Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Int J Mol Sci ; 23(21)2022 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-36361734

RESUMEN

The objective of the present study was to define whether inhaled tetrandrine (TET) could be a promising way to achieve the local effect on its therapeutic efficacy based on biodistribution features using the LPS-treated acute lung injury (ALI) model. The tissue distribution profiles of inhaled TET in normal and ALI mouse models showed that pulmonary inflammation led to an altered distribution in a tissue-specific way. More TET accumulated in almost all tissues including in the blood. Among them, the increased exposure in the lungs was significantly higher than in the other tissues. However, there was a negative increase in the brain. In vitro turnover rates of TET in mouse liver microsomes (MLM) from normal and LPS-treated mice showed significant differences. In the presence of NADPH, TET demonstrated relatively low hepatic clearance (89 mL/h/kg) in that of normal MLM (140 mL/h/kg). Intracellular uptakes of TET in A549, HepG2, RAW264.7, and C8-D1A cells were significantly inhibited by monensin, indicating that the intracellular accumulation of TET is driven by lysosomal trapping. However, in the presence of LPS, only the lysosomal pH partitioning of TET in A549 cell lines increased (~30%). Bidirectional transport of TET across LLC-PK1 cell expressing MDR1 showed that MDR1 is responsible for the low brain exposure via effluxion (ER = 32.46). From the observed overall agreement between the in vitro and in vivo results, we concluded that the downregulation of the CYP3A together with strengthened pulmometry lysosomal trapping magnified the retention of inhaled TET in the lung. These results therefore open the possibility of prolonging the duration of the local anti-inflammation effect against respiratory disorders.


Asunto(s)
Lesión Pulmonar Aguda , Bencilisoquinolinas , Neumonía , Animales , Ratones , Lipopolisacáridos/toxicidad , Distribución Tisular , Bencilisoquinolinas/farmacología , Bencilisoquinolinas/uso terapéutico , Lisosomas , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/tratamiento farmacológico , Neumonía/tratamiento farmacológico
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.
Exp Ther Med ; 25(2): 98, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36761001

RESUMEN

The present study aimed to compare the clinical effects of vitamin E and vitamin D on a rat model of dextran sulfate sodium (DSS)-induced ulcerative colitis (UC), and to elucidate the underlying mechanisms associated with changes in the levels of cytokines. After successful establishment of the rat model of DSS-induced UC, prednisolone (1 mg/kg), vitamin D (50 ng) and vitamin E (6, 30 and 150 IU/kg) were orally administered for 1 week. The pharmacodynamics were evaluated by a daily combination of clinical observation (CO) scores, histopathological evaluations and assessment of molecular markers of inflammation. Administration of vitamin D, vitamin E (30 and 150 IU/kg), prednisolone, and the combination of vitamin D and vitamin E resulted in a decrease in CO scores. The severity of inflammation of the colon was markedly alleviated in the treatment groups compared with that in the untreated DSS group according to the results of histopathological examination; however, they showed different inhibitory effects on the levels of some cytokines. In conclusion, the present results indicated that oral administration of vitamin E could promote recovery of DSS-induced UC by the inhibition of proinflammatory cytokines, and that its underlying mechanism may differ from that of vitamin D and glucocorticoid drugs.

5.
Toxics ; 11(3)2023 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-36976981

RESUMEN

Di-(2-Ethylhexyl) phthalate (DEHP) and bisphenol A (BPA) present significant environmental endocrine-disrupting chemical properties. Although studies have implied reproductive impairment from exposure to BPA and DEHP, no study to date has shown the effect and mechanism of hepatic function after gestational and lactational co-exposure to DEHP and BPA in offspring. A total of 36 perinatal rats were randomly divided into four groups, DEHP (600 mg/kg/day), BPA (80 mg/kg/day), DEHP combined with BPA (600 mg/kg/day + 80 mg/kg/day), and control. Notably, 11 chemical targets were screened after identifying eight substances associated with chemically-induced hepatic damage. Molecular docking simulations revealed a high-scoring combination of eight metabolic components and targets of the PI3K/AKT/FOXO1 signaling pathway. The DEHP and BPA combination disrupted hepatic steatosis, ultimately affecting systemic the glucose and the lipid metabolic homeostasis with significant toxicity. Mechanistically, co-exposure to DEHP and BPA causes liver dysfunction and hepatic insulin resistance via PI3K/AKT/FOXO1 pathway in offspring. This is the first study of the hepatic function and mechanism of co-exposure to DEHP and BPA that combines metabolomics, molecular docking, and traditional toxicity assessment methods.

6.
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.

7.
Zhongguo Wei Zhong Bing Ji Jiu Yi Xue ; 21(4): 219-21, 2009 Apr.
Artículo en Zh | 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
8.
J Occup Health ; 49(2): 95-103, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17429166

RESUMEN

Perfluoroisobutylene (PFIB) is produced as a main by-product in large quantities by the fluoropolymer industry. As a highly toxic compound, even the case of brief inhalation of PFIB can result in acute lung injury (ALI), pulmonary edema and even death. To test for any preventive or therapeutic effects of pyrrolidine dithiocarbamate (PDTC), a NF-kappaB activation inhibitor, against PFIB inhalation-induced ALI, mice were exposed in a flow-past exposure system to PFIB and the prophylactic and therapeutic effects of PDTC were studied. The inhibitory effects of PDTC on ALI, the activation of NF-kappaB, as well as the expression of cytokines (IL-1beta and IL-8) after PFIB exposure were evaluated. The results demonstrated that pretreatment with PDTC (120 mg/kg, 30 min before PFIB exposure) could significantly lower the lung coefficient (wet lung-to-body weight ratio, dry lung-to-body weight ratio, water content in the lung, and lung wet-to-dry weight ratio) and protein content in bronchoalveolar lavage fluid (BALF), but no effects of PDTC were found when PDTC was treated after PFIB inhalation, suggesting a preventative effect rather than a therapeutic effect of PDTC. Furthermore, the above preventative effects of PDTC (when given at 30 min before PFIB exposure) on PFIB-induced lung injury were achieved in a dose-dependent manner. In support of these preventive effects of PDTC, our toxicological studies demonstrated that PFIB-inhalation induced a quick activation of NF-kappaB (0.5 h post PFIB exposure) and expression of IL-1beta and IL-8 (0.5 h and 1 h post PFIB exposure, respectively). Pretreatment with PDTC (120 mg/kg, 30 min before PFIB exposure) resulted in a significant inhibitive effect on the activation of NF-kappaB (0.5 h post PFIB exposure) and expression of IL-1beta and IL-8 (1 h post PFIB exposure). The mortality, the extent of lung injury of the mice indexed by lung coefficients, the content of total protein and albumin in BALF, as well as the lung histopathologic changes, were dramatically alleviated in PFIB exposure after pretreatment with PDTC, clearly suggesting that PDTC has a prophylactic role against PFIB inhalation-induced ALI, and that NF-kappaB activation might play a central role in initiating an acute inflammatory response and in causing injury to the lungs after PFIB inhalation.


Asunto(s)
Antioxidantes/farmacología , Fluorocarburos/toxicidad , Enfermedades Pulmonares/inducido químicamente , Enfermedades Pulmonares/prevención & control , Pirrolidinas/farmacología , Tiocarbamatos/farmacología , Administración por Inhalación , Análisis de Varianza , Animales , Líquido del Lavado Bronquioalveolar/química , Fluorocarburos/administración & dosificación , Masculino , Ratones
9.
J Occup Health ; 47(4): 277-85, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16096351

RESUMEN

Perfluoroisobutylene (PFIB) is a kind of fluoro-olefin that is ten times more toxic than phosgene. The mechanisms of the acute lung injury (ALI) induced by PFIB inhalation remain unclear. To find possible pharmacological interventions, mice and rats were exposed to PFIB, and the prophylactic or therapeutic effects of 3-quinuclidinyl benzilate (QNB) and anisodamine were studied and confirmed. It was observed that the wet lung/body weight and the dry lung/body weight ratios at 24 h after PFIB exposure (130 mg/m(3) for 5 min) were significantly decreased when a single dose of QNB (5 mg/kg) was administered intraperitoneally either 30 min before exposure or 10 h after exposure. Anisodamine was without any prophylactic or therapeutic effects at single doses below 30 mg/kg. The effects of QNB against PFIB inhalation induced ALI were well evidenced by the significantly decreased mice mortality at 72 h, the total protein concentration in bronchoalveolar lavage fluid at 24 h after the PFIB exposure, as well as the ultrastructural observations. The analysis of the time courses of lung sulfhydryl concentration, myeloperoxidase (MPO) activity and hemorheology assay showed that the toxicity of PFIB may be due to consumption of lung protein sulfhydryl, influx of polymorphonuclear leukocytes (PMNs) into the lung, and increased peripheral blood viscosity at a low shear rate, all of which were partially blocked by QNB intervention except for PMN influx. The results suggest that cholinolytics might have prophylactic and therapeutic roles in PFIB inhalation induced ALI.


Asunto(s)
Antagonistas Colinérgicos/uso terapéutico , Fluorocarburos/toxicidad , Exposición por Inhalación , Síndrome de Dificultad Respiratoria/prevención & control , Animales , Peso Corporal/efectos de los fármacos , China , Antagonistas Colinérgicos/administración & dosificación , Pulmón/ultraestructura , Masculino , Ratones , Exposición Profesional , Tamaño de los Órganos/efectos de los fármacos , Quinuclidinil Bencilato/administración & dosificación , Quinuclidinil Bencilato/efectos adversos , Ratas , Ratas Wistar , Síndrome de Dificultad Respiratoria/tratamiento farmacológico , Síndrome de Dificultad Respiratoria/fisiopatología , Alcaloides Solanáceos/administración & dosificación , Alcaloides Solanáceos/efectos adversos
10.
Exp Toxicol Pathol ; 67(10): 533-8, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26388042

RESUMEN

Optimized lung preparation for detailed structural evaluation is required to improve consistency in preclinical safety evaluation, differences of opinion exist among regulatory agency personnel regarding the optimal methods for routine formalin fixation of lungs from rodent toxicology studies. The simple tracheal ligation fixation method emphasizes tracheal ligation before opening the thorax instead of attempting to re-inflate after lung collapse when opening the thorax. Photomicrographs of this method demonstrated an unprecedented ability to maintain the natural lung architecture, in contrast to the unavoidable changes in the alveolar environment by the intratracheal instillation and vascular perfusion methods. In addition, a comparison of fixation methods on lung morphology in a rodent model of LPS-induced acute lung injury demonstrated that the tracheal ligation fixation method may provide a standard approach for morphometry. Additionally, a TUNEL assay was used to determine the degree of autolysis, which revealed that the autolysis was insignificant in the central areas of each lobe of the lung compared to the lung periphery by tracheal ligation fixation. In conclusion, our novel modified method, which avoids the disadvantages of generating artifacts, fulfills the requirement of preserving the clear, natural morphology of the lung making it suitable and worthy of recommendation for toxicological studies in a good laboratory practice (GLP) lab.


Asunto(s)
Pulmón , Patología/métodos , Fijación del Tejido/métodos , Toxicología/métodos , Animales , Apoptosis , Artefactos , Formaldehído , Etiquetado Corte-Fin in Situ , Ligadura , Masculino , Ratas , Ratas Sprague-Dawley , Tráquea
11.
Nanotoxicology ; 9(4): 513-22, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25119417

RESUMEN

After exposing rat embryonic cells to 20 µg/mL of silver nanoparticle (NP) suspension and their released ions for different time periods, silver nanoparticles were found in cellular nuclei, mitochondria, cytoplasm and lysosomes by transmission electron microscopy (TEM). We also observed mitochondrial destruction, distension of endoplasmic reticulum and apoptotic bodies. Global gene expression analysis showed a total of 279 genes that were up-regulated and 389 genes that were down-regulated in the silver-NP suspension exposure group, while 3 genes were up-regulated and 41 genes were down-regulated in the silver ion exposure group. Further, the GO pathway analysis suggested that these differentially expressed genes are involved in several biological processes, such as energy metabolism, oxygen transport, enzyme activities, molecular binding, etc. It is possible that inhibition of oxygen transport is mediated by the significant down-regulation of genes of the globin family, which might play an important role in silver ion-induced toxicity. KEGG pathway analysis showed that there were 23 signal pathways that were affected in the cells after exposure to silver-NP suspension, but not silver ion alone. The most significant change concerned inflammatory signal pathways, which were only found in silver-NP suspension exposed cells, indicating that inflammatory response might play an important role in the mechanism(s) of silver-NP-induced toxicity. The significant up-regulation of matrix metalloproteinases 3 and 9 suggests that silver NPs could induce extracellular matrix degradation via an inflammatory signaling pathway. The significant up-regulation of secretory leukocyte peptidase inhibitor and serine protease inhibitor 2c was considered to be an embryonic cellular defense mechanism in response to silver-NP-induced inflammation.


Asunto(s)
Embrión de Mamíferos/efectos de los fármacos , Perfilación de la Expresión Génica , Nanopartículas del Metal/toxicidad , Plata/química , Animales , Nanopartículas del Metal/química , Microscopía Electrónica de Transmisión , Ratas
12.
Neurotoxicol Teratol ; 26(4): 587-92, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15203181

RESUMEN

gamma-Aminobutyric acid (GABA) is a major inhibitory neurotransmitter in the mammalian central nervous system. Gene targeting of GABA-synthetic glutamic acid decarboxylase (GAD) 67 and GABAA receptor beta(3) subunit induces cleft palate in the mouse. These findings appear to contradict previous pharmacological investigations using benzodiazepines and GABA itself, which indicate that GABA suppresses palatogenesis. Therefore, the effects of picrotoxin and 3-mercaptopropionic acid (3-MP) on palate formation were investigated in the present study. Picrotoxin and 3-MP impair GABA functions by blocking the GABA receptor and synthesis, respectively. Pregnant mice in the critical period [Embryonic Day (E) 11-15] of palatogenesis were administered these substances by subcutaneous injection or continuous infusion at subconvulsive doses, and their fetuses at E17-18 were investigated. A complete cleft in the secondary palate was observed in 15% of 333 embryos in 28 litters. In the remaining fetuses, a complete cleft palate was not observed, but microscopic examination of serial sections revealed partial defects of the palate. Furthermore, rescue from cleft palate in GAD67-deficient mice was attempted by GABA infusion. Horizontal elevation of palatal shelves, which is not observed in nontreated mice, did occur after the infusion in all 14 GABA-infused GAD67-deficient fetuses, although cleft palate still persisted. These results indicate that GABA is required for palatogenesis and is consistent with findings in gene knockout mice.


Asunto(s)
Ácido 3-Mercaptopropiónico/toxicidad , Fisura del Paladar/inducido químicamente , Antagonistas del GABA/toxicidad , Picrotoxina/toxicidad , Ácido gamma-Aminobutírico/deficiencia , Animales , Conducta Animal , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/embriología , Corteza Cerebral/metabolismo , Fisura del Paladar/tratamiento farmacológico , Embrión de Mamíferos , Inhibidores Enzimáticos/toxicidad , Glutamato Descarboxilasa/antagonistas & inhibidores , Isoenzimas/antagonistas & inhibidores , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Ratones Noqueados , Hueso Paladar/efectos de los fármacos , Hueso Paladar/embriología , Ácido gamma-Aminobutírico/uso terapéutico
13.
Zhonghua Nei Ke Za Zhi ; 43(8): 611-3, 2004 Aug.
Artículo en Zh | MEDLINE | ID: mdl-15355669

RESUMEN

OBJECTIVE: To investigate the ability of hemoperfusion to remove some drugs or toxin from the body and its clinical efficacy. METHODS: Sixty-nine cases of poisoning due to tetramine or other drugs poisoning in our hospital between July 1990 and December 2003 were studied. Thirty-four patients among them received conventional treatment (including early gastric lavage, hepatoprotection, diuresis or respiratory support), and the remaining were given conventional treatment and hemoperfusion. Toxin concentration changes in blood before and after hemoperfusion, survival rate, time from come onset to regaining consciousness and convulsion termination and duration of clinical course were compared. RESULTS: In the hemoperfusion group, three patients died, the survival rate was 91.4%, whereas in the non-hemoperfusion group, the survival rate was 85.3% (P > 0.05). Meanwhile the clinical course was markedly shortened in the hemoperfusion group (P < 0.05). The time from coma onset to regaining consciousness and convulsion termination in the hemoperfusion group was significantly shorter than that in the non-hemoperfusion group. The clearance rate was different for different poisons, among them the benzodiazepines had an excellent clearance. CONCLUSIONS: Hemoperfusion could adsorb from blood a different amount of poisons. There is obvious efficacy in shortening clinical course and reducing complications. It could possibly raise survival rate in serious poisoning.


Asunto(s)
Hemoperfusión , Intoxicación/terapia , Rodenticidas/envenenamiento , Adolescente , Adulto , Anciano , Niño , Femenino , Humanos , Masculino , Persona de Mediana Edad , Intoxicación/mortalidad , Rodenticidas/sangre , Tasa de Supervivencia , Resultado del Tratamiento
14.
Artículo en Zh | 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
15.
PLoS One ; 9(4): e94034, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24739881

RESUMEN

The objective of our study was to profile and compare the systematic changes between orally administered artesunate and intramuscularly injected artemether at a low dose over a 3-month period (92 consecutive days) in dogs. Intramuscular administration of 6 mg kg-1 artemether induced a decreased red blood cell (RBC) count (anemia), concurrent extramedullary hematopoiesis in the spleen and inhibition of erythropoiesis in the bone marrow. We also observed a prolonged QT interval and neuropathic changes in the central nervous system, which demonstrated the cortex and motor neuron vulnerability, but no behavioral changes. Following treatment with artesunate, we observed a decreased heart rate, which was most likely due to cardiac conduction system damage, as well as a deceased RBC count, extramedullary hematopoiesis in the spleen and inhibition of erythropoiesis in the bone marrow. However, in contrast to treatment with artemether, neurotoxicity was not observed following treatment with artesunate. In addition, ultra-structural examination by transmission electron microscopy showed mitochondrial damage following treatment with artesunate. These findings demonstrated the spectrum of toxic changes that result upon treatment with artesunate and artemether and show that the prolonged administration of low doses of these derivatives result in diverse toxicity profiles.


Asunto(s)
Artemisininas/toxicidad , Administración Oral , Animales , Arritmias Cardíacas/inducido químicamente , Arteméter , Artemisininas/administración & dosificación , Artesunato , Perros , Recuento de Eritrocitos , Eritropoyesis/efectos de los fármacos , Femenino , Hematopoyesis Extramedular/efectos de los fármacos , Inyecciones Intramusculares , Masculino , Microscopía Electrónica de Transmisión , Mitocondrias/efectos de los fármacos , Mitocondrias/ultraestructura , Pruebas de Toxicidad
16.
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
19.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA