Kajian Farmakoinformatika Senyawa Caulerchlorin dan Racemosin sebagai kandidat Antimalaria
Caulerchlorin and racemosin were bioactive compounds containing in Caulerpa racemosa macroalgae. Previous study, those compounds promoted therapeutical target on metabolic syndrome. However, the therapeutical activity on Malaria not yet defined. Therefore, this study investigated potential activity of caulerchlorin and racemosin from Caulerpa racemosa as antimalarial activity through pharmacoinformatic study. Caulerchlorin and racemosin structures were retrieved from PubChem NCBI and the Plasmodium falciparum dihydroorotate dehydrogenase (PfDHODH) structure was taken out from Protein database with ID 5GJG. Orotic acid was used as native ligand or control. Ligands were predicted their antiplasmodial activity and toxicity. Ligands and protein were interacted by redocking using Molegro Virtual Docker version 6.0 and analyzed by using PyMol 2.3 and Discovery Studio version 21.1.1. Two bioactive compounds also showed antiplasmodial activity and were not mutagenicity and hepatotoxicity affections. Molecular docking performed that caulerchlorin and racemosin exhibited same active residues as well as orotic acid as native substrate. Binding energy revealed two compounds, caulerchlorin and racemosin showed lower binding energy than orotic acid. In summary caulerchlorin and racemosin were potentially inhibited PfDHODH activity by binding substrate sites of PfDHODH protein.
- Research Article
19
- 10.1080/1062936x.2015.1134652
- Feb 1, 2016
- SAR and QSAR in Environmental Research
Plasmodium falciparum, the most fatal parasite that causes malaria, is responsible for over one million deaths per year. P. falciparum dihydroorotate dehydrogenase (PfDHODH) has been validated as a promising drug development target for antimalarial therapy since it catalyzes the rate-limiting step for DNA and RNA biosynthesis. In this study, we investigated the quantitative structure–activity relationships (QSAR) of the antimalarial activity of PfDHODH inhibitors by generating four computational models using a multilinear regression (MLR) and a support vector machine (SVM) based on a dataset of 255 PfDHODH inhibitors. All the models display good prediction quality with a leave-one-out q2 >0.66, a correlation coefficient (r) >0.85 on both training sets and test sets, and a mean square error (MSE) <0.32 on training sets and <0.37 on test sets, respectively. The study indicated that the hydrogen bonding ability, atom polarizabilities and ring complexity are predominant factors for inhibitors’ antimalarial activity. The models are capable of predicting inhibitors’ antimalarial activity and the molecular descriptors for building the models could be helpful in the development of new antimalarial drugs.
- Research Article
4
- 10.1016/j.sciaf.2023.e01944
- Oct 14, 2023
- Scientific African
In vitro antiplasmodial activity, LC-MS analysis, and molecular docking studies of bioactive compounds from Tetrapleura tetraptera (Fabaceae) fruits
- Research Article
20
- 10.1080/1062936x.2016.1189959
- Jun 2, 2016
- SAR and QSAR in Environmental Research
Plasmodium falciparum dihydroorotate dehydrogenase (PfDHODH) catalyses the fourth reaction of de novo pyrimidine biosynthesis in parasites, and represents an important target for the treatment of malaria. In this study, we describe pharmacophore-based virtual screening combined with docking study and biological evaluation as a rational strategy for identification of novel hits as antimalarial agents. Pharmacophore models were established from known PfDHODH inhibitors using the GALAHAD module with IC50 values ranging from 0.033 μM to 142 μM. The best pharmacophore model consisted of three hydrogen bond acceptor, one hydrogen bond donor and one hydrophobic features. The pharmacophore models were validated through receiver operating characteristic and Günere–Henry scoring methods. The best pharmacophore model as a 3D search query was searched against the IBS database. Several compounds with different structures (scaffolds) were retrieved as hit molecules. Among these compounds, those with a QFIT value of more than 81 were docked in the PfDHODH enzyme to further explore the binding modes of these compounds. In silico pharmacokinetic and toxicities were predicted for the best docked molecules. Finally, the identified hits were evaluated in vivo for their antimalarial activity in a parasite inhibition assay. The hits reported here showed good potential to become novel antimalarial agents.
- Research Article
2
- 10.2174/1573409916666200527133126
- May 27, 2020
- Current computer-aided drug design
Leflunomide (LFM) and its active metabolite, teriflunomide (TFM), have drawn a lot of attention for their anticancer activities, treatment of rheumatoid arthritis and malaria due to their capability to inhibit dihydroorotate dehydrogenase (DHODH) and Plasmodium falciparum dihydroorotate dehydrogenase (PfDHODH) enzyme. In this investigation, the strength of intramolecular hydrogen bond (IHB) in five analogs of TFM (ATFM) was analyzed employing density functional theory (DFT) using B3LYP/6-311++G (d, p) level and molecular orbital analysis in the gas phase and water solution. A detailed electronic structure study was performed using the quantum theory of atoms in molecules (QTAIM) and the hydrogen bond energies (EHB) of stable conformer obtained in the range of 76-97 kJ/mol, as a medium hydrogen bond. The effect of substitution on the IHB nature was studied by natural bond orbital analysis (NBO). 1H NMR calculations showed an upward trend in the proton chemical shift of the enolic proton in the chelated ring (14.5 to 15.7ppm) by increasing the IHB strength. All the calculations confirmed the strongest IHB in 5-F-ATFM and the weakest IHB in 2-FATFM. Molecular orbital analysis, including the HOMO-LUMO gap and chemical hardness, was performed to compare the reactivity of inhibitors. Finally, molecular docking analysis was carried out to identify the potency of inhibition of these compounds against PfDHODH enzyme. TFM acts as an inhibitor of dihydroorotate dehydrogenase (DHODH) and Plasmodium falciparum dihydroorotate dehydrogenase (PfDHODH) enzyme. Leflunomide and its active metabolite teriflunomide have been identified as drugs for treatment of some diseases, such as multiple sclerosis (MS), rheumatoid arthritis (RA), malaria, and cancer. Hydrogen bonds play a key role in the interaction between drugs and enzymes. The aim of the present work is to investigate the effect of the strength of intramolecular hydrogen bonds (IHBs) in the active metabolite analogs of leflunomide or analogs of teriflunomide (ATFMs) and study the interaction of these inhibitors against the PfDHODH enzyme using quantum mechanical methods. At first, intramolecular hydrogen bonds in five ATFMs were evaluated by the DFT method, quantum theory of atoms in molecules (QTAIM), nuclear magnetic resonance (NMR), natural bond orbital (NBO), and molecular orbital (MO) analyses. Then, the interaction of these inhibitors against the PfDHODH enzyme were compared using molecular docking study. All the computed results confirm the following trend in the intramolecular hydrogen bond strength in five mono-halo-substituted 2-cyano-3-hydroxy-N-phenylbut-2-enamide (ATFM): 5-FATFM> 4-Br-ATFM ≈ 3-Br-ATFM>3-Cl-ATFM>TFM-Z>2-F-ATFM which is in agreement with QTAIM, NMR, and NBO results. Docking results show that 5-F-ATFM (EHB=97kJ/mol) has the minimum MolDock score due to its considerable IHB strength. For strong IHBs (EHB>100kJ/mol), C=O and O-H group are involved in the intramolecular interactions and do not contribute to the external interactions. Also, the docking study revealed maximum binding energy between TFM-Z and PfDHODH enzyme.
- Research Article
121
- 10.1074/jbc.m109.028589
- Sep 1, 2009
- Journal of Biological Chemistry
Malaria remains a major global health burden and current drug therapies are compromised by resistance. Plasmodium falciparum dihydroorotate dehydrogenase (PfDHODH) was validated as a new drug target through the identification of potent and selective triazolopyrimidine-based DHODH inhibitors with anti-malarial activity in vivo. Here we report x-ray structure determination of PfDHODH bound to three inhibitors from this series, representing the first of the enzyme bound to malaria specific inhibitors. We demonstrate that conformational flexibility results in an unexpected binding mode identifying a new hydrophobic pocket on the enzyme. Importantly this plasticity allows PfDHODH to bind inhibitors from different chemical classes and to accommodate inhibitor modifications during lead optimization, increasing the value of PfDHODH as a drug target. A second discovery, based on small molecule crystallography, is that the triazolopyrimidines populate a resonance form that promotes charge separation. These intrinsic dipoles allow formation of energetically favorable H-bond interactions with the enzyme. The importance of delocalization to binding affinity was supported by site-directed mutagenesis and the demonstration that triazolopyrimidine analogs that lack this intrinsic dipole are inactive. Finally, the PfDHODH-triazolopyrimidine bound structures provide considerable new insight into species-selective inhibitor binding in this enzyme family. Together, these studies will directly impact efforts to exploit PfDHODH for the development of anti-malarial chemotherapy.
- Research Article
7
- 10.2174/1570180819666220422120707
- Mar 1, 2023
- Letters in Drug Design & Discovery
Background: Plasmodium falciparum dihydroorotate dehydrogenase (PfDODH) is one of the enzymes currently explored in the treatment of malaria. Although there is currently no clinically approved drug targeting PfDODH, many of the compounds in clinical trials have [1, 2, 4,] triazolo [1, 5-a] pyrimidin- 7-amine backbone structure. Objective: This study sought to design new compounds from the fragments of known experimental inhibitors of PfDODH. Methods: Nine experimental compounds retrieved from Drug Bank online were downloaded and broken into fragments using the Schrodinger power shell; the fragments were recombined to generate new ligand structures using the BREED algorithm. The new compounds were docked with PfDODH crystal structure, after which the compounds were filtered with extensive drug-likeness and toxicity parameters. A 2D-QSAR model was built using the multiple linear regression method and externally validated. The electronic properties of the compounds were calculated using the density functional theory method. Results: Structural investigation of the six designed compounds, which had superior binding energies than the standard inhibitors, showed that five of them had [1, 2, 4,] triazolo [1, 5-a] pyrimidin-7-amine moieties and interacted with essential residues at the PfDODH binding site. In addition to their drug-like and pharmacokinetic properties, they also showed minimal toxicities. The externally validated 2D-QSAR model with R2 and Q2 values of 0.6852 and 0.6691 confirmed the inhibitory prowess of these compounds against PfDODH. The DFT calculations showed regions of the molecules prone to electrophilic and nucleophilic attacks. Conclusion: The current study thus provides insight into the development of a new set of potent PfDODH inhibitors.
- Research Article
19
- 10.1111/cbdd.13197
- May 12, 2018
- Chemical Biology & Drug Design
Plasmodium falciparum dihydroorotate dehydrogenase (PfDHODH) is a promising drug target for antimalarial chemotherapy. In our continuous efforts to develop more potent PfDHODH inhibitors, a unique library of active ingredients from traditional Chinese medicine (TCM) has been collected and was screened in this study. Through the initial screening, we found that coptisine, a natural alkaloid from TCM Coptidis Rhizoma, was a novel and potent inhibitor of PfDHODH with an IC50 value of 1.83±0.08μm. At the same time, enzyme kinetic analysis using Lineweaver-Burk plot indicated that coptisine is an uncompetitive inhibitor for PfDHODH. Thermal shift assay and molecular docking simulation research reveal that coptisine is capable of binding with PfDHODH. Moreover, coptisine exhibits weak inhibition activity against human DHODH, indicating that coptisine is a selective inhibitor of PfDHODH. Taken together, our study highlights the potential of active ingredients in TCM as valuable resource for discovering novel chemical scaffolds for PfDHODH.
- Research Article
8
- 10.1016/j.compbiolchem.2022.107787
- Nov 8, 2022
- Computational Biology and Chemistry
Integrated structure-guided computational design of novel substituted quinolizin-4-ones as Plasmodium falciparum dihydroorotate dehydrogenase (PfDHODH) inhibitors
- Research Article
38
- 10.1080/07391102.2017.1404935
- Nov 27, 2017
- Journal of Biomolecular Structure and Dynamics
The structural motifs of chalcones, flavones, and triazoles with varied substitutions have been studied for the antimalarial activity. In this study, 25 novel derivatives of chalcone and flavone hybrid derivatives with 1, 2, 3-triazole linkage are docked with Plasmodium falciparum dihydroorotate dehydrogenase to establish their inhibitory activity against Plasmodium falciparum. The best binding conformation of the ligands at the catalytic site of dihydroorotate dehydrogenase are selected to characterize the best bound ligand using the best consensus score and the number of hydrogen bond interactions. The ligand namely (2E)-3-(4-{[1-(3-chloro-4-fluorophenyl)-1H-1, 2, 3-triazol-4-yl]methoxy}-3-methoxyphenyl-1-(2-hydroxy-4,6-dimethoxyphenyl)prop-2-en-1-one, is one the among the five best docked ligands, which interacts with the protein through nine hydrogen bonds and with a consensus score of five. To refine and confirm the docking study results, the stability of complexes is verified using Molecular Dynamics Simulations, Molecular Mechanics /Poisson–Boltzmann Surface Area free binding energy analysis, and per residue contribution for the binding energy. The study implies that the best docked Plasmodium falciparum dihydroorotate dehydrogenase–ligand complex is having high negative binding energy, most stable, compact, and rigid with nine hydrogen bonds. The study provides insight for the optimization of chalcone and flavone hybrids with 1, 2, 3-triazole linkage as potent inhibitors.
- Research Article
15
- 10.1007/s00044-010-9323-4
- Mar 4, 2010
- Medicinal Chemistry Research
The search for new antimalarial agents is necessary as current drugs in the market have become vulnerable due to the emergence of resistant strains of Plasmodium falciparum (Pf). The enzyme dihydroorotatedehydrogenase (PfDHODH) is a validated target for development of antimalarial agents. PfDHODH is a crucial enzyme in the de novo pyrimidine biosynthesis pathway and is essential for the growth of the parasite. In this article, we report the design, synthesis and evaluation of benzanilides as inhibitors of PfDHODH. From the pool of molecules designed using molecular modeling techniques, candidates were shortlisted for further evaluation based on docking scores and 3D-QSAR studies. The activities of these shortlisted analogs were predicted from CoMFA and CoMSIA models. The most promising molecules were synthesized using solvent-free microwave-assisted synthesis and their structures characterized by spectroscopic techniques. The molecules were screened for in vitro antimalarial activity by the whole cell assay method. Two molecules viz. KMC-3 and KMC-15 were found to be active at 8.7 and 5.7 μM concentrations, respectively.
- Research Article
65
- 10.1021/acs.jcim.5b00680
- Mar 8, 2016
- Journal of Chemical Information and Modeling
Plasmodium falciparum dihydroorotate dehydrogenase (PfDHODH), a key enzyme in the de novo pyrimidine biosynthesis pathway, which the Plasmodium falciparum relies on exclusively for survival, has emerged as a promising target for antimalarial drugs. In an effort to discover new and potent PfDHODH inhibitors, 3D-QSAR pharmacophore models were developed based on the structures of known PfDHODH inhibitors and the validated Hypo1 model was used as a 3D search query for virtual screening of the National Cancer Institute database. The virtual hit compounds were further filtered based on molecular docking and Molecular Mechanics/Generalized Born Surface Area binding energy calculations. The combination of the pharmacophore and structure-based virtual screening resulted in the identification of nine new compounds that showed >25% inhibition of PfDHODH at a concentration of 10 μM, three of which exhibited IC50 values in the range of 0.38-20 μM. The most active compound, NSC336047, displayed species-selectivity for PfDHODH over human DHODH and inhibited parasite growth with an IC50 of 26 μM. In addition to this, 13 compounds inhibited parasite growth with IC50 values of ≤ 50 μM, 4 of which showed IC50 values in the range of 5-12 μM. These compounds could be further explored in the identification and development of more potent PfDHODH and parasite growth inhibitors.
- Book Chapter
3
- 10.1007/978-3-030-05282-9_6
- Jan 1, 2019
Structure-based drug design (SBDD) is being efficiently used for the design of antimalarial agents. It is a very effective tool for challenges like drug selectivity and resistance. Over the past decade, a considerable number of druggable targets have been explored—these include Na+ ATPase 4 ion channel, cytochrome bc1, mitochondrial electron transport chain, phosphatidylinositol 4-kinase (PfPI4 K), dihydroorotate dehydrogenase, hemozoin formation, dihydrofolate reductase inhibitors, etc. Among these, Plasmodium falciparum dihydroorotate dehydrogenase (PfDHODH) is a new and very promising target. PfDHODH has shown considerable potential in arresting growth of the parasite at blood stage by inhibiting pyrimidine biosynthesis. This chapter provides a review of all the SBDD efforts for the development of inhibitors against PfDHODH.
- Research Article
- 10.1080/1062936x.2025.2523386
- Jun 3, 2025
- SAR and QSAR in Environmental Research
Plasmodium falciparum dihydroorotate dehydrogenase (PfDHODH) is a well-established target for developing novel antimalarial agents. Novel imidazo[1,2-a]pyrimidines were designed as PfDHODH inhibitors using a fragment-based drug design (FADD) approach. A library of active molecules targeting PfDHODH was analysed to generate fragments using the RDKit BRICS module. These fragments were screened by docking them into the active site of the PfDHODH enzyme. Among them, the lead fragment, fragment-11, demonstrated a significant binding affinity of −6.895 kcal/mol. This fragment was optimized using a fragment-growing approach via the FragGrow webserver. From the 471 generated molecules, two showed binding scores of −7.9 and −7.0 kcal/mol. These molecules were further optimized, resulting in a lead molecule with a binding score of −11.3 kcal/mol. Based on the results from the FragGrow webserver, 216 novel imidazo[1,2-a]pyrimidines were designed using the scaffold-hopping approach. The ADMET properties of these compounds revealing that all the designed compounds exhibited drug-like properties. Docking studies indicated that compounds 28d, 46d, and 49d had strong binding affinities, with 28d showing the highest score of −10.41 kcal/mol. Furthermore, molecular dynamics (MD) simulations of 28d demonstrated good stability in the enzyme-ligand complex. This comprehensive in silico study suggests that imidazo[1,2-a]pyrimidines can serve as potent PfDHODH inhibitors.
- Research Article
5
- 10.1007/s42452-020-2955-1
- Jun 5, 2020
- SN Applied Sciences
Malaria, a disease caused by one of the world’s fatal parasites Plasmodium falciparum, is responsible for over a million death annually. P. falciparum dihydroorotate dehydrogenase (PfDHODH) is a validated target of this deadly parasite. Quantitative structure–activity relationship and molecular docking in silico methods were employed in the discovery of unique PfDHODH inhibitors from the computational design derivatives of indolyl-3-ethanone-α-thioethers through models generation via a genetic function algorithm methods. The best model indicates good power of prediction with coefficient of determination, R2 = 0.9482, adjusted coefficient of determination ({text{R}}_{text{adj}}^{2}) = 0.9288, Leave one out cross-validation coefficient (Q2) = 0.9201 and the external validation ({text{R}}_{text{pred}}^{2}) = 0.6467. The contribution of every descriptor in the model was investigated through finding their mean effect to (pIC50) the activities of the compounds. With MATS5m (− 0.11725), RDF75m (− 0.12097), VE3_Dzp (0.14697), and MLFER_BH (1.08528) contributing more to the model, while AATSC8p (− 0.04833) and minHBa (0.05430) contributed the least to the model. Hence, the mean effect indicated MLFER_BH to be the most relevant descriptor, which aided the design of five derivatives of indolyl-3-ethanone-α-thioethers. All the designed antimalarial compounds were deeply docked within the binding region thereby forming several hydrogens and hydrophobic bonds leading to the generation of better binding affinity and high binding scores (− 156.181 kcal/mol) compared to the design template (− 138.201 kcal/mol) and the standard drug (− 128.467 kcal/mol). Furthermore, all the five designed antimalarial compounds were found to be better bonded to the binding pocket of PfDHODH than other compounds reported by other researchers.
- Research Article
29
- 10.1074/jbc.273.48.31837
- Nov 1, 1998
- Journal of Biological Chemistry
To determine if recognition of the Yersinia pseudotuberculosis invasin protein and natural substrates requires identical integrin residues, a region of the human alpha3 integrin chain predicted to be involved in substrate adhesion was targeted for mutation. One point mutation located in a region of the third N-terminal repeat of the alpha3 chain, alpha3-W220A, failed to promote adhesion to the natural alpha3 beta1 substrate epiligrin but maintained near wild type levels of adhesion to invasin. A second nearby mutation, alpha3-Y218A, which showed no detectable adhesion to epiligrin, was only partially attenuated for invasin binding as well as invasin-mediated bacterial uptake. A third substitution, alpha3-D154A, predicted to be in the second N-terminal repeat not known to be implicated in cell adhesion, was competent for invasin-promoted adhesion events and appeared to encode a receptor of increased activity, as it had a higher efficiency than wild type receptor for adhesion to epiligrin. Cell lines expressing this derivative were not recognized by a function blocking anti-alpha3 antibody, indicating that the second and third repeats of the alpha3 chain are either closely linked in space or the second repeat can modulate activity of the third. Differential effects on substrate adhesion do not appear to be associated with all integrin alpha chain mutations, as alpha4 chain mutations affecting the divalent cation binding domains depressed adhesion to invasin to a significant extent.
- Research Article
- 10.29080/biotropic.v8i2.2080
- Sep 21, 2024
- Biotropic : The Journal of Tropical Biology
- Journal Issue
- 10.29080/biotropic.v8i2
- Sep 21, 2024
- Biotropic : The Journal of Tropical Biology
- Research Article
- 10.29080/biotropic.v8i2.2142
- Sep 21, 2024
- Biotropic : The Journal of Tropical Biology
- Research Article
- 10.29080/biotropic.v8i2.2114
- Sep 21, 2024
- Biotropic : The Journal of Tropical Biology
- Research Article
- 10.29080/biotropic.v8i2.2077
- Sep 21, 2024
- Biotropic : The Journal of Tropical Biology
- Research Article
- 10.29080/biotropic.v8i2.2138
- Sep 21, 2024
- Biotropic : The Journal of Tropical Biology
- Research Article
- 10.29080/biotropic.v8i2.2135
- Sep 21, 2024
- Biotropic : The Journal of Tropical Biology
- Journal Issue
- 10.29080/biotropic.v8i1
- Apr 18, 2024
- Biotropic : The Journal of Tropical Biology
- Research Article
- 10.29080/biotropic.v8i1.1968
- Feb 28, 2024
- Biotropic : The Journal of Tropical Biology
- Research Article
- 10.29080/biotropic.v8i1.2082
- Feb 28, 2024
- Biotropic : The Journal of Tropical Biology
- Ask R Discovery
- Chat PDF
AI summaries and top papers from 250M+ research sources.