Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros











Intervalo de año de publicación
1.
Mol Cell Biochem ; 465(1-2): 175-185, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31853800

RESUMEN

Cutaneous changes like rash and hair loss, as well as other neurogenic inflammation side effects, occur frequently during anticancer treatment with the epidermal growth factor receptor-tyrosine kinase inhibitor (EGFR-TKI), erlotinib. These adverse events may be so severe that they impair the patient's compliance with the treatment or even cause its discontinuation. In the current preclinical study, rats (9.2 weeks) were treated with erlotinib (10 mg/kg/day) ± aprepitant (2 mg/kg/day) for 12 weeks. Visual changes in the development of facial skin lesions/hair loss and SP-receptor expression (immunohistochemically) in facial skin tissue were assessed; also changes in plasma magnesium, 8-isoprostane, substance P (SP), neutrophil superoxide production, and cardiac function (echocardiography) were measured. Erlotinib lowered plasma magnesium 14%, elevated SP 65%, caused 3.7-fold higher basal superoxide production, 2.5-fold higher 8-isoprostane levels, 11.6% lower cardiac systolic, and 10.9% lower diastolic function. Facial dermatological changes (alopecia, skin reddening, scabbing, nose crusting) occurred by 4 weeks (± + to ++) in erlotinib-treated rats, and progressively worsened (±++ to +++) by week 12. Facial skin SP-receptor upregulation (78% higher) occurred in epidermal and hair follicle cells. All adverse effects were substantially and significantly mitigated by aprepitant, including a 62% lowering of skin SP-receptors (p < 0.05). Elevated SP levels mediated the side effects of erlotinib treatment, but aprepitant's significant prevention of the systemic and cutaneous adverse events indicates a novel potential therapy against the side effects of this anticancer treatment.


Asunto(s)
Aprepitant/farmacología , Erupciones por Medicamentos , Clorhidrato de Erlotinib/efectos adversos , Enfermedades del Sistema Nervioso , Antagonistas del Receptor de Neuroquinina-1/farmacología , Animales , Erupciones por Medicamentos/tratamiento farmacológico , Erupciones por Medicamentos/metabolismo , Erupciones por Medicamentos/patología , Clorhidrato de Erlotinib/farmacología , Masculino , Enfermedades del Sistema Nervioso/inducido químicamente , Enfermedades del Sistema Nervioso/tratamiento farmacológico , Enfermedades del Sistema Nervioso/metabolismo , Enfermedades del Sistema Nervioso/patología , Ratas , Ratas Sprague-Dawley
2.
J Cardiovasc Pharmacol ; 65(1): 54-61, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25343568

RESUMEN

To determine whether the epidermal growth factor receptor tyrosine kinase inhibitor, erlotinib may cause hypomagnesemia, inflammation, and cardiac stress, erlotinib was administered to rats (10 mg · kg(-1)· d(-1)) for 9 weeks. Plasma magnesium decreased progressively between 3 and 9 weeks (-9% to -26%). Modest increases in plasma substance P (SP) occurred at 3 (27%) and 9 (25%) weeks. Neutrophil superoxide-generating activity increased 3-fold, and plasma 8-isoprostane rose 210%, along with noticeable appearance of cardiac perivascular nitrotyrosine. The neurokinin-1 (NK-1) receptor antagonist, aprepitant (2 mg · kg(-1) · d(-1)), attenuated erlotinib-induced hypomagnesemia up to 42%, reduced circulating SP, suppressed neutrophil superoxide activity and 8-isoprostane elevations; cardiac nitrotyrosine was diminished. Echocardiography revealed mild to moderately decreased left ventricular ejection fraction (-11%) and % fractional shortening (-17%) by 7 weeks of erlotinib treatment and significant reduction (-17.5%) in mitral valve E/A ratio at week 9 indicative of systolic and early diastolic dysfunction. Mild thinning of the left ventricular posterior wall suggested early dilated cardiomyopathy. Aprepitant completely prevented the erlotinib-induced systolic and diastolic dysfunction and partially attenuated the anatomical changes. Thus, chronic erlotinib treatment does induce moderate hypomagnesemia, triggering SP-mediated oxidative/inflammation stress and mild-to-moderate cardiac dysfunction, which can largely be corrected by the administration of the SP receptor blocker.


Asunto(s)
Receptores ErbB/antagonistas & inhibidores , Estrés Oxidativo/efectos de los fármacos , Inhibidores de Proteínas Quinasas/toxicidad , Quinazolinas/toxicidad , Animales , Aprepitant , Ecocardiografía , Clorhidrato de Erlotinib , Magnesio/sangre , Masculino , Morfolinas/farmacología , Antagonistas del Receptor de Neuroquinina-1/farmacología , Ratas , Ratas Sprague-Dawley , Receptores de Neuroquinina-1/efectos de los fármacos , Receptores de Neuroquinina-1/metabolismo , Sustancia P/sangre , Función Ventricular Izquierda/efectos de los fármacos
3.
Int J Biol Sci ; 9(4): 350-60, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23630447

RESUMEN

BACKGROUND: Anthracyclines, such as doxorubicin (Adriamycin), are highly effective chemotherapeutic agents, but are well known to cause myocardial dysfunction and life-threatening congestive heart failure (CHF) in some patients. METHODS: To generate new hypotheses about its etiology, genome-wide transcript analysis was performed on whole blood RNA from women that received doxorubicin-based chemotherapy and either did, or did not develop CHF, as defined by ejection fractions (EF)≤40%. Women with non-ischemic cardiomyopathy unrelated to chemotherapy were compared to breast cancer patients prior to chemo with normal EF to identify heart failure-related transcripts in women not receiving chemotherapy. Byproducts of oxidative stress in plasma were measured in a subset of patients. RESULTS: The results indicate that patients treated with doxorubicin showed sustained elevations in oxidative byproducts in plasma. At the RNA level, women who exhibited low EFs after chemotherapy had 260 transcripts that differed >2-fold (p<0.05) compared to women who received chemo but maintained normal EFs. Most of these transcripts (201) were not altered in non-chemotherapy patients with low EFs. Pathway analysis of the differentially expressed genes indicated enrichment in apoptosis-related transcripts. Notably, women with chemo-induced low EFs had a 4.8-fold decrease in T-cell leukemia/lymphoma 1A (TCL1A) transcripts. TCL1A is expressed in both cardiac and skeletal muscle, and is a known co-activator for AKT, one of the major pro-survival factors for cardiomyocytes. Further, women who developed low EFs had a 2-fold lower level of ABCB1 transcript, encoding the multidrug resistance protein 1 (MDR1), which is an efflux pump for doxorubicin, potentially leading to higher cardiac levels of drug. In vitro studies confirmed that inhibition of MDR1 by verapamil in rat H9C2 cardiomyocytes increased their susceptibility to doxorubicin-induced toxicity. CONCLUSIONS: It is proposed that chemo-induced cardiomyopathy may be due to a reduction in TCL1A levels, thereby causing increased apoptotic sensitivity, and leading to reduced cardiac MDR1 levels, causing higher cardiac levels of doxorubicin and intracellular free radicals. If so, screening for TCL1A and MDR1 SNPs or expression level in blood, might identify women at greatest risk of chemo-induced heart failure.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/deficiencia , Antineoplásicos/efectos adversos , Proteínas Proto-Oncogénicas/deficiencia , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Animales , Antraciclinas/efectos adversos , Antraciclinas/uso terapéutico , Antineoplásicos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Cardiomiopatías/inducido químicamente , Cardiomiopatías/metabolismo , Células Cultivadas , Doxorrubicina/efectos adversos , Doxorrubicina/uso terapéutico , Femenino , Humanos , Proteínas Proto-Oncogénicas/metabolismo , Ratas
4.
Can J Physiol Pharmacol ; 90(10): 1413-8, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23067376

RESUMEN

Angiotensin may promote endothelial dysfunction through iron accumulation. To research this, bovine endothelial cells (ECs) were incubated with iron (30 µmol·L⁻¹) with or without angiotensin II (100 nmol·L⁻¹). After incubation for 6 h, it was observed that the addition of angiotensin enhanced EC iron accumulation by 5.1-fold compared with a 1.8-fold increase for cells incubated with iron only. This enhanced iron uptake was attenuated by losartan (100 nmol·L⁻¹), d-propranolol (10 µmol·L⁻¹), 4-HO-propranolol (5 µmol·L⁻¹), and methylamine, but not by vitamin E or atenolol. After 6 h of incubation, angiotensin plus iron provoked intracellular oxidant formation (2'7'-dichlorofluorescein diacetate (DCF-DA) fluorescence) and elevated oxidized glutathione; significant loss of cell viability occurred at 48 h. Stimulated prostacyclin release decreased by 38% (6 h) and NO synthesis was reduced by 41% (24 h). Both oxidative events and functional impairment were substantially attenuated by losartan or d-propranolol. It is concluded that angiotensin promoted non-transferrin-bound iron uptake via AT-1 receptor activation, leading to EC oxidative functional impairment. The protective effects of d-propranolol and 4-HO-propranolol may be related to their lysosomotropic properties.


Asunto(s)
Angiotensina II/metabolismo , Endotelio Vascular/metabolismo , Epoprostenol/metabolismo , Hierro/metabolismo , Losartán/farmacología , Óxido Nítrico/metabolismo , Propranolol/análogos & derivados , Antagonistas Adrenérgicos beta/química , Antagonistas Adrenérgicos beta/farmacología , Antagonistas Adrenérgicos beta/uso terapéutico , Angiotensina II/química , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Bloqueadores del Receptor Tipo 1 de Angiotensina II/uso terapéutico , Animales , Aorta/citología , Aorta/efectos de los fármacos , Aorta/metabolismo , Bovinos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Regulación hacia Abajo/efectos de los fármacos , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Epoprostenol/agonistas , Epoprostenol/antagonistas & inhibidores , Glutatión/metabolismo , Hierro/efectos adversos , Sobrecarga de Hierro/prevención & control , Losartán/uso terapéutico , Óxido Nítrico/agonistas , Óxido Nítrico/antagonistas & inhibidores , Oxidación-Reducción , Estrés Oxidativo/efectos de los fármacos , Propranolol/farmacología , Propranolol/uso terapéutico , Receptor de Angiotensina Tipo 1/metabolismo
5.
Can J Physiol Pharmacol ; 90(9): 1257-68, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22913465

RESUMEN

d-Propranolol (d-Pro: 2-8 mg·(kg body mass)(-1)·day(-1)) protected against cardiac dysfunction and oxidative stress during 3-5 weeks of iron overload (2 mg Fe-dextran·(g body mass)(-1)·week(-1)) in Sprague-Dawley rats. At 3 weeks, hearts were perfused in working mode to obtain baseline function; red blood cell glutathione, plasma 8-isoprostane, neutrophil basal superoxide production, lysosomal-derived plasma N-acetyl-ß-galactosaminidase (NAGA) activity, ventricular iron content, and cardiac iron deposition were assessed. Hearts from the Fe-treated group of rats exhibited lower cardiac work (26%) and output (CO, 24%); end-diastolic pressure rose 1.8-fold. Further, glutathione levels increased 2-fold, isoprostane levels increased 2.5-fold, neutrophil superoxide increased 3-fold, NAGA increased 4-fold, ventricular Fe increased 4.9-fold; and substantial atrial and ventricular Fe-deposition occurred. d-Pro (8 mg) restored heart function to the control levels, protected against oxidative stress, and decreased cardiac Fe levels. After 5 weeks of Fe treatment, echocardiography revealed that the following were depressed: percent fractional shortening (%FS, 31% lower); left ventricular (LV) ejection fraction (LVEF, 17%), CO (25%); and aortic pressure maximum (P(max), 24%). Mitral valve E/A declined by 18%, indicating diastolic dysfunction. Cardiac CD11b+ infiltrates were elevated. Low d-Pro (2 mg) provided modest protection, whereas 4-8 mg greatly improved LVEF (54%-75%), %FS (51%-81%), CO (43%-78%), P(max) (56%-100%), and E/A >100%; 8 mg decreased cardiac inflammation. Since d-Pro is an antioxidant and reduces cardiac Fe uptake as well as inflammation, these properties may preserve cardiac function during Fe overload.


Asunto(s)
Antagonistas Adrenérgicos beta/uso terapéutico , Cardiopatías/prevención & control , Sobrecarga de Hierro/tratamiento farmacológico , Estrés Oxidativo/efectos de los fármacos , Propranolol/uso terapéutico , Acetilglucosaminidasa/sangre , Antagonistas Adrenérgicos beta/administración & dosificación , Antagonistas Adrenérgicos beta/química , Animales , Gasto Cardíaco/efectos de los fármacos , Progresión de la Enfermedad , Relación Dosis-Respuesta a Droga , Ecocardiografía , Eritrocitos/efectos de los fármacos , Eritrocitos/metabolismo , Glutatión , Cardiopatías/sangre , Cardiopatías/etiología , Cardiopatías/metabolismo , Sobrecarga de Hierro/sangre , Sobrecarga de Hierro/complicaciones , Sobrecarga de Hierro/metabolismo , Masculino , Miocardio/metabolismo , Neutrófilos/efectos de los fármacos , Neutrófilos/enzimología , Perfusión , Propranolol/administración & dosificación , Propranolol/química , Ratas , Ratas Sprague-Dawley , Estereoisomerismo , Superóxido Dismutasa/metabolismo , Resultado del Tratamiento
6.
Can J Physiol Pharmacol ; 90(8): 1145-9, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22646904

RESUMEN

We determined whether the epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI) N-​(3-​chlorophenyl)-​6,​7-​dimethoxy-​4-​quinazolinamine (tyrphostin AG-1478) causes hypomagnesemia and cardiac dysfunction in rats. Tyrphostin was administered (3 times per week, intraperitoneal injection, to achieve 21.4 mg·(kg body mass)(-1)·day(-1)) to normomagnesemic rats for 5 weeks. Levels of magnesium in the plasma of the tyrphostin-treated rats decreased significantly by the following amount: 17% at week 1, 27% at week 2, and 26%-35% between weeks 3 to 5. Levels of the plasma lipid peroxidation marker 8-isoprostane rose significantly: by 58% at week 1, 168% at week 3, and 113% at week 5. At week 5, blood neutrophils from the tyrphostin-treated group displayed a 2.26-fold higher basal level of O(2)(·-) generation; the ratio of oxidized glutathione (glutathione disulfide; GSSG) to reduced glutathione (GSH) in the red blood cells increased 2.5-fold. At week 5, echocardiography revealed that TKI treatment resulted in significant cardiac systolic dysfunction, with impaired diastolic function and dilated cardiomyopathy. Since hypomagnesemia alone can trigger oxidative stress and cardiac injury, we suggest that inhibition of EGFR-TK caused magnesium wasting, which partly contributed to decreased cardiac contractility.


Asunto(s)
Ecocardiografía/efectos de los fármacos , Inhibidores Enzimáticos/efectos adversos , Receptores ErbB/antagonistas & inhibidores , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Quinazolinas/efectos adversos , Defectos Congénitos del Transporte Tubular Renal/inducido químicamente , Tirfostinos/efectos adversos , Animales , Biomarcadores/sangre , Calcio/sangre , Dinoprost/análogos & derivados , Dinoprost/sangre , Ecocardiografía/métodos , Ecocardiografía/estadística & datos numéricos , Glutatión/sangre , Masculino , Estrés Oxidativo , Ratas , Ratas Sprague-Dawley , Defectos Congénitos del Transporte Tubular Renal/sangre , Superóxidos/sangre
7.
Am J Med Sci ; 342(2): 125-8, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21747282

RESUMEN

In rodents with dietary magnesium deficiency (Mg deficiency), hypomagnesemia, occurs leading to a rise in circulating substance P from neuronal tissues to trigger systemic inflammatory stress in cardiac and intestinal tissues. Sustained elevations of substance P may result from impaired neutral endopeptidase (NEP) activity due to reactive oxygen and reactive nitrogen species. Associated increase in intestinal permeability includes infiltration of WBC and endotoxemia, which can further amplify the systemic inflammatory response that leads to impaired contractile function associated with up-regulation of the cardiac CD14 endotoxin receptor. The neurogenic signal transduction pathways that we have identified in the pro-oxidant/pro-inflammatory processes found with prolonged hypomagnesemia are described in this report.


Asunto(s)
Sistema Cardiovascular/metabolismo , Mucosa Intestinal/metabolismo , Deficiencia de Magnesio , Estrés Oxidativo/fisiología , Animales , Sistema Cardiovascular/fisiopatología , Humanos , Inflamación/metabolismo , Inflamación/fisiopatología , Intestinos/fisiopatología , Deficiencia de Magnesio/metabolismo , Deficiencia de Magnesio/fisiopatología , Neprilisina/metabolismo , Ratas , Especies de Nitrógeno Reactivo/metabolismo , Receptores Inmunológicos/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Sustancia P/fisiología
8.
Magnes Res ; 22(3): 167S-173S, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19780404

RESUMEN

During dietary deficiency of magnesium neurogenic inflammation is mediated, primarily, by elevated levels of substance P (SP). The enzyme most specific for degrading this neuropeptide is neutral endopeptidase (NEP). In recent studies we found that pharmacological inhibition of NEP by phosphoramidon resulted in elevated plasma levels of SP and greater oxidative stress. We also observed that hypomagnesemia reduced cardiac and intestinal expression of NEP. In these magnesium-deficient rats increased intestinal permeability and impaired cardiac contractility occurred. In our colony of genetically-engineered NEP knockout mice that have reduced ability to degrade SP, we found increased oxidative stress that was prevented by SP (neurokinin-1) receptor blockade. Thus, we submit that inhibition of NEP by pharmacological, genetic and dietary approaches (magnesium restriction), causes greater neurogenic inflammation that may result in increased intestinal and cardiac dysfunction.


Asunto(s)
Deficiencia de Magnesio/complicaciones , Neprilisina/antagonistas & inhibidores , Inflamación Neurogénica/etiología , Sustancia P/metabolismo , Animales , Glicopéptidos/farmacología , Corazón/efectos de los fármacos , Inmunohistoquímica , Intestinos/efectos de los fármacos , Inflamación Neurogénica/metabolismo , Inhibidores de Proteasas/farmacología , Ratas , Regulación hacia Arriba
9.
Am J Med Sci ; 338(1): 22-7, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19593099

RESUMEN

Hypomagnesemia continues to be a significant clinical disorder that is present in patients with diabetes mellitus, alcoholism, and treatment with magnesuric drugs (diuretics, cancer chemotherapy agents, etc.). To determine the role of magnesium in cardiovascular pathophysiology, we have used dietary restriction of this cation in animal models. This review highlights some key observations that helped formulate the hypothesis that release of substance P (SP) during experimental dietary Mg deficiency (MgD) may initiate a cascade of deleterious inflammatory, oxidative, and nitrosative events, which ultimately promote cardiomyopathy, in situ cardiac dysfunction, and myocardial intolerance to secondary stresses. SP acts primarily through neurokinin-1 receptors of inflammatory and endothelial cells, and may induce production of reactive oxygen and nitrogen species (superoxide anion, NO*, peroxynitrite, hydroxyl radical), leading to enhanced consumption of tissue antioxidants; stimulate release of inflammatory mediators; promote tissue adhesion molecule expression; and enhance inflammatory cell tissue infiltration and cardiovascular lesion formation. These SP-mediated events may predispose the heart to injury if faced with subsequent oxidative stressors (ischemia/reperfusion, certain drugs) or facilitate development of in situ cardiac dysfunction, especially with prolonged dietary Mg restriction. Significant protection against most of these MgD-mediated events has been observed with interventions that modulate neuronal SP release or its bioactivity, and with several antioxidants (vitamin E, probucol, epicaptopril, d-propranolol). In view of the clinical prevalence of hypomagnesemia, new treatments, beyond magnesium repletion, may be needed to diminish deleterious neurogenic and prooxidative components described in this article.


Asunto(s)
Cardiomiopatías , Deficiencia de Magnesio/complicaciones , Inflamación Neurogénica , Animales , Cardiomiopatías/etiología , Cardiomiopatías/fisiopatología , Dieta , Endotoxemia/metabolismo , Humanos , Daño por Reperfusión Miocárdica/metabolismo , Inflamación Neurogénica/etiología , Inflamación Neurogénica/fisiopatología , Neuropéptidos/metabolismo , Estrés Oxidativo , Ratas , Receptores de N-Metil-D-Aspartato/metabolismo , Receptores de Neuroquinina-1/metabolismo
10.
Exp Biol Med (Maywood) ; 231(4): 473-84, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16565443

RESUMEN

The benefits of acute D-propranolol (D-Pro, non-beta-adrenergic receptor blocker) pretreatment against enhanced ischemia/reperfusion (I/R) injury of hearts from moderate iron-overloaded rats were examined. Perfused hearts from iron-dextran-treated rats (450 mg/kg/week for 3 weeks, intraperitoneal administration) exhibited normal control function, despite iron treatment that elevated plasma iron and conjugated diene levels by 8.1-and 2.5-fold, respectively. However, these hearts were more susceptible to 25 mins of global I/R stress compared with non-loaded hearts; the coronary flow rate, aortic output, cardiac work, left ventricular systolic pressure, positive differential left ventricular pressure (dP/dt), and left ventricular developed pressure displayed 38%, 60%, 55%, 13%, 41%, and 15% lower recoveries, respectively, and a 6.5-fold increase in left ventricular end-diastolic pressure. Postischemic hearts from iron-loaded rats also exhibited 5.6-, 3.48-, 2.43-, and 3.45-fold increases in total effluent iron content, conjugated diene levels, lactate dehydrogenase (LDH) activity, and lysosomal N-acetyl-beta-glucosaminidase (NAGA) activity, respectively, compared with similarly stressed non-loaded hearts. A comparison of detection time profiles during reperfusion suggests that most of the oxidative injury (conjugated diene) in hearts from iron-loaded rats occurred at later times of reperfusion (8.5-15 mins), and this corresponded with heightened tissue iron and NAGA release. D-Pro (2 microM infused for 30 mins) pretreatment before ischemia protected all parameters compared with the untreated iron-loaded group; pressure indices improved 1.2- to 1.6-fold, flow parameters improved 1.70- to 2.96-fold, cardiac work improved 2.87-fold, and end-diastolic pressure was reduced 56%. D-Pro lowered total release of tissue iron, conjugated diene content, LDH activity, and NAGA activity 4.59-, 2.55-, 3.04-, and 4.14-fold, respectively, in the effluent of I/R hearts from the iron-loaded group. These findings suggest that the enhanced postischemic dysfunction and tissue injury of hearts from iron-loaded rats was caused by excessive iron-catalyzed free radical stress, and that the membrane antioxidant properties of D-Pro and its stabilization of sequestered lysosomal iron by D-Pro may contribute to the cardioprotective actions of D-Pro.


Asunto(s)
Corazón/efectos de los fármacos , Complejo Hierro-Dextran/farmacología , Daño por Reperfusión Miocárdica/prevención & control , Propranolol/farmacología , Animales , Antioxidantes/farmacología , Presión Sanguínea/efectos de los fármacos , Gasto Cardíaco/efectos de los fármacos , Circulación Coronaria/efectos de los fármacos , Corazón/fisiología , Técnicas In Vitro , Complejo Hierro-Dextran/sangre , Hígado/metabolismo , Lisosomas/metabolismo , Masculino , Ratas , Ratas Sprague-Dawley
11.
J Pharmacol Exp Ther ; 317(2): 522-8, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16456084

RESUMEN

The influence of selected beta-receptor blockers on iron overload and oxidative stress in endothelial cells (ECs) was assessed. Confluent bovine ECs were loaded with iron dextran (15 muM) for 24 h and then exposed to dihydroxyfumarate (DHF), a source of reactive oxygen species, for up to 2 h. Intracellular oxidant formation, monitored by fluorescence of 2',7'-dichlorofluorescin (DCF; 30 microM), increased and peaked at 30 min; total glutathione decreased by 52 +/- 5% (p < 0.01) at 60 min. When the ECs were pretreated 30 min before iron loading with 1.25 to 10 microM d-propranolol, glutathione losses were attenuated 15 to 80%, with EC(50) = 3.1 microM. d-Propranolol partially inhibited the DCF intensity increase, but atenolol up to 10 microM was ineffective. At 2 h, caspase 3 activity was elevated 3.2 +/- 0.3-fold (p < 0.01) in the iron-loaded and DHF-treated ECs, and cell survival, determined 24 h later, decreased 47 +/- 6% (p < 0.01). Ten micromoles of d-propranolol suppressed the caspase 3 activation by 63% (p < 0.05) and preserved cell survival back to 88% of control (p < 0.01). In separate experiments, 24-h iron loading resulted in a 3.6 +/- 0.8-fold increase in total EC iron determined by atomic absorption spectroscopy; d-propranolol at 5 microM reduced this increase to 1.5 +/- 0.4-fold (p < 0.01) of controls. Microscopic observation by Perls' staining revealed that the excessive iron accumulated in vesicular endosomal/lysosomal structures, which were substantially diminished by d-propranolol. We previously showed that propranolol could readily concentrate into the lysosomes and raise the intralysosomal pH; it is suggested that the lysosomotropic properties of d-propranolol retarded the EC iron accumulation and thereby conferred the protective effects against iron load-mediated cytotoxicity.


Asunto(s)
Antagonistas Adrenérgicos beta/farmacología , Células Endoteliales/efectos de los fármacos , Hierro/metabolismo , Lisosomas/efectos de los fármacos , Estrés Oxidativo , Propranolol/farmacología , Animales , Caspasa 3 , Caspasas/metabolismo , Bovinos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Células Endoteliales/enzimología , Células Endoteliales/metabolismo , Glutatión/metabolismo , Lisosomas/enzimología , Lisosomas/metabolismo , Especies Reactivas de Oxígeno/metabolismo
12.
Cardiovasc Toxicol ; 4(2): 109-15, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15371628

RESUMEN

The effects of zidovudine (AZT) and AZT-monophosphate (AZT-MP) on lipid peroxidation and oxidative cell injury were studied. When microsomal membranes from rat livers were peroxidized by a superoxide-driven, Fe-catalyzed oxy-radical system (ORS), both AZT-MP and, to a lesser extent AZT, but not thymidine, concentration dependently (2-100 microM) enhanced lipid peroxidation (TBARS formation) up to 51% above control. Significance (p < 0.05) was achieved by 6.7 microM AZT-MP. When cultured bovine aortic endothelial cells were incubated with the ORS for 60 min, total glutathione (GSH) decreased by 40% and 24-h cell survival, determined by the tetrazolium salt MTT assay, decreased by 38%. Using this cell system, AZT-MP (7-100 microM) promoted cell death further; at 20 microM 50% (p < 0.01), cell death was induced. In comparison, AZT was less effective. Concurrently, AZT-MP significantly promoted ORS-mediated loss of GSH. These cytotoxic effects were further exacerbated by low extracellular magnesium. Interestingly, when the endothelial cells were exposed to an iron-independent peroxynitrite generating system (SIN-1), the AZT-MP effects were absent. We propose that these pro-oxidant properties of AZT-MP are iron dependent. Because AZT-MP is a major phosphorylated metabolite, the data suggest that potential pro-oxidative activities may be associated with AZT use when catalytic iron is present.


Asunto(s)
Molsidomina/análogos & derivados , Oxidantes/toxicidad , Nucleótidos de Timina/toxicidad , Zidovudina/análogos & derivados , Zidovudina/toxicidad , Animales , Aorta/citología , Bovinos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Didesoxinucleótidos , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Radicales Libres/metabolismo , Glutatión/metabolismo , Técnicas In Vitro , Membranas Intracelulares/efectos de los fármacos , Membranas Intracelulares/metabolismo , Peroxidación de Lípido/efectos de los fármacos , Magnesio/metabolismo , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/metabolismo , Molsidomina/farmacología , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Timidina/farmacología
13.
Cardiovasc Toxicol ; 4(2): 169-77, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15371632

RESUMEN

Treatment of HIV with AZT (zidovudine) may have toxic side effects as a result of multiple mechanisms. It is known that patients with AIDS may suffer from magnesium deficiency (MgD). We studied selected biochemical and histopathologic consequences of AZT administration (0.7 mg/mL in drinking water) with concurrent Mg-deficient (20% of normal) diet in male C57Bl/6N mice for 3 wk. Significant decreases in red blood cell glutathione (GSH) were evident in the Mg-deficient mice with or without AZT treatment, suggesting compromised antioxidant capacity in the blood. Although MgD alone led to a 1.9-fold increase in plasma thromboxane B(2) (TXB(2), derived from the highly vasoconstrictive TXA(2)), AZT + MgD increased the TXB(2) level 3.5-fold. AZT (+/-MgD) provoked prominent hepatic damage expressed by distortion of lobular architecture, nuclear and cellular swelling, and inflammatory lesions and loss of hepatocytes. AZT alone caused mild cardiac lesions, resulting in partial cardiac fibrosis, especially in the atrium. AZT + MgD caused only scattered small-size cardiac lesions consisting of microscopic foci of inflammatory infiltrates in the ventricles but led to more prominent lesions, fibrosis, and scars in the atrium. MgD or AZT alone caused varying degrees of skeletal muscle degeneration; in combination, more intense degeneration and regeneration of muscle cells were evident. In conclusion, it is suggested that both the decreased blood GSH and elevated plasma TXA(2) might contribute, at least in part, to the aggravated pathological damages observed in the atrium and skeletal muscle of the AZT-treated Mg-deficient mice.


Asunto(s)
Fármacos Anti-VIH/toxicidad , Cardiopatías/inducido químicamente , Deficiencia de Magnesio/metabolismo , Miocardio/patología , Zidovudina/toxicidad , Animales , Glutatión/sangre , Cardiopatías/patología , Hígado/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/patología , Tromboxano B2/sangre
14.
J Pharmacol Exp Ther ; 308(1): 85-90, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14569064

RESUMEN

The antioxidant properties of 4-HO-propranolol (4HOP), a major metabolite of propranolol, were studied and compared with that of propranolol and vitamin E (Trolox). When isolated hepatic microsomal membranes were peroxidized by an iron-catalyzed.OH-generating system [dihydroxyfumarate +Fe (III)], 4HOP potently and concentration-dependently inhibited lipid peroxidation; the IC50 value was 1.1 microM, whereas those for Trolox and propranolol were 4.3 and 168 microM, respectively. When isolated human low-density lipoprotein (LDL) was oxidized by 7.5 microM Cu(II) for 9 h, 4HOP at 3 microM delayed the lag phase significantly by 108 min, which was comparable with that of probucol (98-min delay) but was far greater than that provided by propranolol (6 min) or Trolox (47 min). At 1 microM 4HOP, the delay was 45 min. When confluent cultured bovine aortic endothelial cells were exposed to the Fe-catalyzed oxy-radical system, acute loss of glutathione occurred (55% decrease in 50 min). Pretreatment of the cells with 0.067 to 6.7 microM 4HOP for 30 min provided increasing degrees of protection against the glutathione loss; the EC50 value was 1.2 microM, whereas those for Trolox and propranolol were 7.9 and 49 microM, respectively. The loss of cell survival due to the radical stress was also effectively preserved by 4HOP. In separate experiments, when the endothelial glutathione was oxidatively depleted by a peroxynitrite-generating system (3-morpholinosydnonimine), 4HOP also provided potent protective activities. In conclusion, 4HOP is 4- to 8-fold more potent than vitamin E and >100-fold more active than propranolol as a "chain-breaking" antiperoxidatant against membrane and LDL oxidation and can provide superior endothelial cytoprotective efficacy against oxygen- or nitrogen-derived oxidant-mediated cell injury. Being a major metabolite in human and with its plasma level approaching that of propranolol, 4-HO-propranolol may contribute, in part, to the cardiovascular therapeutic benefits of propranolol.


Asunto(s)
Antioxidantes/farmacología , Lipoproteínas LDL/metabolismo , Molsidomina/análogos & derivados , Propranolol/análogos & derivados , Propranolol/farmacología , Animales , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Interacciones Farmacológicas , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Glutatión/metabolismo , Humanos , Radical Hidroxilo/metabolismo , Masculino , Molsidomina/antagonistas & inhibidores , Molsidomina/farmacología , Oxidación-Reducción/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/farmacología
15.
Magnes Res ; 16(2): 91-7, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12892378

RESUMEN

The regulatory role of substance-P (SP) on neutrophil and endothelium activation as well as nitric oxide (NO) production induced by Mg-deficiency was examined. Male Sprague-Dawley rats (180 g) were fed either a Mg-deficient (MgD) or Mg-sufficient (MgS) diet for 3 weeks. Enriched neutrophil fractions (> 85%) isolated from whole blood of the Mg-deficient rats displayed an 11-fold (p < 0.001) higher basal superoxide anion producing activity (assayed as SOD-inhibitable cytochrome c reduction) compared to that obtained from the MgS rats. Treatment of the MgD rats with the specific SP-receptor (SPR) blocker, L-703,606 (1 mg/kg/day as s.c. implanted sustained-release pellets) attenuated the superoxide anion producing activity by 75% (p < 0.025). In parallel, circulating prostacyclin (PGI2) level (assayed as 6-keto-PGF-1alpha) was elevated 13-fold in the MgD rats, but was reduced 90% by L-703,606 treatment. Concomitantly, plasma NO products (nitrate + nitrite), which increased 2.2-fold during Mg-deficiency, were completely suppressed by the SPR blockade. When the isolated hearts were subjected to ischemia/ reperfusion stress, NO products were elevated 2.4-fold in the effluent of the MgD group compared to MgS; such heightened NO release was also attenuated after in vivo treatment with the SPR blocker. In conclusion, SP plays a direct role in promoting activation of the neutrophil and endothelium as well as induction of NO production; these processes might participate in the oxidative stress that contributes to the depletion of blood glutathione and cardiac pathology.


Asunto(s)
Células Endoteliales/citología , Deficiencia de Magnesio , Antagonistas del Receptor de Neuroquinina-1 , Neutrófilos/citología , Animales , Aniones , Epoprostenol/metabolismo , Glutatión/sangre , Magnesio/metabolismo , Masculino , Neutrófilos/metabolismo , Óxido Nítrico/metabolismo , Perfusión , Quinuclidinas/farmacología , Ratas , Ratas Sprague-Dawley , Superóxidos
16.
Exp Biol Med (Maywood) ; 228(6): 665-73, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12773697

RESUMEN

Severe dietary Mg restriction (Mg(9), 9% of recommended daily allowance [RDA], plasma Mg = 0.25 mM) induces a pro-inflammatory neurogenic response in rats (substance P [SP]), and the associated increases in oxidative stress in vivo and cardiac susceptibility to ischemia/reperfusion (I/R) injury were previously shown to be attenuated by SP receptor blockade and antioxidant treatment. The present study assessed if less severe dietary Mg restriction modulates the extent of both the neurogenic/oxidative responses in vivo and I/R injury in vitro. Male Sprague-Dawley rats maintained on Mg(40) (40% RDA, plasma Mg = 0.6 mM) or Mg(100) (100% RDA, plasma Mg = 0.8 mM) diets were assessed for plasma SP levels (CHEM-ELISA) during the first 3 weeks and were compared with the Mg(9) group; red blood cell (RBC) glutathione and plasma malondialdehyde levels were compared at 3 weeks in Mg(9), Mg(20) (plasma Mg = 0.4 mM), Mg(40), and Mg(100) rats; and 40-min global ischemia/30-min reperfusion hearts from 7-week-old Mg(20), Mg(40), and Mg(100) rats were compared with respect to functional recovery (cardiac work, and diastolic, systolic, and developed pressures), tissue LDH release, and free radical production (ESR spectroscopy and alpha-phenyl-N-tert butylnitrone [PBN; 3 mM] spin trapping). The Mg(40) diet induced smaller elevations in plasma SP (50% lower) compared with Mg(9), but with a nearly identical time course. RBC glutathione and plasma malondialdehyde levels revealed a direct relationship between the severity of oxidative stress and hypomagnesemia. The dominant lipid free radical species detected in all I/R groups was the alkoxyl radical (PBN/alkoxyl: alpha(H) = 1.93 G, alpha(N) = 13.63 G); however, Mg(40) and Mg(20) hearts exhibited 2.7- and 3.9-fold higher alkoxyl levels, 40% and 65% greater LDH release, and lower functional recovery (Mg(20) < Mg(40)) compared with Mg(100). Our data suggest that varying dietary Mg intake directly influences the magnitude of the neurogenic/oxidative responses in vivo and the resultant myocardial tolerance to I/R stress.


Asunto(s)
Deficiencia de Magnesio/sangre , Deficiencia de Magnesio/complicaciones , Magnesio/farmacología , Isquemia Miocárdica/sangre , Daño por Reperfusión Miocárdica/sangre , Inflamación Neurogénica/sangre , Sustancia P/sangre , Animales , Modelos Animales de Enfermedad , Espectroscopía de Resonancia por Spin del Electrón , Ensayo de Inmunoadsorción Enzimática/métodos , Eritrocitos/efectos de los fármacos , Eritrocitos/metabolismo , Glutatión/sangre , Hemodinámica/efectos de los fármacos , Hemodinámica/fisiología , Magnesio/sangre , Masculino , Malondialdehído/sangre , Isquemia Miocárdica/fisiopatología , Estrés Oxidativo , Ratas , Ratas Sprague-Dawley , Detección de Spin
17.
Pharmacol Res ; 45(1): 27-33, 2002 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11820858

RESUMEN

Nitric oxide (NO) and superoxide anions (*O(2)(-) ), which are known to be generated by inflammatory cells under certain pathological conditions, may be cytotoxic to the endothelial cells (ECs) due to peroxynitrite formation. We reported previously that certain lipophilic dihydropyridine (DHP) Ca-blockers exhibit antioxidant activities. In the present study, the extent of antioxidant protection by nisoldipine against combined NO/*O(2)(-) or peroxynitrite-mediated EC injury was assessed and compared with nicardipine, nifedipine and Trolox (water-soluble vitamin E). When confluent bovine aortic ECs were exposed to combined NO/*O(2)(-) (generated from 0.25 mM SIN-1), dramatic loss of cell GSH (53 +/- 8%) occurred in 60 min; cell survival/viability, determined 24 h later by the tetrazolium MTT assay, decreased by 45 +/- 6%. NO alone or O(2)(-) alone were ineffective. Nisoldipine pretreatment (30 min) of the cells concentration dependently (0.3-10 micro M) attenuated the SIN-1-induced GSH loss: the EC(50)value was 4.7 micro M and the corresponding values for nicardipine and nifedipine were 7.8 micro M and >20 micro M, respectively, and that for Trolox was 5.2 micro M. These agents (10 micro M) also protected against the loss of cell viability: nisoldipine, 86 plus minus 8%; nicardipine, 60 +/- 7 %; nifedipine, 35 +/- 5 %, and Trolox, 78 +/- 9%. In addition, significant losses of GSH and viability could be induced by incubation of the EC monolayers with purified peroxynitrite (25 micro M). Attenuation of these peroxynitrite-mediated GSH and viability losses was observed with the following order of efficacy: nisoldipine > or = Trolox > nicardipine >> nifedipine. In a cell-free system containing 0.05 mM GSH, none of the agents (10 micro M) were able to inhibit SIN-1- (0.25 mM) or peroxynitrite- (25 micro M) induced depletion (approximately 50%) of GSH. However, with a purified microsomal membrane system, all four agents inhibited the SIN-1- (or peroxynitrite-) induced lipid peroxidation (TBARS) with the following IC(50)values: nisoldipine, 6.3 micro M; nicardipine, 10.6 micro M; nifedipine, >20 micro M, and Trolox, 6.5 micro M. In conclusion, nisoldipine, a vascular selective DHP calcium channel-blocker, demonstrated the greatest protection against the EC injury induced either by SIN-1 or peroxynitrite. The protective mechanism against the cytotoxicity is most likely through a lipophilic 'chain-breaking' antiperoxidative action against the 'OH-like species' released from peroxynitrite or SIN-1.


Asunto(s)
Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio Tipo L/metabolismo , Endotelio Vascular/efectos de los fármacos , Molsidomina/análogos & derivados , Óxido Nítrico/farmacología , Superóxidos/farmacología , Animales , Bovinos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Cromanos/farmacología , Relación Dosis-Respuesta a Droga , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Glutatión/metabolismo , Peroxidación de Lípido/efectos de los fármacos , Molsidomina/farmacología , Nicardipino/farmacología , Nifedipino/farmacología , Nisoldipino/farmacología , Estrés Oxidativo/efectos de los fármacos , Ácido Peroxinitroso/farmacología , Sustancias Reactivas al Ácido Tiobarbitúrico/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA