Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Curr Alzheimer Res ; 20(2): 120-129, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37278042

RESUMEN

BACKGROUND: Alzheimer's disease (AD) and Multiple sclerosis (MS) lead to neurodegenerative processes negatively affecting millions of people worldwide. Their treatment is still difficult and practically incomplete. One of the most commonly used drugs against these neurodegenerative diseases is 4-aminopyridine. However, its use is confined by the high toxicity. OBJECTIVES: The aim of this work is to obtain new peptide derivatives of 4-aminopyridine with decreased toxicity compared to 4-aminopyridine. METHODS: Synthesis was conducted in solution using a consecutive condensation approach. The new derivatives were characterized by melting points, NMR, and Mass spectra. Important ADME (absorption, distribution, metabolism, and excretion) properties have been studied in silico using ACD/Percepta v.2020.2.0 software. Acute toxicity was determined in mice according to a Standard protocol. All new derivatives were tested in vitro for cytotoxic activity in a panel of human (HEP-G2, BV-173) and murine (NEURO 2A) tumor cell lines via a standard MTT-based colorimetric method. ß-secretase inhibitory activity was determined by applying the fluorescent method. RESULTS: New derivatives of 4-aminopyridine containing analogues of the ß-secretase inhibitory peptide (Boc-Val-Asn-Leu-Ala-OH) were obtained. The in vivo toxicity of the tested compounds was found to be as high as 1500 mg/kg. Cell toxicity screening against tumor cell lines of different origins showed negligible growth-inhibitory effects of all investigated 4-aminopyridine analogues. CONCLUSION: Synthesis of new peptide derivatives of 4-aminopyridine is reported. Acute toxicity studies revealed a ca. 150 times lower toxicity of the new compounds as compared to 4-aminopyridine that may be ascribed to their peptide fragment.


Asunto(s)
4-Aminopiridina , Enfermedad de Alzheimer , Ratones , Humanos , Animales , 4-Aminopiridina/toxicidad , 4-Aminopiridina/uso terapéutico , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Enfermedad de Alzheimer/tratamiento farmacológico , Péptidos/farmacología , Línea Celular Tumoral
2.
Molecules ; 27(10)2022 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-35630751

RESUMEN

The increased use of polyphenols nowadays poses the need for identification of their new pharmacological targets. Recently, structure similarity-based virtual screening of DrugBank outlined pseudopurpurin, a hydroxyanthraquinone from Rubia cordifolia spp., as similar to gatifloxacin, a synthetic antibacterial agent. This suggested the bacterial DNA gyrase and DNA topoisomerase IV as potential pharmacological targets of pseudopurpurin. In this study, estimation of structural similarity to referent antibacterial agents and molecular docking in the DNA gyrase and DNA topoisomerase IV complexes were performed for a homologous series of four hydroxyanthraquinones. Estimation of shape- and chemical feature-based similarity with (S)-gatifloxacin, a DNA gyrase inhibitor, and (S)-levofloxacin, a DNA topoisomerase IV inhibitor, outlined pseudopurpurin and munjistin as the most similar structures. The docking simulations supported the hypothesis for a plausible antibacterial activity of hydroxyanthraquinones. The predicted docking poses were grouped into 13 binding modes based on spatial similarities in the active site. The simultaneous presence of 1-OH and 3-COOH substituents in the anthraquinone scaffold were emphasized as relevant features for the binding modes' variability and ability of the compounds to strongly bind in the DNA-enzyme complexes. The results reveal new potential pharmacological targets of the studied polyphenols and help in their prioritization as drug candidates and dietary supplements.


Asunto(s)
Topoisomerasa de ADN IV , Rubia , Antibacterianos/química , Antibacterianos/farmacología , Girasa de ADN/química , Gatifloxacina , Simulación del Acoplamiento Molecular , Polifenoles
3.
Front Pharmacol ; 13: 831791, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35321325

RESUMEN

Sdox is a hydrogen sulfide (H2S)-releasing doxorubicin effective in P-glycoprotein-overexpressing/doxorubicin-resistant tumor models and not cytotoxic, as the parental drug, in H9c2 cardiomyocytes. The aim of this study was the assessment of Sdox drug-like features and its absorption, distribution, metabolism, and excretion (ADME)/toxicity properties, by a multi- and transdisciplinary in silico, in vitro, and in vivo approach. Doxorubicin was used as the reference compound. The in silico profiling suggested that Sdox possesses higher lipophilicity and lower solubility compared to doxorubicin, and the off-targets prediction revealed relevant differences between Dox and Sdox towards several cancer targets, suggesting different toxicological profiles. In vitro data showed that Sdox is a substrate with lower affinity for P-glycoprotein, less hepatotoxic, and causes less oxidative damage than doxorubicin. Both anthracyclines inhibited CYP3A4, but not hERG currents. Unlike doxorubicin, the percentage of zebrafish live embryos at 72 hpf was not affected by Sdox treatment. In conclusion, these findings demonstrate that Sdox displays a more favorable drug-like ADME/toxicity profile than doxorubicin, different selectivity towards cancer targets, along with a greater preclinical efficacy in resistant tumors. Therefore, Sdox represents a prototype of innovative anthracyclines, worthy of further investigations in clinical settings.

4.
Molecules ; 26(21)2021 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-34770768

RESUMEN

The aim of this study was to investigate the chemical space and interactions of natural compounds with sulfotransferases (SULTs) using ligand- and structure-based in silico methods. An in-house library of natural ligands (hormones, neurotransmitters, plant-derived compounds and their metabolites) reported to interact with SULTs was created. Their chemical structures and properties were compared to those of compounds of non-natural (synthetic) origin, known to interact with SULTs. The natural ligands interacting with SULTs were further compared to other natural products for which interactions with SULTs were not known. Various descriptors of the molecular structures were calculated and analyzed. Statistical methods (ANOVA, PCA, and clustering) were used to explore the chemical space of the studied compounds. Similarity search between the compounds in the different groups was performed with the ROCS software. The interactions with SULTs were additionally analyzed by docking into different experimental and modeled conformations of SULT1A1. Natural products with potentially strong interactions with SULTs were outlined. Our results contribute to a better understanding of chemical space and interactions of natural compounds with SULT enzymes and help to outline new potential ligands of these enzymes.


Asunto(s)
Productos Biológicos/química , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Sulfotransferasas/química , Productos Biológicos/farmacología , Análisis por Conglomerados , Flavonoides , Ligandos , Estructura Molecular , Polifenoles , Relación Estructura-Actividad , Sulfotransferasas/metabolismo
5.
Cancers (Basel) ; 13(21)2021 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-34771471

RESUMEN

Tyrosine kinase inhibitors (TKIs) often interact with the multidrug resistant (MDR) phenotype of cancer cells. In some cases, TKIs increase the susceptibility of MDR cancer cells to chemotherapy. As the overexpression of membrane transporter P-glycoprotein (P-gp) is the most common alteration in MDR cancer cells, we investigated the effects of TKI pyrazolo[3,4-d]pyrimidines on P-gp inhibition in two cellular models comprising sensitive and corresponding MDR cancer cells (human non-small cell lung carcinoma and colorectal adenocarcinoma). Tested TKIs showed collateral sensitivity by inducing stronger inhibition of MDR cancer cell line viability. Moreover, TKIs directly interacted with P-gp and inhibited its ATPase activity. Their potential P-gp binding site was proposed by molecular docking simulations. TKIs reversed resistance to doxorubicin and paclitaxel in a concentration-dependent manner. The expression studies excluded the indirect effect of TKIs on P-gp through regulation of its expression. A kinetics study showed that TKIs decreased P-gp activity and this effect was sustained for seven days in both MDR models. Therefore, pyrazolo[3,4-d]pyrimidines with potential for reversing P-gp-mediated MDR even in prolonged treatments can be considered a new therapeutic strategy for overcoming cancer MDR.

6.
Toxics ; 9(5)2021 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-33919268

RESUMEN

The cytotoxicity and microbicidal capacity of seven organic solvents commonly applied for studying plant extracts and bioactive compounds were systematically investigated based on international standards. Four cell lines of normal (CCL-1, HaCaT) or tumor (A-375, A-431) tissue origin, seven bacterial and one fungal strain were used. The impact of the least toxic solvents in the determination of in vitro cytotoxicity was evaluated using a standardized extract from Vaccinium macrocarpon containing 54.2% v/v proanthocyanidins (CystiCran®). The solvents ethanol, methoxyethanol and polyethylene glycol were the least cytotoxic to all cell lines, with a maximum tolerated concentration (MTC) between 1 and 2% v/v. Ethanol, methanol and polyethylene glycol were mostly suitable for antimicrobial susceptibility testing, with minimum inhibitory concentrations (MICs) ≥ 25% v/v. The MTC values of the solvents dimethyl sulfoxide, dimethoxyethane and dimethylformamide varied from 0.03% to 1.09% v/v. The MICs of dimethyl sulfoxide, methoxyethanol and dimethoxyethane were in the range of 3.125-25% v/v. The cytotoxic effects of CystiCran® on eukaryotic cell lines were directly proportional to the superimposed effect of the solvents used. The results of this study can be useful for selecting the appropriate solvents for in vitro estimation of the cytotoxic and growth inhibitory effects of bioactive molecules in eukaryotic and prokaryotic cells.

7.
Drug Resist Updat ; 52: 100713, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32615525

RESUMEN

Overcoming multidrug resistance represents a major challenge for cancer treatment. In the search for new chemotherapeutics to treat malignant diseases, drug repurposing gained a tremendous interest during the past years. Repositioning candidates have often emerged through several stages of clinical drug development, and may even be marketed, thus attracting the attention and interest of pharmaceutical companies as well as regulatory agencies. Typically, drug repositioning has been serendipitous, using undesired side effects of small molecule drugs to exploit new disease indications. As bioinformatics gain increasing popularity as an integral component of drug discovery, more rational approaches are needed. Herein, we show some practical examples of in silico approaches such as pharmacophore modelling, as well as pharmacophore- and docking-based virtual screening for a fast and cost-effective repurposing of small molecule drugs against multidrug resistant cancers. We provide a timely and comprehensive overview of compounds with considerable potential to be repositioned for cancer therapeutics. These drugs are from diverse chemotherapeutic classes. We emphasize the scope and limitations of anthelmintics, antibiotics, antifungals, antivirals, antimalarials, antihypertensives, psychopharmaceuticals and antidiabetics that have shown extensive immunomodulatory, antiproliferative, pro-apoptotic, and antimetastatic potential. These drugs, either used alone or in combination with existing anticancer chemotherapeutics, represent strong candidates to prevent or overcome drug resistance. We particularly focus on outcomes and future perspectives of drug repositioning for the treatment of multidrug resistant tumors and discuss current possibilities and limitations of preclinical and clinical investigations.


Asunto(s)
Antineoplásicos/farmacología , Reposicionamiento de Medicamentos , Resistencia a Múltiples Medicamentos/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Antineoplásicos/uso terapéutico , Biología Computacional , Simulación por Computador , Descubrimiento de Drogas/métodos , Humanos , Neoplasias/patología
8.
Antioxidants (Basel) ; 9(5)2020 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-32380762

RESUMEN

Silymarin is the standardized extract from the fruits of Silybum marianum (L.) Gaertn., a well-known hepatoprotectant and antioxidant. Recently, bioactive compounds of silymarin, i.e., silybins and their 2,3-dehydro derivatives, have been shown to exert anticancer activities, yet with unclear mechanisms. This study combines in silico and in vitro methods to reveal the potential interactions of optically pure silybins and dehydrosilybins with novel protein targets. The shape and chemical similarity with approved drugs were evaluated in silico, and the potential for interaction with the Hedgehog pathway receptor Smoothened (SMO) and BRAF kinase was confirmed by molecular docking. In vitro studies on SMO and BRAF V600E kinase activity and in BRAF V600E A-375 human melanoma cell lines were further performed to examine their effects on these proteins and cancer cell lines and to corroborate computational predictions. Our in silico results direct to new potential targets of silymarin constituents as dual inhibitors of BRAF and SMO, two major targets in anticancer therapy. The experimental studies confirm that BRAF kinase and SMO may be involved in mechanisms of anticancer activities, demonstrating dose-dependent profiles, with dehydrosilybins showing stronger effects than silybins. The results of this work outline the dual SMO/BRAF effect of flavonolignans from Silybum marianum with potential clinical significance. Our approach can be applied to other natural products to reveal their potential targets and mechanism of action.

9.
Drug Resist Updat ; 50: 100682, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32087558

RESUMEN

Multidrug resistance (MDR) is the dominant cause of the failure of cancer chemotherapy. The design of antitumor drugs that are able to evade MDR is rapidly evolving, showing that this area of biomedical research attracts great interest in the scientific community. The current review explores promising recent approaches that have been developed with the aim of circumventing or overcoming MDR. Encouraging results have been obtained in the investigation of the MDR-modulating properties of various classes of natural compounds and their analogues. Inhibition of P-gp or downregulation of its expression have proven to be the main mechanisms by which MDR can be surmounted. The use of hybrid molecules that are able to simultaneously interact with two or more cancer cell targets is currently being explored as a means to circumvent drug resistance. This strategy is based on the design of hybrid compounds that are obtained either by merging the structural features of separate drugs, or by conjugating two drugs or pharmacophores via cleavable/non-cleavable linkers. The approach is highly promising due to the pharmacokinetic and pharmacodynamic advantages that can be achieved over the independent administration of the two individual components. However, it should be stressed that the task of obtaining successful multivalent drugs is a very challenging one. The conjugation of anticancer agents with nitric oxide (NO) donors has recently been developed, creating a particular class of hybrid that can combat tumor drug resistance. Appropriate NO donors have been shown to reverse drug resistance via nitration of ABC transporters and by interfering with a number of metabolic enzymes and signaling pathways. In fact, hybrid compounds that are produced by covalently attaching NO-donors and antitumor drugs have been shown to elicit a synergistic cytotoxic effect in a variety of drug resistant cancer cell lines. Another strategy to circumvent MDR is based on nanocarrier-mediated transport and the controlled release of chemotherapeutic drugs and P-gp inhibitors. Their pharmacokinetics are governed by the nanoparticle or polymer carrier and make use of the enhanced permeation and retention (EPR) effect, which can increase selective delivery to cancer cells. These systems are usually internalized by cancer cells via endocytosis and accumulate in endosomes and lysosomes, thus preventing rapid efflux. Other modalities to combat MDR are described in this review, including the pharmaco-modulation of acridine, which is a well-known scaffold in the development of bioactive compounds, the use of natural compounds as means to reverse MDR, and the conjugation of anticancer drugs with carriers that target specific tumor-cell components. Finally, the outstanding potential of in silico structure-based methods as a means to evaluate the ability of antitumor drugs to interact with drug transporters is also highlighted in this review. Structure-based design methods, which utilize 3D structural data of proteins and their complexes with ligands, are the most effective of the in silico methods available, as they provide a prediction regarding the interaction between transport proteins and their substrates and inhibitors. The recently resolved X-ray structure of human P-gp can help predict the interaction sites of designed compounds, providing insight into their binding mode and directing possible rational modifications to prevent them from becoming P-gp drug substrates. In summary, although major efforts were invested in the search for new tools to combat drug resistant tumors, they all require further implementation and methodological development. Further investigation and progress in the abovementioned strategies will provide significant advances in the rational combat against cancer MDR.


Asunto(s)
Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Diseño de Fármacos , Resistencia a Antineoplásicos/fisiología , Neoplasias/tratamiento farmacológico , Tecnología Farmacéutica/métodos , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/antagonistas & inhibidores , Transportadoras de Casetes de Unión a ATP/metabolismo , Acridinas/farmacología , Antineoplásicos/química , Antineoplásicos/farmacocinética , Antineoplásicos Inmunológicos/farmacología , Antineoplásicos Inmunológicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Glicoconjugados/química , Humanos , Nanopartículas , Óxido Nítrico/metabolismo , Preparaciones de Plantas/farmacología , Preparaciones de Plantas/uso terapéutico , Polímeros/química
10.
Int J Mol Sci ; 20(18)2019 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-31527404

RESUMEN

Heat Shock Protein 90 (Hsp90) chaperone interacts with a broad range of client proteins involved in cancerogenesis and cancer progression. However, Hsp90 inhibitors were unsuccessful as anticancer agents due to their high toxicity, lack of selectivity against cancer cells and extrusion by membrane transporters responsible for multidrug resistance (MDR) such as P-glycoprotein (P-gp). Recognizing the potential of new compounds to inhibit P-gp function and/or expression is essential in the search for effective anticancer drugs. Eleven Hsp90 inhibitors containing an isoxazolonaphtoquinone core were synthesized and evaluated in two MDR models comprised of sensitive and corresponding resistant cancer cells with P-gp overexpression (human non-small cell lung carcinoma and colorectal adenocarcinoma). We investigated the effect of Hsp90 inhibitors on cell growth inhibition, P-gp activity and P-gp expression. Structure-activity relationship analysis was performed in respect to cell growth and P-gp inhibition. Compounds 5, 7, and 9 directly interacted with P-gp and inhibited its ATPase activity. Their potential P-gp binding site was identified by molecular docking studies. In addition, these compounds downregulated P-gp expression in MDR colorectal carcinoma cells, showed good relative selectivity towards cancer cells, while compound 5 reversed resistance to doxorubicin and paclitaxel in concentration-dependent manner. Therefore, compounds 5, 7 and 9 could be promising candidates for treating cancers with P-gp overexpression.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Antineoplásicos/farmacología , Resistencia a Múltiples Medicamentos/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/química , Antineoplásicos/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Perfilación de la Expresión Génica , Proteínas HSP90 de Choque Térmico/química , Proteínas HSP90 de Choque Térmico/genética , Humanos , Modelos Moleculares , Conformación Molecular , Estructura Molecular , Relación Estructura-Actividad
11.
Nat Prod Commun ; 12(2): 175-178, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30428204

RESUMEN

Silymarin, the active constituent of Silybum marianum (milk thistle), and its main component, silybin, are products with well-known hepatoprotective, cytoprotective, antioxidant, and chemopreventative properties. Despite substantial in vitro and in vivo investigations of these flavonolignans, their mechanisms of action and potential toxic effects are not fully defined. In this study we explored important ADME/Tox properties and biochemical interactions of selected flavonolignans using in silico methods. A quantitative structure-activity relationship (QSAR) model based on data from a parallel artificial membrane permeability assay (PAMPA) was used to estimate bioavailability after oral administration. Toxic effects and metabolic transformations were predicted using the knowledge-based expert systems Derek Nexus and Meteor Nexus (Lhasa Ltd). Potential estrogenic activity of the studied silybin congeners was outlined. To address further the stereospecificity of this effect the stereoisomeric forms of silybin were docked into the ligand-binding domain of the human estrogen receptor alpha (ERa) (MOE software, CCG). According to our results both stereoisomers can be accommodated into the ERa active site, but different poses and interactions were observed for silybin A and silybin B.


Asunto(s)
Silibina/farmacocinética , Humanos , Absorción Intestinal , Modelos Moleculares , Simulación del Acoplamiento Molecular , Silibina/química , Silibina/toxicidad
12.
Toxicology ; 392: 140-154, 2017 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-26836498

RESUMEN

The aim of this paper was to provide a proof of concept demonstrating that molecular modelling methodologies can be employed as a part of an integrated strategy to support toxicity prediction consistent with the mode of action/adverse outcome pathway (MoA/AOP) framework. To illustrate the role of molecular modelling in predictive toxicology, a case study was undertaken in which molecular modelling methodologies were employed to predict the activation of the peroxisome proliferator-activated nuclear receptor γ (PPARγ) as a potential molecular initiating event (MIE) for liver steatosis. A stepwise procedure combining different in silico approaches (virtual screening based on docking and pharmacophore filtering, and molecular field analysis) was developed to screen for PPARγ full agonists and to predict their transactivation activity (EC50). The performance metrics of the classification model to predict PPARγ full agonists were balanced accuracy=81%, sensitivity=85% and specificity=76%. The 3D QSAR model developed to predict EC50 of PPARγ full agonists had the following statistical parameters: q2cv=0.610, Nopt=7, SEPcv=0.505, r2pr=0.552. To support the linkage of PPARγ agonism predictions to prosteatotic potential, molecular modelling was combined with independently performed mechanistic mining of available in vivo toxicity data followed by ToxPrint chemotypes analysis. The approaches investigated demonstrated a potential to predict the MIE, to facilitate the process of MoA/AOP elaboration, to increase the scientific confidence in AOP, and to become a basis for 3D chemotype development.


Asunto(s)
Modelos Moleculares , PPAR gamma/metabolismo , Pruebas de Toxicidad/métodos , Animales , Sitios de Unión , Células COS , Línea Celular Tumoral , Chlorocebus aethiops , Cricetinae , Bases de Datos de Proteínas , Hígado Graso/metabolismo , Hígado Graso/patología , Estudios de Factibilidad , Células HEK293 , Haplorrinos , Células Hep G2 , Humanos , Ligandos , Simulación del Acoplamiento Molecular , Estructura Molecular , PPAR gamma/genética , Unión Proteica , Relación Estructura-Actividad Cuantitativa , Reproducibilidad de los Resultados , Medición de Riesgo , Sensibilidad y Especificidad
13.
Curr Top Med Chem ; 15(2): 85-104, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25547098

RESUMEN

For more than half a century free radical-induced alterations at cellular and organ levels have been investigated as a probable underlying mechanism of a number of adverse health conditions. Consequently, significant research efforts have been spent for discovering more effective and potent antioxidants / free radical scavengers for treatment of these adverse conditions. Being by far the most used antioxidants among natural and synthetic compounds, mono- and polyphenols have been the focus of both experimental and computational research on mechanisms of free radical scavenging. Quantum chemical studies have provided a significant amount of data on mechanisms of reactions between phenolic compounds and free radicals outlining a number of properties with a key role for the radical scavenging activity and capacity of phenolics. The obtained quantum chemical parameters together with other molecular descriptors have been used in quantitative structure-activity relationship (QSAR) analyses for the design of new more effective phenolic antioxidants and for identification of the most useful natural antioxidant phenolics. This review aims at presenting the state of the art in quantum chemical and QSAR studies of phenolic antioxidants and at analysing the trends observed in the field in the last decade.


Asunto(s)
Depuradores de Radicales Libres/química , Depuradores de Radicales Libres/farmacología , Fenoles/química , Fenoles/farmacología , Teoría Cuántica , Estructura Molecular , Relación Estructura-Actividad Cuantitativa
14.
Curr Top Med Chem ; 2014 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-25496275

RESUMEN

For more than half a century free radical-induced alterations at cellular and organ levels have been investigated as a probable underlying mechanism of a number of adverse health conditions. Consequently, significant research efforts have been spent for discovering more effective and potent antioxidants / free radical scavengers for treatment of these adverse conditions. Being by far the most used antioxidants among natural and synthetic compounds, mono- and polyphenols have been the focus of both experimental and computational research on mechanisms of free radical scavenging. Quantum chemical studies have provided a significant amount of data on mechanisms of reactions between phenolic compounds and free radicals outlining a number of properties with a key role for the radical scavenging activity and capacity of phenolics. The obtained quantum chemical parameters together with other molecular descriptors have been used in quantitative structure-activity relationship (QSAR) analyses for the design of new more effective phenolic antioxidants and for identification of the most useful natural antioxidant phenolics. This review aims at presenting the state of the art in quantum chemical and QSAR studies of phenolic antioxidants and at analysing the trends observed in the field in the last decade.

15.
Adv Protein Chem Struct Biol ; 85: 217-51, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21920325

RESUMEN

In the past two decades, there has been increasing concern about the potentially adverse effects of exogenous endocrine active substances (EAS) that alter the function of the endocrine system by interfering with hormone regulation. The mechanistic pathways by which EAS may elicit adverse effects, such as developmental and reproductive toxicity, often involve direct binding to nuclear hormone receptors. Certainly, the best studied nuclear receptor is the estrogen receptor (ER). Large-scale in vitro and in vivo methods have been developed to assess the estrogenic toxicity of chemicals. However, there are financial and animal welfare concerns related to their application. Quantitative structure-activity relationship (QSAR) approaches have proven their utility as a priority setting tool in the risk assessment of EAS. In addition, the models help to clarify the binding mode of the interacting substances. As estrogen-mediated effects are usually related to ligand-receptor interactions, and as there have been comprehensive structural studies on the ER, molecular modeling together with other in silico approaches provide a suitable means of studying these estrogenic effects. This chapter provides an overview of the molecular modeling approaches applied to ligand-ER interactions. The progress in the field is outlined, and some critical issues are analyzed based on recently published models where these approaches are applied.


Asunto(s)
Disruptores Endocrinos/toxicidad , Modelos Moleculares , Receptores de Estrógenos/metabolismo , Animales , Disruptores Endocrinos/química , Humanos , Ligandos , Conformación Proteica , Relación Estructura-Actividad Cuantitativa , Receptores de Estrógenos/química
16.
Environ Mol Mutagen ; 48(9): 754-71, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18008355

RESUMEN

Because of its environmental and industrial importance, the aromatic amines are the single chemical class most studied for its ability to induce mutations and cancer. The large database of mutagenicity and carcinogenicity results has been studied with Quantitative Structure-Activity Relationship (QSAR) approaches by several authors, leading to models for the following: (a) the mutagenic potency in Salmonella thyphimurium; (b) the carcinogenic potency in rodents; and (c) the discrimination between rodent carcinogens and noncarcinogens. However, satisfactory models for the discrimination between mutagens and nonmutagens are lacking. The present work provides new QSARs for mutagenic/nonmutagenic homocyclic aromatic amines in S. typhimurium strains TA98 and TA100. The two new models are validated by checking their ability to predict the mutagenicity of further aromatic amines not included in the training set, and not used to generate the QSAR models. In addition, we also validated previous QSAR models for the carcinogenicity/noncarcinogenicity of the aromatic amines with external data. The mechanistic implications of the models are discussed in light of the other QSARs for the aromatic amines. The results of the analysis point to two QSAR models (one for mutagenicity and one for rodent carcinogenicity) as reliable tools for the in silico characterization of the risk posed by the aromatic amines.


Asunto(s)
Aminas/toxicidad , Carcinógenos/toxicidad , Mutágenos/toxicidad , Modelos Químicos , Relación Estructura-Actividad Cuantitativa , Salmonella typhimurium/genética
17.
Pharm Res ; 22(5): 806-15, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15906177

RESUMEN

PURPOSE: To elucidate the differences in the interaction of chiralic ibuprofen (IBP) and naproxen (NAP) with poly(vinylpyrrolidone) (PVP) in a solid state. METHODS: Drugs/PVP physical mixtures and solid dispersions were characterized by scanning electron microscope (SEM), Fourier transform infrared spectrometry (FT-IR), solid state (13)C NMR spectroscopy, and x-ray diffractometry. Molecular modeling study of the crystal structures and PVP was performed. RESULTS: A spontaneous conversion of IBP/PVP physical mixtures in a stable glasslike form (solid dispersion) was observed after storage. The enantiomer reacted more strongly than the racemate. NAP did not interact with PVP. The crystal structures of drugs showed differences in the hydrogen bonding, aromatic interactions, molecular energies, and distances inside the crystals. The trimer structure of PVP was built and optimized. It was proposed that each PVP monomer could interact with one IBP dimmer in contrast to NAP, where two out of three PVP monomers faced the catemer carboxylic groups. CONCLUSIONS: The differences in the interaction of PVP with racemic IBP, enantiomer IBP, and NAP can be related to the differences in their crystal structures. The correlation between the experimental data and the results of the molecular modeling analysis suggest that the IBP dimmer structures are likely to perform HB and aromatic interactions with PVP.


Asunto(s)
Interacciones Farmacológicas , Ibuprofeno/farmacología , Modelos Moleculares , Naproxeno/farmacología , Povidona/farmacología , Rastreo Diferencial de Calorimetría/métodos , Química Farmacéutica/métodos , Imagen por Resonancia Magnética/métodos , Microscopía Electrónica de Rastreo/métodos , Espectroscopía Infrarroja por Transformada de Fourier/métodos , Estereoisomerismo , Tecnología Farmacéutica/métodos , Difracción de Rayos X/métodos
18.
Bioorg Med Chem ; 11(13): 2889-99, 2003 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-12788359

RESUMEN

A series of 25 phenothiazines and structurally related compounds was investigated by QSAR (quantitative structure activity relationship) and 3D-QSAR methods with respect to their MDR (multidrug resistance) reversing activity in P388/ADR- murine leukemia cell line resistant to ADR (adriamycin). The objective was to outline structural properties important for the investigated activity. Different measures for MDR reversal were used and compared. Two 3D-QSAR approaches were applied-CoMFA (comparative molecular field analysis) and CoMSIA (comparative molecular similarity indices analysis). Both, neutral and protonated forms of the compounds were investigated. Molecular models with good predictive power were derived using a hydrophobic field alone and a combination of steric, hydrophobic, and hydrogen bond acceptor fields of the compounds. In the combined models highest contribution of the hydrogen bond acceptor field was noticed. Thus, the dominant role of the hydrophobic and hydrogen bond acceptor fields for MDR reversing activity of the investigated compounds was demonstrated. The structural regions responsible for the differences in anti-MDR activity were analyzed in respect to their hydrophobic, hydrogen bond acceptor and steric nature. The results may direct design of new phenothiazines and related compounds as MDR modulators.


Asunto(s)
Resistencia a Múltiples Medicamentos/efectos de los fármacos , Fenotiazinas/química , Fenotiazinas/farmacología , Relación Estructura-Actividad Cuantitativa , Animales , Antibióticos Antineoplásicos/farmacología , Línea Celular Tumoral , Doxorrubicina/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Enlace de Hidrógeno , Interacciones Hidrofóbicas e Hidrofílicas , Ratones , Modelos Moleculares
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA