Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Toxicol Appl Pharmacol ; 355: 269-285, 2018 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-30009776

RESUMEN

Triptolide (TP), a major active component of Tripterygium wilfordii Hook f., is widely used in the treatment of inflammation and autoimmune disorders. Its clinical application is limited by severe adverse effects, especially cardiotoxicity. Accumulative evidences indicate that TP induces DNA damage by inhibiting RNA polymerase. Considering the relationship among DNA damage, p53, and the role of p53 in mitochondria-dependent apoptosis, we speculate that TP-induced cardiotoxicity results from p53 activation. In this study, the role of p53 in TP-induced cardiotoxicity was investigated in H9c2 cells, primary cardiomyocytes, and C57BL/6 genetic background p53-/- mice. p53 protein level was elevated by TP in vitro and in acute heart injury models. With TP administration (1.2 mg/kg), p53 deficiency prevented heart histology injury and decreased serum cardiac troponin I (cTn-I) and apoptotic proteins. Mechanistically, immunoblotting and immunofluorescence staining identified that TP-induced toxicity is dependent on p53 nuclear translocation and transactivation of Bcl2 family genes, leading to mitochondrial outer membrane permeabilization (MOMP) and mitochondria dysfunction. Consistently, p53 antagonist PFTα counteracted TP-induced p53 overexpression and regulation of Bcl2 family transcription, which improved mitochondrial membrane integrity and prevented apoptosis. Moreover, Bax antagonist Bax inhibitor peptide (BIP) V5 ameliorated TP-induced apoptosis through suppressing membrane depolarization and ROS accumulation. These results suggest that TP-induced cardiotoxicity is p53-dependent by promoting Bax-induced mitochondria-mediated apoptosis.


Asunto(s)
Antiinflamatorios no Esteroideos/toxicidad , Diterpenos/toxicidad , Cardiopatías/inducido químicamente , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias Cardíacas/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Fenantrenos/toxicidad , Proteína p53 Supresora de Tumor/metabolismo , Animales , Apoptosis/efectos de los fármacos , Línea Celular , Diterpenos/antagonistas & inhibidores , Compuestos Epoxi/antagonistas & inhibidores , Compuestos Epoxi/toxicidad , Cardiopatías/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocardio/patología , Fenantrenos/antagonistas & inhibidores , Especies Reactivas de Oxígeno/metabolismo , Proteína p53 Supresora de Tumor/deficiencia , Proteína p53 Supresora de Tumor/genética , Proteína X Asociada a bcl-2/antagonistas & inhibidores
2.
Pharmazie ; 71(7): 394-397, 2016 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-29441915

RESUMEN

Isoliquiritigenin, a flavonoid found in licorice, has been considered as an antioxidive and hepato-protective agent. Recent studies have shown that a possible mechanism for triptolide-induced hepatotoxicity is related to oxidative damage induced by reactive oxygen species. This study was done to investigate the protection effect of isoliquiritigenin against triptolide-induced hepatotoxicity and the mechanism involved. An acute liver injury model was established by intraperitoneal injection of triptolide (1.0 mg · kg-1) in mice. Different doses of isoliquiritigenin (12.5, 25 and 50 mg · kg-1) were employed as protection. The activities of AST, ALT, ALP and LDH in serum and levels of GSH, GPx, SOD, CAT and MDA in liver tissue were detected. The histopathological changes of liver tissues were observed after HE staining. The protein expression of Nrf2 was detected by western blot. Pretreatment with isoliquiritigenin significantly prevented the triptolide-induced hepatotoxicity indicated by reduced activities of AST, ALT, ALP and LDH. Moreover, isoliquiritigenin pretreatment also prevented from triptolide-induced hepatotoxicity by inhibiting MDA and restoring the levels of GSH, GPx, SOD and CAT. In addition, isoliquiritigenin could attenuate histopathological changes induced by triptolide. Furthermore, the results indicated that isoliquiritigenin pretreatment caused an increase in the protein expression of Nrf2. These results indicated that isoliquiritigenin could protect against triptolide-induced hepatotoxicity via activation of the Nrf2 pathway.


Asunto(s)
Chalconas/farmacología , Enfermedad Hepática Inducida por Sustancias y Drogas/prevención & control , Diterpenos/antagonistas & inhibidores , Diterpenos/toxicidad , Factor 2 Relacionado con NF-E2/metabolismo , Fenantrenos/antagonistas & inhibidores , Fenantrenos/toxicidad , Sustancias Protectoras/farmacología , Animales , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Compuestos Epoxi/antagonistas & inhibidores , Compuestos Epoxi/toxicidad , Hígado/efectos de los fármacos , Hígado/metabolismo , Pruebas de Función Hepática , Masculino , Malondialdehído/antagonistas & inhibidores , Ratones , Ratones Endogámicos ICR , Factor 2 Relacionado con NF-E2/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
3.
Pharmazie ; 71(9): 514-523, 2016 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-29441847

RESUMEN

Clinical application of triptolide (TP), a main active ingredient of the traditional Chinese herb Tripterygium wilfordii Hook f. (TWHF), is limited by a series of severe toxicities, including cardiotoxicity. In previous studies, we found the activation of sirtuin 3 (SIRT3) attenuated TP-induced toxicity in cardiomyocytes. Resveratrol (RSV), a polyphenol from the skins of grapes and red wine, is an activator of SIRT3. The current study aimed to investigate the protective effect of RSV against TP-induced cardiotoxicity and the underlying mechanisms. Mice were treated with a single dose of TP (2.5 mg/kg) via the intragastric (i.g.) route. After 24 h, TP induced abnormal changes of serum biochemistry, activity decrease of antioxidant enzymes and damage of heart tissue such as myocardial fiber rupture, cell swelling and interstitial congestion. In contrast, administration with RSV (50 mg/kg i.g. 12 h before and 2 h after the administration of TP) attenuated the detrimental effects induced by TP in BALB/c mice. Moreover, the cardiomyocyte protective effects of RSV on TP-induced heart injury were associated with the activation of SIRT3 and its downstream targets. In vitro study also indicated that RSV counteracted TP-induced cardiotoxicity through SIRT3-FOXO3 signaling pathway in H9c2 cells. Collectively, these findings suggest the potential of RSV as a promising agent in protecting heart from TP-induced damage.


Asunto(s)
Antioxidantes/farmacología , Antioxidantes/uso terapéutico , Cardiotónicos/farmacología , Cardiotónicos/uso terapéutico , Cardiotoxicidad , Diterpenos/toxicidad , Cardiopatías/inducido químicamente , Cardiopatías/prevención & control , Fenantrenos/toxicidad , Sirtuina 3/efectos de los fármacos , Estilbenos/farmacología , Estilbenos/uso terapéutico , Animales , Apoptosis/efectos de los fármacos , Línea Celular , Diterpenos/antagonistas & inhibidores , Compuestos Epoxi/antagonistas & inhibidores , Compuestos Epoxi/toxicidad , Femenino , Proteína Forkhead Box O3/efectos de los fármacos , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Miocardio/enzimología , Miocardio/patología , Miocitos Cardíacos/efectos de los fármacos , Fenantrenos/antagonistas & inhibidores , Resveratrol , Transducción de Señal/efectos de los fármacos
4.
Amino Acids ; 39(5): 1217-26, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20300786

RESUMEN

In this study, we demonstrate a stimulatory effect of tanshinone IIA isolated from the root of Salvia miltiorrhiza on the commitment of bi-potential mesenchymal precursor C2C12 cells into osteoblasts in the presence of bone morphogenetic protein (BMP)-2. At low concentrations, tanshinone IIA enhanced BMP-2-stimulated induction of alkaline phosphatase (ALP), an early phase biomarker of osteoblast differentiation, and mRNA expression of BMPs. ALP induction was inhibited by the BMP antagonist noggin, suggesting that tanshinone IIA enhances the osteogenic activity of BMP signaling. Furthermore, considering the tanshinone IIA-mediated enhancement of BMP-2-stimulated Smad-Runx2 activities, tanshinone IIA could enhance the osteogenic activity of BMP-2 via acceleration of Smad-Runx2 activation. Additionally, pharmacologic inhibition studies suggest the possible involvement of p38 in the action of tanshinone IIA. The p38 inhibitor SB202190 strongly and dose-dependently inhibited tanshinone IIA-enhanced ALP induction. SB202190 also dose-dependently inhibited the tanshinone IIA-induced p38 activation and combined tanshinone IIA-BMP-2-induced Smad activation. In conclusion, tanshinone IIA enhances the commitment of C2C12 cells into osteoblasts and their differentiation through synergistic cross talk between tanshinone IIA-induced p38 activation and BMP-2-induced Smad activation. These activations could subsequently induce the activation of Runx2, which induces osteogenesis via regulation of the osteogenic factors BMP and ALP expression.


Asunto(s)
Proteína Morfogenética Ósea 2/metabolismo , Osteoblastos/metabolismo , Fenantrenos/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Abietanos , Animales , Proteína Morfogenética Ósea 2/antagonistas & inhibidores , Proteínas Portadoras/farmacología , Células Cultivadas , Relación Dosis-Respuesta a Droga , Imidazoles/farmacología , Ratones , Estructura Molecular , Osteoblastos/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Fenantrenos/antagonistas & inhibidores , Fenantrenos/química , Piridinas/farmacología , Relación Estructura-Actividad , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores
5.
Drug Des Devel Ther ; 13: 2057-2066, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31296984

RESUMEN

BACKGROUND: Ginsenoside Rg3 has been reported to exert protection function on germ cells. However, the mechanisms by which Rg3 regulates apoptosis in mouse Leydig cells remain unclear. In addition, triptolide (TP) has been reported to induce infertility in male rats. Thus, this study aimed to investigate the protective effect of Rg3 against TP-induced toxicity in MLTC-1 cells. METHODS: CCK-8, immunofluorescence assay, Western blotting and flow cytometry were used to detect cell proliferation and cell apoptosis, respectively. In addition, the dual luciferase reporter system assay was used to detect the interaction between miR-26a and GSK3ß in MLTC-1 cells. RESULTS: TP significantly inhibited the proliferation of MLTC-1 cells, while the inhibitory effect of TP was reversed by Rg3. In addition, TP markedly induced apoptosis in MLTC-1 cells via increasing the expressions of Bax, active caspase 3, Cyto c and active caspase 9, and decreasing the level of Bcl-2. However, Rg3 alleviated TP-induced apoptosis of MLTC-1 cells. Moreover, the level of miR-26a was obviously downregulated by Rg3 treatment. The protective effect of Rg3 against TP-induced toxicity in MLTC-1 cells was abolished by miR-26a upregulation. Meanwhile, dual-luciferase assay showed GSK3ß was the direct target of miR-26a in MLTC-1 cells. Overexpression of miR-26a markedly decreased the level of GSK3ß. As expected, upregulation of miR-26a could abrogate the protective effects of Rg3 against TP-induced cytotoxicity via inhibiting the expression of GSK3ß. CONCLUSION: These results indicated that Rg3 could protect MLTC-1 against TP by downregulation of miR-26a. Therefore, Rg3 might serve as a potential agent for the treatment of male hypogonadism.


Asunto(s)
Antiespermatogénicos/antagonistas & inhibidores , Diterpenos/antagonistas & inhibidores , Regulación hacia Abajo/efectos de los fármacos , Ginsenósidos/farmacología , Células Intersticiales del Testículo/efectos de los fármacos , MicroARNs/biosíntesis , Fenantrenos/antagonistas & inhibidores , Sustancias Protectoras/farmacología , Animales , Antiespermatogénicos/farmacología , Supervivencia Celular/efectos de los fármacos , Diterpenos/farmacología , Relación Dosis-Respuesta a Droga , Compuestos Epoxi/antagonistas & inhibidores , Compuestos Epoxi/farmacología , Ginsenósidos/química , Masculino , Ratones , MicroARNs/genética , Conformación Molecular , Fenantrenos/farmacología , Sustancias Protectoras/química , Relación Estructura-Actividad
6.
Mol Med Rep ; 19(6): 4841-4851, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30942423

RESUMEN

Anastomotic fibrosis is highly likely to lead to reoperation in Crohn's disease (CD) patients. Triptolide (TPL) is considered to have anti­inflammatory and antifibrotic effects in a variety of autoimmune diseases, including CD. The present study aimed to investigate the effects of TPL on fibroblasts from strictured ileocolonic anastomosis of patients with CD and its underlying mechanism. Primary fibroblasts were obtained from strictured anastomosis tissue (SAT) samples and matched anastomosis­adjacent normal tissue (NT) samples which were collected from 10 CD patients who underwent reoperation because of anastomotic stricture. Reverse transcription­quantitative polymerase chain reaction (RT­qPCR) was used to measure miR­16­1 and heat shock protein 70 (HSP70) levels. Western blotting was conducted to determine expression of HSP70, collagen I (Col­I), collagen III (Col­III) and α­smooth muscle actin (α­SMA) proteins. Agomir­16­1 and antagomir­16­1 were used to up and downregulate the expression of miR­16­1, respectively. Small interfering RNA (siRNA) was employed to inhibit the expression of HSP70. A wound healing assay was performed to measure the migration of fibroblasts. Cell proliferation was evaluated by MTT and 5­bromo­2­deoxyrudidine assays. Cell apoptosis was determined by caspase­3 activity and TUNEL assays. The results demonstrated that the levels of Col­I, Col­III and α­SMA were all significantly upregulated in SAT compared with NT. miR­16­1 levels in the SAT group were significantly compared with the NT group; conversely, the expression levels of HSP70 mRNA and protein in the SAT group were significantly lower compared with the NT group. Next, fibroblasts were treated with TPL to examine its effect on the miR­16­1/HSP70 pathway. The results demonstrated that the elevated expression of miR­16­1 in the SAT group was effectively inhibited by TPL treatment. Compared with the NT group, both the mRNA and protein levels of HSP70 were significantly downregulated in the SAT group cells, while TPL exhibited a strong promoting effect on HSP70 synthesis. Furthermore, upregulation of miR­16­1 reversed the effect of TPL on the miR­16­1/HSP70 pathway in fibroblasts from SAT. Overexpression of miR­16­1 significantly reversed the inhibitory effects of TPL treatment on migration, proliferation and extracellular matrix (ECM)­associated protein expression of fibroblasts from SAT. Finally, downregulation of miR­16­1 caused similar effects to the fibroblasts as the TPL treatment; however, the inhibitory effects on cell biological functions induced by antagomir­16­1 were all significantly reversed by HSP70 silencing. The present findings indicated that TPL may be a potential therapeutic option for postoperative anastomosis fibrosis of patients with CD. The miR­16­1/HSP70 pathway had a substantial role in the inhibitory effects of TPL on migration, proliferation and ECM synthesis rate of fibroblasts from strictured anastomosis tissues.


Asunto(s)
Anastomosis Quirúrgica , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Enfermedad de Crohn/tratamiento farmacológico , Diterpenos/antagonistas & inhibidores , Fibroblastos/efectos de los fármacos , Proteínas HSP70 de Choque Térmico/metabolismo , MicroARNs/metabolismo , Fenantrenos/antagonistas & inhibidores , Actinas/metabolismo , Adulto , Antagomirs/farmacología , Antiinflamatorios/farmacología , Apoptosis/efectos de los fármacos , China , Colágeno Tipo I/metabolismo , Colágeno Tipo III/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Compuestos Epoxi/antagonistas & inhibidores , Femenino , Fibroblastos/patología , Humanos , Masculino , Persona de Mediana Edad , ARN Mensajero , ARN Interferente Pequeño/metabolismo , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
7.
Nan Fang Yi Ke Da Xue Xue Bao ; 38(8): 949-955, 2018 Jul 30.
Artículo en Zh | MEDLINE | ID: mdl-30187882

RESUMEN

OBJECTIVE: To investigate the protective effect of monoside against triptolide-induced liver injury and explore its molecular mechanism. METHODS: BALB/C mice treated with gastric lavage with triptolide and monoside, either alone or in combination, were examined for changes of hepatic biochemical parameters using the serological method. The growth inhibition rate of HepG2 cells treated with triptolide or monoside or both was assessed with MTT assay, and the cell morphological changes were observed using laser confocal microscopy; the expressions of the target proteins in the antioxidative stress pathway were detected using flow cytometry and Western blotting. RESULTS: In BALB/C mice, gastric lavage of triptolide induced obvious hepatic damage. In HepG2 cells, treatment with triptolide significantly inhibited the cell growth, resulting in a cell viability as low as 72.83% at 24 h; triptolide also induced obvious cell apoptosis and cell nucleus deformation, causing an apoptosis rate of 43.1% in the cells at 24 h. Triptolide significantly reduced the expressions of Nrf2 and HO-1 proteins related with the oxidative stress pathway. Combined treatment with morroniside obviously reversed these changes, resulting in significantly decreased hepatic biochemical parameters and the liver index in BALB/C mice and in significantly lowered cell apoptosis rate, improved cell morphology, and increased Nrf2 and HO-1 protein expressions in HepG2 cells. CONCLUSIONS: Monoside protects against triptolide-induced liver injury possibly by relieving oxidative stress.


Asunto(s)
Apoptosis/efectos de los fármacos , Diterpenos/farmacología , Glicósidos/farmacología , Hepatocitos/efectos de los fármacos , Fenantrenos/farmacología , Animales , Aumento de la Célula/efectos de los fármacos , Núcleo Celular/efectos de los fármacos , Diterpenos/antagonistas & inhibidores , Compuestos Epoxi/antagonistas & inhibidores , Compuestos Epoxi/farmacología , Lavado Gástrico , Hemo-Oxigenasa 1/metabolismo , Células Hep G2 , Hepatocitos/fisiología , Humanos , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos BALB C , Factor 2 Relacionado con NF-E2/metabolismo , Estrés Oxidativo , Fenantrenos/antagonistas & inhibidores
8.
Mol Cell Endocrinol ; 259(1-2): 30-42, 2006 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-16962702

RESUMEN

Phytoestrogens are plant-derived, non-steroidal constituents of our diets. They can act as agonists or antagonists of estrogen receptors, and they can modulate the activities of the key enzymes in estrogen biosynthesis. Much less is known about their actions on the androgen and progesterone metabolizing enzymes. We have examined the inhibitory action of phytoestrogens on the key human progesterone-metabolizing enzyme, 20alpha-hydroxysteroid dehydrogenase (AKR1C1). This enzyme inactivates progesterone and the neuroactive 3alpha,5alpha-tetrahydroprogesterone, to form their less active counterparts, 20alpha-hydroxyprogesterone and 5alpha-pregnane-3alpha,20alpha-diol, respectively. We overexpressed recombinant human AKR1C1 in Escherichia coli, purified it to homogeneity, and examined the selected phytoestrogens as inhibitors of NADPH-dependent reduction of a common AKR substrate, 9,10-phenantrenequinone, and progesterone. The most potent inhibitors were 7-hydroxyflavone, 3,7-dihydroxyflavone and flavanone naringenin with IC(50) values in the low microM range. Docking of the flavones in the active site of AKR1C1 revealed their possible binding modes, in which they are sandwiched between the Leu308 and Trp227 of AKR1C1.


Asunto(s)
20-Hidroxiesteroide Deshidrogenasas/antagonistas & inhibidores , Fitoestrógenos/farmacología , Progesterona/metabolismo , 3-Hidroxiesteroide Deshidrogenasas/metabolismo , Miembro C3 de la Familia 1 de las Aldo-Ceto Reductasas , Sitios de Unión , Simulación por Computador , Cumarinas/farmacología , Inhibidores Enzimáticos/farmacología , Congéneres del Estradiol/farmacología , Moduladores de los Receptores de Estrógeno/farmacología , Flavanonas/farmacología , Flavonas/química , Flavonas/farmacología , Humanos , Hidroxiprostaglandina Deshidrogenasas/metabolismo , Isoflavonas/farmacología , Modelos Biológicos , Modelos Moleculares , Fenantrenos/antagonistas & inhibidores , Progesterona Reductasa/metabolismo , Unión Proteica , Proteínas Recombinantes/aislamiento & purificación , Estilbenos/farmacología , Zearalenona/farmacología
9.
Cancer Res ; 36(12): 4387-93, 1976 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-1000489

RESUMEN

After metabolic activation the carcinogen 15,16-dihydro-11-[3H]methylcyclopenta[a]phenanthren-17-one binds to DNA in vitro, and this binding is prevented by 7,8-benzoflavone. Radioactivity cannot be removed from the DNA with organic solvents or by chromatography on Sephadex G-50, even after heat denaturation of the DNA. Enzymatic hydrolysis yields radioactive fractions, which elute from a column of Sephadex LH-20 immediately after the natural nucleosides. At least two species of reactive metabolites are involved in this bending, those with a half-life of a few hr and others with greater stability. After extraction from the aqueous incubation mixture, they could be detected in discrete polar fractions from separations of the complex metabolite mixture by high-pressure liquid chromatography. Their ability to bind to DNA decreased with time at ambient temperature, and they were rapidly deactivated by acid. 7,8-Benzolflavone acted by suppressing the formation of polar metabolites derived from enzymatic oxidation of the aromatic double bonds. The inhibitor had no effect on the enzymes hydroxylating saturated carbon; hence it is unlikely that metabolism of the methyl group is important in conversion of this carcinogen to its proximate form, although the presence of the 11-methyl group is essential for carcinogenic activity in this series.


Asunto(s)
Carcinógenos/metabolismo , ADN/metabolismo , Fenantrenos/metabolismo , Animales , Hidrocarburo de Aril Hidroxilasas/antagonistas & inhibidores , Hidrocarburo de Aril Hidroxilasas/biosíntesis , Fenómenos Químicos , Química , Cromatografía Líquida de Alta Presión , Inducción Enzimática/efectos de los fármacos , Flavonoides/farmacología , Técnicas In Vitro , Cinética , Masculino , Metilación , Microsomas Hepáticos/metabolismo , Fenantrenos/antagonistas & inhibidores , Ratas
10.
Biochim Biophys Acta ; 1126(3): 319-26, 1992 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-1322178

RESUMEN

Aristolochic acid and PGBx, two structurally unrelated, protein-targeted inhibitors of isolated phospholipases A2, are effective antagonists of calcium ionophore A23187-stimulated mobilization of [3H]arachidonate from human neutrophils. We now report that preincubation of neutrophils with oleoylacetylglycerol (OAG, 15 microM) substantially reverses the inhibitory effect of 200 microM aristolochic acid (from 70 to 24% inhibition). Similarly, OAG increases the IC50 for PGBx from 2.5 to greater than 20 microM. The effects of OAG on inhibition by either aristolochic acid or PGBx are dose-dependent, with an ED50 of 2.5 microM. Protection against inhibition by either aristolochic acid or PGBx is also observed with phorbol myristate acetate (PMA, ED50 3 nM), but not 4-alpha-phorbol didecanoate. Aristolochic acid and PGBx do not inhibit PMA-stimulated superoxide generation, and are thus not protein kinase C inhibitors. Furthermore, neither aristolochic acid nor PGBx inhibit diglyceride generation through the phospholipase D/phosphatidate phosphohydrolase pathway. A23187-stimulated [3H]arachidonate mobilization is increased by 20-50% when neutrophils are preincubated with OAG or PMA. The present results indicate that OAG and PMA also modulate the A23187-stimulated [3H]arachidonate mobilization so as to render it less sensitive to inhibitors of phospholipase A2.


Asunto(s)
Ácido Araquidónico/metabolismo , Ácidos Aristolóquicos , Neutrófilos/efectos de los fármacos , Fenantrenos/farmacología , Fosfolipasas A/antagonistas & inhibidores , Polímeros/farmacología , Prostaglandinas B/farmacología , Calcimicina/farmacología , Diglicéridos/farmacología , Humanos , N-Formilmetionina Leucil-Fenilalanina/farmacología , Neutrófilos/metabolismo , Fenantrenos/antagonistas & inhibidores , Fosfatidato Fosfatasa/metabolismo , Fosfolipasa D/metabolismo , Fosfolipasas A2 , Prostaglandinas B/antagonistas & inhibidores , Esfingosina/farmacología , Superóxidos/metabolismo , Acetato de Tetradecanoilforbol/farmacología
11.
Free Radic Biol Med ; 38(3): 388-95, 2005 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-15629867

RESUMEN

9,10-Phenanthraquinone (PQ), a major quinone contained in diesel exhaust particles and atmospheric PM(2.5), undergoes one-electron reduction by flavin enzymes such as NADPH-cytochrome P450 reductase, leading to production of reactive oxygen species in vitro. We have detected an ESR signal for superoxide (O(2)(-)) and hydroxyl radicals ((.)OH) by the spin trap method when PQ was mixed with P450 reductase, NADPH, and iron(III). When we examined the effects of PQ on A549 human pulmonary epithelial cells, PQ induced apoptosis with a LC(50) of approximately 7 microM. Formation of protein carbonyls was also detected in cells after treatment with PQ, suggesting that PQ induces oxidative damage. Iron chelators such as 1,10-phenanthroline (OP), desferrioxamine mesylate, and deferiprone respectively afforded protection against the toxic effects of PQ. Furthermore, treatment of A549 cells with 10-20 microM PQ for 12 h specifically down-regulated protein levels of Cu,Zn-superoxide dismutase (Cu,Zn-SOD) and heme oxygenase-1 (HO-1) by more than 50%. Pretreatment of cells with OP (10 microM) markedly reduced the down-regulation of Cu,Zn-SOD and HO-1 and protein carbonyl formation in response to PQ. The inhibitor of Cu,Zn-SOD, diethyldithiocarbamate, enhanced the toxic effects of 5 microM PQ. The present findings suggest that PQ causes iron-mediated oxidative damage that is exacerbated by the concomitant down-regulation of Cu,Zn-SOD.


Asunto(s)
Células Epiteliales/efectos de los fármacos , Depuradores de Radicales Libres/farmacología , Hemo Oxigenasa (Desciclizante)/efectos de los fármacos , Quelantes del Hierro/farmacología , Fenantrenos/toxicidad , Superóxido Dismutasa/efectos de los fármacos , Apoptosis/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Sistema Enzimático del Citocromo P-450/metabolismo , Ditiocarba/farmacología , Espectroscopía de Resonancia por Spin del Electrón , Depuradores de Radicales Libres/química , Radicales Libres/química , Hemo Oxigenasa (Desciclizante)/metabolismo , Hemo-Oxigenasa 1 , Humanos , Quelantes del Hierro/química , Proteínas de la Membrana , Fenantrenos/antagonistas & inhibidores , Fenantrolinas/farmacología , Alveolos Pulmonares/citología , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo , Emisiones de Vehículos/toxicidad
12.
FASEB J ; 16(11): 1463-4, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12205049

RESUMEN

Smoking is a major risk factor for endothelial cell injury and subsequent coronary artery disease. Epidemiological studies implicate the phospholipase A2/arachidonic acid cascade in the mechanism by which smoking causes heart disease. However, specific components of cigarette smoke that activate this pathway have not been identified. The purpose of this study was to investigate the effects of polycyclic aromatic hydrocarbons contained in cigarette smoke on phospholipase A2 (PLA2) activity and apoptosis of human coronary artery endothelial cells. 1-methylanthracene (1-MA), phenanthrene (PA), and benzo(a)pyrene (B(a)P) caused significant release of 3H-arachidonate from endothelial cells. 1-MA and PA, but not B(a)P, also caused significant release of 3H-linoleic acid. Release of fatty acids from membrane phospholipids preceded the onset of apoptosis. 3H-arachidonate release and apoptosis induced by 1-MA, B(a)P, and PA were inhibited by methylarachidonoyl-fluorophosphonate, an inhibitor of Groups IV and VI PLA2s. Bromoenol lactone, an inhibitor of Group VI enzymes, inhibited both 3H-arachidonate release and apoptosis induced by 1-MA and PA, but not B(a)P. MJ33, an inhibitor of the acidic calcium-independent PLA2, attenuated 3H-arachidonate release and apoptosis by PA, but not 1-MA or B(a)P. The presence of Groups IV and VI and the acidic iPLA2 in endothelial cells was demonstrated by reverse transcriptase-polymerase chain reaction and Western analysis. These data suggest that 1-MA, B(a)P and PA induce apoptosis of endothelial cells by a mechanism that involves activation of these three distinct isoforms of PLA2.


Asunto(s)
Apoptosis , Vasos Coronarios/enzimología , Endotelio Vascular/enzimología , Fosfolipasas A/fisiología , Hidrocarburos Policíclicos Aromáticos/farmacología , Antracenos/farmacología , Ácido Araquidónico/metabolismo , Benzo(a)pireno/antagonistas & inhibidores , Benzo(a)pireno/farmacología , Células Cultivadas , Vasos Coronarios/citología , Vasos Coronarios/metabolismo , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Inhibidores Enzimáticos/farmacología , Humanos , Ácido Linoleico/metabolismo , Modelos Biológicos , Fenantrenos/antagonistas & inhibidores , Fenantrenos/farmacología , Fosfolipasas A/antagonistas & inhibidores , Fosfolipasas A2 , Hidrocarburos Policíclicos Aromáticos/análisis , Fumar
13.
Toxicology ; 268(3): 191-7, 2010 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-20026164

RESUMEN

It has been widely recognized that nitric oxide (NO) suppresses oxidative damage of endothelial cell, but little is known about its pathophysiological role in apoptotic induction by 9,10-phenanthrenequinone (9,10-PQ), a major quinone component in diesel exhaust particles. Here, we have investigated the change in NO level in human aortic endothelial cells and the effect of NO in each step of apoptotic signaling initiated by 9,10-PQ. Treatment with 9,10-PQ evoked a bell-shaped production of NO, which was presumably due to increase in an active form of endothelial NO synthase. Pretreatment with exogenous NO decreased the susceptibility of the cells to 9,10-PQ, and retrieved from apoptotic signaling (reactive oxygen species generation, glutathione depletion and caspase activation) induced during exposure to high concentrations of 9,10-PQ. In addition, inhibition of endogenous NO production augmented the toxicity of 9,10-PQ. Interestingly, the 9,10-PQ treatment resulted in marked decreases in the proteasomal activities, which were partially abrogated by NO and a cell-permeable cGMP analog. These results indicate that proteasomal dysfunction by oxidative stress participates in the 9,10-PQ-induced apoptotic signaling and is ameliorated by NO via a cGMP-dependent pathway, thereby suggesting the protective role of NO in vascular damage caused by 9,10-PQ.


Asunto(s)
Apoptosis/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Mutágenos/toxicidad , Óxido Nítrico/farmacología , Fenantrenos/antagonistas & inhibidores , Fenantrenos/toxicidad , Complejo de la Endopetidasa Proteasomal/efectos de los fármacos , Western Blotting , Línea Celular , Supervivencia Celular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Glutatión/metabolismo , Guanilato Ciclasa/metabolismo , Humanos , Peróxido de Hidrógeno/farmacología , Óxido Nítrico/metabolismo , Donantes de Óxido Nítrico/farmacología , Óxido Nítrico Sintasa/antagonistas & inhibidores , Estrés Oxidativo/efectos de los fármacos
15.
Vet Hum Toxicol ; 29(2): 113-7, 1987 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-3576942

RESUMEN

Male Sprague-Dawley rats were treated ip with beta-naphthoflavone (BNF, 40 mg/kg/day) in dimethylsulfoxide (DMSO, 26.7 mg BNF/ml) for three days. At 24 hr after the pretreatment DMSO (3.0 ml/kg), phenanthrene (150 mg/kg), ozonized or nitrated products of phenanthrene (150 mg/kg), pyrene (150 mg/kg), or ozonized or nitrated products of pyrene (150 mg/kg) were injected ip. Phenanthrene, pyrene, and their ozonized or nitrated products were dissolved in DMSO (50 mg/ml). No increase in the level of aspartate aminotransferase (AST), alanine aminotransferase (ALT) or sorbitol dehydrogenase (SDH) was seen in the pretreated rats 48 hr after the treatment. This is in contrast to what was seen in previous work without the BNF pretreatment. BNF pretreatment induced a small but significant increase in gamma-glutamyl transpeptidase (GGTP) levels. No treatment group receiving BNF differed from another with respect to GGTP. A decrease in lactate dehydrogenase (LDH) levels was noted in the nitro-PAH treatment groups; the same phenomenon was observed earlier in rats treated with nitro-PAH without BNF treatment. These results suggest that the mixed-function oxidase systems specifically induced by BNF have a protective effect against the hepatotoxicity of the oxonized or nitrated products of phenanthrene and pyrene.


Asunto(s)
Benzoflavonas/farmacología , Flavonoides/farmacología , Hepatopatías/prevención & control , Fenantrenos/antagonistas & inhibidores , Pirenos/antagonistas & inhibidores , Animales , Enfermedad Hepática Inducida por Sustancias y Drogas , L-Lactato Deshidrogenasa/sangre , Masculino , Dióxido de Nitrógeno , Ozono , Fenantrenos/toxicidad , Pirenos/toxicidad , Ratas , Ratas Endogámicas , beta-naftoflavona
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA