Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 24(13)2023 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-37446386

RESUMEN

Connexin 43 (Cx43) is expressed in the left and right ventricles and is primarily responsible for conducting physiological responses in microvasculature. Studies have demonstrated that NADPH oxidase (NOX) enzymes are essential in cardiac redox biology and are responsible for the generation of reactive oxygen species (ROS). NOX2 is linked to left ventricular remodeling following myocardial infarction (MI). It was hypothesized that conjugated linoleic acid (cLA) treatment increases NOX-2 levels in heart tissue and disrupts connexins between the myocytes in the ventricle. Data herein demonstrate that cLA treatment significantly decreases survival in a murine model of MI. The observance of cLA-induced ventricular tachyarrhythmia's (VT) led to the subsequent investigation of the underlying mechanism in this MI model. Mice were treated with cLA for 12 h, 24 h, 48 h, or 72 h to determine possible time-dependent changes in NOX and Cx43 signaling pathways in isolated left ventricles (LV) extracted from cardiac tissue. The results suggest that ROS generation, through the stimulation of NOX2 in the LV, triggers a decrease in Cx43 levels, causing dysfunction of the gap junctions following treatment with cLA. This cascade of events may initiate VT and subsequent death during MI. Taken together, individuals at risk of MI should use caution regarding cLA consumption.


Asunto(s)
Ácidos Linoleicos Conjugados , Infarto del Miocardio , Ratones , Animales , Conexina 43/metabolismo , Ácidos Linoleicos Conjugados/farmacología , Especies Reactivas de Oxígeno/metabolismo , Infarto del Miocardio/metabolismo , NADPH Oxidasas/metabolismo , Conexinas/metabolismo , Muerte Súbita , Remodelación Ventricular
2.
J Cell Physiol ; 233(7): 5214-5221, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29150941

RESUMEN

The occurrence of myocardial infarction (MI) increases appreciably with age. In the Framingham Heart Study, the incidence of MI more than doubles for men and increases more than five-fold in women (ages 55-64 years compared to 85-94 years). MicroRNAs (miRNAs) quantitatively regulate their target's expression post-transcriptionally by either silencing action through binding at the 3'UTR domains or degrading the messages at their coding regions. In either case, these regulations affect the cardiac transcriptional output and cardiac function. Among the known cardiac associated miRNA, miRNA-1, miRNA-133a, and miRNA-34a have been shown to induce adverse structural remodeling to impair cardiac contractile function. In the present study, an in vivo model of MI in young (3 month) and old (22 month) mice is used to investigate the possible role whereby these three miRNAs exert negative effects on heart function following MI. Herein we demonstrate that in older mouse heart, all three microRNAs show increased levels of expression, while miRNA-1 shows a further increase in old mouse heart following MI, which corresponds to left ventricular (LV) wall thinning. These structural changes in cardiac tissue may causes downstream LV dilation and subsequent LV dysfunction. Results presented here suggest that significantly elevated levels of miRNA-1 in post-MI old heart could be predictive of cardiac injury in older mice as the high risk biomarker for MI in older individuals.


Asunto(s)
Envejecimiento/genética , Lesiones Cardíacas/genética , MicroARNs/genética , Infarto del Miocardio/genética , Anciano , Anciano de 80 o más Años , Animales , Modelos Animales de Enfermedad , Femenino , Lesiones Cardíacas/fisiopatología , Humanos , Masculino , Ratones , Persona de Mediana Edad , Infarto del Miocardio/fisiopatología , Caracteres Sexuales , Función Ventricular Izquierda/genética
3.
Exp Cell Res ; 347(1): 222-231, 2016 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-27515002

RESUMEN

Acquired tamoxifen (TAM) resistance is a significant clinical problem in treating patients with estrogen receptor α (ERα)+ breast cancer. We reported that ERα increases nuclear respiratory factor-1 (NRF-1), which regulates nuclear-encoded mitochondrial gene transcription, in MCF-7 breast cancer cells and NRF-1 knockdown stimulates apoptosis. Whether NRF-1 and target gene expression is altered in endocrine resistant breast cancer cells is unknown. We measured NRF-1and metabolic features in a cell model of progressive TAM-resistance. NRF-1 and its target mitochondrial transcription factor A (TFAM) were higher in TAM-resistant LCC2 and LCC9 cells than TAM-sensitive MCF-7 cells. Using extracellular flux assays we observed that LCC1, LCC2, and LCC9 cells showed similar oxygen consumption rate (OCR), but lower mitochondrial reserve capacity which was correlated with lower Succinate Dehydrogenase Complex, Subunit B in LCC1 and LCC2 cells. Complex III activity was lower in LCC9 than MCF-7 cells. LCC1, LCC2, and LCC9 cells had higher basal extracellular acidification (ECAR), indicating higher aerobic glycolysis, relative to MCF-7 cells. Mitochondrial bioenergetic responses to estradiol and 4-hydroxytamoxifen were reduced in the endocrine-resistant cells compared to MCF-7 cells. These results suggest the acquisition of altered metabolic phenotypes in response to long term antiestrogen treatment may increase vulnerability to metabolic stress.


Asunto(s)
Neoplasias de la Mama/metabolismo , Resistencia a Antineoplásicos/efectos de los fármacos , Metabolismo Energético , Factor Nuclear 1 de Respiración/metabolismo , Tamoxifeno/farmacología , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Respiración de la Célula/efectos de los fármacos , ADN Mitocondrial/metabolismo , Proteínas de Unión al ADN/metabolismo , Complejo III de Transporte de Electrones/metabolismo , Estradiol/farmacología , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Ontología de Genes , Humanos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Fosforilación Oxidativa/efectos de los fármacos , Subunidades de Proteína/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Análisis de Secuencia de ARN , Tamoxifeno/análogos & derivados , Factores de Transcripción/metabolismo
4.
Neurobiol Dis ; 84: 4-21, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26024962

RESUMEN

Aerobes require oxygen for metabolism and normal free radical formation. As a result, maintaining the redox homeostasis is essential for brain cell survival due to their high metabolic energy requirement to sustain electrochemical gradients, neurotransmitter release, and membrane lipid stability. Further, brain antioxidant levels are limited compared to other organs and less able to compensate for reactive oxygen and nitrogen species (ROS/RNS) generation which contribute oxidative/nitrative stress (OS/NS). Antioxidant treatments such as vitamin E, minocycline, and resveratrol mediate neuroprotection by prolonging the incidence of or reversing OS and NS conditions. Redox imbalance occurs when the antioxidant capacity is overwhelmed, consequently leading to activation of alternate pathways that remain quiescent under normal conditions. If OS/NS fails to lead to adaptation, tissue damage and injury ensue, resulting in cell death and/or disease. The progression of OS/NS-mediated neurodegeneration along with contributions from microglial activation, dopamine metabolism, and diabetes comprise a detailed interconnected pathway. This review proposes a significant role for OS/NS and more specifically, lipid peroxidation (LPO) and other lipid modifications, by triggering microglial activation to elicit a neuroinflammatory state potentiated by diabetes or abnormal dopamine metabolism. Subsequently, sustained stress in the neuroinflammatory state overwhelms cellular defenses and prompts neurotoxicity resulting in the onset or amplification of brain damage.


Asunto(s)
Enfermedades Neurodegenerativas/metabolismo , Estrés Oxidativo/fisiología , Animales , Encéfalo/metabolismo , Humanos
5.
J Biol Chem ; 287(53): 44071-82, 2012 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-23144452

RESUMEN

The oxidation and nitration of unsaturated fatty acids by oxides of nitrogen yield electrophilic derivatives that can modulate protein function via post-translational protein modifications. The biological mechanisms accounting for fatty acid nitration and the specific structural characteristics of products remain to be defined. Herein, conjugated linoleic acid (CLA) is identified as the primary endogenous substrate for fatty acid nitration in vitro and in vivo, yielding up to 10(5) greater extent of nitration products as compared with bis-allylic linoleic acid. Multiple enzymatic and cellular mechanisms account for CLA nitration, including reactions catalyzed by mitochondria, activated macrophages, and gastric acidification. Nitroalkene derivatives of CLA and their metabolites are detected in the plasma of healthy humans and are increased in tissues undergoing episodes of ischemia reperfusion. Dietary CLA and nitrite supplementation in rodents elevates NO(2)-CLA levels in plasma, urine, and tissues, which in turn induces heme oxygenase-1 (HO-1) expression in the colonic epithelium. These results affirm that metabolic and inflammatory reactions yield electrophilic products that can modulate adaptive cell signaling mechanisms.


Asunto(s)
Ácidos Grasos/metabolismo , Ácido Linoleico/metabolismo , Nitratos/metabolismo , Nitritos/metabolismo , Animales , Línea Celular , Humanos , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Óxido Nítrico/metabolismo , Transducción de Señal
6.
Nat Chem Biol ; 6(6): 433-41, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20436486

RESUMEN

Electrophilic fatty acids are generated during inflammation by non-enzymatic reactions and can modulate inflammatory responses. We used a new mass spectrometry-based electrophile capture strategy to reveal the formation of electrophilic oxo-derivatives (EFOX) from the omega-3 fatty acids docosahexaenoic acid (DHA) and docosapentaenoic acid (DPA). These EFOX were generated by a cyclooxygenase-2 (COX-2)-catalyzed mechanism in activated macrophages. Modulation of COX-2 activity by aspirin increased the rate of EFOX production and their intracellular levels. Owing to their electrophilic nature, EFOX adducted to cysteine and histidine residues of proteins and activated Nrf2-dependent anti-oxidant gene expression. We confirmed the anti-inflammatory nature of DHA- and DPA-derived EFOX by showing that they can act as peroxisome proliferator-activated receptor-gamma (PPAR gamma) agonists and inhibit pro-inflammatory cytokine and nitric oxide production, all within biological concentration ranges. These data support the idea that EFOX are signaling mediators that transduce the beneficial clinical effects of omega-3 fatty acids, COX-2 and aspirin.


Asunto(s)
Antiinflamatorios/síntesis química , Ciclooxigenasa 2/metabolismo , Ácidos Grasos Omega-3/metabolismo , Antiinflamatorios/metabolismo , Antiinflamatorios/farmacología , Borohidruros/farmacología , Línea Celular , Membrana Celular/metabolismo , Ácidos Docosahexaenoicos/química , Ácidos Docosahexaenoicos/metabolismo , Ácidos Grasos Insaturados/metabolismo , Ácidos Grasos Insaturados/farmacología , Glutatión/metabolismo , Humanos , Hidroxilación , Interleucina-10/genética , Interleucina-6/genética , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , PPAR gamma/metabolismo , PPAR gamma/farmacología
7.
Circ Res ; 107(7): 877-87, 2010 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-20705923

RESUMEN

RATIONALE: Low-dose acetylsalicylic acid (aspirin) is widely used in the treatment and prevention of vascular atherothrombosis. Cardiovascular doses of aspirin also reduce systemic blood pressure and improve endothelium-dependent vasorelaxation in patients with atherosclerosis or risk factors for atherosclerosis. Aspirin can acetylate proteins, other than its pharmacological target cyclooxygenase, at lysine residues. The role of lysine acetylation in mediating the effects of low-dose aspirin on the endothelium is not known. OBJECTIVE: To determine the role of lysine acetylation of endothelial nitric oxide synthase (eNOS) in the regulation of endothelial NO production by low-dose aspirin and to examine whether the lysine deacetylase histone deacetylase (HDAC)3 antagonizes the effect of low-dose aspirin on endothelial NO production by reversing acetylation of functionally critical eNOS lysine residues. METHODS AND RESULTS: Low concentrations of aspirin induce lysine acetylation of eNOS, stimulating eNOS enzymatic activity and endothelial NO production in a cyclooxygenase-1-independent fashion. Low-dose aspirin in vivo also increases bioavailable vascular NO in an eNOS-dependent and cyclooxygenase-1-independent manner. Low-dose aspirin promotes the binding of eNOS to calmodulin. Lysine 609 in the calmodulin autoinhibitory domain of bovine eNOS mediates aspirin-stimulated binding of eNOS to calmodulin and eNOS-derived NO production. HDAC3 inhibits aspirin-stimulated (1) lysine acetylation of eNOS, (2) eNOS enzymatic activity, (3) eNOS-derived NO, and (4) binding of eNOS to calmodulin. Conversely, downregulation of HDAC3 promotes lysine acetylation of eNOS and endothelial NO generation. CONCLUSIONS: Lysine acetylation of eNOS is a posttranslational protein modification supporting low-dose aspirin-induced vasoprotection. HDAC3, by deacetylating aspirin-acetylated eNOS, antagonizes aspirin-stimulated endothelial production of NO.


Asunto(s)
Aspirina/farmacología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/enzimología , Histona Desacetilasas/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Acetilación/efectos de los fármacos , Animales , Calmodulina/metabolismo , Bovinos , Línea Celular , Relación Dosis-Respuesta a Droga , Células Endoteliales/citología , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Humanos , Riñón/citología , Lisina/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Óxido Nítrico Sintasa de Tipo III/genética , Inhibidores de Agregación Plaquetaria/farmacología , Procesamiento Proteico-Postraduccional/fisiología , Venas Umbilicales/citología
8.
Circ Res ; 107(4): 540-8, 2010 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-20558825

RESUMEN

RATIONALE: Nitro-oleic acid (OA-NO(2)) is a bioactive, nitric-oxide derived fatty acid with physiologically relevant vasculoprotective properties in vivo. OA-NO(2) exerts cell signaling actions as a result of its strong electrophilic nature and mediates pleiotropic cell responses in the vasculature. OBJECTIVE: The present study sought to investigate the protective role of OA-NO(2) in angiotensin (Ang) II-induced hypertension. METHODS AND RESULTS: We show that systemic administration of OA-NO(2) results in a sustained reduction of Ang II-induced hypertension in mice and exerts a significant blood pressure lowering effect on preexisting hypertension established by Ang II infusion. OA-NO(2) significantly inhibits Ang II contractile response as compared to oleic acid (OA) in mesenteric vessels. The improved vasoconstriction is specific for the Ang II type 1 receptor (AT(1)R)-mediated signaling because vascular contraction by other G-protein-coupled receptors is not altered in response to OA-NO(2) treatment. From the mechanistic viewpoint, OA-NO(2) lowers Ang II-induced hypertension independently of peroxisome proliferation-activated receptor (PPAR)gamma activation. Rather, OA-NO(2), but not OA, specifically binds to the AT(1)R, reduces heterotrimeric G-protein coupling, and inhibits IP(3) (inositol-1,4,5-trisphosphate) and calcium mobilization, without inhibiting Ang II binding to the receptor. CONCLUSIONS: These results demonstrate that OA-NO(2) diminishes the pressor response to Ang II and inhibits AT(1)R-dependent vasoconstriction, revealing OA-NO(2) as a novel antagonist of Ang II-induced hypertension.


Asunto(s)
Angiotensina II/antagonistas & inhibidores , Angiotensina II/toxicidad , Hipertensión/inducido químicamente , Hipertensión/prevención & control , Nitrocompuestos/uso terapéutico , Ácido Oléico/uso terapéutico , Angiotensina II/fisiología , Animales , Presión Sanguínea/efectos de los fármacos , Presión Sanguínea/fisiología , Hipertensión/fisiopatología , Ácidos Linoleicos/uso terapéutico , Ratones , Ratones Endogámicos C57BL , Ácidos Oléicos/uso terapéutico , Ratas , Ratas Sprague-Dawley
9.
J Biol Chem ; 285(16): 12321-33, 2010 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-20097754

RESUMEN

The peroxisome proliferator-activated receptor-gamma (PPARgamma) binds diverse ligands to transcriptionally regulate metabolism and inflammation. Activators of PPARgamma include lipids and anti-hyperglycemic drugs such as thiazolidinediones (TZDs). Recently, TZDs have raised concern after being linked with increased risk of peripheral edema, weight gain, and adverse cardiovascular events. Most reported endogenous PPARgamma ligands are intermediates of lipid metabolism and oxidation that bind PPARgamma with very low affinity. In contrast, nitro derivatives of unsaturated fatty acids (NO(2)-FA) are endogenous products of nitric oxide ((*)NO) and nitrite (NO(2)(-))-mediated redox reactions that activate PPARgamma at nanomolar concentrations. We report that NO(2)-FA act as partial agonists of PPARgamma and covalently bind PPARgamma at Cys-285 via Michael addition. NO(2)-FA show selective PPARgamma modulator characteristics by inducing coregulator protein interactions, PPARgamma-dependent expression of key target genes, and lipid accumulation is distinctively different from responses induced by the TZD rosiglitazone. Administration of this class of signaling mediators to ob/ob mice revealed that NO(2)-FA lower insulin and glucose levels without inducing adverse side effects such as the increased weight gain induced by TZDs.


Asunto(s)
Ácidos Grasos Insaturados/farmacología , Hipoglucemiantes/farmacología , Nitrocompuestos/farmacología , PPAR gamma/agonistas , PPAR gamma/metabolismo , Células 3T3-L1 , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Secuencia de Bases , Glucemia/metabolismo , Línea Celular , Cartilla de ADN/metabolismo , Diabetes Mellitus/tratamiento farmacológico , Diabetes Mellitus/metabolismo , Ácidos Grasos Insaturados/química , Humanos , Hipoglucemiantes/química , Técnicas In Vitro , Insulina/sangre , Ligandos , Metabolismo de los Lípidos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Nitrocompuestos/química , Ácido Oléico/química , Ácido Oléico/farmacología , PPAR gamma/química , PPAR gamma/genética , Unión Proteica , Procesamiento Proteico-Postraduccional , Estructura Terciaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Rosiglitazona , Transducción de Señal , Espectrometría de Masas en Tándem , Tiazolidinedionas/farmacología
10.
Circ Res ; 105(10): 965-72, 2009 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-19797175

RESUMEN

RATIONALE: Fatty acid nitroalkenes are endogenously generated electrophilic byproducts of nitric oxide and nitrite-dependent oxidative inflammatory reactions. Existing evidence indicates nitroalkenes support posttranslational protein modifications and transcriptional activation that promote the resolution of inflammation. OBJECTIVE: The aim of this study was to assess whether in vivo administration of a synthetic nitroalkene could elicit antiinflammatory actions in vivo using a murine model of vascular injury. METHODS AND RESULTS: The in vivo administration (21 days) of nitro-oleic acid (OA-NO(2)) inhibited neointimal hyperplasia after wire injury of the femoral artery in a murine model (OA-NO(2) treatment resulted in reduced intimal area and intima to media ratio versus vehicle- or oleic acid (OA)-treated animals,P<0.0001). Increased heme oxygenase (HO)-1 expression accounted for much of the vascular protection induced by OA-NO(2) in both cultured aortic smooth muscle cells and in vivo. Inhibition of HO by Sn(IV)-protoporphyrin or HO-1 small interfering RNA reversed OA-NO(2)-induced inhibition of platelet-derived growth factor-stimulated rat aortic smooth muscle cell migration. The upregulation of HO-1 expression also accounted for the antistenotic actions of OA-NO(2) in vivo, because inhibition of neointimal hyperplasia following femoral artery injury was abolished in HO-1(-/-) mice (OA-NO(2)-treated wild-type versus HO-1(-/-) mice, P=0.016). CONCLUSIONS: In summary, electrophilic nitro-fatty acids induce salutary gene expression and cell functional responses that are manifested by a clinically significant outcome, inhibition of neointimal hyperplasia induced by arterial injury.


Asunto(s)
Arteria Femoral/enzimología , Arteria Femoral/lesiones , Hemo Oxigenasa (Desciclizante)/biosíntesis , Nitrocompuestos/farmacología , Ácidos Oléicos/farmacología , Túnica Íntima/enzimología , Animales , Movimiento Celular/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Inflamación/metabolismo , Ratones , Ratones Noqueados , Óxido Nítrico/metabolismo , Nitrocompuestos/metabolismo , Ácidos Oléicos/metabolismo , Oxidación-Reducción/efectos de los fármacos , Factor de Crecimiento Derivado de Plaquetas/farmacología , Ratas , Regulación hacia Arriba/efectos de los fármacos
11.
Arterioscler Thromb Vasc Biol ; 30(5): 938-45, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20167658

RESUMEN

OBJECTIVE: Inflammatory processes and foam cell formation are key determinants in the initiation and progression of atherosclerosis. Electrophilic nitro-fatty acids, byproducts of nitric oxide- and nitrite-dependent redox reactions of unsaturated fatty acids, exhibit antiinflammatory signaling actions in inflammatory and vascular cell model systems. The in vivo action of nitro-fatty acids in chronic inflammatory processes such as atherosclerosis remains to be elucidated. METHODS AND RESULTS: Herein, we demonstrate that subcutaneously administered 9- and 10-nitro-octadecenoic acid (nitro-oleic acid) potently reduced atherosclerotic lesion formation in apolipoprotein E-deficient mice. Nitro-fatty acids did not modulate serum lipoprotein profiles. Immunostaining and gene expression analyses revealed that nitro-oleic acid attenuated lesion formation by suppressing tissue oxidant generation, inhibiting adhesion molecule expression, and decreasing vessel wall infiltration of inflammatory cells. In addition, nitro-oleic acid reduced foam cell formation by attenuating oxidized low-density lipoprotein-induced phosphorylation of signal transducer and activator of transcription-1, a transcription factor linked to foam cell formation in atherosclerotic plaques. Atherosclerotic lesions of nitro-oleic acid-treated animals also showed an increased content of collagen and alpha-smooth muscle actin, suggesting conferral of higher plaque stability. CONCLUSION: These results reveal the antiatherogenic actions of electrophilic nitro-fatty acids in a murine model of atherosclerosis.


Asunto(s)
Antiinflamatorios/farmacología , Antioxidantes/farmacología , Enfermedades de la Aorta/prevención & control , Apolipoproteínas E/deficiencia , Aterosclerosis/prevención & control , Ácidos Oléicos/farmacología , Actinas/metabolismo , Animales , Antiinflamatorios/administración & dosificación , Antioxidantes/administración & dosificación , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/metabolismo , Enfermedades de la Aorta/patología , Apolipoproteínas E/genética , Aterosclerosis/genética , Aterosclerosis/metabolismo , Aterosclerosis/patología , Moléculas de Adhesión Celular/metabolismo , Células Cultivadas , Quimiocina CCL2/metabolismo , Colágeno/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Células Espumosas/efectos de los fármacos , Células Espumosas/metabolismo , Inyecciones Subcutáneas , Lipoproteínas LDL/metabolismo , Masculino , Ratones , Ratones Noqueados , Ácidos Oléicos/administración & dosificación , Oxidantes/metabolismo , Estrés Oxidativo/efectos de los fármacos , Fosforilación , Factor de Transcripción STAT1/metabolismo , Transducción de Señal/efectos de los fármacos
12.
Circ Res ; 103(12): 1441-50, 2008 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-18988897

RESUMEN

The transcription factor, p53, and the adaptor protein, p66shc, both play essential roles in promoting oxidative stress in the vascular system. However, the relationship between the two in the context of endothelium-dependent vascular tone is unknown. Here, we report a novel, evolutionarily conserved, p53-mediated transcriptional mechanism that regulates p66shc expression and identify p53 as an important determinant of endothelium-dependent vasomotor function. We provide evidence of a p53 response element in the promoter of p66shc and show that angiotensin II-induced upregulation of p66shc in endothelial cells is dependent on p53. In addition, we demonstrate that downregulation of p66shc expression, as well as inhibition of p53 function in mice, mitigates angiotensin II-induced impairment of endothelium-dependent vasorelaxation, decrease in bioavailable nitric oxide, and hypertension. These findings reveal a novel p53-dependent transcriptional mechanism for the regulation of p66shc expression that is operative in the vascular endothelium and suggest that this mechanism is important in impairing endothelium-dependent vascular relaxation.


Asunto(s)
Endotelio Vascular/fisiopatología , Proteínas Adaptadoras de la Señalización Shc/biosíntesis , Transcripción Genética/fisiología , Proteína p53 Supresora de Tumor/fisiología , Regulación hacia Arriba/fisiología , Sistema Vasomotor/fisiopatología , Animales , Línea Celular , Endotelio Vascular/metabolismo , Endotelio Vascular/fisiología , Humanos , Ratones , Ratas , Ratas Endogámicas WKY , Proteínas Adaptadoras de la Señalización Shc/genética , Proteínas Adaptadoras de la Señalización Shc/fisiología , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src , Vasodilatación/genética , Vasodilatación/fisiología , Sistema Vasomotor/fisiología
13.
Free Radic Biol Med ; 44(8): 1610-6, 2008 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-18280257

RESUMEN

Retinoids are signaling molecules that are involved in proliferation, differentiation, and apoptosis during development. Retinoids exert their effects, in part, by binding to nuclear receptors, thereby altering gene expression. Clinical use of retinoids in the treatment of neuroblastoma is of interest due to their success in management of acute promyelocytic leukemia. Using the SK-N-SH human neuroblastoma cell line we investigated the effects of the differentiation agent all-trans-retinoic acid (ATRA) on the expression of manganese superoxide dismutase (MnSOD), an enzyme previously shown to enhance differentiation in vitro. Manganese superoxide dismutase mRNA, protein, and activity levels increased in a time-dependent manner upon treatment with ATRA. Nuclear levels of the NF-kappaB proteins p50 and p65 increased within 24 h of ATRA administration. This increase paralleled the degradation of the cytoplasmic inhibitor IkappaB-beta. Furthermore an increase in DNA binding to a NF-kappaB element occurred within a 342-bp enhancer (I2E) of the SOD2 gene with 10 microM ATRA treatment. Reporter analysis showed that ATRA-mediated I2E-dependent luciferase expression was attenuated upon mutation of the NF-kappaB element, suggesting a contribution of this transcription factor to retinoid-mediated upregulation of MnSOD. This study identifies SOD2 as a retinoid-responsive gene and demonstrates activation of the NF-kappaB pathway in response to ATRA treatment of SK-N-SH cells. These results suggest that signaling events involving NF-kappaB and SOD2 may contribute to the effects of retinoids used in cancer therapy.


Asunto(s)
FN-kappa B/metabolismo , Neuroblastoma/enzimología , Superóxido Dismutasa/biosíntesis , Tretinoina/farmacología , Inducción Enzimática , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Humanos , Proteínas I-kappa B/metabolismo , Neuroblastoma/metabolismo , ARN Mensajero/efectos de los fármacos , Células Tumorales Cultivadas , Regulación hacia Arriba
14.
Free Radic Biol Med ; 42(8): 1193-200, 2007 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-17382200

RESUMEN

The Mn(III) meso-tetrakis(N-ethylpyridinium-2-yl)porphyrin, MnIIITE-2-PyP5+ (AEOL-10113) has proven effective in treating oxidative stress-induced conditions including cancer, radiation damage, diabetes, and central nervous system trauma. The ortho cationic pyridyl nitrogens of MnTE-2-PyP5+ are essential for its high antioxidant potency. The exceptional ability of MnIIITE-2-PyP5+ to dismute O2.- parallels its ability to reduce ONOO- and CO3-. Decreasing levels of these species are considered its predominant mode of action, which may also involve redox regulation of signaling pathways. Recently, Ferrer-Sueta at al. (Free Radic. Biol. Med. 41:503-512; 2006) showed, with submitochondrial particles, that>or=3 microM MnIIITE-2-PyP5+ was able to protect components of the mitochondrial electron transport chain from peroxynitrite-mediated damage. Our study complements their data in showing, for the first time that micromolar mitochondrial concentrations of MnIIITE-2-PyP5+ are obtainable in vivo. For this study we have developed a new and sensitive method for MnIIITE-2-PyP5+ determination in tissues. The method is based on the exchange of porphyrin Mn2+ with Zn2+, followed by the HPLC/fluorescence detection of ZnIITE-2-PyP4+. At 4 and 7 h after a single 10 mg/kg intraperitoneal administration of MnIIITE-2-PyP5+, the mice (8 in total) were anesthetized and perfused with saline. Mitochondria were then isolated by the method of Mela and Seitz (Methods Enzymol.55:39-46; 1979). We found MnIIITE-2-PyP5+ localized in heart mitochondria to 2.95 ng/mg protein. Given the average value of mitochondrial volume of 0.6 microL/mg protein, the calculated MnIIITE-2-PyP5+ concentration is 5.1 microM, which is sufficient to protect mitochondria from oxidative damage. This study establishes, for the first time, that MnIIITE-2-PyP5+, a highly charged metalloporphyrin, is capable of entering mitochondria in vivo at levels sufficient to exert there its antioxidant action; such a result encourages its development as a prospective therapeutic agent.


Asunto(s)
Antioxidantes/farmacología , Metaloporfirinas/farmacología , Mitocondrias Cardíacas/fisiología , Superóxido Dismutasa/metabolismo , Animales , Antioxidantes/química , Manganeso/metabolismo , Ratones , Mitocondrias Cardíacas/efectos de los fármacos , Modelos Moleculares , Espectrofotometría , Zinc/metabolismo
15.
Antioxid Redox Signal ; 9(7): 1001-8, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17508921

RESUMEN

Reactive oxygen (ROS) and nitrogen species (RNS) generation have been proposed to be an important mechanism of doxorubicin (Adriamycin; ADR)-induced cardiotoxicity and cardiomyocyte apoptosis, processes that may be mediated by p53 protein. We note that ADR treatment resulted in increased levels of p53 protein in cardiomyocyte mitochondria and nuclei. Modulation of the cardiomyocyte redox state in genetically engineered mice by modulation of enzymes involved in metabolism of ROS/RNS, manganese superoxide dismutase (MnSOD), or inducible nitric oxide synthase (iNOS), or a combination of these, regulated levels of mitochondrial/nuclear p53 in cardiomyocytes after ADR administration. These observations led to the hypothesis that mitochondrial/nuclear p53 localization and function in the cardiomyocyte response to ADR may be regulated through redox-dependent mechanism(s).


Asunto(s)
Núcleo Celular/metabolismo , Mitocondrias/metabolismo , Miocitos Cardíacos/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Antibióticos Antineoplásicos/farmacología , Doxorrubicina/farmacología , Humanos , Modelos Biológicos , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos , Oxidación-Reducción/efectos de los fármacos
16.
Mol Cancer Ther ; 5(11): 2851-60, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17121932

RESUMEN

Multidrug resistance-associated protein 1 (MRP1) mediates the ATP-dependent efflux of endobiotics and xenobiotics, including estradiol 17-(beta-d-glucuronide), leukotriene C(4), and the reduced glutathione conjugate of 4-hydroxy-2-nonenal (HNE), a highly reactive product of lipid peroxidation. Adriamycin is an effective cancer chemotherapeutic drug whose use is limited by cardiotoxicity. Adriamycin induces oxidative stress and production of HNE in cardiac tissue, which may contribute to cardiomyopathy. We investigated the role of Mrp1 in Adriamycin-induced oxidative stress in cardiac tissue. Mice were treated with Adriamycin (20 mg/kg, i.p.), and heart homogenate and sarcolemma membranes were assayed for Mrp1 expression and ATP-dependent transport activity. Expression of Mrp1 was increased at 6 and 24 hours after Adriamycin treatment compared with saline treatment. HNE-adducted proteins were significantly increased (P < 0.001) in the homogenates at 6 hours after Adriamycin treatment and accumulated further with time; HNE adduction of a 190-kDa protein was evident 3 days after Adriamycin treatment. Mrp1 was localized predominately in sarcolemma as shown by confocal and Western blot analysis. Sarcolemma membrane vesicles transported leukotriene C(4) with a K(m) and V(max) of 51.8 nmol/L and 94.1 pmol/min/mg, respectively, and MK571 (10 micromol/L) inhibited the transport activity by 65%. Exposure of HEK(Mrp1) membranes to HNE (10 micromol/L) significantly decreased the V(max) for estradiol 17-(beta-d-glucuronide) transport by 50%. These results show that expression of Mrp1 in the mouse heart is localized predominantly in sarcolemma. Adriamycin treatment increased Mrp1 expression and HNE adduction of Mrp1. Cardiac Mrp1 may play a role in protecting the heart from Adriamycin-induced cardiomyopathy by effluxing HNE conjugates.


Asunto(s)
Aldehídos/metabolismo , Antibióticos Antineoplásicos/farmacología , Doxorrubicina/farmacología , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Miocardio/metabolismo , Aldehídos/farmacología , Animales , Antibióticos Antineoplásicos/toxicidad , Transporte Biológico/efectos de los fármacos , Línea Celular , Doxorrubicina/toxicidad , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Estrés Oxidativo
17.
Cardiovasc Res ; 69(1): 186-97, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16157314

RESUMEN

OBJECTIVE: Treatment with adriamycin (ADR) is associated with cardiotoxicity mediated through the generation of superoxide (O2*-). Because nitric oxide (*NO) reacts with O2*-, generating peroxynitrite, we hypothesized that decreased *NO production would lead to protection in acute cardiac injury. METHODS: We investigated the role of decreased *NO levels in exacerbation of ADR-induced cardiotoxicity in vivo using iNOS (-/-) mice. Pathology, biochemical injury markers, and cardiac function were used to assess ADR-induced cardiac injury. RESULTS: Ultrastructural analysis demonstrated that iNOS (-/-) mice exhibited extensive cytoplasmic swelling and degeneration of mitochondria when compared to wildtype mice following treatment with ADR. Mice lacking iNOS exhibited a decrease in resting indices of cardiac function as well as an impairment in the positive inotropic actions of isoproterenol following treatment with ADR compared to nTg mice. Cardiac troponin, creatine phosphokinase, and lactate dehydrogenase levels were significantly increased after treatment in iNOS (-/-) mice as compared to controls and wildtype mice. CONCLUSIONS: These results indicate that a lack of *NO production by iNOS caused significantly enhanced cardiac injury. However, when iNOS (-/-) mice were crossed with manganese superoxide dismutase (MnSOD)-overexpressing animals, mitochondrial injury was ameliorated to the level of the wild type. These findings suggest that reduction of *NO levels mediated by ADR treatment leads to increased cardiac mitochondrial injury that can be attenuated by a compensatory increase in MnSOD.


Asunto(s)
Antibióticos Antineoplásicos/efectos adversos , Cardiomiopatías/inducido químicamente , Doxorrubicina/efectos adversos , Depuradores de Radicales Libres/metabolismo , Óxido Nítrico/metabolismo , Superóxido Dismutasa/metabolismo , Animales , Cardiomiopatías/metabolismo , Cardiomiopatías/prevención & control , Cardiotónicos/farmacología , Citoplasma/patología , Isoproterenol/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Mitocondrias Cardíacas/patología , Miocardio/metabolismo , Miocardio/patología , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Superóxido Dismutasa/genética
18.
Mol Cancer Ther ; 4(7): 1056-64, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16020663

RESUMEN

In the present study, we used genetically engineered B6C3 mice [mice overexpressing manganese superoxide dismutase (TgM(+/+)), mice in which inducible nitric oxide synthase had been inactivated (iNOSKO(-/-)), and crosses of these two genotypes] to study the role of manganese superoxide dismutase (MnSOD) and inducible nitric oxide synthase (iNOS) in the development of acute Adriamycin-induced cardiotoxicity. Both nontransgenic and genetically engineered mice were treated with 20 mg/kg Adriamycin and cardiac left ventricular tissues studied at 0, 3, 6, and 24 hours. Ultrastructural damage and levels of 4-hydroxy-2-nonenal (4HNE) protein adducts and 3-nitrotyrosine (3NT) were determined in cardiomyocytes using immunogold ultrastructural techniques. Our previous results showed that Adriamycin caused mitochondrial injury without significant nuclear or cytoplasmic damage at early time points. Interestingly, overexpression of MnSOD protected against acute mitochondrial injury, whereas deficiency in iNOS potentiated mitochondrial injury in comparison with levels of injury present in cardiomyocyte mitochondria of nontransgenic mice. In TgM(+/+) mice, there was a significant inverse correlation between mitochondrial injury and 4HNE/3NT levels at all time points analyzed, suggesting that reactive oxygen species/reactive nitrogen species damage products directly regulated acute Adriamycin-induced mitochondrial injury in these mice. The present studies are the first to directly quantify the effects of MnSOD and iNOS on mitochondrial injury during acute Adriamycin-induced cardiotoxicity and show extensive and specific patterns of posttranslational modifications of mitochondrial proteins following Adriamycin treatment.


Asunto(s)
Doxorrubicina/toxicidad , Mitocondrias Cardíacas/patología , Estrés Oxidativo , Superóxido Dismutasa/metabolismo , Actinas/efectos de los fármacos , Actinas/metabolismo , Aldehídos/análisis , Aldehídos/metabolismo , Animales , Southern Blotting , Western Blotting , Masculino , Ratones , Ratones Endogámicos , Ratones Mutantes , Mitocondrias Cardíacas/efectos de los fármacos , Mitocondrias Cardíacas/metabolismo , Mitocondrias Cardíacas/ultraestructura , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Nitratos/sangre , Especies Reactivas de Oxígeno/metabolismo , Superóxido Dismutasa/efectos de los fármacos , Superóxido Dismutasa/genética , Tirosina/análogos & derivados , Tirosina/metabolismo
19.
J Nutr Biochem ; 34: 8-16, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27156147

RESUMEN

Cardiovascular health is influenced by dietary composition and the western diet is composed of varying types/amounts of fat. Conjugated linoleic acid (cLA) is an abundant dietary unsaturated fatty acid associated with health benefits but its biological signaling is not well understood. Nitrite is enriched in vegetables within the diet and can impact signaling of unsaturated fatty acids; however, its role on cLA signaling is not well understood. Elucidating how nitrite may impact the biological signaling of cLA is important due to the dietary consumption of both cLA and nitrite in the western diet. Since co-administration of cLA and nitrite results in cardioprotection during myocardial infarction (MI), it was hypothesized that cLA and nitrite may affect cardiac mitochondrial respiratory function and complex activity in MI. C57BL/6J mice were treated with cLA and nitrite for either 10 or 13days, where MI was induced on day 3. Following treatment, respiration and complex activity were measured. Among the major findings of this study, cLA treatment (10days) decreases state 3 respiration in vivo. Following MI, nitrite alone and in combination with cLA attenuates increased state 3 respiration and decreases hydrogen peroxide levels. Further, nitrite and cLA co-treatment attenuates increased complex III activity after MI. These results suggest that cLA, nitrite and the combination significantly alter cardiac mitochondrial respiratory and electron transport chain activity in vivo and following MI. Overall, the daily consumption of cLA and nitrite in the diet can have diverse cardiovascular implications, some of which occur at the mitochondrial level.


Asunto(s)
Cardiotónicos/uso terapéutico , Suplementos Dietéticos , Modelos Animales de Enfermedad , Ácidos Linoleicos Conjugados/uso terapéutico , Mitocondrias Cardíacas/metabolismo , Daño por Reperfusión Miocárdica/prevención & control , Nitrito de Sodio/uso terapéutico , Animales , Cardiotónicos/administración & dosificación , Ecocardiografía , Transporte de Electrón , Complejo I de Transporte de Electrón/metabolismo , Complejo II de Transporte de Electrones/metabolismo , Complejo III de Transporte de Electrones/antagonistas & inhibidores , Complejo III de Transporte de Electrones/metabolismo , Complejo IV de Transporte de Electrones/metabolismo , Corazón/diagnóstico por imagen , Peróxido de Hidrógeno/antagonistas & inhibidores , Peróxido de Hidrógeno/metabolismo , Ácidos Linoleicos Conjugados/administración & dosificación , Masculino , Ratones Endogámicos C57BL , Mitocondrias Cardíacas/enzimología , ATPasas de Translocación de Protón Mitocondriales/metabolismo , Daño por Reperfusión Miocárdica/diagnóstico por imagen , Daño por Reperfusión Miocárdica/enzimología , Daño por Reperfusión Miocárdica/metabolismo , Factores de Acoplamiento de la Fosforilación Oxidativa/metabolismo , Estrés Oxidativo
20.
Neurotoxicology ; 26(3): 407-16, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15869795

RESUMEN

Environmental exposure to mercurials continues to be a public health issue due to their deleterious effects on immune, renal and neurological function. Recently the safety of thimerosal, an ethyl mercury-containing preservative used in vaccines, has been questioned due to exposure of infants during immunization. Mercurials have been reported to cause apoptosis in cultured neurons; however, the signaling pathways resulting in cell death have not been well characterized. Therefore, the objective of this study was to identify the mode of cell death in an in vitro model of thimerosal-induced neurotoxicity, and more specifically, to elucidate signaling pathways which might serve as pharmacological targets. Within 2 h of thimerosal exposure (5 microM) to the human neuroblastoma cell line, SK-N-SH, morphological changes, including membrane alterations and cell shrinkage, were observed. Cell viability, assessed by measurement of lactate dehydrogenase (LDH) activity in the medium, as well as the 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide (MTT) assay, showed a time- and concentration-dependent decrease in cell survival upon thimerosal exposure. In cells treated for 24 h with thimerosal, fluorescence microscopy indicated cells undergoing both apoptosis and oncosis/necrosis. To identify the apoptotic pathway associated with thimerosal-mediated cell death, we first evaluated the mitochondrial cascade, as both inorganic and organic mercurials have been reported to accumulate in the organelle. Cytochrome c was shown to leak from the mitochondria, followed by caspase 9 cleavage within 8 h of treatment. In addition, poly(ADP-ribose) polymerase (PARP) was cleaved to form a 85 kDa fragment following maximal caspase 3 activation at 24 h. Taken together these findings suggest deleterious effects on the cytoarchitecture by thimerosal and initiation of mitochondrial-mediated apoptosis.


Asunto(s)
Antifúngicos/toxicidad , Apoptosis/efectos de los fármacos , Neoplasias Encefálicas/patología , Mitocondrias/efectos de los fármacos , Neuroblastoma/patología , Timerosal/toxicidad , Western Blotting , Caspasa 3 , Caspasa 9 , Caspasas/metabolismo , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Citocromos c/metabolismo , Humanos , L-Lactato Deshidrogenasa/metabolismo , Microscopía Fluorescente , Poli(ADP-Ribosa) Polimerasas/metabolismo , Fracciones Subcelulares/enzimología , Fracciones Subcelulares/metabolismo , Sales de Tetrazolio , Tiazoles , Timerosal/análisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA