Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
BMC Pharmacol Toxicol ; 21(1): 79, 2020 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-33213519

RESUMEN

BACKGROUND: Although two novel synthesized compounds with tri-aryl structures; 3-(4-chlorophenyl)-5-(4-fluorophenyl)-4-phenyl-4,5-dihydro-1,2,4-oxadiazole (A) and 3,5-bis-(4-chlorophenyl)-4-phenyl-4,5-dihydro-1,2,4-oxadiazole (B) have been previously demonstrated to possess remarkable anti-breast cancer activity, their cardiotoxicity remains a major concern due to their mechanism of action. To address this concern, we assessed the ability of these compounds to cause toxicity towards H9c2 cardiomyocytes as an in vitro model of cardiotoxicity. METHODS: Cytotoxic activity of both compounds was explored in vitro on H9c2 cells using MTT assay. Annexin V/PI method, intracellular ROS determination and mitochondrial membrane potential assay were applied to elucidate the mechanism of action of the cell death. RESULTS: MTT assay revealed a concentration- and time-dependent cardiotoxicity. Findings of apoptosis by double staining with annexin V and propidium iodide divulged no cell death including apoptosis and necrosis at the concentration that were effective to inhibit cancer cells proliferation (10 µM) at 24 and 48 h. Furthermore, flow cytometric measurement of membrane potential and ROS determination using DCFH-DA verified the safe concentration of the compounds against H9c2 cells with no cardiotoxic effect. However, the higher concentration of the compounds could induce cell death through ROS-mediated mitochondrial dysfunction. CONCLUSIONS: Altogether, the results represented two novel chemical molecules possessing anti-breast cancer activity with minimum cardiac side effect.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/toxicidad , Cardiotoxinas/química , Cardiotoxinas/toxicidad , Animales , Línea Celular , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Relación Dosis-Respuesta a Droga , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Potencial de la Membrana Mitocondrial/fisiología , Ratas
2.
Toxins (Basel) ; 12(4)2020 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-32325789

RESUMEN

It is widely accepted that snake venom cardiotoxins (CTXs) target the plasma membranes of cells. In the present study, we investigated the role of Asp residues in the interaction of Naja atra cardiotoxin 1 (CTX1) and cardiotoxin 3 (CTX3) with phospholipid bilayers using chemical modification. CTX1 contains three Asp residues at positions 29, 40, and 57; CTX3 contains two Asp residues at positions 40 and 57. Compared to Asp29 and Asp40, Asp57 was sparingly modified with semi-carbazide, as revealed by matrix-assisted laser desorption ionization-time of flight (MALDI-TOF) mass and mass/mass analyses. Thus, semi-carbazide-modified CTX1 (SEM-CTX1) mainly contained modified Asp29 and Asp40, while SEM-CTX3 contained modified Asp40. Compared to that of native toxins, trifluoroethanol easily induced structural transition of SEM-CTX1 and SEM-CTX3, suggesting that the structural flexibility of CTXs was constrained by Asp40. Modification of Asp29 and Asp40 markedly promoted the ability of CTX1 to induce permeability of cell membranes and lipid vesicles; CTX3 and SEM-CTX3 showed similar membrane-damaging activity. Modification of Asp residues did not affect the membrane-binding capability of CTXs. Circular dichroism spectra of SEM-CTX3 and CTX3 were similar, while the gross conformation of SEM-CTX1 was distinct from that of CTX1. The interaction of CTX1 with membrane was distinctly changed by Asp modification. Collectively, our data suggest that Asp29 of CTX1 suppresses the optimization of membrane-bound conformation to a fully active state and that the function of Asp40 in the structural constraints of CTX1 and CTX3 is not important for the manifestation of membrane-perturbing activity.


Asunto(s)
Ácido Aspártico/química , Cardiotoxinas , Proteínas Cardiotóxicas de Elápidos , Membrana Dobles de Lípidos/metabolismo , Naja naja , Secuencia de Aminoácidos , Animales , Cardiotoxinas/química , Cardiotoxinas/farmacología , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Proteínas Cardiotóxicas de Elápidos/química , Proteínas Cardiotóxicas de Elápidos/farmacología , Humanos , Células K562 , Permeabilidad/efectos de los fármacos
3.
Circ Res ; 126(8): 947-964, 2020 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-32091972

RESUMEN

RATIONALE: Drug-induced proarrhythmia is so tightly associated with prolongation of the QT interval that QT prolongation is an accepted surrogate marker for arrhythmia. But QT interval is too sensitive a marker and not selective, resulting in many useful drugs eliminated in drug discovery. OBJECTIVE: To predict the impact of a drug from the drug chemistry on the cardiac rhythm. METHODS AND RESULTS: In a new linkage, we connected atomistic scale information to protein, cell, and tissue scales by predicting drug-binding affinities and rates from simulation of ion channel and drug structure interactions and then used these values to model drug effects on the hERG channel. Model components were integrated into predictive models at the cell and tissue scales to expose fundamental arrhythmia vulnerability mechanisms and complex interactions underlying emergent behaviors. Human clinical data were used for model framework validation and showed excellent agreement, demonstrating feasibility of a new approach for cardiotoxicity prediction. CONCLUSIONS: We present a multiscale model framework to predict electrotoxicity in the heart from the atom to the rhythm. Novel mechanistic insights emerged at all scales of the system, from the specific nature of proarrhythmic drug interaction with the hERG channel, to the fundamental cellular and tissue-level arrhythmia mechanisms. Applications of machine learning indicate necessary and sufficient parameters that predict arrhythmia vulnerability. We expect that the model framework may be expanded to make an impact in drug discovery, drug safety screening for a variety of compounds and targets, and in a variety of regulatory processes.


Asunto(s)
Antiarrítmicos/química , Arritmias Cardíacas/tratamiento farmacológico , Cardiotoxinas/química , Simulación por Computador , Descubrimiento de Drogas/métodos , Canal de Potasio ERG1/química , Antiarrítmicos/metabolismo , Antiarrítmicos/uso terapéutico , Arritmias Cardíacas/metabolismo , Cardiotoxicidad/metabolismo , Cardiotoxicidad/prevención & control , Cardiotoxinas/efectos adversos , Cardiotoxinas/metabolismo , Descubrimiento de Drogas/tendencias , Canal de Potasio ERG1/metabolismo , Femenino , Humanos , Síndrome de QT Prolongado/tratamiento farmacológico , Síndrome de QT Prolongado/metabolismo , Aprendizaje Automático , Masculino , Moxifloxacino/química , Moxifloxacino/metabolismo , Moxifloxacino/uso terapéutico , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/fisiología , Fenetilaminas/química , Fenetilaminas/metabolismo , Fenetilaminas/uso terapéutico , Estructura Secundaria de Proteína , Sulfonamidas/química , Sulfonamidas/metabolismo , Sulfonamidas/uso terapéutico , Inhibidores de Topoisomerasa II/química , Inhibidores de Topoisomerasa II/metabolismo , Inhibidores de Topoisomerasa II/uso terapéutico
4.
Bioorg Med Chem Lett ; 30(3): 126890, 2020 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-31870648

RESUMEN

Antibacterial activity of the three-finger toxins from cobra venom, including cytotoxin 3 from N. kaouthia, cardiotoxin-like basic polypeptide A5 from N. naja (CLBP), and alpha-neurotoxin from N. oxiana venom, was investigated. All toxins failed to influence Gram-negative bacteria. The most pronounced activity against Bacillus subtilis was demonstrated by CLBP. The latter is ascribed to the presence of additional Lys-residues within the membrane-binding motif of this toxin.


Asunto(s)
Antibacterianos/química , Venenos Elapídicos/metabolismo , Péptidos/química , Secuencia de Aminoácidos , Animales , Antibacterianos/aislamiento & purificación , Antibacterianos/farmacología , Bacillus subtilis/efectos de los fármacos , Cardiotoxinas/química , Elapidae/metabolismo , Escherichia coli/efectos de los fármacos , Pruebas de Sensibilidad Microbiana , Péptidos/aislamiento & purificación , Péptidos/farmacología , Estructura Terciaria de Proteína , Staphylococcus aureus/efectos de los fármacos
5.
Electrophoresis ; 40(23-24): 3108-3116, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31650569

RESUMEN

Accurate profiling of the lipophilicity of amphoteric compounds might be complex and laborious. In the present work the lipophilicity of 12 anthracycline antibiotics-four parent drugs: doxorubicin, daunorubicin, epidoxorubicin, and epidaunorubicin and eight novel formamidyne derivatives with attached morpholine, hexamethylenoimine or piperidine rings-was determined based on novel approach using MEEKC. In the second stage, lipophilicity was correlated with anthracycline toxicity towards two cell lines. In rat cardiomyoblast cell line (h9c2) a significant correlation between the logP and toxicity was found. The anthracycline lipophilicity was not correlated with toxicity towards the endothelial hybrid cell line (EAhy.926). In conclusion, the lipophilicity of anthracyclines seems to determine their toxicity towards cardiomyoblasts but not on endothelial cells, suggesting a different mechanism of anthracyclines intercellular transport or extrusion in cardiomyoblast and endothelial cells.


Asunto(s)
Antraciclinas , Antibacterianos , Cardiotoxinas , Cromatografía Capilar Electrocinética Micelar/métodos , Animales , Antraciclinas/análisis , Antraciclinas/química , Antraciclinas/toxicidad , Antibacterianos/análisis , Antibacterianos/química , Antibacterianos/toxicidad , Cardiotoxinas/análisis , Cardiotoxinas/química , Cardiotoxinas/toxicidad , Línea Celular , Supervivencia Celular/efectos de los fármacos , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Endotelio Vascular/citología , Interacciones Hidrofóbicas e Hidrofílicas , Ratas
6.
Int J Biol Macromol ; 136: 512-520, 2019 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-31199971

RESUMEN

Snake venom cardiotoxins (CTXs) present diverse pharmacological functions. Previous studies have reported that CTXs affect the activity of some serine proteases, namely, chymotrypsin, subtilisin, trypsin, and acetylcholinesterase. To elucidate the mode of action of CTXs, the interaction of CTXs with chymotrypsin was thus investigated. It was found that Naja atra CTX isotoxins concentration-dependently enhanced chymotrypsin activity. The capability of CTX1 and CTX5 in increasing chymotrypsin activity was higher than that of CTX2, CTX3, and CTX4. Removal of the molecular beacon-bound CTXs by chymotrypsin, circular dichroism measurement, and acrylamide quenching of Trp fluorescence indicated that CTXs bound to chymotrypsin. Chemical modification of Lys, Arg, or Met residues of CTX1 attenuated its capability to enhance chymotrypsin activity without impairing their bond with chymotrypsin. Catalytically inactive chymotrypsin retained the binding affinity for native and modified CTX1. CTX1 and chemically modified CTX1 differently altered the global conformation of chymotrypsin and inactivated chymotrypsin. Moreover, CTX1 did not reduce the interaction of 2-(p-toluidino)-naphthalene-6-sulfonate (TNS) with chymotrypsin and inactivated chymotrypsin. Together with previous results revealing that TNS can bind at the hydrophobic region of active site in chymotrypsin, our data suggest that CTXs can enhance chymotrypsin activity by binding to the region outside the enzyme's active site.


Asunto(s)
Cardiotoxinas/farmacología , Quimotripsina/metabolismo , Naja naja , Secuencia de Aminoácidos , Animales , Sitios de Unión , Cardiotoxinas/química , Cardiotoxinas/metabolismo , Quimotripsina/química , Simulación del Acoplamiento Molecular , Conformación Proteica/efectos de los fármacos
7.
Zebrafish ; 16(4): 379-387, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31145051

RESUMEN

Envenomation by the Venezuelan bushmaster snake (Lachesis muta muta) (Serpentes: Viperidae) is characterized by local and cardiac alterations. This study investigates the in vivo cardiac dysfunction, tissue destruction, and cellular processes triggered by Lachesis muta muta snake crude venom and a C-type lectin (CTL)-like toxin named Mutacytin-1 (MC-1). The 28 kDa MC-1 was obtained by molecular exclusion, ion exchange, and C-18 (checking pureness) reverse-phase chromatographies. N-terminal sequencing of the first eight amino acids (NNCPQ LLM) revealed 100% identity with Mutina (CTL-like) isolated from Lachesis stenophrys, which is a Ca2+-dependent-type galactoside-binding lectin from Bothrops jararaca and CTL BpLec from Bothrops pauloensis. The cardiotoxicity in zebrafish of MC-1 was evaluated by means of specific phenotypic expressions and larvae behavior at 5, 15, 30, 40 and 60 min post-treatment. The L. muta muta venom and MC-1 also produced heart rate/rhythm alterations, circulation modifications, and the presence of thrombus and apoptotic phenomenon with pericardial damages. Acridine orange (100 µg/mL) was used to visualize apoptosis cellular process in control and treated whole embryos. The cardiotoxic alterations happened in more than 90% of all larvae under the action of L. muta muta venom and MC-1. The findings have demonstrated the potential cardiotoxicity by L. muta muta venom, suggesting the possibility of cardiovascular damages to patients after bushmaster envenoming.


Asunto(s)
Cardiotoxicidad/embriología , Cardiotoxinas/farmacología , Crotalinae , Lectinas Tipo C , Proteínas de Reptiles/química , Venenos de Serpiente/química , Pez Cebra/embriología , Animales , Cardiotoxinas/química , Crotalinae/embriología , Embrión no Mamífero/efectos de los fármacos , Lectinas Tipo C/química , Proteínas de Reptiles/farmacología
8.
Protein Sci ; 28(5): 952-963, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30891862

RESUMEN

ß-Cardiotoxin is a novel member of the snake venom three-finger toxin (3FTX) family. This is the first exogenous protein to antagonize ß-adrenergic receptors and thereby causing reduction in heart rates (bradycardia) when administered into animals, unlike the conventional cardiotoxins as reported earlier. 3FTXs are stable all ß-sheet peptides with 60-80 amino acid residues. Here, we describe the three-dimensional crystal structure of ß-cardiotoxin together with the identification of a molten globule intermediate in the unfolding pathway of this protein. In spite of the overall structural similarity of this protein with conventional cardiotoxins, there are notable differences observed at the loop region and in the charge distribution on the surface, which are known to be critical for cytolytic activity of cardiotoxins. The molten globule intermediate state present in the thermal unfolding pathway of ß-cardiotoxin was however not observed during the chemical denaturation of the protein. Interestingly, circular dichroism (CD) and NMR studies revealed the presence of α-helical secondary structure in the molten globule intermediate. These results point to substantial conformational plasticity of ß-cardiotoxin, which might aid the protein in responding to the sometimes conflicting demands of structure, stability, and function during its biological lifetime.


Asunto(s)
Cardiotoxinas/química , Ophiophagus hannah/metabolismo , Venenos de Serpiente/metabolismo , Animales , Dicroismo Circular , Espectroscopía de Resonancia Magnética , Modelos Moleculares , Desnaturalización Proteica , Pliegue de Proteína , Estructura Secundaria de Proteína , Venenos de Serpiente/química
9.
Clin Pharmacol Ther ; 105(3): 614-624, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30460992

RESUMEN

Anthracycline-induced cardiotoxicity (ACT) is a severe adverse drug reaction for a subset of children treated with anthracyclines as part of chemotherapy protocols. The identification of genetic markers associated with increased ACT susceptibility has clinical significance toward improving patient care and our understanding of the molecular mechanisms involved in ACT. Human-induced pluripotent stem cell-derived cardiomyocytes represent a novel approach to determine the pharmacogenomics of ACT and guide the development of genetic screening tests.


Asunto(s)
Antraciclinas/efectos adversos , Antibióticos Antineoplásicos/efectos adversos , Cardiotoxinas/efectos adversos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Farmacogenética/tendencias , Antraciclinas/química , Antibióticos Antineoplásicos/química , Cardiotoxinas/química , Niño , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Miocitos Cardíacos/metabolismo
10.
Expert Rev Proteomics ; 15(11): 873-886, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30328726

RESUMEN

INTRODUCTION: Being important representatives of various proteomes, membrane-active cationic peptides (CPs) are attractive objects as lead compounds in the design of new antibacterial, anticancer, antifungal, and antiviral molecules. Numerous CPs are found in insect and snake venoms, where many of them reveal cytolytic properties. Due to advances in omics technologies, the number of such peptides is growing dramatically. Areas covered: To understand structure-function relationships for CPs in a living cell, detailed analysis of their hydrophobic/hydrophilic properties is indispensable. We consider two structural classes of membrane-active CPs: latarcins (Ltc) from spider and cardiotoxins (CTXs) from snake venoms. While the former are void off disulfide bonds and conformationally flexible, the latter are structurally rigid and cross-linked with disulfide bonds. In order to elucidate structure-activity relationships behind their antibacterial, anticancer, and hemolytic effects, the properties of these polypeptides are considered on a side-by-side basis. Expert commentary: An ever-increasing number of venom-derived membrane-active polypeptides require new methods for identification of their functional propensities and sequence-based design of novel pharmacological substances. We address these issues considering a number of the designed peptides, based either on Ltc or CTX sequences. Experimental and computer modeling techniques required for these purposes are delineated.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/farmacología , Cardiotoxinas/farmacología , Péptidos/química , Péptidos/farmacología , Animales , Antibacterianos/química , Antibacterianos/farmacología , Antifúngicos/química , Antifúngicos/farmacología , Péptidos Catiónicos Antimicrobianos/química , Antineoplásicos/química , Antineoplásicos/farmacología , Cardiotoxinas/química , Disulfuros/química , Diseño de Fármacos , Hemolíticos/química , Hemolíticos/farmacología , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Péptidos/metabolismo , Venenos de Araña/química , Relación Estructura-Actividad
11.
Sci Rep ; 8(1): 10160, 2018 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-29976997

RESUMEN

Pharmaceutical agents despite their efficacy to treat disease can cause additional unwanted cardiovascular side effects. Cardiotoxicity is characterized by changes in either the function and/or structure of the myocardium. Over recent years, functional cardiotoxicity has received much attention, however morphological damage to the myocardium and/or loss of viability still requires improved detection and mechanistic insights. A human 3D cardiac microtissue containing human induced pluripotent stem cell-derived cardiomyocytes (hiPS-CMs), cardiac endothelial cells and cardiac fibroblasts was used to assess their suitability to detect drug induced changes in cardiac structure. Histology and clinical pathology confirmed these cardiac microtissues were morphologically intact, lacked a necrotic/apoptotic core and contained all relevant cell constituents. High-throughput methods to assess mitochondrial membrane potential, endoplasmic reticulum integrity and cellular viability were developed and 15 FDA approved structural cardiotoxins and 14 FDA approved non-structural cardiotoxins were evaluated. We report that cardiac microtissues provide a high-throughput experimental model that is both able to detect changes in cardiac structure at clinically relevant concentrations and provide insights into the phenotypic mechanisms of this liability.


Asunto(s)
Imagenología Tridimensional , Miocardio/patología , Antineoplásicos/farmacología , Biomarcadores/metabolismo , Cardiotoxinas/química , Supervivencia Celular/efectos de los fármacos , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Supervivencia Tisular/efectos de los fármacos
12.
J Atheroscler Thromb ; 25(7): 557-565, 2018 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-29731506

RESUMEN

Patients with Stage A heart failure (HF) show no HF symptoms but have related comorbid diseases with a high risk of progressing to HF. Screening for comorbid diseases warrants closer attention because of the growing interest in addressing Stage A HF as the best means of preventing eventual progression to overt HF such as Stages C and D. The identification of individuals of Stage A HF is potentially useful for the implementation of HF-prevention strategies; however, not all Stage A HF patients develop left ventricular (LV) structural heart disease or symptomatic HF, which lead to advanced HF stages. Therefore, Stage A HF requires management with the long-term goal of avoiding HF development; likewise, Stage B HF patients are ideal targets for HF prevention. Although the early detection of subclinical LV dysfunction is, thus, essential for delaying the progression to HF, the assessment of subclinical LV dysfunction can be challenging. Global longitudinal strain (GLS) as assessed by speckle-tracking echocardiography has recently been reported to be a sensitive marker of early subtle LV myocardial abnormalities, helpful for the prediction of the outcomes for various cardiac diseases, and superior to conventional echocardiographic indices. GLS reflects LV longitudinal myocardial systolic function, and can be assessed usually by means of two-dimensional speckle-tracking. This article reviews the importance of the assessment of subclinical LV dysfunction in Stage A HF patients by means of GLS, and its current potential to prevent progression to later stage HF.


Asunto(s)
Cardiología/tendencias , Insuficiencia Cardíaca/terapia , Envejecimiento , Cardiotoxinas/química , Comorbilidad , Diagnóstico por Computador , Progresión de la Enfermedad , Ecocardiografía , Ventrículos Cardíacos/patología , Humanos , Hipercolesterolemia/patología , Miocardio/patología , Factores de Riesgo , Sístole
13.
PLoS One ; 13(4): e0195577, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29630634

RESUMEN

Vandetanib, a multi-kinase inhibitor used for the treatment of various cancers, has been reported to induce several adverse cardiac effects. However, the underlying mechanisms of vandetanib-induced cardiotoxicity are unclear. This study aimed to investigate the mechanism of vandetanib-induced cardiotoxicity using intracellular electrophysiological recordings on human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs), rabbit Purkinje fibers, and HEK293 cells transiently expressing human ether-a-go-go-related gene (hERG; the rapidly activating delayed rectifier K+ channel, IKr), KCNQ1/KCNE1 (the slowly activating delayed rectifier K+ current, IKs), KCNJ2 (the inwardly rectifying K+ current, IK1) or SCN5A (the inward Na+ current, INa). Purkinje fiber assays and ion channel studies showed that vandetanib at concentrations of 1 and 3 µM inhibited the hERG currents and prolonged the action potential duration. Alanine scanning and in silico hERG docking studies demonstrated that Y652 and F656 in the hERG S6 domain play critical roles in vandetanib binding. In hiPSC-CMs, vandetanib markedly reduced the maximum rate of depolarization during the AP upstroke. Ion channel studies revealed that hiPSC-CMs were more sensitive to inhibition of the INa by vandetanib than in a heterogeneously expressed HEK293 cell model, consistent with the changes in the AP parameters of hiPSC-CMs. The subclasses of Class I antiarrhythmic drugs inhibited INa currents in a dose-dependent manner in hiPSC-CMs and SCN5A-encoded HEK293 cells. The inhibitory potency of vandetanib for INa was much higher in hiPSC-CMs (IC50: 2.72 µM) than in HEK293 cells (IC50: 36.63 µM). These data suggest that AP and INa assays using hiPSC-CMs are useful electrophysiological models for prediction of drug-induced cardiotoxicity.


Asunto(s)
Cardiotoxicidad/fisiopatología , Cardiotoxinas/toxicidad , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/fisiología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/fisiología , Piperidinas/toxicidad , Ramos Subendocárdicos/efectos de los fármacos , Ramos Subendocárdicos/fisiopatología , Quinazolinas/toxicidad , Potenciales de Acción/efectos de los fármacos , Animales , Cardiotoxinas/química , Canal de Potasio ERG1/química , Canal de Potasio ERG1/genética , Canal de Potasio ERG1/metabolismo , Fenómenos Electrofisiológicos , Femenino , Células HEK293 , Humanos , Técnicas In Vitro , Células Madre Pluripotentes Inducidas/citología , Canal de Potasio KCNQ1/genética , Canal de Potasio KCNQ1/metabolismo , Modelos Moleculares , Miocitos Cardíacos/citología , Canal de Sodio Activado por Voltaje NAV1.5/genética , Canal de Sodio Activado por Voltaje NAV1.5/metabolismo , Técnicas de Placa-Clamp , Piperidinas/química , Canales de Potasio de Rectificación Interna/genética , Canales de Potasio de Rectificación Interna/metabolismo , Canales de Potasio con Entrada de Voltaje/genética , Canales de Potasio con Entrada de Voltaje/metabolismo , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/toxicidad , Quinazolinas/química , Conejos
14.
Molecules ; 23(2)2018 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-29463054

RESUMEN

This study presents two sensitive fluorescent assays for sensing heparin on the basis of the electrostatic interaction between heparin and Naja naja atra cardiotoxin 3 (CTX3). Owing to CTX3-induced folded structure of an adenosine-based molecular beacon (MB) or a DNA aptamer against CTX3, a reduction in the fluorescent signal of the aptamer or MB 5'-end labeled with carboxyfluorescein (FAM) and 3'-end labeled with 4-([4-(dimethylamino)phenyl]azo)-benzoic acid (DABCYL) was observed upon the addition of CTX3. The presence of heparin and formation of the CTX3-heparin complex caused CTX3 detachment from the MB or aptamer, and restoration of FAM fluorescence of the 5'-FAM-and-3'-DABCYL-labeled MB and aptamer was subsequently noted. Moreover, the detection of heparin with these CTX3-aptamer and CTX3-MB sensors showed high sensitivity and selectivity toward heparin over chondroitin sulfate and hyaluronic acid regardless of the presence of plasma. The limit of detection for heparin in plasma was determined to be 16 ng/mL and 15 ng/mL, respectively, at a signal-to-noise ratio of 3. This study validates the practical utility of the CTX3-aptamer and CTX3-MB systems for determining the concentration of heparin in a biological matrix.


Asunto(s)
Aptámeros de Nucleótidos/química , Técnicas Biosensibles , Cardiotoxinas/química , Heparina/aislamiento & purificación , Adenosina/química , Animales , Elapidae , Fluorescencia
15.
Artículo en Inglés | MEDLINE | ID: mdl-29382576

RESUMEN

Anuran toxins released from the skin glands are involved in defence against predators and microorganisms. Secretion from parotoid macroglands of bufonid toads is a rich source of bioactive compounds with the cytotoxic, cardiotoxic and hemolytic activity. Bufadienolides are considered the most toxic components of the toad poison, whereas the protein properties are largely unknown. In the present work, we analysed the cardio-, myo-, and neurotropic activity of extract and the selected proteins from Bufo bufo parotoids in in vitro physiological bioassays carried out on two standard model organisms: beetles and frogs. Our results demonstrate a strong cardioactivity of B. bufo gland extract. The toad poison stimulates (by 16%) the contractility of the insect heart and displays the cardioinhibitory effect on the frog heartbeat frequency (a 27% decrease), coupled with an irreversible cardiac arrest. The gland extract also exhibits significant myotropic properties (a 10% decrease in the muscle contraction force), whereas its neuroactivity remains low (a 4% decrease in the nerve conduction velocity). Among identified peptides present in the B. bufo parotoid extract are serine proteases, muscle creatine kinase, phospholipid hydroperoxide glutathione peroxidase, cytotoxic T-lymphocyte protein, etc. Some proteins contribute to the cardioinhibitory effect. Certain compounds display the paralytic (myo- and neurotropic) properties. As the toad gland extract exhibits a strong cardiotoxic activity, we conclude that the poison is a potent agent capable of slaying a predator. Our results also provide the guides for the use of toad poison-peptides in therapeutics and new drug development.


Asunto(s)
Proteínas Anfibias/toxicidad , Venenos de Anfibios/toxicidad , Bufo bufo/fisiología , Cardiotoxinas/toxicidad , Bloqueantes Neuromusculares/toxicidad , Neurotoxinas/toxicidad , Piel/metabolismo , Proteínas Anfibias/química , Proteínas Anfibias/aislamiento & purificación , Proteínas Anfibias/metabolismo , Venenos de Anfibios/química , Venenos de Anfibios/aislamiento & purificación , Venenos de Anfibios/metabolismo , Animales , Bufo bufo/crecimiento & desarrollo , Cardiotoxinas/química , Cardiotoxinas/aislamiento & purificación , Cardiotoxinas/metabolismo , Femenino , Jardines , Corazón/efectos de los fármacos , Corazón/fisiología , Frecuencia Cardíaca/efectos de los fármacos , Miembro Posterior , Técnicas In Vitro , Contracción Muscular/efectos de los fármacos , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/fisiología , Conducción Nerviosa/efectos de los fármacos , Bloqueantes Neuromusculares/química , Bloqueantes Neuromusculares/aislamiento & purificación , Bloqueantes Neuromusculares/metabolismo , Neurotoxinas/química , Neurotoxinas/aislamiento & purificación , Neurotoxinas/metabolismo , Parques Recreativos , Polonia , Proteómica/métodos , Ranidae , Nervio Ciático/efectos de los fármacos , Nervio Ciático/fisiología , Tenebrio
16.
J Chem Inf Model ; 57(11): 2799-2810, 2017 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-29053272

RESUMEN

Cardiotoxins (CTs) from snake venoms are a family of homologous highly basic proteins that have extended hydrophobic patterns on their molecular surfaces. CTs are folded into three ß-structured loops stabilized by four disulfide bridges. Being well-structured in aqueous solution, most of these proteins are membrane-active, although the exact molecular mechanisms of CT-induced cell damage are still poorly understood. To elucidate the structure-function relationships in CTs, a detailed knowledge of their spatial organization and local conformational dynamics is required. Protein domain motions can be either derived from a set of experimental structures or generated via molecular dynamics (MD). At the same time, traditional clustering algorithms in the Cartesian coordinate space often fail to properly take into account the local large-scale dihedral angle transitions that occur in MD simulations. This is because such perturbations are usually offset by changes in the neighboring dihedrals, thus preserving the overall protein fold. States with a "locally perturbed" backbone were found in experimental 3D models of some globular proteins and have been shown to be functionally meaningful. In this work, the possibility of large-scale dihedral angle transitions in the course of long-term MD in explicit water was explored for three CTs with different membrane activities: CT 1, 2 (Naja oxiana) and CT A3 (Naja atra). Analysis of the MD-derived distributions of backbone torsion angles revealed several important common and specific features in the structural/dynamic behavior of these proteins. First, large-amplitude transitions were detected in some residues located in the functionally important loop I region. The K5/L6 pair of residues was found to induce a perturbation of the hydrophobic patterns on the molecular surface of CTs-reversible breaking of a large nonpolar zone ("bottom") into two smaller ones and their subsequent association. Second, the characteristic sizes of these patterns perfectly coincided with the dimensions of the nonpolar zones on the surfaces of model two-component (zwitterionic/anionic) membranes. Taken together with experimental data on the CT-induced leakage of fluorescent dye from such membranes, these results allowed us to formulate a two-stage mechanism of CT-membrane binding. The principal finding of this study is that even local conformational dynamics of CTs can seriously affect their functional activity via a tuning of the membrane binding site - specific "hot spots" (like the K5/L6 pair) in the protein structure.


Asunto(s)
Cardiotoxinas/química , Cardiotoxinas/metabolismo , Simulación de Dinámica Molecular , Secuencia de Aminoácidos , Animales , Membrana Celular/metabolismo , Interacciones Hidrofóbicas e Hidrofílicas , Membrana Dobles de Lípidos/metabolismo , Naja naja , Conformación Proteica en Lámina beta
17.
Biomed Chromatogr ; 31(12)2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28544073

RESUMEN

The acute cardiotoxicity induced by Veratrum nigrum (VN) is explored by analyzing heart tissue metabolic profiles in mouse models and applying reversed-phase liquid chromatography mass spectrometry and hydrophilic interaction liquid chromatography mass spectrometry that are based on ultra-high-performance liquid chromatography quadrupole time-of-flight mass spectrometry. An animal model of acute heart injury was established in mice via intra-gastric administration of VN. Then, electrocardiogram and echocardiograph monitoring of cardiac function and pathological examination were performed on mice in both the control and VN groups, and it was verified that acute heart injury was caused. Meanwhile, comparing the results of the control and VN groups, we detected 36 differential endogenous metabolites of heart tissue, including taurine, riboflavin, purine and lipids, which are related to many possible pathways such as purine metabolism, taurine and hypotaurine metabolism and energy metabolism. Our study provides a scientific approach for evaluating and revealing the mechanisms of VN-induced cardiotoxicity via the metabolomic strategy.


Asunto(s)
Cardiotoxinas/toxicidad , Cromatografía Líquida de Alta Presión/métodos , Metaboloma/efectos de los fármacos , Extractos Vegetales/toxicidad , Veratrum/química , Animales , Cardiotoxicidad/metabolismo , Cardiotoxinas/química , Modelos Animales de Enfermedad , Corazón/efectos de los fármacos , Interacciones Hidrofóbicas e Hidrofílicas , Masculino , Espectrometría de Masas/métodos , Redes y Vías Metabólicas , Metabolómica/métodos , Ratones , Ratones Endogámicos C57BL , Miocardio/metabolismo , Miocardio/patología , Extractos Vegetales/química
18.
J Mol Model ; 22(10): 238, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27628673

RESUMEN

Cardiotoxins (CTXs) belonging to the three-finger toxin superfamily of snake venoms are one of principal toxic components and the protein toxins exhibit membrane lytic activities when the venoms are injected into victims. In the present study, complex formations between CTX VI (a P-type CTX from Naja atra) and CTX1 (an S-type CTX from Naja naja) on zwitterionic POPC bilayers (a major lipid component of cell membranes) have been studied in near physiological conditions for a total dynamic time scale of 1.35 µs using all-atom molecular dynamics (MD) simulations. Comprehensive analyses of the MD data revealed that residues such as Leu1, Lys2, Tyr11, Lys31, Asp57 and Arg58 of CTX VI, and Ala16, Lys30 and Arg58 of CTX1 were crucial for establishing interactions with the POPC bilayer. Moreover, loop I, along with globular head and loop II of CTX VI, and loop II of CTX1 were found to be the structural regions chiefly governing complex formation of the respective proteins with POPC. Rationalizations for the differential binding modes of CTXs and implications of the findings for designing small molecular inhibitors to the toxins are also discussed. Graphical Abstract Binding modes of a P-type CTX and an S-type CTX towards the POPC bilayer.


Asunto(s)
Cardiotoxinas/química , Membrana Celular/química , Venenos de Serpiente/química , Animales , Membrana Dobles de Lípidos/química , Simulación de Dinámica Molecular , Naja naja/metabolismo , Conformación Proteica
19.
Chem Res Toxicol ; 29(6): 981-90, 2016 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-27104637

RESUMEN

Yessotoxin (YTX) is a marine phycotoxin produced by dinoflagellates and accumulated in filter feeding shellfish. Although no human intoxication episodes have been reported, YTX content in shellfish is regulated by many food safety authorities due to their worldwide distribution. YTXs have been related to ultrastructural heart damage in vivo, but the functional consequences in the long term have not been evaluated. In this study, we explored the accumulative cardiotoxic potential of YTX in vitro and in vivo. Preliminary in vitro evaluation of cardiotoxicity was based on the effect on hERG (human ether-a-go-go related gene) channel trafficking. In vivo experiments were performed in rats that received repeated administrations of YTX followed by recordings of electrocardiograms, arterial blood pressure, plasmatic cardiac biomarkers, and analysis of myocardium structure and ultrastructure. Our results showed that an exposure to 100 nM YTX for 12 or 24 h caused an increase of extracellular surface hERG channels. Furthermore, remarkable bradycardia and hypotension, structural heart alterations, and increased plasma levels of tissue inhibitor of metalloproteinases-1 were observed in rats after four intraperitoneal injections of YTX at doses of 50 or 70 µg/kg that were administered every 4 days along a period of 15 days. Therefore, and for the first time, YTX-induced subacute cardiotoxicity is supported by evidence of cardiovascular function alterations related to its repeated administration. Considering international criteria for marine toxin risk estimation and that the regulatory limit for YTX has been recently raised in many countries, YTX cardiotoxicity might pose a health risk to humans and especially to people with previous cardiovascular risk.


Asunto(s)
Cardiotoxinas/toxicidad , Enfermedades Cardiovasculares/metabolismo , Corazón/efectos de los fármacos , Oxocinas/toxicidad , Animales , Células CHO , Cardiotoxicidad , Cardiotoxinas/administración & dosificación , Cardiotoxinas/química , Células Cultivadas , Cricetulus , Canal de Potasio ERG1/metabolismo , Humanos , Inyecciones Intraperitoneales , Conformación Molecular , Venenos de Moluscos , Oxocinas/administración & dosificación , Oxocinas/química , Ratas , Ratas Sprague-Dawley
20.
PLoS One ; 11(1): e0147049, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26751696

RESUMEN

Glycosylated α-dystroglycan provides an essential link between extracellular matrix proteins, like laminin, and the cellular cytoskeleton via the dystrophin-glycoprotein complex. In secondary dystroglycanopathy muscular dystrophy, glycosylation abnormalities disrupt a complex O-mannose glycan necessary for muscle structural integrity and signaling. Fktn-deficient dystroglycanopathy mice develop moderate to severe muscular dystrophy with skeletal muscle developmental and/or regeneration defects. To gain insight into the role of glycosylated α-dystroglycan in these processes, we performed muscle fiber typing in young (2, 4 and 8 week old) and regenerated muscle. In mice with Fktn disruption during skeletal muscle specification (Myf5/Fktn KO), newly regenerated fibers (embryonic myosin heavy chain positive) peaked at 4 weeks old, while total regenerated fibers (centrally nucleated) were highest at 8 weeks old in tibialis anterior (TA) and iliopsoas, indicating peak degeneration/regeneration activity around 4 weeks of age. In contrast, mature fiber type specification at 2, 4 and 8 weeks old was relatively unchanged. Fourteen days after necrotic toxin-induced injury, there was a divergence in muscle fiber types between Myf5/Fktn KO (skeletal-muscle specific) and whole animal knockout induced with tamoxifen post-development (Tam/Fktn KO) despite equivalent time after gene deletion. Notably, Tam/Fktn KO retained higher levels of embryonic myosin heavy chain expression after injury, suggesting a delay or abnormality in differentiation programs. In mature fiber type specification post-injury, there were significant interactions between genotype and toxin parameters for type 1, 2a, and 2x fibers, and a difference between Myf5/Fktn and Tam/Fktn study groups in type 2b fibers. These data suggest that functionally glycosylated α-dystroglycan has a unique role in muscle regeneration and may influence fiber type specification post-injury.


Asunto(s)
Distroglicanos/genética , Fibras Musculares Esqueléticas/fisiología , Distrofia Muscular Animal/genética , Proteínas/genética , Regeneración , Animales , Cardiotoxinas/química , Modelos Animales de Enfermedad , Distroglicanos/metabolismo , Exones , Eliminación de Gen , Genotipo , Glicosilación , Laminina/metabolismo , Ratones , Ratones Noqueados , Músculo Esquelético/fisiología , Distrofia Muscular Animal/metabolismo , Necrosis , Transducción de Señal , Transferasas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA