrugarh, Assam, India
| | - Bibhuti Bhushan Kakoti
- DBT-Bioinformatics Infrastructure Facility, Centre for Biotechnology and Bioinformatics, School of Science and Engineering, Dibrugarh University, Dibrugarh, Assam, India
| | - Diganta Sarma
- Department of Chemistry, School of Science and Engineering, Dibrugarh University, Dibrugarh, Assam, India
| | - Alak Kumar Buragohain
- DBT-Bioinformatics Infrastructure Facility, Centre for Biotechnology and Bioinformatics, School of Science and Engineering, Dibrugarh University, Dibrugarh, Assam, India.
| |
Collapse
|
39
|
Panigrahi GK, Suthar MK, Verma N, Asthana S, Tripathi A, Gupta SK, Saxena JK, Raisuddin S, Das M. Investigation of the interaction of anthraquinones of Cassia occidentalis seeds with bovine serum albumin by molecular docking and spectroscopic analysis: Correlation to their in vitro cytotoxic potential. Food Res Int 2015. [DOI: 10.1016/j.foodres.2015.08.022] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
40
|
Sigdel S, Hui G, Smith TJ, Murrell JC, Lee JK. Molecular dynamics simulation to rationalize regioselective hydroxylation of aromatic substrates by soluble methane monooxygenase. Bioorg Med Chem Lett 2015; 25:1611-5. [DOI: 10.1016/j.bmcl.2015.01.069] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Revised: 01/27/2015] [Accepted: 01/30/2015] [Indexed: 10/24/2022]
|