Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
1.
Chem Biol Interact ; 347: 109618, 2021 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-34364836

RESUMEN

The toxicity of hybrid nanoparticles, consisting of non-toxic components, zirconium dioxide nanoparticles (ZrO2 NPs), and caffeic acid (CA), was examined against four different cell lines (HTR-8 SV/Neo, JEG-3, JAR, and HeLa). Stable aqueous ZrO2 sol, synthesized by forced hydrolysis, consists of 3-4 nm in size primary particles organized in 30-60 nm in size snowflake-like particles, as determined by transmission electron microscopy and direct light scattering measurements. The surface modification of ZrO2 NPs with CA leads to the formation of an interfacial charge transfer (ICT) complex followed by the appearance of absorption in the visible spectral range. The spectroscopic observations are complemented with the density functional theory calculations using a cluster model. The ZrO2 NPs and CA are non-toxic against four different cell lines in investigated concentration range. Also, ZrO2 NPs promote the proliferation of HTR-8 SV/Neo, JAR, and HeLa cells. On the other hand, hybrid ZrO2/CA NPs induced a significant reduction of the viability of the JEG-3 cells (39 %) for the high concentration of components (1.6 mM ZrO2 and 0.4 mM CA).


Asunto(s)
Ácidos Cafeicos/toxicidad , Nanopartículas del Metal/toxicidad , Placenta/efectos de los fármacos , Circonio/toxicidad , Ácidos Cafeicos/química , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Teoría Funcional de la Densidad , Femenino , Humanos , Nanopartículas del Metal/química , Modelos Químicos , Tamaño de la Partícula , Placenta/patología , Embarazo , Pruebas de Toxicidad , Circonio/química
2.
Regul Toxicol Pharmacol ; 124: 104974, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34139276

RESUMEN

Sida rhombifolia (Malvaceae) is popularly used as a treatment for several pathological conditions; however, there is a lack of studies that identify its compounds and that evaluate comprehensively the safety of its consumption. Therefore, the aim of this study was to determinate the phytochemical constitution of the crude extract of Sida rhombifolia (CESR), and its safety in models of acute and repeated doses (28 days) toxicity. The tested dose for the model of acute toxicity was 2000 mg/kg doses for the repeated dose model were 150, 300 e 600 mg/kg. Hematological, biochemical, histopathological and oxidative markers were investigated. HPLC-DAD-MS analysis evidenced the presence of caffeic acid, coumarin, and rutin. In the acute toxicity model the only altered parameters were tissue ROS, and AST and BUN in serum. As for the repeated dose experiment both hematological and biochemical markers remained within the values of reference for the species. Obtained results demonstrate that the CESR did not present significant toxic effects when administrated orally to male and female rats in acute and repeated doses.


Asunto(s)
Malvaceae/química , Extractos Vegetales/toxicidad , Administración Oral , Animales , Ácidos Cafeicos/análisis , Ácidos Cafeicos/toxicidad , Cumarinas/análisis , Cumarinas/toxicidad , Femenino , Masculino , Componentes Aéreos de las Plantas/química , Extractos Vegetales/administración & dosificación , Extractos Vegetales/química , Ratas , Rutina/análisis , Rutina/toxicidad , Pruebas de Toxicidad Aguda , Pruebas de Toxicidad Subaguda
3.
ACS Appl Mater Interfaces ; 13(9): 10705-10718, 2021 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-33635046

RESUMEN

The validation of metal-phenolic nanoparticles (MPNs) in preclinical imaging studies represents a growing field of interest due to their versatility in forming predesigned structures with unique properties. Before MPNs can be used in medicine, their pharmacokinetics must be optimized so that accumulation in nontargeted organs is prevented and toxicity is minimized. Here, we report the fabrication of MPNs made of a coordination polymer core that combines In(III), Cu(II), and a mixture of the imidazole 1,4-bis(imidazole-1-ylmethyl)-benzene and the catechol 3,4-dihydroxycinnamic acid ligands. Furthermore, a phenolic-based coating was used as an anchoring platform to attach poly(ethylene glycol) (PEG). The resulting MPNs, with effective hydrodynamic diameters of around 120 nm, could be further derivatized with surface-embedded molecules, such as folic acid, to facilitate in vivo targeting and multifunctionality. The prepared MPNs were evaluated for in vitro plasma stability, cytotoxicity, and cell internalization and found to be biocompatible under physiological conditions. First, biomedical evaluations were then performed by intrinsically incorporating trace amounts of the radioactive metals 111In or 64Cu during the MPN synthesis directly into their polymeric matrix. The resulting particles, which had identical physicochemical properties to their nonradioactive counterparts, were used to perform in vivo single-photon emission computed tomography (SPECT) and positron emission tomography (PET) in tumor-bearing mice. The ability to incorporate multiple metals and radiometals into MPNs illustrates the diverse range of functional nanoparticles that can be prepared with this approach and broadens the scope of these nanoconstructs as multimodal preclinical imaging agents.


Asunto(s)
Ácidos Cafeicos/química , Nanopartículas del Metal/química , Neoplasias/diagnóstico por imagen , Radiofármacos/química , Animales , Ácidos Cafeicos/farmacocinética , Ácidos Cafeicos/toxicidad , Línea Celular Tumoral , Radioisótopos de Cobre/química , Radioisótopos de Cobre/farmacocinética , Radioisótopos de Cobre/toxicidad , Femenino , Humanos , Imidazoles/química , Imidazoles/farmacocinética , Imidazoles/toxicidad , Radioisótopos de Indio/química , Radioisótopos de Indio/farmacocinética , Radioisótopos de Indio/toxicidad , Ligandos , Nanopartículas del Metal/toxicidad , Ratones Endogámicos BALB C , Imagen Multimodal , Tomografía de Emisión de Positrones , Prueba de Estudio Conceptual , Radiofármacos/farmacocinética , Radiofármacos/toxicidad , Tomografía Computarizada de Emisión de Fotón Único , Tomografía Computarizada por Rayos X
4.
Pharm Dev Technol ; 25(10): 1271-1280, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32892659

RESUMEN

Caffeic acid phenethyl ester (CAPE), a major pharmacologically active component of poplar type propolis, is known for its proapoptotic, anti-inflammatory, antioxidant, antiviral, and enzyme inhibiting activities. The aim of this study was to perform an in vitro and in vivo safety assessment of a micellar system based on a newly synthesized copolymer, consisting of polyglycidol and poly(allyl glycidyl ether) (C12-PAGE-PG) as a drug delivery platform for CAPE. The in vitro studies on HepG2 and L929 cells by MTT and LDH assays after treatment with the empty and CAPE-loaded micelles showed no cytotoxic effects of the empty micelles and retained cytotoxic activity of CAPE loaded in the micelles. No hemolysis or stimulation of mouse lymphocytes or macrophages was observed in vitro. In vivo hematological, biochemical, and histological assays on rats, treated with the empty (2580 and 5160 µg/kg) or CAPE-loaded (375 and 750 µg CAPE/kg) micelles did not reveal pathological changes of any of the parameters assayed after 14-days' treatment. In conclusion, initial toxicological data characterize C12-PAGE-PG as a non-toxic and promising copolymer for development of micellar drug delivery systems, particularly for a hydrophobic active substance as CAPE.


Asunto(s)
Ácidos Cafeicos/administración & dosificación , Sistemas de Liberación de Medicamentos , Alcohol Feniletílico/análogos & derivados , Polímeros/química , Animales , Ácidos Cafeicos/toxicidad , Línea Celular , Compuestos Epoxi/química , Células Hep G2 , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Masculino , Ratones , Micelas , Alcohol Feniletílico/administración & dosificación , Alcohol Feniletílico/toxicidad , Glicoles de Propileno/química , Ratas , Ratas Wistar
5.
Eur J Pharmacol ; 887: 173464, 2020 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-32781171

RESUMEN

DNA methylation represents an important epigenetic regulation of the genome. Earlier studies have suggested that dietary phenolic compounds including those contained in coffee, tea and soy products may modulate the level of DNA methylation. In this study, we first characterize the effect of caffeic acid phenethyl ester (CAPE) and other dietary phenolic compounds on DNA methylation in vitro. The IC50 values of CAPE, daidzein, isorhamnetin and genistein are 7.6, 6.9, 6.2, and 4.3 µM, respectively, in an in-vitro enzymatic assay system. Computational analysis indicates that CAPE, daidzein, isorhamnetin and genistein can bind inside the DNA substrate-binding site in human DNMT1 with a favorable binding energy. In an animal study, we find that maternal CAPE treatment shifts the coat color distribution of the 21-day-old Avy/a offspring towards the yellow phenotype, indicating that CAPE inhibits the methylation of the agouti gene promoter sequence in vivo. The results from this study may shed light on the potential epigenetic effect in the offspring resulting from maternal intake of certain coffee phenolics during pregnancy.


Asunto(s)
Ácidos Cafeicos/farmacología , Café , Metilación de ADN/efectos de los fármacos , Epigénesis Genética/efectos de los fármacos , Simulación del Acoplamiento Molecular/métodos , Alcohol Feniletílico/análogos & derivados , Polifenoles/farmacología , Animales , Ácidos Cafeicos/química , Ácidos Cafeicos/toxicidad , Café/efectos adversos , Metilación de ADN/fisiología , Relación Dosis-Respuesta a Droga , Epigénesis Genética/fisiología , Femenino , Células HT29 , Humanos , Masculino , Ratones , Ratones Transgénicos , Alcohol Feniletílico/química , Alcohol Feniletílico/farmacología , Alcohol Feniletílico/toxicidad , Polifenoles/química , Polifenoles/toxicidad , Embarazo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/patología , Estructura Secundaria de Proteína , Salmón
6.
Eur J Med Chem ; 177: 259-268, 2019 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-31158743

RESUMEN

Unexpected inhibitory effects against eeAChE could be found for a newly synthesized class of caffeic acid phenethyl ester (CAPE) derivatives. Thus, phenethyl-(E)-3-(3,5-dimethoxy-4-phenethoxyphenyl)-acrylate (Ki = 1.97 ±â€¯0.38 µM, Ki´â€¯= 2.44 ±â€¯0.07 µM) and 4-(2-(((E)-3-(3,4-bis(benzyloxy)phenyl)acryloyl)oxy)ethyl)-1,2-phenylene (2E,2'E)-bis(3-(3,4-bis(benzyloxy)phenyl)acrylate) (Ki = 0.72 ±â€¯0.31 µM, Ki´â€¯= 1.80 ±â€¯0.21 µM) showed very good inhibition of eeAChE, while being non cytotoxic for malignant human cancer cells and non-malignant mouse fibroblasts. Also, they are weak inhibitors for BChE (from equine serum).


Asunto(s)
Ácidos Cafeicos/farmacología , Inhibidores de la Colinesterasa/farmacología , Alcohol Feniletílico/análogos & derivados , Alcohol Feniletílico/farmacología , Acetilcolinesterasa/química , Acetilcolinesterasa/metabolismo , Animales , Butirilcolinesterasa/metabolismo , Ácidos Cafeicos/síntesis química , Ácidos Cafeicos/química , Ácidos Cafeicos/toxicidad , Dominio Catalítico , Línea Celular Tumoral , Inhibidores de la Colinesterasa/síntesis química , Inhibidores de la Colinesterasa/química , Inhibidores de la Colinesterasa/toxicidad , Electrophorus , Caballos , Humanos , Ratones , Simulación del Acoplamiento Molecular , Células 3T3 NIH , Alcohol Feniletílico/síntesis química , Alcohol Feniletílico/toxicidad
7.
Biochem Biophys Res Commun ; 505(2): 612-617, 2018 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-30278886

RESUMEN

Caffeic acid is a natural phytochemical structurally similar to other cinnamic acids. In this study we found caffeic acid (CA) but not ferulic, sinapic or cinnamic acids inhibited proliferation of hepatocellular carcinoma cells (HCC) and reduced cell numbers by inducing apoptosis. Only transient exposure to CA was required for these lethal effects that are associated with disruption of mitochondrial membrane potential and induction of reactive oxygen species. By comparison, primary hepatocytes resisted CA toxicity for nearly 48 h, consistent with selective sensitivity of HCC to CA. These results support use of CA as an anti-tumor agent to inhibit HCC, especially if delivered by locoregional catheterization in an embolization procedure.


Asunto(s)
Antineoplásicos/farmacología , Ácidos Cafeicos/farmacología , Neoplasias Hepáticas Experimentales/tratamiento farmacológico , Animales , Antineoplásicos/uso terapéutico , Antineoplásicos/toxicidad , Ácidos Cafeicos/uso terapéutico , Ácidos Cafeicos/toxicidad , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Hepatocitos/citología , Hepatocitos/efectos de los fármacos , Neoplasias Hepáticas Experimentales/patología , Marmota , Potencial de la Membrana Mitocondrial/efectos de los fármacos
8.
Int J Mol Sci ; 19(3)2018 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-29510500

RESUMEN

Chlorogenic acid (CGA) decreases colon cancer-cell proliferation but the combined anti-cancer effects of CGA with its major colonic microbial metabolites, caffeic acid (CA), 3-phenylpropionic acid (3-PPA) and benzoic acid (BA), needs elucidation as they occur together in colonic digesta. Caco-2 cancer cells were treated for 24 h with the four compounds individually (50-1000 µM) and as an equimolar ratio (1:1:1:1; MIX). The effective concentration to decrease cell proliferation by 50% (EC50) was lower for MIX (431 ± 51.84 µM) and CA (460 ± 21.88) versus CGA (758 ± 19.09 µM). The EC50 for cytotoxicity measured by lactate dehydrogenase release in MIX (527 ± 75.34 µM) showed more potency than CA (740 ± 38.68 µM). Cell proliferation was decreased by 3-PPA and BA at 1000 µM with no cytotoxicity. Cell-cycle arrest was induced at the S-phase by CA (100 µM), MIX (100 µM), CGA (250 µM) and 3-PPA (500 µM) with activation of caspase-3 by CGA, CA, MIX (500 and 1000 µM). Mitochondrial DNA content was reduced by 3-PPA (1000 µM). The anti-cancer effects occurred at markedly lower concentrations of each compound within MIX than when provided singly, indicating that they function together to enhance anti-colon cancer activities.


Asunto(s)
Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Ácido Clorogénico/farmacología , Puntos de Control de la Fase S del Ciclo Celular/efectos de los fármacos , Ácido Benzoico/farmacología , Ácido Benzoico/toxicidad , Células CACO-2 , Ácidos Cafeicos/farmacología , Ácidos Cafeicos/toxicidad , Ácido Clorogénico/toxicidad , Humanos , Fenilpropionatos/farmacología , Fenilpropionatos/toxicidad
9.
J Med Chem ; 60(16): 7084-7098, 2017 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-28745898

RESUMEN

Targeting mitochondrial oxidative stress is an effective therapeutic strategy. In this context, a rational design of mitochondriotropic antioxidants (compounds 22-27) based on a dietary antioxidant (caffeic acid) was performed. Jointly named as AntiOxCINs, these molecules take advantage of the known ability of the triphenylphosphonium cation to target active molecules to mitochondria. The study was guided by structure-activity-toxicity-property relationships, and we demonstrate in this work that the novel AntiOxCINs act as mitochondriotropic antioxidants. In general, AntiOxCINs derivatives prevented lipid peroxidation and acted as inhibitors of the mitochondrial permeability transition pore. AntiOxCINs toxicity profile was found to be dependent on the structural modifications performed on the dietary antioxidant. On the basis of mitochondrial and cytotoxicity/antioxidant cellular data, compound 25 emerged as a potential candidate for the development of a drug candidate with therapeutic application in mitochondrial oxidative stress-related diseases. Compound 25 increased GSH intracellular levels and showed no toxicity on mitochondrial morphology and function.


Asunto(s)
Antioxidantes/farmacología , Ácidos Cafeicos/farmacología , Mitocondrias/metabolismo , Estrés Oxidativo/efectos de los fármacos , Animales , Antioxidantes/síntesis química , Antioxidantes/toxicidad , Ácidos Cafeicos/síntesis química , Ácidos Cafeicos/toxicidad , Ácido Glutámico/metabolismo , Glutatión/metabolismo , Células Hep G2 , Humanos , Hierro/química , Quelantes del Hierro/síntesis química , Quelantes del Hierro/farmacología , Quelantes del Hierro/toxicidad , Peroxidación de Lípido/efectos de los fármacos , Malatos/metabolismo , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Microsomas Hepáticos/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/antagonistas & inhibidores , Poro de Transición de la Permeabilidad Mitocondrial , Ratas , Ácido Succínico/metabolismo
10.
Toxicol Appl Pharmacol ; 324: 26-35, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28363435

RESUMEN

Glucocorticoid-induced osteoporosis (GIO) is one of the most common causes of secondary osteoporosis. Given that glucocorticoids are considered as a main component of the treatment protocols for a variety of inflammation and immune-mediated diseases besides its use as adjuvant to several chemotherapeutic agents, it is crucial to find ways to overcome this critical adverse effect. Caffeic acid phenethyl ester (CAPE), which is a natural compound derived from honeybee propolis displayed promising antiosteoporotic effects against mechanical bone injury in various studies. The current work aimed at investigating the potential protective effect of CAPE against GIO in vivo with emphasis on the modulation of oxidative status and receptor activator of NF-kB ligand (RANKL)/osteoprotegrin (OPG) signaling. The results showed that CAPE opposed dexamethasone (DEX)-mediated alterations in bone histology and tartarate-resistant acid phosphatase (TRAP) activity. In addition, CAPE restored oxidative balance, Runt-related transcription factor 2 (RunX2) expression and reduced caspase-3 activity in femur tissues. Co-administration of CAPE with DEX normalized RANKL/OPG ratio and Akt activation indicating a reduction in DEX-osteoclastogenesis. In conclusion, concurrent treatment of CAPE with DEX exhibited promising effects in the protection against DEX-induced osteoporosis through opposing osteoclastogenesis and protecting osteoblasts. The potent antioxidant activity of CAPE is, at least in part, involved in its anti-apoptotic effects and modulation of RunX2 and RANKL/OPG signals. The use of CAPE-enriched propolis formulas is strongly recommended for patients on chronic glucocorticoid therapy to help in the attenuation of GIO.


Asunto(s)
Ácidos Cafeicos/toxicidad , Glucocorticoides/toxicidad , Osteoporosis/metabolismo , Osteoprotegerina/metabolismo , Estrés Oxidativo/efectos de los fármacos , Alcohol Feniletílico/análogos & derivados , Ligando RANK/metabolismo , Animales , Masculino , Osteoporosis/inducido químicamente , Osteoporosis/prevención & control , Estrés Oxidativo/fisiología , Alcohol Feniletílico/toxicidad , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley
11.
Chem Biol Drug Des ; 89(5): 655-662, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-27863053

RESUMEN

Caffeic acid phenethyl ester (CAPE) is the bioactive constituent of propolis from honeybee hives and is well known for its anti-inflammatory, anticarcinogenic, antioxidant, and immunomodulatory properties. Herein, we revisited the cellular mechanism underlying the diverse biological effects of CAPE. We demonstrated that XPO1/CRM1, a major nuclear export receptor, is a cellular target of CAPE. Through nuclear export functional assay, we observed a clear shift of XPO1 cargo proteins from a cytoplasmic localization to nucleus when treated with CAPE. In particular, we showed that CAPE could specifically target the non-catalytic and conserved Cys528 of XPO1 through the means of mass spectrometric analysis. In addition, we demonstrated that the mutation of Cys528 residue in XPO1 could rescue the nuclear export defects caused by CAPE. Furthermore, we performed position-restraint molecular dynamics simulation to show that the Michael acceptor moiety of CAPE is the warhead to enable covalent binding with Cys528 residue of XPO1. The covalent modulation of nuclear export by CAPE may explain its diverse biological effects. Our findings may have general implications for further investigation of CAPE and its structural analogs.


Asunto(s)
Ácidos Cafeicos/química , Carioferinas/metabolismo , Alcohol Feniletílico/análogos & derivados , Receptores Citoplasmáticos y Nucleares/metabolismo , Sitios de Unión , Ácidos Cafeicos/toxicidad , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Células HeLa , Humanos , Carioferinas/antagonistas & inhibidores , Carioferinas/genética , Células MCF-7 , Espectrometría de Masas , Microscopía Confocal , Simulación de Dinámica Molecular , Péptidos/análisis , Alcohol Feniletílico/química , Alcohol Feniletílico/toxicidad , Estructura Terciaria de Proteína , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores , Receptores Citoplasmáticos y Nucleares/genética , Relación Estructura-Actividad , Proteína Exportina 1
12.
Toxins (Basel) ; 8(7)2016 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-27399778

RESUMEN

Epidemiological studies have found a positive association between coffee consumption and a lower risk of cardiovascular disorders, some cancers, diabetes, Parkinson and Alzheimer disease. Coffee consumption, however, has also been linked to an increased risk of developing some types of cancer, including bladder cancer in adults and leukemia in children of mothers who drink coffee during pregnancy. Since cancer is driven by the accumulation of DNA alterations, the ability of the coffee constituent caffeic acid to induce DNA damage in cells may play a role in the carcinogenic potential of this beverage. This carcinogenic potential may be exacerbated in cells with DNA repair defects. People with the genetic disease Fanconi Anemia have DNA repair deficiencies and are predisposed to several cancers, particularly acute myeloid leukemia. Defects in the DNA repair protein Fanconi Anemia D2 (FANCD2) also play an important role in the development of a variety of cancers (e.g., bladder cancer) in people without this genetic disease. This communication shows that cells deficient in FANCD2 are hypersensitive to the cytotoxicity (clonogenic assay) and DNA damage (γ-H2AX and 53BP1 focus assay) induced by caffeic acid and by a commercial lyophilized coffee extract. These data suggest that people with Fanconi Anemia, or healthy people who develop sporadic mutations in FANCD2, may be hypersensitive to the carcinogenic activity of coffee.


Asunto(s)
Ácidos Cafeicos/toxicidad , Café/toxicidad , Daño del ADN , Reparación del ADN , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/deficiencia , Anemia de Fanconi/patología , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Anemia de Fanconi/genética , Anemia de Fanconi/metabolismo , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/genética , Histonas/metabolismo , Humanos , Proteína 1 de Unión al Supresor Tumoral P53/metabolismo
13.
Acta Pharmacol Sin ; 36(5): 627-43, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25891082

RESUMEN

AIM: Tanshinol is an important catechol in the antianginal herb Salvia miltiorrhiza roots (Danshen). This study aimed to characterize tanshinol methylation. METHODS: Metabolites of tanshinol were analyzed by liquid chromatography/mass spectrometry. Metabolism was assessed in vitro with rat and human enzymes. The major metabolites were synthesized for studying their interactions with drug metabolizing enzymes and transporters and their vasodilatory properties. Dose-related tanshinol methylation and its influences on tanshinol pharmacokinetics were also studied in rats. RESULTS: Methylation, preferentially in the 3-hydroxyl group, was the major metabolic pathway of tanshinol. In rats, tanshinol also underwent considerable 3-O-sulfation, which appeared to be poor in human liver. These metabolites were mainly eliminated via renal excretion, which involved tubular secretion mainly by organic anion transporter (OAT) 1. The methylated metabolites had no vasodilatory activity. Entacapone-impaired methylation did not considerably increase systemic exposure to tanshinol in rats. The saturation of tanshinol methylation in rat liver could be predicted from the Michaelis constant of tanshinol for catechol-O-methyltransferase (COMT). Tanshinol had low affinity for human COMT and OATs; its methylated metabolites also had low affinity for the transporters. Tanshinol and its major human metabolite (3-O-methyltanshinol) exhibited negligible inhibitory activities against human cytochrome P450 enzymes, organic anion transporting polypeptides 1B1/1B3, multidrug resistance protein 1, multidrug resistance-associated protein 2, and breast cancer resistance protein. CONCLUSION: Tanshinol is mainly metabolized via methylation. Tanshinol and its major human metabolite have low potential for pharmacokinetic interactions with synthetic antianginal agents. This study will help define the risk of hyperhomocysteinemia related to tanshinol methylation.


Asunto(s)
Ácidos Cafeicos/farmacocinética , Fármacos Cardiovasculares/farmacocinética , Medicamentos Herbarios Chinos/farmacocinética , Hígado/enzimología , Salvia miltiorrhiza/química , Administración Oral , Animales , Biotransformación , Ácidos Cafeicos/administración & dosificación , Ácidos Cafeicos/aislamiento & purificación , Ácidos Cafeicos/toxicidad , Fármacos Cardiovasculares/administración & dosificación , Fármacos Cardiovasculares/aislamiento & purificación , Fármacos Cardiovasculares/toxicidad , Catecol O-Metiltransferasa/metabolismo , Cromatografía Liquida , Sistema Enzimático del Citocromo P-450/metabolismo , Medicamentos Herbarios Chinos/administración & dosificación , Medicamentos Herbarios Chinos/aislamiento & purificación , Medicamentos Herbarios Chinos/toxicidad , Interacciones de Hierba-Droga , Humanos , Inyecciones Intravenosas , Túbulos Renales/metabolismo , Masculino , Espectrometría de Masas , Proteínas de Transporte de Membrana/metabolismo , Metilación , Microsomas Hepáticos/enzimología , Proteína 1 de Transporte de Anión Orgánico/metabolismo , Fitoterapia , Raíces de Plantas , Plantas Medicinales , Ratas Sprague-Dawley , Eliminación Renal , Sulfatos/metabolismo
14.
Bioorg Med Chem ; 21(22): 7182-93, 2013 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-24080105

RESUMEN

Caffeic acid and its naturally occurring derivative caffeic acid phenethyl ester (CAPE) have antiproliferative and cytotoxic properties in a variety of cancer cell lines without displaying significant toxicity toward healthy cells, and are considered to be potential anticancer agents. However, little is known about their effects on prostate cancer cells. We synthesized and evaluated the effects of caffeic acid, CAPE (2) and 18 synthetic derivatives on cell viability and androgen-dependent cell proliferation, subcellular localisation and expression of androgen receptor (AR) and secretion of prostate-specific antigen (PSA) in LNCaP human hormone-dependent prostate cancer cells. Several synthetic derivatives of CAPE were strong, concentration-dependent cytotoxic agents in LNCaP cells with IC50 values in the 6.8-26.6 µM range, potencies that were up to five-fold greater than that of CAPE (33.7±4.0 µM). A number of caffeic acid derivatives were inhibitors of androgen-stimulated LNCaP cell proliferation with concomitant inhibition of DHT-stimulated PSA secretion. Compound 24 was the most cytotoxic and antiproliferative caffeic acid derivative (IC50 values of 6.8±0.3 and 2.4±0.8 µM, respectively) inhibiting DHT-stimulated cell proliferation and PSA secretion statistically significantly at concentrations as low as 0.3 µM. Exposure to DHT increased cytoplasmic and nuclear AR levels and co-treatment with increasing concentrations of compound 24 or CAPE (2), notably, further increased these levels. In conclusion, a number of synthetic derivatives of caffeic acid are potent inhibitors of androgen-dependent prostate cancer cell proliferation and viability, acting, at least in part, via an antiandrogenic mechanism that involves increased nuclear accumulation of (presumably inactive) AR.


Asunto(s)
Antineoplásicos , Ácidos Cafeicos , Cinamatos/química , Antineoplásicos/síntesis química , Antineoplásicos/química , Antineoplásicos/farmacología , Antineoplásicos/toxicidad , Ácidos Cafeicos/síntesis química , Ácidos Cafeicos/química , Ácidos Cafeicos/farmacología , Ácidos Cafeicos/toxicidad , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Cinamatos/síntesis química , Cinamatos/farmacología , Cinamatos/toxicidad , Humanos , Masculino , Antígeno Prostático Específico/metabolismo , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Receptores Androgénicos/metabolismo , Relación Estructura-Actividad
15.
Cancer Biol Ther ; 14(10): 907-15, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-23917379

RESUMEN

Free radicals are formed as a result of cellular processes and play a key role in predisposition to and development of numerous diseases and of premature aging. Recently, we reported the syntheses of a number of novel phenolic antioxidants for possible application in food industry. In the present study, analyses of the cellular processes and molecular gene expression effects of some of the novel antioxidants in normal human tissues and in cancer cells were undertaken. Results indicated that whereas the examined antioxidants showed no effects on morphology and gene expression of normal human oral and gingival epithelial tissues, they exerted a profound cell killing effect on breast cancer cells, including on chemotherapy-resistant breast cancer cells and on oral squamous carcinoma cells. Among the tested antioxidants, N-decyl-N-(3-methoxy-4-hydroxybenzyl)-3-(3,4-dihydroxyphenyl) propanamide and N-decyl-N-(3,5-dimethoxy-4-hydroxybenzyl)-3-(3,4-dihydroxyphenyl) propanamide were the most promising, with excellent potential for cancer treatment. Moreover, our gene expression databases can be used as a roadmap for future analysis of mechanisms of antioxidant action.


Asunto(s)
Antineoplásicos/toxicidad , Antioxidantes/toxicidad , Lípidos/toxicidad , Antineoplásicos/farmacología , Antioxidantes/farmacología , Ácidos Cafeicos/farmacología , Ácidos Cafeicos/toxicidad , Caspasa 3/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Proteínas de Unión al ADN/metabolismo , Ensayos de Selección de Medicamentos Antitumorales , Expresión Génica/efectos de los fármacos , Histonas/metabolismo , Humanos , Hidroxibenzoatos/farmacología , Hidroxibenzoatos/toxicidad , Lípidos/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Células MCF-7 , Proteína Homóloga de MRE11 , Antígeno Nuclear de Célula en Proliferación/genética , Antígeno Nuclear de Célula en Proliferación/metabolismo
16.
J Ethnopharmacol ; 149(1): 84-92, 2013 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-23770030

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Propolis is a bee product with numerous biological and pharmacological properties, such as immunomodulatory and anti-inflammatory activities. It has been used in folk medicine as a healthy drink and in food to improve health and prevent inflammatory diseases. However, little is known about its mechanism of action. Thus, the goal of this study was to verify the antioxidant activity and to explore the anti-inflammatory properties of propolis by addressing its intracellular mechanism of action. Caffeic acid was investigated as a possible compound responsible for propolis action. MATERIALS AND METHODS: The antioxidant properties of propolis and caffeic acid were evaluated by using the 2,2-Diphenyl-1-picrylhydrazyl free radical (DPPH) scavenging method. To analyze the anti-inflammatory activity, Raw 264.7 macrophages were treated with different concentrations of propolis or caffeic acid, and nitric oxide (NO) production, a strong pro-inflammatory mediator, was evaluated by the Griess reaction. The concentrations of propolis and caffeic acid that inhibited NO production were evaluated on intracellular signaling pathways triggered during inflammation, namely p38 mitogen-activated protein kinase (MAPK), c-jun NH2-terminal kinase (JNK1/2), the transcription nuclear factor (NF)-κB and extracellular signal-regulated kinase (ERK1/2), through Western blot using specific antibodies. A possible effect of propolis on the cytotoxicity of hepatocytes was also evaluated, since this product can be used in human diets. RESULTS: Caffeic acid showed a higher antioxidant activity than propolis extract. Propolis and caffeic acid inhibited NO production in macrophages, at concentrations without cytotoxicity. Furthermore, both propolis and caffeic acid suppressed LPS-induced signaling pathways, namely p38 MAPK, JNK1/2 and NF-κB. ERK1/2 was not affected by propolis extract and caffeic acid. In addition, propolis and caffeic acid did not induce hepatotoxicity at concentrations with strong anti-inflammatory potential. CONCLUSIONS: Propolis exerted an antioxidant and anti-inflammatory action and caffeic acid may be involved in its inhibitory effects on NO production and intracellular signaling cascades, suggesting its use as a natural source of safe anti-inflammatory drugs.


Asunto(s)
Antiinflamatorios/farmacología , Ácidos Cafeicos/farmacología , Depuradores de Radicales Libres/farmacología , Macrófagos/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , FN-kappa B/antagonistas & inhibidores , Própolis/farmacología , Animales , Antiinflamatorios/aislamiento & purificación , Antiinflamatorios/toxicidad , Ácidos Cafeicos/aislamiento & purificación , Ácidos Cafeicos/toxicidad , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Etnofarmacología , Depuradores de Radicales Libres/aislamiento & purificación , Depuradores de Radicales Libres/toxicidad , Lipopolisacáridos/farmacología , Macrófagos/inmunología , Ratones , Óxido Nítrico/antagonistas & inhibidores , Óxido Nítrico/biosíntesis , Própolis/química , Própolis/toxicidad
17.
Genet Mol Res ; 10(2): 1130-40, 2011 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-21710465

RESUMEN

Phenols are a large and diverse class of compounds, many of which occur naturally in a variety of food plants; they exhibit a wide range of biological effects, including antibacterial, anti-inflammatory, antiallergic, hepatoprotective, antithrombotic, antiviral, anticarcinogenic, and vasodilatory actions. We examined the genotoxic and clastogenic potential of three phenolic compounds: caffeic, cinnamic and ferulic acids, using the comet and micronucleus assays in vitro. Drug-metabolizing rat hepatoma tissue cells (HTCs) were used. Three different concentrations (50, 500 and 1500 µM) of these phenolic acids were tested on the HTCs for 24 h. The caffeic, cinnamic and ferulic acids were not genotoxic by the comet assay (P > 0.05). However, the micronucleus test showed an increase in the frequency of micronucleated cells for the three compounds, indicating that these substances have clastogenic effects in HTC.


Asunto(s)
Ácidos Cafeicos/toxicidad , Cinamatos/toxicidad , Ácidos Cumáricos/toxicidad , Mutágenos/toxicidad , Animales , Ensayo Cometa , Técnicas In Vitro , Ratas , Células Tumorales Cultivadas
18.
Chem Biol Interact ; 192(3): 243-56, 2011 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-21458432

RESUMEN

Glutathione S-transferase (GST) and multidrug resistance-associated proteins (MRPs) play major roles in drug resistance in melanoma. In this study, we investigated caffeic acid phenethyl ester (CAPE) as a selective GST inhibitor in the presence of tyrosinase, which is abundant in melanoma cells. Tyrosinase bioactivates CAPE to an o-quinone, which reacts with glutathione to form CAPE-SG conjugate. Our findings indicate that 90% CAPE was metabolized by tyrosinase after a 60-min incubation. LC-MS/MS analyses identified a CAPE-SG conjugate as a major metabolite. In the presence of tyrosinase, CAPE (10-25µM) showed 70-84% GST inhibition; whereas in the absence of tyrosinase, CAPE did not inhibit GST. CAPE-SG conjugate and CAPE-quinone (25µM) demonstrated ⩾85% GST inhibition via reversible and irreversible mechanisms, respectively. Comparing with CDNB and GSH, the non-substrate CAPE acted as a weak, reversible GST inhibitor at concentrations >50µM. Furthermore, MK-571, a selective MRP inhibitor, and probenecid, a non-selective MRP inhibitor, decrease the IC(50) of CAPE (15µM) by 13% and 21%, apoptotic cell death by 3% and 13%, and mitochondrial membrane potential in human SK-MEL-28 melanoma cells by 10% and 56%, respectively. Moreover, computational docking analyses suggest that CAPE binds to the GST catalytic active site. Caffeic acid, a hydrolyzed product of CAPE, showed a similar GST inhibition in the presence of tyrosinase. Although, as controls, 4-hydroxyanisole and L-tyrosine were metabolized by tyrosinase to form quinones and glutathione conjugates, they exhibited no GST inhibition in the absence and presence of tyrosinase. In conclusion, both CAPE and caffeic acid selectively inhibited GST in the presence of tyrosinase. Our results suggest that intracellularly formed quinones and glutathione conjugates of caffeic acid and CAPE may play major roles in the selective inhibition of GST in SK-MEL-28 melanoma cells. Moreover, the inhibition of MRP enhances CAPE-induced toxicity in the SK-MEL-28 melanoma cells.


Asunto(s)
Ácidos Cafeicos/metabolismo , Inhibidores Enzimáticos/metabolismo , Glutatión Transferasa/antagonistas & inhibidores , Melanoma/metabolismo , Monofenol Monooxigenasa/metabolismo , Alcohol Feniletílico/análogos & derivados , Ácidos Cafeicos/toxicidad , Línea Celular Tumoral , Inhibidores Enzimáticos/toxicidad , Glutatión Transferasa/metabolismo , Humanos , Melanoma/patología , Alcohol Feniletílico/metabolismo , Alcohol Feniletílico/toxicidad
19.
Toxicol Appl Pharmacol ; 250(1): 69-77, 2011 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-20950636

RESUMEN

Arsenic trioxide (ATO) treatment is a useful therapy against human acute promyelocytic leukemia (APL), however, it concomitantly brings potential adverse consequences including serious side effect, human carcinogenicity and possible development of resistance. This investigation revealed that those problems might be relaxed by simultaneous application with (-)-epigallocatechin-3-gallate (EGCG), one of the major components from green tea. EGCG significantly lowered down the ATO concentration required for an effective control of APL cells, HL-60. The simultaneous treatment of ATO with EGCG induced a mitochondria-dependent apoptosis in HL-60 cells significantly, which accounted for more than 70% of the cell death in the treatment. The mechanism of apoptosis induction was elucidated. EGCG in HL-60 cells acted as a pro-oxidant enhancing intracellular hydrogen peroxide significantly. ATO, on the other hand, induced heme oxygenase-1 (HO-1) to catalyze heme degradation, thereby provided ferrous iron for EGCG-induced hydrogen peroxide to precede Fenton reaction, which in turn generated deleterious reactive oxygen species to damage cell. In addition, EGCG inhibited expression of ferritin, which supposedly to sequester harmful ferrous iron, thereby augmented the occurrence of Fenton reaction. This investigation also provided evidence that ATO, since mainly acted to induce HO-1 in simultaneous treatment with EGCG, could be replaced by other HO-1 inducer with much less human toxicity. Furthermore, several of our preliminary investigations revealed that the enhanced cytotoxicity induced by combining heme degradation and Fenton reaction is selectively toxic to malignant but not non-malignant cells.


Asunto(s)
Antineoplásicos/toxicidad , Catequina/análogos & derivados , Citotoxinas/toxicidad , Ferritinas/antagonistas & inhibidores , Óxidos/toxicidad , Apoptosis/efectos de los fármacos , Trióxido de Arsénico , Arsenicales , Ácidos Cafeicos/toxicidad , Catequina/toxicidad , Línea Celular Tumoral , Sinergismo Farmacológico , Ferritinas/metabolismo , Células HL-60 , Hemo Oxigenasa (Desciclizante)/metabolismo , Humanos , Neoplasias/enzimología , Neoplasias/metabolismo , Neoplasias/patología , Alcohol Feniletílico/análogos & derivados , Alcohol Feniletílico/toxicidad
20.
Chem Biol Interact ; 188(1): 1-14, 2010 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-20685355

RESUMEN

In the current work, we investigated the in vitro biochemical mechanism of Caffeic Acid Phenylethyl Ester (CAPE) toxicity and eight hydroxycinnamic/caffeic acid derivatives in vitro, using tyrosinase enzyme as a molecular target in human SK-MEL-28 melanoma cells. Enzymatic reaction models using tyrosinase/O(2) and HRP/H(2)O(2) were used to delineate the role of one- and two-electron oxidation. Ascorbic acid (AA), NADH and GSH depletion were used as markers of quinone formation and oxidative stress in CAPE induced toxicity in melanoma cells. Ethylenediamine, an o-quinone trap, prevented the formation of o-quinone and oxidations of AA and NADH mediated by tyrosinase bioactivation of CAPE. The IC(50) of CAPE towards SK-MEL-28 melanoma cells was 15muM. Dicoumarol, a diaphorase inhibitor, and 1-bromoheptane, a GSH depleting agent, increased CAPE's toxicity towards SK-MEL-28 cells indicating quinone formation played an important role in CAPE induced cell toxicity. Cyclosporin-A and trifluoperazine, inhibitors of the mitochondrial membrane permeability transition pore (PTP), prevented CAPE toxicity towards melanoma cells. We further investigated the role of tyrosinase in CAPE toxicity in the presence of a shRNA plasmid, targeting tyrosinase mRNA. Results from tyrosinase shRNA experiments showed that CAPE led to negligible anti-proliferative effect, apoptotic cell death and ROS formation in shRNA plasmid treated cells. Furthermore, it was also found that CAPE selectively caused escalation in the ROS formation and intracellular GSH (ICG) depletion in melanocytic human SK-MEL-28 cells which express functional tyrosinase. In contrast, CAPE did not lead to ROS formation and ICG depletion in amelanotic C32 melanoma cells, which do not express functional tyrosinase. These findings suggest that tyrosinase plays a major role in CAPE's selective toxicity towards melanocytic melanoma cell lines. Our findings suggest that the mechanisms of CAPE toxicity in SK-MEL-28 melanoma cells mediated by tyrosinase bioactivation of CAPE included quinone formation, ROS formation, intracellular GSH depletion and induced mitochondrial toxicity.


Asunto(s)
Apoptosis/efectos de los fármacos , Ácidos Cafeicos/toxicidad , Melanoma Experimental/patología , Alcohol Feniletílico/análogos & derivados , Animales , Ácido Ascórbico/metabolismo , Línea Celular Tumoral , Glutatión/metabolismo , Masculino , Melanoma Experimental/enzimología , Monofenol Monooxigenasa/metabolismo , NAD/metabolismo , Alcohol Feniletílico/toxicidad , Ratas , Ratas Sprague-Dawley , Espectrofotometría Ultravioleta
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA