Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros











Intervalo de año de publicación
1.
Front Pharmacol ; 13: 1011065, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36172182

RESUMEN

Iron oxide nanoparticles (IONPs) are the first generation of nanomaterials approved by the Food and Drug Administration for use as imaging agents and for the treatment of iron deficiency in chronic kidney disease. However, several IONPs-based imaging agents have been withdrawn because of toxic effects and the poor understanding of the underlying mechanisms. This study aimed to evaluate IONPs toxicity and to elucidate the underlying mechanism after intravenous administration in rats. Seven-week-old rats were intravenously administered IONPs at doses of 0, 10, 30, and 90 mg/kg body weight for 14 consecutive days. Toxicity and molecular perturbations were evaluated using traditional toxicological assessment methods and proteomics approaches, respectively. The administration of 90 mg/kg IONPs induced mild toxic effects, including abnormal clinical signs, lower body weight gain, changes in serum biochemical and hematological parameters, and increased organ coefficients in the spleen, liver, heart, and kidneys. Toxicokinetics, tissue distribution, histopathological, and transmission electron microscopy analyses revealed that the spleen was the primary organ for IONPs elimination from the systemic circulation and that the macrophage lysosomes were the main organelles of IONPs accumulation after intravenous administration. We identified 197 upregulated and 75 downregulated proteins in the spleen following IONPs administration by proteomics. Mechanically, the AKT/mTOR/TFEB signaling pathway facilitated autophagy and lysosomal activation in splenic macrophages. This is the first study to elucidate the mechanism of IONPs toxicity by combining proteomics with traditional methods for toxicity assessment.

2.
Toxicol Appl Pharmacol ; 284(2): 134-41, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25727309

RESUMEN

OBJECTIVE: To characterize the mechanism of action of thiazolidinedione (TZD)-induced liver mitochondrial toxicity caused by troglitazone, rosiglitazone, and pioglitazone in HepaRG cells. METHODS: Human hepatoma cells (HepaRG) were treated with troglitazone, rosiglitazone, or pioglitazone (12.5, 25, and 50µM) for 48h. The Seahorse Biosciences XF24 Flux Analyzer was used to measure mitochondrial oxygen consumption. The effect of TZDs on reactive oxygen species (ROS) and mitochondrial membrane potential (MMP) were detected by flow cytometry. The mitochondrial ultrastructure of HepaRG cells was observed under a transmission electrical microscope (TEM). mtDNA content was evaluated by real-time PCR, and ATP content and mitochondrial respiratory chain (MRC) complex I, II, III, IV activity were measured via chemiluminescence. Results were considered statistically significant at p<0.05. RESULTS: Among the three drugs, troglitazone exhibited the highest potency, followed by rosiglitazone, and then pioglitazone. The TZDs caused varying degrees of mitochondrial respiratory function disorders including decreases in oxygen consumption, MRC activity, and ATP level, and an elevation in ROS level. TZD treatment resulted in mtDNA content decline, reduction in MMP, and alterations of mitochondrial structure. CONCLUSION: All investigated TZDs show a certain degree of mitochondrial toxicity, with troglitazone exhibiting the highest potency. The underlying mechanism of TZD-induced hepatotoxicity may be associated with alterations in mitochondrial respiratory function disorders, oxidative stress, and changes in membrane permeability. These parameters may be used early in drug development to further optimize risk:benefit profiles.


Asunto(s)
Hígado/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Tiazolidinedionas/toxicidad , Línea Celular Tumoral , Cromanos/toxicidad , ADN Mitocondrial/genética , Transporte de Electrón/efectos de los fármacos , Humanos , Hipoglucemiantes/toxicidad , Hígado/metabolismo , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias/metabolismo , Estrés Oxidativo/efectos de los fármacos , Pioglitazona , Especies Reactivas de Oxígeno/metabolismo , Rosiglitazona , Troglitazona
3.
Biomed Chromatogr ; 29(8): 1259-66, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25616210

RESUMEN

Meditinib (ME) is a novel tyrosine kinase inhibitor used as an antichronic myeloid leukemia drug. A simple, sensitive and specific LC/MS/MS method was developed and validated for the analysis of ME and its metabolite demethylation meditinib (PI) in monkey plasma using naltrexone as the internal standard. Sample preparation involved protein precipitation with methanol. The analysis was carried out on an Agilent C8 column (3.5 µm, 2.1 × 50 mm). Elution was achieved with a mobile phase gradient varying the proportion of a water solution containing 0.1% formic acid (solvent A) and a 0.1% formic acid in methanol solution (solvent B) at a flow rate of 300 µL/min. The method had a linear calibration curve over the concentration range of 2-1000 ng/mL for ME and 2-1000 ng/mL for PI. The lower limits of quantification of ME and PI were 2 and 2 ng/mL, respectively. The intra- and inter-day precision values were <15% and accuracy values were within ±10.0%. The mean recoveries of ME and PI from plasma were >85%. The assay has been successfully used for pharmacokinetic evaluation of ME and PI using the monkey as an animal model, and those data are reported for the first time.


Asunto(s)
Antineoplásicos/sangre , Haplorrinos/sangre , Inhibidores de Proteínas Quinasas/sangre , Espectrometría de Masas en Tándem/métodos , Animales , Antineoplásicos/metabolismo , Femenino , Leucemia Mielógena Crónica BCR-ABL Positiva/sangre , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Límite de Detección , Masculino , Inhibidores de Proteínas Quinasas/metabolismo
4.
J Occup Health ; 53(4): 250-7, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21670563

RESUMEN

OBJECTIVES: To investigate the injury of cell tight junctions and change in actin level in the alveolus epithelial cells of the lung after perfluoroisobutylene (PFIB) exposure and the role of myosin light chain kinase (MLCK) in the injury. METHODS: Rats and mice were exposed to a sublethal dose of PFIB. The changes in tight junction zonula occludens-1 (ZO-1), actin and myosin light chain kinase (MLCK) were detected by immunofluorescence at 30 min, 1, 2, 4, 8, 16, 24, 48 and 72 h after PFIB exposure. The role of MLCK was analyzed by lung indices and the actin level. RESULTS: The normal ZO-1 immunofluorescence density and those after PFIB exposure were 71.63, 39.41, 37.59, 35.71, 33.22, 31.34, 31.61, 24.51, 40.03 and 44.71 respectively, The normal actin immunofluorescence density and those after PFIB exposure were 31.82, 36.46, 36.57, 41.60, 40.95, 35.41, 30.69, 19.96, 29.30 and 33.00 respectively, The normal MLCK immunofluorescence density and those after PFIB exposure were 61.21, 50.87, 48.37, 43.65, 41.96, 35.44, 31.77, 30.85, 33.10 and 38.20 respectively. When the MLCK inhibitor ML-7 was given in advance, pulmonary edema and actin degradation were suppressed. CONCLUSIONS: At an earlier stage, the increased permeability of the blood-air barrier after PFIB exposure is probably the result of injury of cell tight junctions that acts in concert with later changes in actin, resulting in an increase in permeability. MLCK could be a potential target for novel drug development for relief of acute lung injury.


Asunto(s)
Actinas/metabolismo , Lesión Pulmonar Aguda/metabolismo , Lesión Pulmonar Aguda/patología , Fluorocarburos/toxicidad , Quinasa de Cadena Ligera de Miosina/metabolismo , Uniones Estrechas/metabolismo , Actinas/efectos de los fármacos , Lesión Pulmonar Aguda/inducido químicamente , Análisis de Varianza , Animales , Barrera Alveolocapilar/efectos de los fármacos , Barrera Alveolocapilar/metabolismo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Quinasa de Cadena Ligera de Miosina/antagonistas & inhibidores , Tamaño de los Órganos , Fosfoproteínas/metabolismo , Edema Pulmonar/inducido químicamente , Edema Pulmonar/patología , Ratas , Ratas Wistar , Uniones Estrechas/efectos de los fármacos , Factores de Tiempo , Proteína de la Zonula Occludens-1
5.
J Occup Health ; 52(1): 48-57, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20035103

RESUMEN

OBJECTIVES: To investigate the complete process of cell injuries in the blood-air barrier after perfluoroisobutylene (PFIB) exposure. METHODS: Rats were exposed to PFIB (140 mg/m(3)) for 5 min. The pathological changes were evaluated by lung wet-to-dry weight ratio, total protein concentration of bronchoalveolar lavage fluid and HE stain. Ultrastructural changes were observed by transmission electron microscope. Apoptosis was detected by in situ apoptosis detection. Changes of actin in the lung tissue were evaluated by western blot assay. RESULTS: No significant pulmonary edema or increased permeability was observed within the first 4 h, post PFIB exposure. However, inflammatory cell infiltration and alveolar wall thickening were observed from 2 h. Destruction of the alveoli constitution integrity, edema and protein leakage were observed at 8 h. The injuries culminated at 24 h and then recovered gradually. The ultrastructural injuries of alveolar type I epithelial cells, alveolar type II epithelial cells and pulmonary microvascular endothelial cells were observed at 30 min post PFIB exposure. Some injuries were similar to apoptosis. Compared with control, more serious injuries were observed in PFIB-exposed rats after 30 min. At 8 h, some signs of cell necrosis were observed. The injuries culminated at 24 h and then ameliorated. The number of apoptotic cells abnormally increased at 30 min post PFIB exposure, the maximum appeared at 24 h, and then ameliorated gradually. Western blot analysis revealed that the level of actin in the lung showed no significant changes within the first 4 h post PFIB exposure. However, it decreased at 8 h, reached a nadir at 24 h, and then recovered gradually. CONCLUSIONS: The pathological processes were in progress persistently post PFIB exposure. The early injuries probably were the result of the direct attack of PFIB and the advanced injuries probably arose from the inflammatory reaction induced by PFIB.


Asunto(s)
Lesión Pulmonar Aguda/inducido químicamente , Apoptosis , Células Epiteliales/efectos de los fármacos , Fluorocarburos/farmacocinética , Fluorocarburos/toxicidad , Actinas/efectos de los fármacos , Enfermedad Aguda , Administración por Inhalación , Animales , Barrera Alveolocapilar/efectos de los fármacos , Barrera Alveolocapilar/ultraestructura , Líquido del Lavado Bronquioalveolar , Células Epiteliales/ultraestructura , Fluorocarburos/administración & dosificación , Masculino , Ratas , Ratas Wistar
6.
Zhongguo Wei Zhong Bing Ji Jiu Yi Xue ; 21(4): 219-21, 2009 Apr.
Artículo en Chino | MEDLINE | ID: mdl-19374789

RESUMEN

OBJECTIVE: To observe the protective effect of insulin-like growth factor-1 (IGF-1) on acute lung injury induced by perfluoroisobutylene (PFIB) inhalation in mice. METHODS: Sixty-four male Kunming mice were randomly divided into normal control (A) group, exposed (B) group, recombinant adenoviruses 5 of IGF-1 (Ad5-IGF-1) intervention (C) group (in which Ad5-IGF-1 was injected into the trachea of the mice), blank vector control (D) group. B, C and D groups were exposed to gaseous PFIB in a flow-past whole-body exposure system. The lung index, concentration of total protein and albumin in bronchoalveolar lavage fluid (BALF), concentration of IGF-1 in serum and lung homogenate were measured. The lung pathologic changes were examined with light microscope, and ultrastructure changes in alveolar type II cells (ATII) with electron microscope. RESULTS: Compared with A group, the lung index, concentration of total protein in BALF were significantly increased in other groups, the lung index and concentration of total protein and albumin of BALF in B and D groups were prominently higher than C group (all P<0.01). The concentration of IGF-1 in serum of B and D groups was lower markedly than that of A group, and the concentration of IGF-1 in serum of C group was distinctly higher than those of A, B, D groups (all P<0.01). The concentration of IGF-1 in lung homogenate of B, C, D groups was higher than that of A group, and the concentration of IGF-1 in lung homogenate of C group was significantly higher than that of B and D groups (all P<0.01). Lung hyaline membrane formation, diffuse alveolar atelectasis, accumulation of edema fluid, red blood cell exudation, were obviously milder in C group, and changes in the ultrastructure of ATII showed a similar result. CONCLUSION: The protective effect of Ad5-IGF-1 against the toxicity of PFIB inhalation is identified. In the mice pretreated with Ad5-IGF-1 is able to significantly lower lung index, the protein concentration in BALF, and the concentration of IGF-1 in serum and lung homogenate is obviously increased. Protection of ATII may be one of the mechanisms.


Asunto(s)
Lesión Pulmonar Aguda/prevención & control , Fluorocarburos/toxicidad , Factor I del Crecimiento Similar a la Insulina/genética , Lesión Pulmonar Aguda/inducido químicamente , Adenoviridae/genética , Animales , Líquido del Lavado Bronquioalveolar/química , Modelos Animales de Enfermedad , Células Epiteliales/patología , Vectores Genéticos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Pulmón/metabolismo , Pulmón/patología , Masculino , Ratones , Alveolos Pulmonares/patología
7.
Artículo en Chino | MEDLINE | ID: mdl-15130444

RESUMEN

OBJECTIVE: To establish of acute respiratory distress syndrome (ARDS) model in canine after inhalation of perfluoroisobutylene (PFIB), and to observe the progressing of lung injury, and to study the mechanisms of injury. METHODS: A device of inhalation of PFIB for canine was made. The concentration of PFIB was 0.30 - 0.32 mg/L. Serum IL-6 and IL-8 were dynamically measured. Clinical manifestations, pathology of organs in canine were observed. RESULTS: (1) During inhalation, the concentration of PFIB remained stable; (2) After inhalation, blood arterial oxygen partial pressure fell gradually, and eventually met the criteria for diagnosing ARDS; (3) The level of IL-8 in serum rises significantly after inhalation (P < 0.05), whereas that of IL-6 was not obviously altered (P > 0.05); (4) Within 6 hours after inhalation, no abnormality in canine was observed, but afterwards symptoms gradually appeared, and typical breath of ARDS, such as high frequency and lower level could be seen in later phase; (5) Pathological examination showed severe congestion, edema and atelectasis in most part of both lungs, and signs of anoxia in other organs. CONCLUSIONS: (1) The device designed is capable of ensuring control of inhalation of PFIB; (2) Exposure to PFIB for 30 mins, canines all met the criteria for diagnosing ARDS 22 hours after inhalation, therefore the modeling is successful; (3) PFIB specifically damages the lung by causing excessive inflammation.


Asunto(s)
Fluorocarburos/toxicidad , Pulmón/patología , Síndrome de Dificultad Respiratoria/inducido químicamente , Administración por Inhalación , Animales , Modelos Animales de Enfermedad , Perros , Femenino , Interleucina-6/sangre , Interleucina-8/sangre , Pulmón/efectos de los fármacos , Masculino , Distribución Aleatoria , Síndrome de Dificultad Respiratoria/sangre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA