1
|
Mili A, Birangal S, Giridhar J, Nandakumar K, Lobo R. Identification of phytomolecules as isoform and mutation specific PI3K-α inhibitor for protection against breast cancer using e-pharmacophore modeling and molecular dynamics simulations. BMC Chem 2024; 18:241. [PMID: 39696683 DOI: 10.1186/s13065-024-01317-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 10/04/2024] [Indexed: 12/20/2024] Open
Abstract
PI3K-α mutation plays a critical role in cancer development, notably in breast cancer, particularly within HR + /HER2- subtypes. These mutations drive tumor growth and survival by activating the PI3K/AKT/mTOR pathway, which is essential for cell proliferation and survival. Our research aimed to identify natural compounds that can inhibit mutant and specific isoforms of PI3K-α to prevent tumor progression. e-Pharmacophore model was generated using Receptor-Ligand complex using the Inavolisib drug (PDB:8EXV) and phase screening was performed using the Molport database of natural compounds. Through molecular docking studies we identified seven promising compounds for further molecular dynamics simulations. Among these, three compounds-STOCK1N-85097, STOCK1N-85998, and STOCK1N-86060-showed significant stability and interaction with PI3K-α. These compounds demonstrated favorable results in several parameters, including RMSD, RMSF, Rg, SASA, PCA, FEL, and total energy evaluations. Therefore, these compounds are projected to function as PI3K-α inhibitors and because of its natural origin it can possess fewer side effects than the conventional medicine, which should be validated by proper in vivo and in vitro models.
Collapse
Affiliation(s)
- Ajay Mili
- Department of Pharmacognosy, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Sumit Birangal
- Department of Pharmaceutical Chemistry, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Jyothi Giridhar
- Department of Pharmaceutical Chemistry, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Krishnadas Nandakumar
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Richard Lobo
- Department of Pharmacognosy, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.
| |
Collapse
|
2
|
de Sousa NF, de Araújo IMA, Rodrigues TCML, da Silva PR, de Moura JP, Scotti MT, Scotti L. Proposition of In silico Pharmacophore Models for Malaria: A Review. Comb Chem High Throughput Screen 2024; 27:2525-2543. [PMID: 37815185 DOI: 10.2174/0113862073247691230925062440] [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: 03/05/2023] [Revised: 07/23/2023] [Accepted: 08/09/2023] [Indexed: 10/11/2023]
Abstract
In the field of medicinal chemistry, the concept of pharmacophore refers to the specific region of a molecule that possesses essential structural and chemical characteristics for binding to a receptor and eliciting biological activity. Understanding the pharmacophore is crucial for drug research and development, as it allows the design of new drugs. Malaria, a widespread disease, is commonly treated with chloroquine and artemisinin, but the emergence of parasite resistance limits their effectiveness. This study aims to explore computer simulations to discover a specific pharmacophore for Malaria, providing new alternatives for its treatment. A literature review was conducted, encompassing articles proposing a pharmacophore for Malaria, gathered from the "Web of Science" database, with a focus on recent publications to ensure up-to-date analysis. The selected articles employed diverse methods, including ligand-based and structurebased approaches, integrating molecular structure and biological activity data to yield comprehensive analyses. Affinity evaluation between the proposed pharmacophore and the target receptor involved calculating free energy to quantify their interaction. Multiple linear regression was commonly utilized, though it is sensitive to multicollinearity issues. Another recurrent methodology was the use of the Schrödinger package, employing tools such as the Phase module and the OPLS force field for interaction analysis. Pharmacophore model proposition allows threedimensional representations guiding the synthesis and design of new biologically active compounds, offering a promising avenue for discovering therapeutic agents to combat Malaria.
Collapse
Affiliation(s)
- Natália Ferreira de Sousa
- Postgraduate Program of Natural and Synthetic Bioactive Products (PgPNSB), Health Sciences Center, Federal University of Paraíba, João Pessoa-PB, Brazil
| | - Igor Mikael Alves de Araújo
- Postgraduate Program of Natural and Synthetic Bioactive Products (PgPNSB), Health Sciences Center, Federal University of Paraíba, João Pessoa-PB, Brazil
| | | | - Pablo Rayff da Silva
- Postgraduate Program of Natural and Synthetic Bioactive Products (PgPNSB), Health Sciences Center, Federal University of Paraíba, João Pessoa-PB, Brazil
| | - Jéssica Paiva de Moura
- Postgraduate Program of Natural and Synthetic Bioactive Products (PgPNSB), Health Sciences Center, Federal University of Paraíba, João Pessoa-PB, Brazil
| | - Marcus Tullius Scotti
- Postgraduate Program of Natural and Synthetic Bioactive Products (PgPNSB), Health Sciences Center, Federal University of Paraíba, João Pessoa-PB, Brazil
| | - Luciana Scotti
- Postgraduate Program of Natural and Synthetic Bioactive Products (PgPNSB), Health Sciences Center, Federal University of Paraíba, João Pessoa-PB, Brazil
| |
Collapse
|
3
|
Siqueira-Neto JL, Wicht KJ, Chibale K, Burrows JN, Fidock DA, Winzeler EA. Antimalarial drug discovery: progress and approaches. Nat Rev Drug Discov 2023; 22:807-826. [PMID: 37652975 PMCID: PMC10543600 DOI: 10.1038/s41573-023-00772-9] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/17/2023] [Indexed: 09/02/2023]
Abstract
Recent antimalarial drug discovery has been a race to produce new medicines that overcome emerging drug resistance, whilst considering safety and improving dosing convenience. Discovery efforts have yielded a variety of new molecules, many with novel modes of action, and the most advanced are in late-stage clinical development. These discoveries have led to a deeper understanding of how antimalarial drugs act, the identification of a new generation of drug targets, and multiple structure-based chemistry initiatives. The limited pool of funding means it is vital to prioritize new drug candidates. They should exhibit high potency, a low propensity for resistance, a pharmacokinetic profile that favours infrequent dosing, low cost, preclinical results that demonstrate safety and tolerability in women and infants, and preferably the ability to block Plasmodium transmission to Anopheles mosquito vectors. In this Review, we describe the approaches that have been successful, progress in preclinical and clinical development, and existing challenges. We illustrate how antimalarial drug discovery can serve as a model for drug discovery in diseases of poverty.
Collapse
Affiliation(s)
| | - Kathryn J Wicht
- Holistic Drug Discovery and Development (H3D) Centre, University of Cape Town, Rondebosch, South Africa
- South African Medical Research Council Drug Discovery and Development Research Unit, Department of Chemistry and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch, South Africa
| | - Kelly Chibale
- Holistic Drug Discovery and Development (H3D) Centre, University of Cape Town, Rondebosch, South Africa
- South African Medical Research Council Drug Discovery and Development Research Unit, Department of Chemistry and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch, South Africa
| | | | - David A Fidock
- Department of Microbiology and Immunology and Center for Malaria Therapeutics and Antimicrobial Resistance, Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | | |
Collapse
|
4
|
Le D, Akiyama T, Weiss D, Kim M. Dissociation kinetics of small-molecule inhibitors in Escherichia coli is coupled to physiological state of cells. Commun Biol 2023; 6:223. [PMID: 36841892 PMCID: PMC9968327 DOI: 10.1038/s42003-023-04604-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 02/16/2023] [Indexed: 02/27/2023] Open
Abstract
Bioactive small-molecule inhibitors represent a treasure chest for future drugs. In vitro high-throughput screening is a common approach to identify the small-molecule inhibitors that bind tightly to purified targets. Here, we investigate the inhibitor-target binding/unbinding kinetics in E. coli cells using a benzimidazole-derivative DNA inhibitor as a model system. We find that its unbinding rate is not constant but depends on cell growth rate. This dependence is mediated by the cellular activity, forming a feedback loop with the inhibitor's activity. In accordance with this feedback, we find cell-to-cell heterogeneity in inhibitor-target interaction, leading to co-existence of two distinct subpopulations: actively growing cells that dissociate the inhibitors from the targets and non-growing cells that do not. We find similar heterogeneity for other clinical DNA inhibitors. Our studies reveal a mechanism that couples inhibitor-target kinetics to cell physiology and demonstrate the significant effect of this coupling on drug efficacy.
Collapse
Affiliation(s)
- Dai Le
- Department of Physics, Emory University, Atlanta, GA, 30322, USA
- Graduate Division of Biological and Biomedical Sciences, Emory University, Atlanta, GA, 30322, USA
| | - Tatsuya Akiyama
- Department of Physics, Emory University, Atlanta, GA, 30322, USA
- Graduate Division of Biological and Biomedical Sciences, Emory University, Atlanta, GA, 30322, USA
| | - David Weiss
- Graduate Division of Biological and Biomedical Sciences, Emory University, Atlanta, GA, 30322, USA
- Division of Infectious Diseases, Department of Medicine, Emory University, Atlanta, GA, 30322, USA
- Antibiotic Research Center, Emory University, Atlanta, GA, 30322, USA
| | - Minsu Kim
- Department of Physics, Emory University, Atlanta, GA, 30322, USA.
- Graduate Division of Biological and Biomedical Sciences, Emory University, Atlanta, GA, 30322, USA.
- Antibiotic Research Center, Emory University, Atlanta, GA, 30322, USA.
| |
Collapse
|
5
|
Kaharudin FA, Zohdi RM, Mukhtar SM, Sidek HM, Bihud NV, Rasol NE, Ahmad FB, Ismail NH. In vitro antiplasmodial and cytotoxicity activities of crude extracts and major compounds from Goniothalamus lanceolatus. JOURNAL OF ETHNOPHARMACOLOGY 2020; 254:112657. [PMID: 32045683 DOI: 10.1016/j.jep.2020.112657] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 02/05/2020] [Accepted: 02/05/2020] [Indexed: 06/10/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Malaria, a devastating infectious disease which was initially recognized as episodic fever, is caused by parasitic protozoan of the genus Plasmodium. Medicinal plants with ethnobotanical information to treat fever and/or malaria has been the key element in identifying potential plant candidates for antimalarial screening. Goniothalamus lanceolatus Miq. (Annonaceae) is used as a folk remedy, particularly to treat fever and skin diseases. AIM OF THE STUDY In this context, supported with previous preliminary data of its antiplasmodial activity, this study was undertaken to determine the in vitro antiplasmodial and cytotoxicity activities of G. lanceolatus crude extracts and its major compounds. MATERIALS AND METHODS The in vitro antiplasmodial activity was determined by parasite lactate dehydrogenase (pLDH) assay on chloroquine-sensitive (3D7) and chloroquine-resistant (K1) strains of Plasmodium falciparum. The cytotoxicity activity was evaluated using the MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay on hepatocellular carcinoma (HepG2) and normal liver (WRL-68) cell lines. RESULTS The root methanol extract possessed potent antiplasmodial activity against both P. falciparum 3D7 and K1 strains (IC50 = 2.7 μg/ml, SI = 140; IC50 = 1.7 μg/ml, SI = 236). Apart from the DCM extract of stem bark and root that were found to be inactive (IC50 > 50 μg/ml) against 3D7 strain, all other tested crude extracts exhibited promising (5< IC50 < 15 μg/ml) to moderate (15< IC50 < 50 μg/ml) antiplasmodial activity against both strain. Additionally, only compound C (Parvistone D) exerted promising antiplasmodial activity against 3D7 strain (IC50 = 7.5 μM, SI = 51) whereas compound A, B and D showed moderate antiplasmodial activity against the same strain (20 < IC50 < 100 μM). Interestingly, when tested on K1 strain, compound A, C and D exhibited promising antiplasmodial activity (2 < IC50 < 20 μM) while compound B exhibited moderate activity (IC50 = 26.9 μM). Cytotoxicity study showed that all tested crude extracts and compounds were non-toxic on WRL-68 and HepG2 cell lines (CC50 > 30 µg/ml, CC50 > 10 µM, respectively), except for the hexane and DCM extracts of root, which exerted mild cytotoxicity on HepG2 cell line (IC50 < 30 μg/ml). CONCLUSIONS This study suggests that the root methanol extract and compound C (Parvistone D) obtained from G. lanceolatus are highly potential for exploitation as source of antimalarial agents. Parvistone D is identified as one of the bioactive styryl lactones found in the plant extract. It is also noteworthy, that the extract and compound were more active against chloroquine-resistant (K1) strain of P. falciparum. Further studies are being carried out to assess their toxicity profile and antimalarial efficacy in animal model.
Collapse
Affiliation(s)
- Fatin Amelina Kaharudin
- Atta-ur-Rahman Institute for Natural Product Discovery, Universiti Teknologi MARA, Selangor Branch, Puncak Alam Campus, 42300, Puncak Alam, Selangor, Malaysia; Faculty of Pharmacy, Universiti Teknologi MARA, Selangor Branch, Puncak Alam Campus, 42300, Puncak Alam, Selangor, Malaysia
| | - Rozaini Mohd Zohdi
- Atta-ur-Rahman Institute for Natural Product Discovery, Universiti Teknologi MARA, Selangor Branch, Puncak Alam Campus, 42300, Puncak Alam, Selangor, Malaysia; Faculty of Pharmacy, Universiti Teknologi MARA, Selangor Branch, Puncak Alam Campus, 42300, Puncak Alam, Selangor, Malaysia.
| | - Shahida Muhamad Mukhtar
- Atta-ur-Rahman Institute for Natural Product Discovery, Universiti Teknologi MARA, Selangor Branch, Puncak Alam Campus, 42300, Puncak Alam, Selangor, Malaysia
| | - Hasidah Mohd Sidek
- School of Biosciences and Biotechnology, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, 43600, Bangi, Selangor, Malaysia
| | - Nur Vicky Bihud
- Atta-ur-Rahman Institute for Natural Product Discovery, Universiti Teknologi MARA, Selangor Branch, Puncak Alam Campus, 42300, Puncak Alam, Selangor, Malaysia; Faculty of Applied Sciences, Universiti Teknologi MARA, 40450, Shah Alam, Selangor, Malaysia
| | - Nurulfazlina Edayah Rasol
- Atta-ur-Rahman Institute for Natural Product Discovery, Universiti Teknologi MARA, Selangor Branch, Puncak Alam Campus, 42300, Puncak Alam, Selangor, Malaysia; Faculty of Applied Sciences, Universiti Teknologi MARA, 40450, Shah Alam, Selangor, Malaysia
| | - Fasihuddin Badruddin Ahmad
- Department of Chemistry, Faculty of Resource Science and Technology, Universiti Malaysia Sarawak, 94300, Kota Samarahan, Sarawak, Malaysia
| | - Nor Hadiani Ismail
- Atta-ur-Rahman Institute for Natural Product Discovery, Universiti Teknologi MARA, Selangor Branch, Puncak Alam Campus, 42300, Puncak Alam, Selangor, Malaysia; Faculty of Applied Sciences, Universiti Teknologi MARA, 40450, Shah Alam, Selangor, Malaysia
| |
Collapse
|
6
|
Yu R, Cheng LP, Li M, Pang W. Discovery of Novel Neuraminidase Inhibitors by Structure-Based Virtual Screening, Structural Optimization, and Bioassay. ACS Med Chem Lett 2019; 10:1667-1673. [PMID: 31857844 DOI: 10.1021/acsmedchemlett.9b00447] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Accepted: 11/25/2019] [Indexed: 12/29/2022] Open
Abstract
Neuraminidase (NA) is a significant therapeutic target for treating influenza. In this study, a new lead NA inhibitor AN-329/10738021 was discovered by structure-based virtual screening, molecular dynamics simulations, and bioassay validation. Optimization of lead AN-329/10738021, which holds a novel scaffold of N'-benzylidene benzohydrazone, leads to discovery of some novel NA inhibitors Y-1-Y-11. Compound Y-1 exerts the best inhibition activity (IC50 = 0.21 μM) against NA, which is better than oseltamivir carboxylate (OSC) (IC50 = 3.04 μM) and lead AN-329/10738021 (IC50 = 1.92 μM). Molecular docking analysis indicates that the good potency of Y-1 may be ascribed to the elongation of the benzylidene moiety of the molecule to the 430-cavity. The results of this study may offer useful reference for development of novel NA inhibitors.
Collapse
Affiliation(s)
- Rao Yu
- School of Chemical and Environmental Engineering, Shanghai Institute of Technology, Shanghai 201418, China
| | - Li Ping Cheng
- School of Chemical and Environmental Engineering, Shanghai Institute of Technology, Shanghai 201418, China
| | - Meng Li
- School of Chemical and Environmental Engineering, Shanghai Institute of Technology, Shanghai 201418, China
| | - Wan Pang
- School of Chemical and Environmental Engineering, Shanghai Institute of Technology, Shanghai 201418, China
| |
Collapse
|
7
|
Lin HY, Ho Y, Liu HL. Structure-Based Pharmacophore Modeling to Discover Novel CCR5 Inhibitors for HIV-1/Cancers Therapy. ACTA ACUST UNITED AC 2019. [DOI: 10.4236/jbise.2019.121002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
8
|
Toviwek B, Suphakun P, Choowongkomon K, Hannongbua S, Gleeson MP. Synthesis and evaluation of the NSCLC anti-cancer activity and physical properties of 4-aryl- N -phenylpyrimidin-2-amines. Bioorg Med Chem Lett 2017; 27:4749-4754. [DOI: 10.1016/j.bmcl.2017.08.063] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 08/19/2017] [Accepted: 08/25/2017] [Indexed: 10/18/2022]
|
9
|
Yao TT, Xie JF, Liu XG, Cheng JL, Zhu CY, Zhao JH, Dong XW. Integration of pharmacophore mapping and molecular docking in sequential virtual screening: towards the discovery of novel JAK2 inhibitors. RSC Adv 2017. [DOI: 10.1039/c6ra24959k] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
An integrated sequential virtual screening protocol by combining molecular docking and pharmacophore mapping was successfully constructed to identify novel small-molecule inhibitors of JAK2.
Collapse
Affiliation(s)
- Ting-Ting Yao
- ZJU-ENS Joint Laboratory of Medicinal Chemistry
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research
- College of Pharmaceutical Sciences
- Zhejiang University
- Hangzhou
| | - Jiang-Feng Xie
- ZJU-ENS Joint Laboratory of Medicinal Chemistry
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research
- College of Pharmaceutical Sciences
- Zhejiang University
- Hangzhou
| | - Xing-Guo Liu
- School of Chemical Engineering and Light Industry
- Guangdong University of Technology
- Guangzhou
- P. R. China
| | - Jing-Li Cheng
- Institute of Pesticide and Environmental Toxicology
- Ministry of Agriculture Key Laboratory of Agricultural Entomology
- Zhejiang University
- Hangzhou 310029
- P. R. China
| | - Cheng-Yuan Zhu
- ZJU-ENS Joint Laboratory of Medicinal Chemistry
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research
- College of Pharmaceutical Sciences
- Zhejiang University
- Hangzhou
| | - Jin-Hao Zhao
- ZJU-ENS Joint Laboratory of Medicinal Chemistry
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research
- College of Pharmaceutical Sciences
- Zhejiang University
- Hangzhou
| | - Xiao-Wu Dong
- ZJU-ENS Joint Laboratory of Medicinal Chemistry
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research
- College of Pharmaceutical Sciences
- Zhejiang University
- Hangzhou
| |
Collapse
|
10
|
Klimek K, Strubińska J, Czernel G, Ginalska G, Gagoś M. In vitro evaluation of antifungal and cytotoxic activities as also the therapeutic safety of the oxidized form of amphotericin B. Chem Biol Interact 2016; 256:47-54. [PMID: 27350166 DOI: 10.1016/j.cbi.2016.06.022] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Revised: 06/14/2016] [Accepted: 06/23/2016] [Indexed: 11/19/2022]
Abstract
The aim of this study was to evaluate the antifungal and cytotoxic activities of the oxidized form of amphotericin B (AmB-Ox) as well as to determine whether oxidation process of AmB is therapeutically beneficial in vitro. The antifungal activity was estimated against Candida albicans ATCC 10231 and Candida parasilosis ATCC 22019 by broth microdilution method according to the NCCLS M27-A2 standards. The in vitro cytotoxicity was evaluated using normal green monkey kidney cells (GMK) by MTT assay. The obtained results demonstrated that AmB-Ox possesses 16-fold decreased antifungal properties against the two Candida strains and 5-fold lower cytotoxic activity towards GMK cells in comparison with AmB. The therapeutic safety in vitro assessed by calculating the ratio between cytotoxicity (CC50 value) to antifungal activity (MIC value) showed that oxidation of AmB is a very unfavourable process in vitro, because leads to formation of derivative (AmB-Ox) that lost antifungal properties much more rapidly than cytotoxic activity. Thus, the process of the oxidation of AmB in vivo (if it occurs) can be also highly harmful for patient.
Collapse
Affiliation(s)
- Katarzyna Klimek
- Chair and Department of Biochemistry and Biotechnology, Medical University of Lublin, Chodzki Street 1, 20-093, Lublin, Poland.
| | - Joanna Strubińska
- Department of Cell Biology, Institute of Biology and Biotechnology, Maria Curie-Skłodowska University, 20-033, Lublin, Poland
| | - Grzegorz Czernel
- Department of Biophysics, University of Life Sciences in Lublin, Akademicka 13, 20-950, Lublin, Poland.
| | - Grazyna Ginalska
- Chair and Department of Biochemistry and Biotechnology, Medical University of Lublin, Chodzki Street 1, 20-093, Lublin, Poland
| | - Mariusz Gagoś
- Department of Cell Biology, Institute of Biology and Biotechnology, Maria Curie-Skłodowska University, 20-033, Lublin, Poland.
| |
Collapse
|
11
|
Mishra R, Paliwal S, Agarwal A, Sharma S. Identification of Structurally Diverse Antimicrobials Through Sequential Application of Pharmacophore Modeling, Virtual Screening, Molecular Docking and In Vitro Microbiological Assay. Interdiscip Sci 2016; 9:332-340. [PMID: 26947220 DOI: 10.1007/s12539-016-0156-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 02/10/2016] [Accepted: 02/16/2016] [Indexed: 10/22/2022]
Abstract
Dihydrofolate reductase enzyme has been an attractive biological target for the design and development of antimicrobials. Considering this, we have attempted to identify novel dihydrofolate reductase inhibitors through our well-defined in silico and in vitro work flow. An accurate and predictive pharmacophore model comprising of one hydrogen bond acceptor, two hydrophobic and one ring aromatic was developed and utilized as a query to search the National Cancer Institute and Maybridge database leading to retrieval of various compounds which were filtered on the basis of estimated activity, fit value and Lipinski's violation. Selected hits NSC3423, KM09759, NSC391, NSC2091 and HTS00630 were subjected to docking studies which resulted into visualization of potential interaction capabilities of hits in line to pharmacophoric features. The identified hits were evaluated for in vitro antimicrobial potential, and the results revealed that among all the five hits, NSC3423 is the most potent compound with activity against E. coli, P. aeruginosa, S. aureus, B. substilis, A. niger and F. oxysporum. On the other hand, KM09759, NSC391, NSC2091 and HTS00630 showed varying degree of activities against gram-positive, gram-negative and fungal strains.
Collapse
Affiliation(s)
- Ruchi Mishra
- Department of Pharmacy, Banasthali University, Tonk, Rajasthan, 304022, India
| | - Sarvesh Paliwal
- Department of Pharmacy, Banasthali University, Tonk, Rajasthan, 304022, India.
| | - Ankita Agarwal
- Department of Pharmacy, Banasthali University, Tonk, Rajasthan, 304022, India
| | - Swapnil Sharma
- Department of Pharmacy, Banasthali University, Tonk, Rajasthan, 304022, India
| |
Collapse
|
12
|
Pérez-Moreno G, Cantizani J, Sánchez-Carrasco P, Ruiz-Pérez LM, Martín J, el Aouad N, Pérez-Victoria I, Tormo JR, González-Menendez V, González I, de Pedro N, Reyes F, Genilloud O, Vicente F, González-Pacanowska D. Discovery of New Compounds Active against Plasmodium falciparum by High Throughput Screening of Microbial Natural Products. PLoS One 2016; 11:e0145812. [PMID: 26735308 PMCID: PMC4703298 DOI: 10.1371/journal.pone.0145812] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 12/09/2015] [Indexed: 12/03/2022] Open
Abstract
Due to the low structural diversity within the set of antimalarial drugs currently available in the clinic and the increasing number of cases of resistance, there is an urgent need to find new compounds with novel modes of action to treat the disease. Microbial natural products are characterized by their large diversity provided in terms of the chemical complexity of the compounds and the novelty of structures. Microbial natural products extracts have been underexplored in the search for new antiparasitic drugs and even more so in the discovery of new antimalarials. Our objective was to find new druggable natural products with antimalarial properties from the MEDINA natural products collection, one of the largest natural product libraries harboring more than 130,000 microbial extracts. In this work, we describe the optimization process and the results of a phenotypic high throughput screen (HTS) based on measurements of Plasmodium lactate dehydrogenase. A subset of more than 20,000 extracts from the MEDINA microbial products collection has been explored, leading to the discovery of 3 new compounds with antimalarial activity. In addition, we report on the novel antiplasmodial activity of 4 previously described natural products.
Collapse
Affiliation(s)
- Guiomar Pérez-Moreno
- Instituto de Parasitología y Biomedicina “López-Neyra”, Consejo Superior de Investigaciones Científicas, Parque Tecnológico de Ciencias de la Salud, Avenida del Conocimiento, s/n, 18016-Armilla (Granada), Spain
| | - Juan Cantizani
- Fundación MEDINA, Parque Tecnológico de Ciencias de la Salud, Avenida del Conocimiento, 34.18016-Armilla (Granada), Spain
| | - Paula Sánchez-Carrasco
- Instituto de Parasitología y Biomedicina “López-Neyra”, Consejo Superior de Investigaciones Científicas, Parque Tecnológico de Ciencias de la Salud, Avenida del Conocimiento, s/n, 18016-Armilla (Granada), Spain
| | - Luis Miguel Ruiz-Pérez
- Instituto de Parasitología y Biomedicina “López-Neyra”, Consejo Superior de Investigaciones Científicas, Parque Tecnológico de Ciencias de la Salud, Avenida del Conocimiento, s/n, 18016-Armilla (Granada), Spain
| | - Jesús Martín
- Fundación MEDINA, Parque Tecnológico de Ciencias de la Salud, Avenida del Conocimiento, 34.18016-Armilla (Granada), Spain
| | - Noureddine el Aouad
- Fundación MEDINA, Parque Tecnológico de Ciencias de la Salud, Avenida del Conocimiento, 34.18016-Armilla (Granada), Spain
| | - Ignacio Pérez-Victoria
- Fundación MEDINA, Parque Tecnológico de Ciencias de la Salud, Avenida del Conocimiento, 34.18016-Armilla (Granada), Spain
| | - José Rubén Tormo
- Fundación MEDINA, Parque Tecnológico de Ciencias de la Salud, Avenida del Conocimiento, 34.18016-Armilla (Granada), Spain
| | - Víctor González-Menendez
- Fundación MEDINA, Parque Tecnológico de Ciencias de la Salud, Avenida del Conocimiento, 34.18016-Armilla (Granada), Spain
| | - Ignacio González
- Fundación MEDINA, Parque Tecnológico de Ciencias de la Salud, Avenida del Conocimiento, 34.18016-Armilla (Granada), Spain
| | - Nuria de Pedro
- Fundación MEDINA, Parque Tecnológico de Ciencias de la Salud, Avenida del Conocimiento, 34.18016-Armilla (Granada), Spain
| | - Fernando Reyes
- Fundación MEDINA, Parque Tecnológico de Ciencias de la Salud, Avenida del Conocimiento, 34.18016-Armilla (Granada), Spain
| | - Olga Genilloud
- Fundación MEDINA, Parque Tecnológico de Ciencias de la Salud, Avenida del Conocimiento, 34.18016-Armilla (Granada), Spain
| | - Francisca Vicente
- Fundación MEDINA, Parque Tecnológico de Ciencias de la Salud, Avenida del Conocimiento, 34.18016-Armilla (Granada), Spain
| | - Dolores González-Pacanowska
- Instituto de Parasitología y Biomedicina “López-Neyra”, Consejo Superior de Investigaciones Científicas, Parque Tecnológico de Ciencias de la Salud, Avenida del Conocimiento, s/n, 18016-Armilla (Granada), Spain
- * E-mail:
| |
Collapse
|