Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 93
Filtrar
1.
JNMA J Nepal Med Assoc ; 56(211): 711-715, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30381772

RESUMEN

Methotrexate is an antimetabolite drug with antineoplastic and immunomodulatory properties, useful as an antineoplastic agent in various haematological and solid tumours. MTX toxicity can occur because of accidental ingestion/overdose by the patient or because of prescription error. The toxic effects manifest as severe mucositis or as organ damage (bone marrow depression, renal/hepatic injury). The toxicity usually results from parenteral overdose or repeated chronic drug ingestion. Acute high dose ingestion does not result in MTX toxicity because of saturable absorption kinetics. We present a case of MTX toxicity occurring as a result of prescription error resulting in repeat daily dosing of the drug, and the challenges associated with the management of the same, in a patient with multiple comorbidities. The present case emphasizes on a note of caution on the part of the prescriber and the suggestions regarding the measures which can be taken to avoid MTX toxicity. Keywords: drug overdose; Methotrexate; mucositis; pancytopenia.


Asunto(s)
Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Errores Médicos/efectos adversos , Metotrexato , Manejo de Atención al Paciente/métodos , Comorbilidad , Enfermedad de la Arteria Coronaria/complicaciones , Diabetes Mellitus Tipo 2/complicaciones , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/diagnóstico , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/etiología , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/fisiopatología , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/terapia , Nutrición Enteral/métodos , Antagonistas del Ácido Fólico/administración & dosificación , Antagonistas del Ácido Fólico/toxicidad , Humanos , Fallo Renal Crónico/complicaciones , Masculino , Metotrexato/administración & dosificación , Metotrexato/toxicidad , Persona de Mediana Edad , Diálisis Renal/métodos
2.
Reprod Toxicol ; 80: 92-104, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29859881

RESUMEN

We discuss the possibilities to prevent the post-exposure teratogenic effects of several teratogens: valproic acid (VPA), diabetes and alcohol. Co-administration of folic acid with VPA reduced the rate of Neural Tube Defects (NTD) and other anomalies in rodents, but apparently not in pregnant women. Antioxidants or the methyl donor S-adenosyl methionine prevented Autism Spectrum Disorder (ASD) like behavior in mice and rats. In vivo and in vitro studies demonstrated that antioxidants, arachidonic acid, myoinositol and nutritional agents may prevent diabetes-embryopathy. Prevention of alcohol-induced embryonic and fetal injuries and neurodevelopmental deficits was achieved by supplementation of zinc, choline, vasoactive intestinal proteins (VIP related peptides), antioxidants and folic acid. While the animal research described in this review is indicative of possible preventions of the different teratogenic effects, this is not yet the focus in human research. Future research should promote further knowledge where our current understanding is the vaguest, human prevention.


Asunto(s)
Anomalías Inducidas por Medicamentos/prevención & control , Antioxidantes/administración & dosificación , Diabetes Gestacional , Ácido Fólico/administración & dosificación , Embarazo en Diabéticas , Efectos Tardíos de la Exposición Prenatal/prevención & control , Teratógenos/toxicidad , Anomalías Inducidas por Medicamentos/etiología , Animales , Antioxidantes/uso terapéutico , Diabetes Gestacional/metabolismo , Etanol/toxicidad , Éteres de Etila , Femenino , Ácido Fólico/uso terapéutico , Antagonistas del Ácido Fólico/toxicidad , Humanos , Estrés Oxidativo , Embarazo , Embarazo en Diabéticas/metabolismo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Compuestos de Sulfhidrilo , Ácido Valproico/toxicidad
3.
Drug Chem Toxicol ; 41(1): 51-61, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28298149

RESUMEN

Methotrexate (MTX) is a folic acid antagonist that is widely used to treat a variety of diseases. One of the most serious side effects of MTX therapy is hepatotoxicity. The potential molecular cytotoxic mechanisms of MTX toward isolated rat hepatocytes were investigated using Accelerated Cytotoxicity Mechanism Screening (ACMS) techniques. A concentration and time dependent increase in cytotoxicity and reactive oxygen species (ROS) formation and a decrease in mitochondrial membrane potential (MMP) were observed with MTX. Furthermore, a significant increase in MTX (300 µM)-induced cytotoxicity and ROS formation were observed when glutathione (GSH)-depleted hepatocytes were used whereas addition of N-acetylcysteine (a GSH precursor) decreased cytotoxicity. Catalase inactivation also increased MTX-induced cytotoxicity, while the direct addition of catalase to the hepatocytes decreased cytotoxicity. MTX treatment in isolated rat mitochondria caused swelling and significantly decreased adenosine triphosphate (ATP) and GSH content, and cytochrome c release. Potent antioxidants such as mesna, resveratrol and Trolox decreased MTX-induced cytotoxicity and ROS formation and increased MMP. This study suggests that MTX-induced cytotoxicity caused by ROS formation and GSH oxidation leads to oxidative stress and mitochondrial injury in rat hepatocytes.


Asunto(s)
Citocromos c/metabolismo , Antagonistas del Ácido Fólico/toxicidad , Hepatocitos/efectos de los fármacos , Metotrexato/toxicidad , Mitocondrias Hepáticas/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Adenosina Trifosfato/metabolismo , Animales , Antioxidantes/farmacología , Catalasa/metabolismo , Relación Dosis-Respuesta a Droga , Glutatión/metabolismo , Hepatocitos/enzimología , Hepatocitos/patología , Peroxidación de Lípido/efectos de los fármacos , Masculino , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias Hepáticas/enzimología , Mitocondrias Hepáticas/patología , Dilatación Mitocondrial/efectos de los fármacos , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Factores de Tiempo
4.
Nucleic Acids Res ; 45(5): 2472-2489, 2017 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-27924000

RESUMEN

The biogenesis of ribosomes in vivo is an essential process for cellular functions. Transcription of ribosomal RNA (rRNA) genes is the rate-limiting step in ribosome biogenesis controlled by environmental conditions. Here, we investigated the role of folate antagonist on changes of DNA double-strand breaks (DSBs) landscape in mouse embryonic stem cells. A significant DSB enhancement was detected in the genome of these cells and a large majority of these DSBs were found in rRNA genes. Furthermore, spontaneous DSBs in cells under folate deficiency conditions were located exclusively within the rRNA gene units, representing a H3K4me1 hallmark. Enrichment H3K4me1 at the hot spots of DSB regions enhanced the recruitment of upstream binding factor (UBF) to rRNA genes, resulting in the increment of rRNA genes transcription. Supplement of folate resulted in a restored UBF binding across DNA breakage sites of rRNA genes, and normal rRNA gene transcription. In samples from neural tube defects (NTDs) with low folate level, up-regulation of rRNA gene transcription was observed, along with aberrant UBF level. Our results present a new view by which alterations in folate levels affects DNA breakage through epigenetic control leading to the regulation of rRNA gene transcription during the early stage of development.


Asunto(s)
Roturas del ADN de Doble Cadena , Deficiencia de Ácido Fólico/genética , Regulación del Desarrollo de la Expresión Génica , Genes de ARNr , Proteínas del Complejo de Iniciación de Transcripción Pol1/metabolismo , Transcripción Genética , Animales , Células Cultivadas , Células Madre Embrionarias/metabolismo , Feto/metabolismo , Antagonistas del Ácido Fólico/toxicidad , Deficiencia de Ácido Fólico/metabolismo , Fase G1/genética , Histonas/metabolismo , Leucovorina/farmacología , Metotrexato/toxicidad , Ratones , Defectos del Tubo Neural/genética , Defectos del Tubo Neural/metabolismo
5.
PLoS One ; 10(3): e0121869, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25822193

RESUMEN

DNA methylation is thought to be involved in the etiology of neural tube defects (NTDs). However, the exact mechanism between DNA methylation and NTDs remains unclear. Herein, we investigated the change of methylation in mouse model of NTDs associated with folate dysmetabolism by use of ultraperformance liquid chromatography tandem mass spectrometry (UPLC/MS/MS), liquid chromatography-electrospray ionization tandem mass spectrometry (LC-MS/MS), microarray, matrix-assisted laser desorption/ionization time-of-flight mass spectrometry and Real time quantitative PCR. Results showed that NTD neural tube tissues had lower concentrations of 5-methyltetrahydrofolate (5-MeTHF, P = 0.005), 5-formyltetrahydrofolate (5-FoTHF, P = 0.040), S-adenosylmethionine (SAM, P = 0.004) and higher concentrations of folic acid (P = 0.041), homocysteine (Hcy, P = 0.006) and S-adenosylhomocysteine (SAH, P = 0.045) compared to control. Methylation levels of genomic DNA decreased significantly in the embryonic neural tube tissue of NTD samples. 132 differentially methylated regions (35 low methylated regions and 97 high methylated regions) were selected by microarray. Two genes (Siah1b, Prkx) in Wnt signal pathway demonstrated lower methylated regions (peak) and higher expression in NTDs (P<0.05; P<0.05). Results suggest that DNA hypomethylation was one of the possible epigenetic variations correlated with the occurrence of NTDs induced by folate dysmetabolism and that Siah1b, Prkx in Wnt pathway may be candidate genes for NTDs.


Asunto(s)
Metilación de ADN/efectos de los fármacos , Antagonistas del Ácido Fólico/toxicidad , Ácido Fólico/metabolismo , Metotrexato/toxicidad , Defectos del Tubo Neural/etiología , Defectos del Tubo Neural/metabolismo , Animales , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Metilación de ADN/genética , Modelos Animales de Enfermedad , Desarrollo Embrionario/efectos de los fármacos , Desarrollo Embrionario/genética , Femenino , Deficiencia de Ácido Fólico/complicaciones , Ratones , Defectos del Tubo Neural/genética , Embarazo , Proteínas Serina-Treonina Quinasas , Espectrometría de Masas en Tándem , Ubiquitina-Proteína Ligasas/genética , Vía de Señalización Wnt/efectos de los fármacos , Vía de Señalización Wnt/genética
6.
Mol Cell Biochem ; 398(1-2): 223-31, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25245820

RESUMEN

The causal metabolic pathway and the underlying mechanism between folate deficiency and neural tube defects (NTDs) remain obscure. Thymidylate (dTMP) is catalyzed by thymidylate synthase (TS) using the folate-derived one-carbon unit as the sole methyl donor. This study aims to examine the role of dTMP biosynthesis in the development of neural tube in mice by inhibition of TS via a specific inhibitor, raltitrexed (RTX). Pregnant mice were intraperitoneally injected with various doses of RTX on gestational day 7.5, and embryos were examined for the presence of NTDs on gestational day 11.5. TS activity and changes of dUMP and dTMP levels were measured following RTX treatment at the optimal dose. DNA damage was determined by detection of phosphorylated replication protein A2 (RPA2) and γ-H2AX in embryos with NTDs induced by RTX. Besides, apoptosis and proliferation were also analyzed in RTX-treated embryos with NTDs. We found that NTDs were highly occurred by the treatment of RTX at the optimal dose of 11.5 mg/kg b/w. RTX treatment significantly inhibited TS activity. Meanwhile, dTMP was decreased associated with the accumulation of dUMP in RTX-treated embryos. Phosphorylated RPA2 and γ-H2AX were significantly increased in RTX-treated embryos with NTDs compared to control. More apoptosis and decreased proliferation were also found in embryos with NTDs induced by RTX. These results indicate that impairment of dTMP biosynthesis caused by RTX led to the development of NTDs in mice. DNA damage and imbalance between apoptosis and proliferation may be potential mechanisms.


Asunto(s)
Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Daño del ADN , Embrión de Mamíferos/efectos de los fármacos , Defectos del Tubo Neural/metabolismo , Quinazolinas/toxicidad , Tiofenos/toxicidad , Animales , Western Blotting , Nucleótidos de Desoxiuracil/metabolismo , Embrión de Mamíferos/metabolismo , Embrión de Mamíferos/patología , Femenino , Antagonistas del Ácido Fólico/administración & dosificación , Antagonistas del Ácido Fólico/toxicidad , Edad Gestacional , Histonas/metabolismo , Inyecciones Intraperitoneales , Masculino , Ratones Endogámicos C57BL , Defectos del Tubo Neural/inducido químicamente , Defectos del Tubo Neural/genética , Fosforilación/efectos de los fármacos , Embarazo , Subunidades de Proteína/metabolismo , Quinazolinas/administración & dosificación , Proteína de Replicación A/metabolismo , Tiofenos/administración & dosificación , Timidina Monofosfato/metabolismo , Timidilato Sintasa/antagonistas & inhibidores , Timidilato Sintasa/metabolismo
7.
J Appl Toxicol ; 34(8): 862-9, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23836430

RESUMEN

Apoptosis and proliferation play important roles in embryonic development and are required for neural tube closure. The antifolate drug methotrexate (MTX) induces folate dysmetabolism by inhibition of dihydrofolate reductase and causes abnormal apoptosis and proliferation. In this study, we established an animal model of neural tube defects (NTDs) using MTX to investigate the role of apoptosis and proliferation in NTDs caused by folate deficiency. Differential gene expressions were studied by microarray and reverse transcription-polymerase chain reaction in the NTD animal model. Results showed that 30.8% of NTDs were caused by using MTX in treatment regimens. Microarray indicated that 166 genes were significantly different between the control and NTD mice, including four apoptosis-related genes (Endog, Trp53, Casp3, Bax) and three proliferation-related genes (Ptch1, Pla2g4a, Foxg1). Levels of Endog, Trp53, Casp3, Bax (fold change>1.5) were upregulated but Ptch1, Pla2g4a, Foxg1 (fold change<0.67) were downregulated (P<0.05). These results were confirmed by reverse transcription-polymerase chain reaction. TUNEL, immunohistochemical assays and Western blot were further used to detect apoptosis and proliferation in the NTD animal model. It was found that apoptosis in neuroepithelial cells was increased as determined by TUNEL (P<0.05). Expressions of caspase-3 were significantly enhanced (P<0.05) but expressions of phosphohistone H3 were greatly decreased (P<0.05). These results concluded that MTX caused a folate and folate-associated dysmetabolism, and further induced abnormal apoptosis and proliferation, which may play a critical role in the occurrence of NTDs caused by folate deficiency.


Asunto(s)
Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Antagonistas del Ácido Fólico/toxicidad , Metotrexato/toxicidad , Neurulación/efectos de los fármacos , Animales , Caspasa 3/genética , Caspasa 3/metabolismo , Modelos Animales de Enfermedad , Regulación hacia Abajo , Desarrollo Embrionario/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Femenino , Ácido Fólico/metabolismo , Deficiencia de Ácido Fólico/complicaciones , Deficiencia de Ácido Fólico/embriología , Histonas/genética , Histonas/metabolismo , Ratones , Ratones Endogámicos C57BL , Análisis por Micromatrices , Defectos del Tubo Neural/embriología , Embarazo , Tetrahidrofolato Deshidrogenasa/metabolismo , Regulación hacia Arriba
8.
J Basic Clin Physiol Pharmacol ; 25(2): 249-53, 2014 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-24285126

RESUMEN

BACKGROUND: Trimethoprim-sulfamethoxazole (TMP-SMX) is a broad-spectrum antibiotic. However, its use is associated with toxic reactions. Virgin coconut oil (VCO), derived from coconut, has been widely used throughout history for its medicinal value. The aim of this study was to investigate the beneficial actions of VCO against TMP-SMX-induced alterations in serum biochemical end points. METHODS: Twenty rats were divided into four groups. Group 1 (control) received no drug, whereas group 2 received TMP-SMX (8/40 mg/kg) twice daily for 7 days. Group 3 was administered coconut oil at a dose of 600 mg/kg body weight per day. The last group was treated with TMP-SMX (8/40 mg/kg) and coconut oil (600 mg/kg) simultaneously. Blood samples were collected from all groups on the 8th day of the experiment for measurement of serum biochemical parameters. Organ weights and coefficients were also evaluated. RESULTS: TMP-SMX caused a significant (p<0.05) increase in the levels of serum total bilirubin, lactate dehydrogenase, and alkaline phosphatase by 192%, 67%, and 41%, respectively, relative to controls. This was followed by a significant reduction in triglyceride and relative kidney weight by 40% and 7%, respectively. There were no significant differences (p>0.05) in the activities of serum aminotransferases, total acid phosphatase, γ-glutamyl transferase, uric acid, cholesterol, albumin, and urea levels. Supplementation of VCO ameliorated TMP-SMX-induced effects by restoring the levels of total bilirubin, alkaline phospahatase, and lactate dehydrogenase. CONCLUSIONS: The results of this study demonstrate that the active components of coconut oil had protective effects against the toxic effects induced by TMP-SMX administration, especially in the liver of rats.


Asunto(s)
Antiinfecciosos/toxicidad , Enfermedad Hepática Inducida por Sustancias y Drogas/prevención & control , Antagonistas del Ácido Fólico/toxicidad , Aceites de Plantas/uso terapéutico , Combinación Trimetoprim y Sulfametoxazol/toxicidad , Animales , Biomarcadores/sangre , Enfermedad Hepática Inducida por Sustancias y Drogas/sangre , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Aceite de Coco , Riñón/efectos de los fármacos , Pruebas de Función Renal , Pruebas de Función Hepática , Masculino , Tamaño de los Órganos/efectos de los fármacos , Aceites de Plantas/administración & dosificación , Ratas Wistar
9.
Drug Des Devel Ther ; 7: 449-75, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23818757

RESUMEN

BACKGROUND: There is a dearth of treatment options for community-acquired and nosocomial Pseudomonas infections due to several rapidly emerging multidrug resistant phenotypes, which show resistance even to combination therapy. As an alternative, developing selective promiscuous hybrid compounds for simultaneous modulation of multiple targets is highly appreciated because it is difficult for the pathogen to develop resistance when an inhibitor has activity against multiple targets. METHODS: In line with our previous work on phytochemical-antibiotic combination assays and knowledge-based methods, using a fragment combination approach we here report a novel drug design strategy of conjugating synergistic phytochemical-antibiotic combinations into a single hybrid entity for multi-inhibition of P. aeruginosa DNA gyrase subunit B (GyrB)/topoisomerase IV subunit B (ParE) and dihydrofolate reductase (DHFR) enzymes. The designed conjugates were evaluated for their multitarget specificity using various computational methods including docking and dynamic simulations, drug-likeness using molecular properties calculations, and pharmacophoric features by stereoelectronic property predictions. RESULTS: Evaluation of the designed hybrid compounds based on their physicochemical properties has indicated that they are promising drug candidates with drug-like pharmacotherapeutic profiles. In addition, the stereoelectronic properties such as HOMO (highest occupied molecular orbital), LUMO (lowest unoccupied molecular orbital), and MEP (molecular electrostatic potential) maps calculated by quantum chemical methods gave a good correlation with the common pharmacophoric features required for multitarget inhibition. Furthermore, docking and dynamics simulations revealed that the designed compounds have favorable binding affinity and stability in both the ATP-binding sites of GyrB/ParE and the folate-binding site of DHFR, by forming strong hydrogen bonds and hydrophobic interactions with key active site residues. CONCLUSION: This new design concept of hybrid "phyto-drug" scaffolds, and their simultaneous perturbation of well-established antibacterial targets from two unrelated pathways, appears to be very promising and could serve as a prospective lead in multitarget drug discovery.


Asunto(s)
Antibacterianos/farmacología , Topoisomerasa de ADN IV/antagonistas & inhibidores , Diseño de Fármacos , Antagonistas del Ácido Fólico/farmacología , Pseudomonas aeruginosa/efectos de los fármacos , Inhibidores de Topoisomerasa II , Adenosina Trifosfato/metabolismo , Antibacterianos/química , Sitios de Unión , Antagonistas del Ácido Fólico/química , Antagonistas del Ácido Fólico/toxicidad , Metotrexato/química , Metotrexato/farmacología , Modelos Moleculares , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Electricidad Estática , Tetrahidrofolato Deshidrogenasa/química
10.
J Appl Toxicol ; 33(9): 915-23, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22806879

RESUMEN

Impaired folate metabolism is considered a risk factor for neural tube defects (NTDs). However, the relationship between folate deficiency and the risk of NTDs remains unclear, because experimentally induced dietary folate deficiency is insufficient to cause NTDs in non-mutant mice. Methotrexate (MTX) is a specific folate antagonist that competitively inhibits dihydrofolate reductase (DHFR) activity. The objective of this study was to develop a folate dysmetabolism murine model, and study the development of NTDs and its mechanism. Pregnant mice were injected with different doses of MTX [0, 0.5, 1.0, 3.0, 4.5 and 6.0 mg kg(-1) body weight (b/w) intraperitoneally (i.p.)] on gestational day 7.5 and sacrificed on gestational day 11.5. DHFR activity in embryonic tissues was detected, and folate concentrations were analyzed using LC/MS/MS. Copy number variations (CNVs) in neural tube tissues were detected using array comparative genomic hybridization (aCGH). A dose of MTX 4.5 mg kg(-1) b/w, resulted in the highest incidence of NTDs (31.4%) compared with the other groups, and DHFR activities, 5-MeTHF and 5-FoTHF concentrations in embryonic tissues decreased significantly after MTX injection. Furthermore, we found three high-confidence CNVs on chromosome X using aCGH, which was confirmed by RT-PCR and MassARRAY. These results indicate that MTX could cause a folate-associated dysmetabolism, which is similar to that of dietary folate deficiency in mice. The presence of CNVs in neural tube tissues was associated with the development of NTDs.


Asunto(s)
Antagonistas del Ácido Fólico/toxicidad , Inestabilidad Genómica/efectos de los fármacos , Metotrexato/toxicidad , Defectos del Tubo Neural/patología , Anomalías Inducidas por Medicamentos/etiología , Anomalías Inducidas por Medicamentos/patología , Animales , Cromatografía Liquida , Hibridación Genómica Comparativa , Variaciones en el Número de Copia de ADN/efectos de los fármacos , Dieta , Relación Dosis-Respuesta a Droga , Desarrollo Embrionario/efectos de los fármacos , Inhibidores Enzimáticos/metabolismo , Femenino , Ácido Fólico/metabolismo , Antagonistas del Ácido Fólico/administración & dosificación , Deficiencia de Ácido Fólico/metabolismo , Deficiencia de Ácido Fólico/patología , Masculino , Metotrexato/administración & dosificación , Ratones , Ratones Endogámicos C57BL , Tubo Neural/efectos de los fármacos , Tubo Neural/embriología , Tubo Neural/patología , Defectos del Tubo Neural/inducido químicamente , Defectos del Tubo Neural/embriología , Embarazo , Reacción en Cadena en Tiempo Real de la Polimerasa , Reproducibilidad de los Resultados , Espectrometría de Masas en Tándem , Tetrahidrofolato Deshidrogenasa/metabolismo
11.
Birth Defects Res A Clin Mol Teratol ; 94(4): 187-207, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22434686

RESUMEN

Methotrexate and aminopterin are folic acid antagonists that inhibit dihydrofolate reductase, resulting in a block in the synthesis of thymidine and inhibition of DNA synthesis. Methotrexate has been used for the treatment of malignancy, rheumatic disorders, and psoriasis and termination of intrauterine pregnancy. Recently, methotrexate has become a standard treatment for ectopic pregnancy. The misdiagnosis of an intrauterine pregnancy as an ectopic pregnancy can result in exposure of a continuing pregnancy to dose levels of methotrexate of 50 mg/m(2) (maternal body surface area). Experimental animal studies have associated methotrexate therapy with embryo death in mice, rats, rabbits, and monkeys. Structural malformations have been most consistently produced in rabbits at a maternal dose level of 19.2 mg/kg. Abnormalities in rabbits include hydrocephalus, microphthalmia, cleft lip and palate, micrognathia, dysplastic sacral and caudal vertebrate, phocomelia, hemimelia, syndactyly, and ectrodactyly. Based on human case reports of methotrexate exposure during pregnancy, a methotrexate embryopathy has been described that includes growth deficiency, microcephaly, hypoplasia of skull bones, wide fontanels, coronal or lambdoidal craniosynostosis, upswept frontal scalp hair, broad nasal bridge, shallow supraorbital ridges, prominent eyes, low-set ears, maxillary hypoplasia, epicanthal folds, short limbs, talipes, hypodactyly, and syndactyly. This syndrome may be associated with exposures between 6 and 8 weeks after conception and dose levels of 10 mg/week or greater. More recent case reports of methotrexate exposure for the misdiagnosis of ectopic pregnancy involve treatment before 6 weeks after conception and have raised the suggestion of a distinct syndrome due to such early exposures. Tetralogy of Fallot and perhaps other neural crest cell-related abnormalities may be features of this early syndrome. A disproportionality analysis of methotrexate and aminopterin case reports and series provides support for pulmonary atresia, craniosynostosis, and limb deficiencies as reported more often than expected in methotrexate-exposed children. Denominator-based data will be welcome to better define elements of a methotrexate embryopathy and possibly to distinguish an early exposure syndrome from anomalies traditionally associated with methotrexate exposure.


Asunto(s)
Anomalías Inducidas por Medicamentos/etiología , Anomalías Múltiples/inducido químicamente , Metotrexato/toxicidad , Teratógenos/toxicidad , Anomalías Inducidas por Medicamentos/patología , Anomalías Múltiples/patología , Aminopterina/toxicidad , Animales , Femenino , Enfermedades Fetales/etiología , Antagonistas del Ácido Fólico/toxicidad , Humanos , Ratones , Ratones Endogámicos C57BL , Embarazo , Complicaciones del Embarazo/tratamiento farmacológico , Embarazo Ectópico/tratamiento farmacológico , Conejos , Ratas , Ratas Sprague-Dawley , Tetrahidrofolato Deshidrogenasa/efectos de los fármacos
12.
Photochem Photobiol Sci ; 11(4): 653-60, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22234658

RESUMEN

Nanoparticle (NP)-based targeted drug delivery involves cell-specific targeting followed by a subsequent therapeutic action from the therapeutic carried by the NP system. NPs conjugated with methotrexate (MTX), a potent inhibitor of dihydrofolate reductase (DHFR) localized in cytosol, have been under investigation as a delivery system to target cancer cells to enhance the therapeutic index of methotrexate, which is otherwise non-selectively cytotoxic. Despite improved therapeutic activity from MTX-conjugated NPs in vitro and in vivo, the therapeutic action of these conjugates following cellular entry is poorly understood; in particular it is unclear whether the therapeutic activity requires release of the MTX. This study investigates whether MTX must be released from a nanoparticle in order to achieve the therapeutic activity. We report herein light-controlled release of methotrexate from a dendrimer-based conjugate and provide evidence suggesting that MTX still attached to the nanoconjugate system is fully able to inhibit the activity of its enzyme target and the growth of cancer cells.


Asunto(s)
Dendrímeros/química , Antagonistas del Ácido Fólico/toxicidad , Metotrexato/toxicidad , Nanoconjugados/química , Tetrahidrofolato Deshidrogenasa/química , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Antagonistas del Ácido Fólico/síntesis química , Antagonistas del Ácido Fólico/química , Humanos , Nanoconjugados/toxicidad , Neoplasias/tratamiento farmacológico , Fotólisis , Espectrofotometría Ultravioleta , Tetrahidrofolato Deshidrogenasa/metabolismo , Rayos Ultravioleta
13.
Bioorg Med Chem ; 18(22): 7773-85, 2010 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-20951595

RESUMEN

In the process of drug discovery the lead-identification phase may be critical due to the likely poor safety profile of the candidates, causing the delay or even the abandonment of a certain project. Nowadays, combining molecular modeling and in vivo cellular evaluation can help to identify compounds with an enhanced safety profile. Previously, two quinoxalines have been identified as inhibitors of the folate-dependent proteins belonging to the thymidylate synthase cycle. Unfortunately, cytotoxic activity against a panel of cisplatin(cDDP)-sensitive ovarian carcinoma cell lines and their resistant counterparts was coupled with toxicity to non-tumorigenic Vero cells. Here we describe the application of a ligand-based virtual screening, and several [1,2,4]triazolo[4,3-a]quinoxalines were optimized to improve their ADME-tox profile. The resulting 4-(trifluoromethyl)-1-p-tolyl-[1,2,4]triazolo[4,3-a]quinoxaline (24), which interferes intracellularly with DHFR and TS reducing the protein levels like 5-FU, but without inducing TS ternary complex formation, was 2-times less toxic in vitro than cisplatin and 5-FU.


Asunto(s)
Antagonistas del Ácido Fólico/química , Ácido Fólico/metabolismo , Ligandos , Quinoxalinas/química , Triazoles/química , Línea Celular Tumoral , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacocinética , Inhibidores Enzimáticos/toxicidad , Ácido Fólico/química , Antagonistas del Ácido Fólico/farmacocinética , Antagonistas del Ácido Fólico/toxicidad , Humanos , Quinoxalinas/farmacocinética , Quinoxalinas/toxicidad , Tetrahidrofolato Deshidrogenasa/química , Tetrahidrofolato Deshidrogenasa/metabolismo , Timidilato Sintasa/antagonistas & inhibidores , Timidilato Sintasa/metabolismo , Triazoles/farmacocinética , Triazoles/toxicidad
14.
Food Chem Toxicol ; 48(7): 1973-9, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20451574

RESUMEN

Methotrexate (MTX) is a folic acid antagonist widely used as a cytotoxic chemotherapeutic agent for leukemia and other malignancies. The purpose of this study was to investigate the damage caused by MTX on liver mitochondria and its protection by using antioxidant properties of lipoic acid. MTX substantially affects mitochondrial function by reducing glutathione levels leading to disturbances in antioxidant enzyme defense system. Lipoic acid occurs naturally in mitochondria as a coenzyme. In various studies lipoic acid has been convincingly shown to exhibit an antioxidant role when supplemented exogenously. We studied the effect of lipoic acid pre-treatment on the toxicity of MTX in mouse liver mitochondria focusing specifically on the oxidative stress. MTX caused a significant rise in the mitochondrial lipid peroxidation (LPO), protein carbonyl (PC) content and superoxide radical generation. It also affected the mitochondrial thiol profile. Pre-treatment of mice with lipoic acid (35 mg/kg) markedly lowered mitochondrial LPO, PC content and superoxide radical generation. It also restored decreased enzymatic and non-enzymatic antioxidants of mitochondria. It is suggested that lipoic acid has a potential role in suppressing MTX-induced mitochondrial toxicity, and it affords protection either by reversing the decline of antioxidants or by the directly scavenging the free radicals.


Asunto(s)
Antagonistas del Ácido Fólico/toxicidad , Metotrexato/antagonistas & inhibidores , Metotrexato/toxicidad , Mitocondrias Hepáticas/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Ácido Tióctico/farmacología , Animales , Peso Corporal/efectos de los fármacos , ADN/metabolismo , Radicales Libres/metabolismo , Glutatión/metabolismo , Glutatión Peroxidasa/metabolismo , Técnicas In Vitro , Metabolismo de los Lípidos/efectos de los fármacos , Ratones , Oxidación-Reducción , Carbonilación Proteica/efectos de los fármacos , Compuestos de Sulfhidrilo/metabolismo , Superóxido Dismutasa/metabolismo , Superóxidos/metabolismo , Sustancias Reactivas al Ácido Tiobarbitúrico/metabolismo
15.
Pharmacol Biochem Behav ; 95(4): 428-33, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20332001

RESUMEN

Although most children with acute lymphoblastic leukemia (ALL) can be cured, a significant subset of survivors manifests focal deficits in cognitive function, even when the treatment regimen does not include cranial radiation. Intrathecal administration of the folate antagonist methotrexate (MTX) is necessary to prevent leukemic relapse within the central nervous system, but is suspected to contribute to treatment-induced cognitive dysfunction. To better elucidate the underlying pathophysiology, we sought to establish a rodent model of the cognitive and neurotoxic effects resulting from direct administration of MTX into the cerebrospinal fluid (CSF). MTX or artificial CSF was injected via transcutaneous puncture at the level of the cisterna magna. Subsequent behavioral tests were designed to assess cognitive domains frequently impaired among children treated for ALL. MTX administration produced both recognition and spatial memory deficits, without altering general activity or motor coordination. In addition, MTX significantly reduced folate levels in both CSF and serum and increased CSF homocysteine. Thus, we have established an animal model that mimics the clinical effects of prophylactic intrathecal MTX on cognitive function. Using this model we can further study the pathophysiology of MTX-induced cognitive dysfunction and test protective interventions.


Asunto(s)
Antimetabolitos Antineoplásicos/administración & dosificación , Antimetabolitos Antineoplásicos/toxicidad , Trastornos del Conocimiento/inducido químicamente , Homocisteína/líquido cefalorraquídeo , Metotrexato/administración & dosificación , Metotrexato/toxicidad , Animales , Conducta Animal/efectos de los fármacos , Cisterna Magna , Trastornos del Conocimiento/sangre , Trastornos del Conocimiento/líquido cefalorraquídeo , Trastornos del Conocimiento/fisiopatología , Modelos Animales de Enfermedad , Ácido Fólico/sangre , Ácido Fólico/líquido cefalorraquídeo , Antagonistas del Ácido Fólico/administración & dosificación , Antagonistas del Ácido Fólico/toxicidad , Mediadores de Inflamación/líquido cefalorraquídeo , Inyecciones Espinales , Masculino , Trastornos de la Memoria/sangre , Trastornos de la Memoria/líquido cefalorraquídeo , Trastornos de la Memoria/inducido químicamente , Actividad Motora/efectos de los fármacos , Pruebas Neuropsicológicas , Desempeño Psicomotor/efectos de los fármacos , Ratas , Ratas Long-Evans , Reconocimiento en Psicología/efectos de los fármacos , Factores de Tiempo
16.
Clin Cancer Res ; 15(9): 3084-93, 2009 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-19383815

RESUMEN

PURPOSE: ABCC2 (MRP2) and ABCG2 (BCRP) transport various endogenous and exogenous compounds, including many anticancer drugs, into bile, feces, and urine. We investigated the possibly overlapping roles of Abcg2 and Abcc2 in the elimination of the anticancer drug methotrexate (MTX) and its toxic metabolite 7-hydroxymethotrexate (7OH-MTX). EXPERIMENTAL DESIGN: We generated and characterized Abcc2;Abcg2(-/-) mice, and used these to determine the overlapping roles of Abcc2 and Abcg2 in the elimination of MTX and 7OH-MTX after i.v. administration of 50 mg/kg MTX. RESULTS: Compared with wild-type, the plasma areas under the curve (AUC) for MTX were 1.6-fold and 2.0-fold higher in Abcg2(-/-) and Abcc2(-/-) mice, respectively, and 3.3-fold increased in Abcc2;Abcg2(-/-) mice. The biliary excretion of MTX was 23-fold reduced in Abcc2;Abcg2(-/-) mice, and the MTX levels in the small intestine were dramatically decreased. Plasma levels of 7OH-MTX were not significantly altered in Abcg2(-/-) mice, but the areas under the curve were 6.2-fold and even 12.4-fold increased in Abcc2(-/-) and Abcc2;Abcg2(-/-) mice, respectively. This indicates that Abcc2 compensates for Abcg2 deficiency but that Abcg2 can only partly compensate for Abcc2 absence. Furthermore, 21-fold decreased biliary 7OH-MTX excretion in Abcc2;Abcg2(-/-) mice and substantial 7OH-MTX accumulation in the liver and kidney were seen. We additionally found that in the absence of Abcc2, Abcg2 mediated substantial urinary excretion of MTX and 7OH-MTX. CONCLUSIONS: Abcc2 and Abcg2 together are major determinants of MTX and 7OH-MTX pharmacokinetics. Variations in ABCC2 and/or ABCG2 activity due to polymorphisms or coadministered inhibitors may therefore substantially affect the therapeutic efficacy and toxicity in patients treated with MTX.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/fisiología , Sistema Biliar/metabolismo , Hígado/metabolismo , Metotrexato/análogos & derivados , Metotrexato/farmacocinética , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/fisiología , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Transportadoras de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Área Bajo la Curva , Western Blotting , Cromatografía Líquida de Alta Presión , Heces/química , Femenino , Antagonistas del Ácido Fólico/farmacocinética , Antagonistas del Ácido Fólico/toxicidad , Antagonistas del Ácido Fólico/orina , Masculino , Metotrexato/toxicidad , Metotrexato/orina , Ratones , Ratones Noqueados , Proteína 2 Asociada a Resistencia a Múltiples Medicamentos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Distribución Tisular
17.
Food Chem Toxicol ; 46(9): 3015-22, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18601970

RESUMEN

Several studies have well confirmed the contribution of oxidative stress in the pathogenesis of methotrexate (MTX)-induced damage in the small intestine. Many agents have been tried experimentally to reduce or inhibit the oxidative stress. To our knowledge, there is no study about apricot consumption on the MTX-induced damage in the small intestine. The aim of this study was to determine the possible protective effects of apricot and beta-carotene on MTX-induced intestinal damage in rats. The rats were randomly divided into seven groups as follows; I-control group; II-apricot group; III-beta-carotene group; IV-MTX group; V-apricot+MTX group; VI-beta-carotene+MTX group and VII-apricot+beta-carotene+MTX group. In the MTX group; fusion and shortening in the villus, epithelial desquamation, crypt loss, inflammatory cell infiltration in the lamina propria, goblet cell depletion and microvillar damage were observed in the small intestine. Parallel to histological results, malondialdehyde (MDA) content and myeloperoxidase (MPO) activity were found to be increased, whereas superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GP-x) activities and glutathione (GSH) content were decreased in the MTX group. However, single or combined application of apricot and beta-carotene ameliorated all of these hazardous effects in antioxidant system in MTX-treated groups. In conclusion, our results demonstrate that apricot and/or beta-carotene treatment may protect the impairment of oxidative stress and ameliorate MTX-induced intestine damage at biochemical and histological levels.


Asunto(s)
Antagonistas del Ácido Fólico/toxicidad , Intestinos/patología , Metotrexato/toxicidad , Estrés Oxidativo/efectos de los fármacos , Sustancias Protectoras , Prunus/química , beta Caroteno/farmacología , Animales , Compuestos de Bifenilo , Catalasa/metabolismo , Depuradores de Radicales Libres/farmacología , Glutatión/metabolismo , Glutatión Peroxidasa/metabolismo , Intestinos/efectos de los fármacos , Peroxidación de Lípido/efectos de los fármacos , Masculino , Microscopía Electrónica , Peroxidasa/metabolismo , Fenoles/farmacología , Picratos , Extractos Vegetales/farmacología , Ratas , Ratas Wistar , Superóxido Dismutasa/metabolismo
18.
Anticancer Res ; 27(2): 825-33, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17465208

RESUMEN

BACKGROUND: Pemetrexed (Alimta) is a new-generation multitargeted antifolate that inhibits several key enzymes in the de novo pathways of pyrimidine and purine biosynthesis, including thymidylate synthase (TS), dihydrofolate reductase (DHFR) and glycinamide ribonucleotide formyltransferase (GARFT). Alimta has demonstrated antitumor activity in a broad array of human malignancies, e.g. breast, non-small cell lung cancer, malignant pleural mesothelioma and pancreatic, colorectal, gastric, bladder, head and neck cancer, and is currently in phase III clinical trials. It has been reported that a dose of 600 mg/m2 of pemetrexed showed toxicity to bone marrow and the gastrointestinal system. The aim of this investigation was to evaluate raloxifene (RAL) in combination with 5-fluorouracil (5-FU)/pemetrered multitargeted antifolate (MTA) to determine the most effective regimens and cellular mechanism of action to mitigate pemetrexed cytotoxicity in human bone marrow cells. MATERIALS AND METHODS: In order to determine the sequence-dependent interaction between MTA, 5-FU and RAL on proliferation, cell viability was carried out using the Quick Cell Proliferation Assay by exposing the HS-5 and MCF-7 cells to (i) MTA, 5-FU and RAL alone, or (ii) RAL 24 h prior to 5-FU followed 2 h later by MTA, or (iii) 5-FU 2 h prior to MTA followed 24 h later by RAL. RESULTS: The growth rate in MCF-7 in early RAL was 69 +/- 8.65% and late RAL was 36 +/- 4.6% of the control whereas in bone marrow early RAL was 78 +/- 8.65% and late RAL was 52 +/- 5.49% of the control. The late RAL exhibits significant protection against MTA cytotoxicity in bone marrow. The findings were further supported by cell flow cytometry, apoptosis and Western blot analysis data. CONCLUSION: This study suggests that sequence-dependent administration of RAL (5FU/MTA/RAL), in combination with 5-FU/MTA, protects against MTA toxicity in human bone marrow while maintaining the maximum inhibitory effect of pemetrexed in breast cancer.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Células de la Médula Ósea/efectos de los fármacos , Enfermedades de la Médula Ósea/inducido químicamente , Antagonistas del Ácido Fólico/toxicidad , Glutamatos/toxicidad , Guanina/análogos & derivados , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/toxicidad , Enfermedades de la Médula Ósea/prevención & control , Neoplasias de la Mama/tratamiento farmacológico , Procesos de Crecimiento Celular/efectos de los fármacos , Línea Celular Tumoral , Esquema de Medicación , Fluorouracilo/administración & dosificación , Fluorouracilo/farmacología , Antagonistas del Ácido Fólico/administración & dosificación , Antagonistas del Ácido Fólico/farmacología , Glutamatos/administración & dosificación , Glutamatos/farmacología , Guanina/administración & dosificación , Guanina/farmacología , Guanina/toxicidad , Humanos , Pemetrexed , Clorhidrato de Raloxifeno/administración & dosificación , Clorhidrato de Raloxifeno/farmacología
19.
Zhongguo Dang Dai Er Ke Za Zhi ; 9(2): 159-63, 2007 Apr.
Artículo en Chino | MEDLINE | ID: mdl-17448316

RESUMEN

OBJECTIVE: To study the effect of methotrexate (MTX), a folic acid antagonist which can lead to folic acid deficient, on the cardiac development and on the expressions of BMP2b and HAS2 in zebrafish. METHODS: The zebrafish embryos at 6-48 hrs post fertilization (hpf) were treated with various concentrations of MTX (0.5 x 10(-3), 1.0 x 10(-3) and 2.0 x 10(-3) M). At 48 hpf, the percentage of cardiac malformation and heart rate were recorded. The zebrafish embryos at 6-10 hpf treated with 1.5 x 10(-3) M MTX were used as the MTX treatment group. At 24 and 48 hpf the cardiac morphology was observed under a microscope. The expressions of BMP2b and HAS2 in zebrafish were detected by in situ antisense RNA hybridization and real-time PCR. RESULTS: 6-12 hpf, the early embryonic developmental stage, was a sensitive period that MTX affected cardiac formation of zebrafish. The retardant cardiac development and the evidently abnormal cardiac morphology was found in the MTX treatment group. The results of in situ antisense RNA hybridization showed that the expressions of BMP2b and HAS2 in the zebrafish heart were reduced in the MTX treatment group at 36 and 48 hpf. The real-time PCR results demonstrated that the BMP2b expression decreased at 12, 24, 36 and 48 hpf, and that the HAS2 expression decreased at 24, 36 and 48 hpf in the treatment group compared with the control group without MTX treatment. CONCLUSIONS: The inhibition of folic acid function may affect cardiac development of early embryos, resulting in a retardant development and a morphological abnormality of the heart in zebrafish, possibly by down-regulating the expressions of BMP2b and HAS2.


Asunto(s)
Anomalías Inducidas por Medicamentos/etiología , Proteínas Morfogenéticas Óseas/genética , Antagonistas del Ácido Fólico/toxicidad , Regulación de la Expresión Génica/efectos de los fármacos , Glucuronosiltransferasa/genética , Cardiopatías Congénitas/inducido químicamente , Metotrexato/toxicidad , Proteínas de Pez Cebra/genética , Animales , Proteína Morfogenética Ósea 2 , Regulación hacia Abajo , Hialuronano Sintasas , Reacción en Cadena de la Polimerasa , Pez Cebra
20.
Biochem Soc Trans ; 35(Pt 2): 386-90, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17371283

RESUMEN

Polyamines play an important role in cell growth and differentiation, while their overproduction has potentially oncogenic consequences. Polyamine homoeostasis, a critical determinant of cell fate, is precisely tuned at the level of biosynthesis, degradation and transport. The enzymes ODC (ornithine decarboxylase), AdoMetDC (S-adenosylmethionine decarboxylase) and SSAT (spermidine/spermine N(1)-acetyltransferase) are critical for polyamine pool maintenance. Our experiments were designed to examine the expression of these enzymes in testosterone-induced hypertrophic and antifolate-induced hyperplastic mouse kidney, characterized by activation of AR (androgen receptor) and HGF (hepatocyte growth factor) membrane receptor c-Met respectively. The expression of these key enzymes was up-regulated by antifolate CB 3717 injury-evoked activation of HGF/c-Met signalling. In contrast, activation of the testosterone/AR pathway remarkably induced a selective increase in ODC expression without affecting other enzymes. Studies in catecholamine-depleted kidneys point to a synergistic interaction between the signalling pathways activated via cell membrane catecholamine receptors and AR, as well as c-Met. We found that this cross-talk modulated the expression of ODC and AdoMetDC, enzymes limiting polyamine biosynthesis, but not SSAT. This is in contrast with the antagonistic cross-talk between AR- and c-Met-mediated signalling which negatively regulated the expression of ODC, but affected neither AdoMetDC nor SSAT.


Asunto(s)
Riñón/fisiología , Poliaminas/metabolismo , Receptores Citoplasmáticos y Nucleares/fisiología , Transducción de Señal/fisiología , Testosterona/farmacología , Adenosilmetionina Descarboxilasa/metabolismo , Animales , Ácido Fólico/análogos & derivados , Ácido Fólico/toxicidad , Antagonistas del Ácido Fólico/toxicidad , Factor de Crecimiento de Hepatocito/genética , Factor de Crecimiento de Hepatocito/fisiología , Hipertrofia , Riñón/efectos de los fármacos , Riñón/patología , Ratones , Ornitina Descarboxilasa/genética , Quinazolinas/toxicidad , ARN Mensajero/genética , Transducción de Señal/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA