Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Pharmaceuticals (Basel) ; 13(6)2020 May 28.
Article in English | MEDLINE | ID: mdl-32481669

ABSTRACT

This work showcased the first physicochemical investigation of psoralen (PSO) binding to double stranded DNA (dsDNA) through electroanalytical methods. Results evidenced that PSO presents one non-reversible anodic peak at electric potential (Epa) ≈ 1.42 V, which is associated with its oxidation and the formation of an epoxide derivative. Moreover, PSO analytical signal (i.e., faradaic current) decreases linearly with the addition of dsDNA, while the electric potential associated to PSO oxidation shifts towards more positive values, indicating thence that dsDNA addition hinders PSO oxidation. These findings were corroborated by the chemoinformatic study, which evidenced that PSO intercalated noncovalently at first between base-pairs of the DNA duplex, and then irreversibly formed adducts with both DNA strands, leading up to the formation of a cross-link which bridges the DNA helix, which explains the linear dependence between the faradaic current generated by PSO oxidation and the concentration of DNA in the test-solution, as well as the dependence between Ep and the addition of dsDNA solution. Therefore, the findings herein reported evidence of the applicability of electroanalytical approaches, such as voltammetry in the study of DNA intercalating agents.

2.
Mol Divers ; 21(3): 677-695, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28523625

ABSTRACT

Malaria, a tropical parasitic disease caused by Plasmodium spp., continues to place a heavy social burden, with almost 200 million cases and more than 580,000 deaths per year. Plasmodium falciparum purine nucleoside phosphorylase (PfPNP) can be targeted for antimalarial drug design since its inhibition kills malaria parasites both in vitro and in vivo. Although the currently known inhibitors of PfPNP, immucillins, are orally available and of low toxicity to animals and humans, to the best of our knowledge, none of these compounds has entered clinical trials for the treatment of malaria. Using a pharmacophore-based virtual screening coupled to a consensual molecular docking approach, we identified 59 potential PfPNP inhibitors that are predicted to be orally absorbed in a Caco-2 cell model. Although most of these compounds are predicted to have high plasma protein binding levels, poor water solubility (except for compound 25) and CYP3A4 metabolic stability (except for 4, 7 and 8), four structures (4, 7, 8 and 25) remain as potential leads because of their plausible interaction with a specific hydrophobic pocket of PfPNP, which would confer them higher selectivity for PfPNP over human PNP. Additionally, both predicted Gibbs free energies for binding and molecular dynamics suggest that compound 4 may form a more stable complex with PfPNP than 5[Formula: see text]-methylthio-immucillin-H, a potent and selective inhibitor of PfPNP.


Subject(s)
Antimalarials/chemistry , Antimalarials/pharmacology , Drug Evaluation, Preclinical/methods , Plasmodium falciparum/enzymology , Purine-Nucleoside Phosphorylase/antagonists & inhibitors , Caco-2 Cells , Computer Simulation , Databases, Pharmaceutical , Humans , Hydrophobic and Hydrophilic Interactions , Models, Molecular , Molecular Docking Simulation , Molecular Dynamics Simulation , Plasmodium falciparum/drug effects , Protozoan Proteins/antagonists & inhibitors , Purines/metabolism
3.
J Biomol Struct Dyn ; 29(6): 702-16, 2012.
Article in English | MEDLINE | ID: mdl-22546000

ABSTRACT

Trypanothione reductase has long been investigated as a promising target for chemotherapeutic intervention in Chagas disease, since it is an enzyme of a unique metabolic pathway that is exclusively present in the pathogen but not in the human host, which has the analog Glutathione reductase. In spite of the present data-set includes a small number of compounds, a combined use of flexible docking, pharmacophore perception, ligand binding site prediction, and Grid-Independent Descriptors GRIND2-based 3D-Quantitative Structure-Activity Relationships (QSAR) procedures allowed us to rationalize the different biological activities of a series of 11 aryl ß-aminocarbonyl derivatives, which are inhibitors of Trypanosoma cruzi trypanothione reductase (TcTR). Three QSAR models were built and validated using different alignments, which are based on docking with the TcTR crystal structure, pharmacophore, and molecular interaction fields. The high statistical significance of the models thus obtained assures the robustness of this second generation of GRIND descriptors here used, which were able to detect the most important residues of such enzyme for binding the aryl ß-aminocarbonyl derivatives, besides to rationalize distances among them. Finally, a revised binding mode has been proposed for our inhibitors and independently supported by the different methodologies here used, allowing further optimization of the lead compounds with such combined structure- and ligand-based approaches in the fight against the Chagas disease.


Subject(s)
Butyrophenones/chemistry , NADH, NADPH Oxidoreductases/antagonists & inhibitors , NADH, NADPH Oxidoreductases/chemistry , Protozoan Proteins/antagonists & inhibitors , Protozoan Proteins/chemistry , Trypanosoma cruzi/enzymology , Binding Sites , Chagas Disease/drug therapy , Ligands , Models, Molecular , NADH, NADPH Oxidoreductases/metabolism , Protozoan Proteins/metabolism , Quantitative Structure-Activity Relationship
4.
Future Med Chem ; 3(8): 947-60, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21707398

ABSTRACT

Alzheimer's disease is a complex neurodegenerative disorder of the central nervous system, characterized by amyloid-ß deposits, τ-protein aggregation, oxidative stress and reduced levels of acetylcholine in the brain. One pharmacological approach is to restore acetylcholine level by inhibiting acetylcholinesterase (AChE) with reversible inhibitors, such as galanthamine, thus helping to improve the cognitive symptoms of the disease. In order to design new galanthamine derivatives and search for novel, potential inhibitors with improved interactions, as well as a suitable pharmacokinetic profile and low toxicity, several molecular modeling techniques were applied. These techniques included the investigation of AChE-drug complexes (1QT1 and 1ACJ Protein Data Bank codes), ligand-binding sites calculation within the active site of the enzyme, pharmacophore perception of galanthamine derivatives, virtual screening, toxicophorical analysis and estimation of pharmacokinetics properties. A total of four galanthamine derivatives having a N-alkyl-phenyl chain were designed, since the tertiary amine substituents could reach the peripheral anionic site that is not occupied by galanthamine. In addition, 12 drug-like compounds from the Ilibdiverse database were selected by virtual screening as novel, hypothetical AChE inhibitors. The toxicophorical analysis revealed that only four proposed inhibitors have chemical groups able to develop mutagenicity and chromosome damage. The remaining compounds showed only mild or none toxicophorical alerts. At least three screened compounds presented theoric parameters consistent with good oral bioavailability. The designed molecules have the potential to become new lead compounds that might guide the design of drugs with optimized pharmacodynamic and pharmacokinetic properties in order to improve the treatment of Alzheimer's disease by creating new pharmacotherapeutic options.


Subject(s)
Acetylcholinesterase/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/enzymology , Cholinesterase Inhibitors/chemistry , Cholinesterase Inhibitors/pharmacokinetics , Drug Design , Animals , Cholinesterase Inhibitors/pharmacology , Cholinesterase Inhibitors/toxicity , Humans , Models, Molecular
5.
Drug Metab Lett ; 5(4): 267-75, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22292473

ABSTRACT

Poor pharmacokinetics and toxicity are responsible for most drug candidate failures. In order to attempt to some degree of ADMET (Absorption, Distribution, Metabolism, Excrection and Toxicity) information, in silico predictions arise currently as an interesting alternative to evaluate prototypes during early stages of the drug design processes, especially for anticancer candidates that constitute a class of therapeutic agents that exhibit substantial toxicity. A benzimidazole and a phenylbenzamide derivatives, previously identified as novel anticancer lead compounds able to prevent DNA binding to hnRNP K protein, were evaluated in silico regarding their metabolic profile and toxicity potential in order to give insights to the design of drug candidates with an adequate pharmaceutical profile. Considering the structure of proposed metabolites for both molecules, the phenylbenzamide derivative seems to be a molecule with better pharmaceutic profile, since its possible metabolites present a milder degree of chemical structure toxic alerts than the benzimidazole derivative that can cause chromosome damage induced by the benzimidazole group. It would be desirable during optimization of the phenylbenzamide derivative to maintain these characteristics during generation of analogues with substituents that are not known as potent toxicophoric groups. For the benzimidazole derivative, if the toxic events are really severe as it seems, one possible strategy would be replace the benzimidazole ring system by bioisosteres with lower toxic potential, hoping to maintain or enhance biological activity.


Subject(s)
Antineoplastic Agents/chemistry , Benzimidazoles/chemistry , Computational Biology , Drug Design , Metabolomics , Animals , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/toxicity , Benzimidazoles/metabolism , Benzimidazoles/pharmacology , Benzimidazoles/toxicity , Binding Sites , Biotransformation , Computer-Aided Design , DNA/metabolism , Heterogeneous-Nuclear Ribonucleoproteins/antagonists & inhibitors , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Humans , Ligands , Molecular Structure , Structure-Activity Relationship
6.
J Mol Graph Model ; 28(6): 513-23, 2010 Feb 26.
Article in English | MEDLINE | ID: mdl-20074987

ABSTRACT

We have used various computational methodologies including molecular dynamics, density functional theory, virtual screening, ADMET predictions and molecular interaction field studies to design and analyze four novel potential inhibitors of farnesyltransferase (FTase). Evaluation of two proposals regarding their drug potential as well as lead compounds have indicated them as novel promising FTase inhibitors, with theoretically interesting pharmacotherapeutic profiles, when compared to the very active and most cited FTase inhibitors that have activity data reported, which are launched drugs or compounds in clinical tests. One of our two proposals appears to be a more promising drug candidate and FTase inhibitor, but both derivative molecules indicate potentially very good pharmacotherapeutic profiles in comparison with Tipifarnib and Lonafarnib, two reference pharmaceuticals. Two other proposals have been selected with virtual screening approaches and investigated by us, which suggest novel and alternatives scaffolds to design future potential FTase inhibitors. Such compounds can be explored as promising molecules to initiate a research protocol in order to discover novel anticancer drug candidates targeting farnesyltransferase, in the fight against cancer.


Subject(s)
Computer-Aided Design , Drug Design , Drug Evaluation, Preclinical/methods , Drug Screening Assays, Antitumor/methods , Enzyme Inhibitors/pharmacology , Farnesyltranstransferase/antagonists & inhibitors , Molecular Dynamics Simulation , Neoplasms/drug therapy , Crystallography, X-Ray , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/therapeutic use , Neoplasms/enzymology , Protein Structure, Secondary
7.
J Pharm Pharm Sci ; 12(3): 378-87, 2009.
Article in English | MEDLINE | ID: mdl-20067712

ABSTRACT

PURPOSE: To discuss the contribution of psoralen and bergapten metabolites on psoralens toxicity. METHODS: Computational chemistry prediction of metabolic reactions and toxicophoric groups based on the expert systems Derek and Meteor. RESULTS: a total of 15 metabolites were suggested for both psoralen and bergapten based on phase 1 and 2 biotransformations until the 3rd generation. Five toxicophoric substructures were shared among psoralen, bergapten and their corresponding metabolites; one toxicophoric marker (resorcinol) was only identified in bergapten and its biotransformation products. CONCLUSION: Although the toxic effects of psoralens are well known and documented, there is little information concerning the role of their metabolites in this process. We believe this work add to the knowledge of which molecular substructures are relevant to the process of metabolism and toxicity induction, thus guiding the search and development of more effective and less toxic drugs to treat vitiligo.


Subject(s)
Expert Systems , Ficusin/metabolism , Ficusin/toxicity , Methoxsalen/analogs & derivatives , Photosensitizing Agents/metabolism , Photosensitizing Agents/toxicity , 5-Methoxypsoralen , Animals , Biotransformation , Computational Biology , Computer Simulation , Drug Discovery/methods , Humans , Methoxsalen/metabolism , Methoxsalen/toxicity , Vitiligo/drug therapy
8.
J Phys Chem A ; 112(38): 8902-10, 2008 Sep 25.
Article in English | MEDLINE | ID: mdl-18698751

ABSTRACT

In this work, we have used molecular dynamics, density functional theory, virtual screening, ADMET predictions, and molecular interaction field studies to design and propose eight novel potential inhibitors of CDK2. The eight molecules proposed showed interesting structural characteristics that are required for inhibiting the CDK2 activity and show potential as drug candidates for the treatment of cancer. The parameters related to the Rule of Five were calculated, and only one of the molecules violated more than one parameter. One of the proposals and one of the drug-like compounds selected by virtual screening indicated to be promising candidates for CDK2-based cancer therapy.


Subject(s)
Cyclin-Dependent Kinase 2/antagonists & inhibitors , Drug Evaluation, Preclinical/methods , Models, Molecular , Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Quantum Theory , Animals , Computational Biology , Cyclin-Dependent Kinase 2/chemistry , Cyclin-Dependent Kinase 2/metabolism , Databases, Factual , Humans , Protein Conformation , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/toxicity , Rats , Thermodynamics
9.
J Pharm Sci ; 97(3): 1089-98, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18214973

ABSTRACT

The addition of computer-aided drug design (CADD) technologies to the research and drug discovery approaches could lead to a reduction of up to 50% in the cost of drug design. Designing a drug is the process of finding or creating a molecule which has a specific activity on a biological organism. Development and drug discovery is a time-consuming, expensive, and interdisciplinary process whereas scientific advancements during the past two decades have altered the way pharmaceutical research produces new bioactive molecules. Advances in computational techniques and hardware solutions have enabled in silico methods to speed up lead optimization and identification. We will review current topics in computer-aided molecular design underscoring some of the most recent approaches and interdisciplinary processes. We will discuss some of the most efficient pathways and design.


Subject(s)
Computer-Aided Design , Drug Design
SELECTION OF CITATIONS
SEARCH DETAIL
...