Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Br J Haematol ; 179(1): 131-141, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28653339

RESUMEN

We aimed to compare hypofibrinogenaemia prevalence in major bleeding patients across all clinical contexts, fibrinogen supplementation practice, and explore the relationship between fibrinogen concentrations and mortality. This cohort study included all adult patients from 20 hospitals across Australia and New Zealand who received massive transfusion between April 2011 and October 2015. Of 3566 patients, 2829 (79%) had fibrinogen concentration recorded, with a median first and lowest concentration of 2·0 g/l (interquartile range [IQR] 1·5-2·7) and 1·8 g/l (IQR 1·3-2·4), respectively. Liver transplant (1·7 g/l, IQR 1·2-2·1), trauma (1·8, IQR 1·3-2·5) and vascular surgery (1·9 g/l, IQR 1·4-2·5) had lower concentrations. Total median fibrinogen dose administered from all products was 7·3 g (IQR 3·3-13·0). Overall, 1732 (61%) received cryoprecipitate and 9 (<1%) fibrinogen concentrate. Time to cryoprecipitate issue in those with initial fibrinogen concentration <1 g/l was 2·5 h (IQR 1·2-4·3 h). After adjustment, initial fibrinogen concentration had a U-shaped association with in-hospital mortality [adjusted odds ratios: fibrinogen <1 g/l, 2·31 (95% confidence interval (CI) 1·48-3·60); 1-1·9 g/l, 1·29 (95% CI 0·99-1·67) and >4 g/l, 2·03 (95% CI 1·35-3·04), 2-4 g/l reference category]. The findings indicate areas for practice improvement including timely administration of cryoprecipitate, which is the most common source of concentrated fibrinogen in Australia and New Zealand.


Asunto(s)
Coagulantes/administración & dosificación , Coagulantes/sangre , Fibrinógeno/administración & dosificación , Hemorragia/terapia , Afibrinogenemia/sangre , Afibrinogenemia/complicaciones , Afibrinogenemia/epidemiología , Anciano , Australia/epidemiología , Transfusión Sanguínea , Estudios de Cohortes , Femenino , Hemorragia/epidemiología , Hemorragia/etiología , Hemorragia/mortalidad , Mortalidad Hospitalaria , Humanos , Masculino , Persona de Mediana Edad , Mortalidad , Nueva Zelanda/epidemiología , Oportunidad Relativa , Prevalencia , Sistema de Registros
2.
Diabetes Obes Metab ; 19(7): 936-943, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28155245

RESUMEN

AIM: To determine the effect of Scriptaid, a compound that can replicate aspects of the exercise adaptive response through disruption of the class IIa histone deacetylase (HDAC) corepressor complex, on muscle insulin action in obesity. MATERIALS AND METHODS: Diet-induced obese mice were administered Scriptaid (1 mg/kg) via daily intraperitoneal injection for 4 weeks. Whole-body and skeletal muscle metabolic phenotyping of mice was performed, in addition to echocardiography, to assess cardiac morphology and function. RESULTS: Scriptaid treatment had no effect on body weight or composition, but did increase energy expenditure, supported by increased lipid oxidation, while food intake was also increased. Scriptaid enhanced the expression of oxidative genes and proteins, increased fatty acid oxidation and reduced triglycerides and diacylglycerides in skeletal muscle. Furthermore, ex vivo insulin-stimulated glucose uptake by skeletal muscle was enhanced. Surprisingly, heart weight was reduced in Scriptaid-treated mice and was associated with enhanced expression of genes involved in oxidative metabolism in the heart. Scriptaid also improved indices of both diastolic and systolic cardiac function. CONCLUSION: These data show that pharmacological targeting of the class IIa HDAC corepressor complex with Scriptaid could be used to enhance muscle insulin action and cardiac function in obesity.


Asunto(s)
Cardiotónicos/uso terapéutico , Metabolismo Energético/efectos de los fármacos , Corazón/efectos de los fármacos , Inhibidores de Histona Desacetilasas/uso terapéutico , Hidroxilaminas/uso terapéutico , Músculo Esquelético/efectos de los fármacos , Obesidad/tratamiento farmacológico , Quinolinas/uso terapéutico , Animales , Fármacos Antiobesidad/efectos adversos , Fármacos Antiobesidad/uso terapéutico , Cardiotónicos/efectos adversos , Dieta Alta en Grasa/efectos adversos , Ecocardiografía , Ecocardiografía Doppler , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Corazón/diagnóstico por imagen , Corazón/fisiopatología , Histona Desacetilasa 2/antagonistas & inhibidores , Histona Desacetilasa 2/metabolismo , Inhibidores de Histona Desacetilasas/efectos adversos , Hidroxilaminas/efectos adversos , Resistencia a la Insulina , Masculino , Ratones Endogámicos C57BL , Músculo Esquelético/metabolismo , Miocardio/patología , Obesidad/etiología , Obesidad/patología , Obesidad/fisiopatología , Tamaño de los Órganos , Quinolinas/efectos adversos
3.
Cytokine ; 69(2): 255-62, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25052838

RESUMEN

Activin A, a member of the transforming growth factor-ß superfamily, is stimulated early in inflammation via the Toll-like receptor (TLR) 4 signalling pathway, which is also activated in myocardial ischaemia-reperfusion. Neutralising activin A by treatment with the activin-binding protein, follistatin, reduces inflammation and mortality in several disease models. This study assesses the regulation of activin A and follistatin in a murine myocardial ischaemia-reperfusion model and determines whether exogenous follistatin treatment is protective against injury. Myocardial activin A and follistatin protein levels were elevated following 30 min of ischaemia and 2h of reperfusion in wild-type mice. Activin A, but not follistatin, gene expression was also up-regulated. Serum activin A did not change significantly, but serum follistatin decreased. These responses to ischaemia-reperfusion were absent in TLR4(-/-) mice. Pre-treatment with follistatin significantly reduced ischaemia-reperfusion induced myocardial infarction. In mouse neonatal cardiomyocyte cultures, activin A exacerbated, while follistatin reduced, cellular injury after 3h of hypoxia and 2h of re-oxygenation. Neither activin A nor follistatin affected hypoxia-reoxygenation induced reactive oxygen species production by these cells. However, activin A reduced cardiomyocyte mitochondrial membrane potential, and follistatin treatment ameliorated the effect of hypoxia-reoxygenation on cardiomyocyte mitochondrial membrane potential. Taken together, these data indicate that myocardial ischaemia-reperfusion, through activation of TLR4 signalling, stimulates local production of activin A, which damages cardiomyocytes independently of increased reactive oxygen species. Blocking activin action by exogenous follistatin reduces this damage.


Asunto(s)
Activinas/metabolismo , Folistatina/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Activinas/sangre , Activinas/genética , Animales , Animales Recién Nacidos , Células Cultivadas , Folistatina/genética , Folistatina/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , L-Lactato Deshidrogenasa/metabolismo , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones Endogámicos C57BL , Daño por Reperfusión Miocárdica/genética , Daño por Reperfusión Miocárdica/patología , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptor Toll-Like 4/deficiencia , Receptor Toll-Like 4/metabolismo
4.
Nat Commun ; 15(1): 258, 2024 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-38225272

RESUMEN

There are epidemiological associations between obesity and type 2 diabetes, cardiovascular disease and Alzheimer's disease. The role of amyloid beta 42 (Aß42) in these diverse chronic diseases is obscure. Here we show that adipose tissue releases Aß42, which is increased from adipose tissue of male mice with obesity and is associated with higher plasma Aß42. Increasing circulating Aß42 levels in male mice without obesity has no effect on systemic glucose homeostasis but has obesity-like effects on the heart, including reduced cardiac glucose clearance and impaired cardiac function. The closely related Aß40 isoform does not have these same effects on the heart. Administration of an Aß-neutralising antibody prevents obesity-induced cardiac dysfunction and hypertrophy. Furthermore, Aß-neutralising antibody administration in established obesity prevents further deterioration of cardiac function. Multi-contrast transcriptomic analyses reveal that Aß42 impacts pathways of mitochondrial metabolism and exposure of cardiomyocytes to Aß42 inhibits mitochondrial complex I. These data reveal a role for systemic Aß42 in the development of cardiac disease in obesity and suggest that therapeutics designed for Alzheimer's disease could be effective in combating obesity-induced heart failure.


Asunto(s)
Enfermedad de Alzheimer , Diabetes Mellitus Tipo 2 , Masculino , Ratones , Animales , Péptidos beta-Amiloides , Diabetes Mellitus Tipo 2/complicaciones , Anticuerpos Neutralizantes , Obesidad/complicaciones , Glucosa , Fragmentos de Péptidos
5.
J Mol Cell Cardiol ; 46(1): 86-92, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18977358

RESUMEN

The endothelium plays a vital role in the maintenance of vascular tone and structural vascular integrity, principally mediated via the actions of nitric oxide (NO). L-arginine is the immediate substrate for NO synthesis, and the availability of extracellular L-arginine is critical for the production of NO. Activation of protein kinase C (PKC) dependent signalling pathways are a feature of a number of cardiovascular disease states, and in this study we aimed to systematically evaluate the mechanism by which PKC regulates L-arginine transport in endothelial cells. In response to PKC activation (PMA 100 nM, 30 min), [(3)H]L-arginine uptake by bovine aortic endothelial cells (BAEC) was reduced to 45+4% of control (p<0.05). This resulted from a 53% reduction in the Vmax (p<0.05), with no change in the K(m) for L-arginine. Western blot analysis and confocal microscopy revealed no change in the expression or membrane distribution of CAT-1, the principal BAEC L-arginine transporter. Moreover in (32)P-labeling studies, PMA exposure did not result in CAT-1 phosphorylation. We therefore explored the possibility that PKC altered and interaction with MARCKS protein, a candidate membrane associated protein. By co-immunoprecipitation we show that CAT-1 interacts with, a membrane associated protein, that was significantly inhibited by PKC activation (p<0.05). Moreover antisense inhibition of MARCKS abolished the PMA effect on L-arginine transport. PKC dependent mechanisms regulate the transport of L-arginine, mediated via process involving MARCKS.


Asunto(s)
Arginina/metabolismo , Transportador de Aminoácidos Catiónicos 1/metabolismo , Endotelio/metabolismo , Péptidos y Proteínas de Señalización Intracelular/fisiología , Proteínas de la Membrana/fisiología , Proteína Quinasa C/metabolismo , Animales , Arginina/química , Transporte Biológico , Bovinos , Membrana Celular/metabolismo , Células Endoteliales/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Cinética , Proteínas de la Membrana/metabolismo , Sustrato de la Proteína Quinasa C Rico en Alanina Miristoilada , Óxido Nítrico Sintasa de Tipo III/metabolismo , Oligonucleótidos Antisentido/química , Fosforilación
6.
J Cell Biochem ; 108(1): 156-68, 2009 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-19544385

RESUMEN

Myocardial injury due to ischemia-reperfusion (I-R) damage remains a major clinical challenge. Its pathogenesis is complex including endothelial dysfunction and heightened oxidative stress although the key driving mechanism remains uncertain. In this study we tested the hypothesis that the I-R process induces a state of insufficient L-arginine availability for NO biosynthesis, and that this is pivotal in the development of myocardial I-R damage. In neonatal rat ventricular cardiomyocytes (NVCM), hypoxia-reoxygenation significantly decreased L-arginine uptake and NO production (42 +/- 2% and 71 +/- 4%, respectively, both P < 0.01), maximal after 2 h reoxygenation. In parallel, mitochondrial membrane potential significantly decreased and ROS production increased (both P < 0.01). NVCMs infected with adenovirus expressing the L-arginine transporter, CAT1, and NVCMs supplemented with L-arginine both exhibited significant (all P < 0.05) improvements in NO generation and mitochondrial membrane potentials, with a concomitant significant fall in ROS production and lactate dehydrogenase release during hypoxia-reoxygenation. In contrast, L-arginine deprived NVCM had significantly worsened responses to hypoxia-reoxygenation. In isolated perfused mouse hearts, L-arginine infusion during reperfusion significantly improved left ventricular function after I-R. These improved contractile responses were not dependent on coronary flow but were associated with a significant decrease in nitrotyrosine formation and increases in phosphorylation of both Akt and troponin I. Collectively, these data strongly implicate reduced L-arginine availability as a key factor in the pathogenesis of I-R injury. Increasing L-arginine availability via increased CAT1 expression or by supplementation improves myocardial responses to I-R. Restoration of L-arginine availability may therefore be a valuable strategy to ameliorate I-R injury.


Asunto(s)
Arginina/metabolismo , Daño por Reperfusión Miocárdica/etiología , Animales , Ratones , Daño por Reperfusión Miocárdica/metabolismo , Miocitos Cardíacos/metabolismo , Óxido Nítrico/metabolismo , Consumo de Oxígeno , Ratas , Ratas Sprague-Dawley , Canales Catiónicos TRPV/genética , Canales Catiónicos TRPV/metabolismo , Transfección
7.
Circulation ; 115(10): 1269-74, 2007 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-17325243

RESUMEN

BACKGROUND: Endothelial dysfunction because of reduced nitric oxide bioavailability is a key feature of essential hypertension. We have found that normotensive siblings of subjects with essential hypertension have impaired endothelial function accompanied by altered arginine metabolism. METHODS AND RESULTS: We have identified a novel C/T polymorphism in the 3'UTR of the principal arginine transporter, solute carrier family 7 (cationic amino acid transporter, y+ system), member 1 gene (SLC7A1). The minor T allele significantly attenuates reporter gene expression (P<0.01) and is impaired in its capacity to form DNA-protein complexes (P<0.05). In 278 hypertensive subjects the frequency of the T allele was 13.3% compared with 7.6% in 498 normotensive subjects (P<0.001). Moreover, the overall genotype distribution observed in hypertensives differed significantly from that in normotensives (P<0.001). To complement these studies, we generated an endothelial-specific transgenic mouse overexpressing L-arginine transporter SLC7A1. The Slc7A1 transgenic mice exhibited significantly enhanced responses to the endothelium-dependent vasodilator acetylcholine (-log EC50 for wild-type versus Slc7A1 transgenic: 6.87+/-0.10 versus 7.56+/-0.13; P<0.001). This was accompanied by elevated production of nitric oxide by isolated aortic endothelial cells. CONCLUSIONS: The present study identifies a key, functionally active polymorphism in the 3'UTR of SLC7A1. As such, this polymorphism may account for the apparent link between altered endothelial function, L-arginine, and nitric oxide metabolism and predisposition to essential hypertension.


Asunto(s)
Región de Flanqueo 3'/genética , Transportador de Aminoácidos Catiónicos 1/genética , Células Endoteliales/metabolismo , Hipertensión/genética , Polimorfismo Genético , Animales , Aorta/efectos de los fármacos , Aorta/fisiopatología , Arginina/metabolismo , Células CHO , Células Cultivadas , Cricetinae , Cricetulus , Ensayo de Cambio de Movilidad Electroforética , Frecuencia de los Genes , Técnicas de Transferencia de Gen , Genes Reporteros , Genotipo , Células HeLa , Humanos , Ratones , Óxido Nítrico , Técnicas de Cultivo de Órganos , Vasodilatación/efectos de los fármacos , Vasodilatación/genética , Vasodilatadores/farmacología
8.
Int J Biochem Cell Biol ; 40(5): 920-8, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18069040

RESUMEN

The action of oxidatively modified low-density lipoprotein on vascular endothelial cells has been proposed to be a crucial process leading to endothelial dysfunction and atherogenesis. However, the biochemical mechanism for such action is not clear. We have previously shown that arginine uptake and metabolism are major determinants of endothelial function in heart failure and hypertension. In the present study we therefore aimed to assess the effects of oxidized LDL, a major pro-atherogenic molecule, on endothelial l-arginine metabolism and its uptake. Endothelial cells were exposed to oxidized LDL or native LDL for 24h, and the resultant effects on (1) the intracellular content of arginine and its major metabolites including citrulline, N(G)-hydroxy-l-arginine, asymmetric dimethylarginine, symmetric dimethylarginine and ornithine, (2) [3H]-l-arginine uptake and, (3) the pattern of distribution of cationic amino acid transporter 1, the principal l-arginine transporter, by confocal microscopy. Oxidized LDL (100 microg/mL) reduced intracellular arginine and N(G)-hydroxy-l-arginine contents by 56 and 71% (P<0.05), respectively, with a concomitant 205% increase in ADMA (P<0.05). In conjunction, oxidized LDL reduced endothelial uptake of [3H]-arginine by 60%. Furthermore, incubation of endothelial cells with oxLDL led to internalization of cationic amino acid transporter 1. We demonstrate a novel mechanism, reduced l-arginine transport, by which oxidized LDL impairs the ability of the endothelium to generate nitric oxide.


Asunto(s)
Arginina/metabolismo , Endotelio Vascular/metabolismo , Lipoproteínas LDL/farmacología , Transporte Biológico/efectos de los fármacos , Transportador de Aminoácidos Catiónicos 1/genética , Transportador de Aminoácidos Catiónicos 1/metabolismo , Línea Celular , Membrana Celular/química , Membrana Celular/metabolismo , Colesterol/metabolismo , Concanavalina A/farmacología , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Humanos , ARN Mensajero/metabolismo
9.
Curr Med Chem ; 14(14): 1539-49, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17584062

RESUMEN

Coronary heart disease (CHD) remains the greatest killer in the Western world, and although the death rate from CHD has been falling, the current increased prevalence of major risk factors including obesity and diabetes, suggests it is likely that CHD incidence will increase over the next 20 years. In conjunction with preventive strategies, major advances in the treatment of acute coronary syndromes and myocardial infarction have occurred over the past 20 years. In particular the ability to rapidly restore blood flow to the myocardium during heart attack, using interventional cardiologic or thrombolytic approaches has been a major step forward. Nevertheless, while 'reperfusion' is a major therapeutic aim, the process of ischemia followed by reperfusion is often followed by the activation of an injurious cascade. While the pathogenesis of ischemia-reperfusion is not completely understood, there is considerable evidence implicating reactive oxygen species (ROS) as an initial cause of the injury. ROS formed during oxidative stress can initiate lipid peroxidation, oxidize proteins to inactive states and cause DNA strand breaks, all potentially damaging to normal cellular function. ROS have been shown to be generated following routine clinical procedures such as coronary bypass surgery and thrombolysis, due to the unavoidable episode of ischemia-reperfusion. Furthermore, they have been associated with poor cardiac recovery post-ischemia, with recent studies supporting a role for them in infarction, necrosis, apoptosis, arrhythmogenesis and endothelial dysfunction following ischemia-reperfusion. In normal physiological condition, ROS production is usually homeostatically controlled by endogenous free radical scavengers such as superoxide dismutase, catalase, and the glutathione peroxidase and thioredoxin reductase systems. Accordingly, targeting the generation of ROS with various antioxidants has been shown to reduce injury following oxidative stress, and improve recovery from ischemia-reperfusion injury. This review summarises the role of myocardial antioxidant enzymes in ischemia-reperfusion injury, particularly the glutathione peroxidase (GPX) and the thioredoxin reductase (TxnRed) systems. GPX and TxnRed are selenocysteine dependent enzymes, and their activity is known to be dependent upon an adequate supply of dietary selenium. Moreover, various studies suggest that the supply of selenium as a cofactor also regulates gene expression of these selenoproteins. As such, dietary selenium supplementation may provide a safe and convenient method for increasing antioxidant protection in aged individuals, particularly those at risk of ischemic heart disease, or in those undergoing clinical procedures involving transient periods of myocardial hypoxia.


Asunto(s)
Daño por Reperfusión Miocárdica/metabolismo , Oxidorreductasas/metabolismo , Selenio/metabolismo , Animales , Humanos , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Selenio/administración & dosificación
10.
Biol Trace Elem Res ; 114(1-3): 197-206, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-17206002

RESUMEN

Cardiac surgery often generates oxidative stress leading to ischemia reperfusion injury (I-R). Antioxidants have been shown to prevent this injury and have been added to cardioplegic solutions to assist in recovery. In this study, we tested the effectiveness of sodium selenite in protecting against ischemia reperfusion injury and investigated the mechanisms behind this protection. Hearts from male Wistar rats were subjected to ischemia reperfusion using the Langendorf model. Krebs-Henseleit perfusion solutions were supplemented with 0, 0.1, 0.5, 1.0, and 10 microM sodium selenite. Hearts were perfused for 30 min and then subjected to 22.5 min of global ischemia followed by 45 min reperfusion. Heart rate, ischemic contracture, end diastolic pressure, and developed ventricular pressure were monitored. At the completion of the experiment, hearts were homogenized and tissue extracts were assayed for glutathione peroxidase (GSH-Px) and thioredoxin reductase (Thx-Red) activity. Sodium selenite, at a concentration of 0.5 microM, demonstrated a protective effect on the recovery of cardiac function following I-R, as evidenced by a lower end diastolic pressure and enhanced recovery of rate pressure product. There was no beneficial effect observed in hearts perfused with 0.1 microM sodium selenite-supplemented buffer, whereas poorer functional recovery was observed in hearts perfused with 10 microM sodium selenite-supplemented buffer. The beneficial effect of sodium selenite was not mediated through increased activity of GSH-Px or Thx-Red. This study demonstrates that the addition of sodium selenite to reperfusion solutions, at an optimal concentration of 0.5 microM, assists in cardiac recovery following ischemia reperfusion.


Asunto(s)
Isquemia Miocárdica/patología , Daño por Reperfusión/tratamiento farmacológico , Selenito de Sodio/uso terapéutico , Animales , Glutatión Peroxidasa/metabolismo , Masculino , Ratas , Ratas Wistar , Reductasa de Tiorredoxina-Disulfuro/metabolismo
11.
PLoS One ; 10(3): e0120934, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25798941

RESUMEN

The development of diabetic cardiomyopathy is a key contributor to heart failure and mortality in obesity and type 2 diabetes (T2D). Current therapeutic interventions for T2D have limited impact on the development of diabetic cardiomyopathy. Clearly, new therapies are urgently needed. A potential therapeutic target is protein kinase D (PKD), which is activated by metabolic insults and implicated in the regulation of cardiac metabolism, contractility and hypertrophy. We therefore hypothesised that PKD inhibition would enhance cardiac function in T2D mice. We first validated the obese and T2D db/db mouse as a model of early stage diabetic cardiomyopathy, which was characterised by both diastolic and systolic dysfunction, without overt alterations in left ventricular morphology. These functional characteristics were also associated with increased PKD2 phosphorylation in the fed state and a gene expression signature characteristic of PKD activation. Acute administration of the PKD inhibitor CID755673 to normal mice reduced both PKD1 and 2 phosphorylation in a time and dose-dependent manner. Chronic CID755673 administration to T2D db/db mice for two weeks reduced expression of the gene expression signature of PKD activation, enhanced indices of both diastolic and systolic left ventricular function and was associated with reduced heart weight. These alterations in cardiac function were independent of changes in glucose homeostasis, insulin action and body composition. These findings suggest that PKD inhibition could be an effective strategy to enhance heart function in obese and diabetic patients and provide an impetus for further mechanistic investigations into the role of PKD in diabetic cardiomyopathy.


Asunto(s)
Azepinas/farmacología , Benzofuranos/farmacología , Cardiomiopatías Diabéticas/fisiopatología , Corazón/efectos de los fármacos , Corazón/fisiopatología , Proteína Quinasa C/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Disfunción Ventricular/fisiopatología , Animales , Azepinas/administración & dosificación , Benzofuranos/administración & dosificación , Diabetes Mellitus Tipo 2/complicaciones , Cardiomiopatías Diabéticas/diagnóstico , Cardiomiopatías Diabéticas/tratamiento farmacológico , Cardiomiopatías Diabéticas/genética , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Activación Enzimática , Masculino , Ratones , Miocardio/metabolismo , Miocardio/patología , Tamaño de los Órganos , Fosforilación , Proteína Quinasa C/metabolismo , Inhibidores de Proteínas Quinasas/administración & dosificación , Transcriptoma , Disfunción Ventricular/tratamiento farmacológico , Disfunción Ventricular/metabolismo , Función Ventricular Izquierda/efectos de los fármacos
12.
Eur J Pharmacol ; 769: 64-70, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26522927

RESUMEN

Reduced nitric oxide (NO) bioavailability plays a central role in the pathogenesis of myocardial ischemia-reperfusion injury (I-R), and reduced l-arginine transport via cationic amino acid transporter-1 is a key contributor to the reduced NO levels. Insulin can increase NO levels by increasing the transport of its substrate l-arginine but insulin alone exerts minimal cardiac protection in I-R. We hypothesized that combined insulin and l-arginine may provide cardioprotective effects in the setting of myocardial I-R. The effect of supplemental insulin, l-arginine and the combination was examined in cardiomyocytes exposed to hypoxia/reoxygenation and in isolated perfused mouse hearts undergoing ischemia/reperfusion. When compared to controls, cardiomyocytes treated upon reoxygenation with 1nM insulin+1mM l-arginine exhibited significant (all P<0.05) improvements in NO generation and mitochondrial membrane potential, with a concomitant fall in reactive oxygen species production and LDH release. Insulin also increased l-arginine uptake following hypoxia-reoxygenation (P<0.05; n=4-6). In langendorff perfused isolated mouse hearts, combined l-arginine-insulin treatment upon reperfusion significantly (all P<0.05; n=9-11) improved recovery of left ventricular developed pressure, rate pressure product and end diastolic pressure following ischemia, independent of any changes in post-ischemic coronary flow, together with significantly lower LDH release. The observed improvements were greater than l-arginine or insulin treatment alone. In isolated cardiomyocytes (n=3-5), 1nM insulin caused cationic amino acid transporter-1 to redistribute to the cellular membrane from the cytosol and the effects of insulin on l-arginine uptake were partially dependent on the PI3K/Akt pathway. l-arginine-insulin treatment may be a novel strategy to ameliorate I-R injury.


Asunto(s)
Arginina/farmacología , Cardiotónicos/farmacología , Insulina/farmacología , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Animales , Arginina/metabolismo , Transporte Biológico/efectos de los fármacos , Hipoxia de la Célula/efectos de los fármacos , Interacciones Farmacológicas , Espacio Intracelular/efectos de los fármacos , Espacio Intracelular/metabolismo , Masculino , Ratones , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Oxígeno/metabolismo , Ratas
13.
Redox Rep ; 9(6): 317-20, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15720825

RESUMEN

Cardiac ischemia--reperfusion injury results in oxidative stress and poor physiological recovery. This study examined the amount of lipid and protein oxidation during ischemia-reperfusion to assess the degree of oxidative stress. Selenium supplementation was used to alter the antioxidant status of rats and the recovery of myocardial function post ischemia-reperfusion was investigated. Male Wistar rats were fed diets containing 0, 50, and 1000 microg/kg sodium selenite for 5 weeks, whilst controls received normal rat food containing 240 microg/kg selenium. Langendorff-perfused hearts were subjected to 22.5 min global ischemia and 45 min reperfusion, with functional recovery assessed. Heart tissues were assayed for the presence of lipid peroxides and protein carbonyls and correlated to cardiac recovery. Following ischemia and reperfusion there was a significant increase in both protein oxidation and lipid peroxidation. Hearts from selenium-deficient animals demonstrated higher levels of both protein carbonyls and lipid peroxides and were more susceptible to ischemia-reperfusion injury when compared to controls (38% versus 47% recovery of rate pressure product (RPP)). Selenium supplementation lowered the levels of protein carbonyls and lipid peroxides and resulted in improved recovery of cardiac function post ischemia-reperfusion (57% recovery of RPP). These data suggest that selenium supplementation may provide an effective method for reducing oxidative damage post cardiac ischemia-reperfusion.


Asunto(s)
Dieta , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Selenio/administración & dosificación , Animales , Peso Corporal/efectos de los fármacos , Corazón/efectos de los fármacos , Peroxidación de Lípido/efectos de los fármacos , Masculino , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/fisiopatología , Miocardio/metabolismo , Oxidación-Reducción , Proteínas/metabolismo , Ratas , Selenio/deficiencia
14.
J Trace Elem Med Biol ; 18(1): 81-8, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15487768

RESUMEN

Glutathione peroxidase and thioredoxin reductase are selenocysteine-dependent enzymes that protect against oxidative injury. This study examined the effects of dietary selenium on the activity of these two enzymes in rats, and investigated the ability of selenium to modulate myocardial function post ischemia-reperfusion. Male wistar rats were fed diets containing 0, 50, 240 and 1000 microg/kg sodium selenite for 5 weeks. Langendorff perfused hearts isolated from these rats were subjected to 22.5 min global ischemia and 45 min reperfusion, with functional recovery assessed. Liver samples were collected at the time of sacrifice, and heart and liver tissues assayed for thioredoxin reductase and glutathione peroxidase activity. Selenium deficiency reduced the activity of both glutathione peroxidase and thioredoxin reductase systemically. Hearts from selenium deficient animals were more susceptible to ischemia-reperfusion injury when compared to normal controls (38% recovery of rate pressure product (RPP) vs. 47% recovery of RPP). Selenium supplementation increased the endogenous activity of thioredoxin reductase and glutathione peroxidase and resulted in improved recovery of cardiac function post ischemia reperfusion (57% recovery of RPP). Endogenous activity of glutathione peroxidase and thioredoxin reductase is dependent on an adequate supply of the micronutrient selenium. Reduced activity of these antioxidant enzymes is associated with significant reductions in myocardial function post ischemia-reperfusion.


Asunto(s)
Dieta , Glutatión Peroxidasa/metabolismo , Isquemia Miocárdica/metabolismo , Reperfusión Miocárdica , Miocardio/metabolismo , Selenio , Reductasa de Tiorredoxina-Disulfuro/metabolismo , Animales , Peso Corporal , Suplementos Dietéticos , Técnicas In Vitro , Hígado/enzimología , Masculino , Contracción Miocárdica/efectos de los fármacos , Distribución Aleatoria , Ratas , Ratas Wistar , Selenio/administración & dosificación , Selenio/metabolismo , Selenio/farmacología
15.
PLoS One ; 9(8): e104643, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25111602

RESUMEN

BACKGROUND: Impaired mitochondrial function is fundamental feature of heart failure (HF) and myocardial ischemia. In addition to the effects of heightened oxidative stress, altered nitric oxide (NO) metabolism, generated by a mitochondrial NO synthase, has also been proposed to impact upon mitochondrial function. However, the mechanism responsible for arginine transport into mitochondria and the effect of HF on such a process is unknown. We therefore aimed to characterize mitochondrial L-arginine transport and to investigate the hypothesis that impaired mitochondrial L-arginine transport plays a key role in the pathogenesis of heart failure and myocardial injury. METHODS AND RESULTS: In mitochondria isolated from failing hearts (sheep rapid pacing model and mouse Mst1 transgenic model) we demonstrated a marked reduction in L-arginine uptake (p<0.05 and p<0.01 respectively) and expression of the principal L-arginine transporter, CAT-1 (p<0.001, p<0.01) compared to controls. This was accompanied by significantly lower NO production and higher 3-nitrotyrosine levels (both p<0.05). The role of mitochondrial L-arginine transport in modulating cardiac stress responses was examined in cardiomyocytes with mitochondrial specific overexpression of CAT-1 (mtCAT1) exposed to hypoxia-reoxygenation stress. mtCAT1 cardiomyocytes had significantly improved mitochondrial membrane potential, respiration and ATP turnover together with significantly decreased reactive oxygen species production and cell death following mitochondrial stress. CONCLUSION: These data provide new insights into the role of L-arginine transport in mitochondrial biology and cardiovascular disease. Augmentation of mitochondrial L-arginine availability may be a novel therapeutic strategy for myocardial disorders involving mitochondrial stress such as heart failure and reperfusion injury.


Asunto(s)
Arginina/metabolismo , Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/metabolismo , Lesiones Cardíacas/etiología , Lesiones Cardíacas/metabolismo , Mitocondrias Cardíacas/metabolismo , Oxígeno/metabolismo , Animales , Transporte Biológico , Regulación de la Expresión Génica , Insuficiencia Cardíaca/patología , Lesiones Cardíacas/patología , Ratones , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Óxido Nítrico/biosíntesis , Estrés Oxidativo , Ovinos , Canales Catiónicos TRPV/metabolismo
16.
Br J Pharmacol ; 168(1): 238-52, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22924634

RESUMEN

BACKGROUND AND PURPOSE: Annexin-A1 (ANX-A1) is an endogenous, glucocorticoid-regulated anti-inflammatory protein. The N-terminal-derived peptide Ac-ANX-A1(2-26) preserves cardiomyocyte viability, but the impact of ANX-A1-peptides on cardiac contractility is unknown. We now test the hypothesis that ANX-A1 preserves post-ischaemic recovery of left ventricular (LV) function. EXPERIMENTAL APPROACH: Ac-ANX-A1(2-26) was administered on reperfusion, to adult rat cardiomyocytes as well as hearts isolated from rats, wild-type mice and mice deficient in endogenous ANX-A1 (ANX-A1(-/-)). Myocardial viability and recovery of LV function were determined. KEY RESULTS: Ischaemia-reperfusion markedly impaired both cardiomyocyte viability and recovery of LV function by 60%. Treatment with exogenous Ac-ANX-A1(2-26) at the onset of reperfusion prevented cardiomyocyte injury and significantly improved recovery of LV function, in both intact rat and wild-type mouse hearts. Ac-ANX-A1(2-26) cardioprotection was abolished by either formyl peptide receptor (FPR)-nonselective or FPR1-selective antagonists, Boc2 and cyclosporin H, but was relatively insensitive to the FPR2-selective antagonist QuinC7. ANX-A1-induced cardioprotection was associated with increased phosphorylation of the cell survival kinase Akt. ANX-A1(-/-) exaggerated impairment of post-ischaemic recovery of LV function, in addition to selective LV FPR1 down-regulation. CONCLUSIONS AND IMPLICATIONS: These data represent the first evidence that ANX-A1 affects myocardial function. Our findings suggest ANX-A1 is an endogenous regulator of post-ischaemic recovery of LV function. Furthermore, the ANX-A1-derived peptide Ac-ANX-A1(2-26) on reperfusion rescues LV function, probably via activation of FPR1. ANX-A1-based therapies may thus represent a novel clinical approach for the prevention and treatment of myocardial reperfusion injury.


Asunto(s)
Anexina A1/metabolismo , Cardiotónicos/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/prevención & control , Miocitos Cardíacos/metabolismo , Fragmentos de Péptidos/farmacología , Disfunción Ventricular Izquierda/prevención & control , Animales , Anexina A1/deficiencia , Anexina A1/farmacología , Regulación hacia Abajo , Femenino , Técnicas In Vitro , Masculino , Ratones , Contracción Miocárdica , Daño por Reperfusión Miocárdica/complicaciones , Fosforilación , Ratas , Ratas Sprague-Dawley , Disfunción Ventricular Izquierda/etiología , Disfunción Ventricular Izquierda/metabolismo
17.
J Hypertens ; 28(4): 780-8, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20186099

RESUMEN

OBJECTIVE: A number of factors contribute to diabetes-associated vascular dysfunction. In the present study, we tested whether exposure to advanced glycation end-products (AGEs) impairs vascular reactivity independently of hyperglycemia and examined the potential mechanisms responsible for diabetes and AGE-associated vascular dysfunction. METHODS: Vasodilator function was studied using infusion of exogenous AGEs into Sprague-Dawley rats as compared with control and streptozotocin-induced diabetic rats all followed for 16 weeks (n = 10 per group). The level of arginine metabolites and expression of endothelial nitric oxide synthase (eNOS) and downstream mediators of nitric oxide-dependent signaling were examined. To further explore these mechanisms, cultured bovine aortic endothelial cells (BAECs) were exposed to AGEs. RESULTS: Both diabetic and animals infused with AGE-modified rat serum albumin (AGE-RSA) had significantly impaired vasodilatory response to acetylcholine. Unlike diabetes-associated endothelial dysfunction, AGE infusion was not associated with changes in plasma arginine metabolites, asymmetric dimethyl-L-arginine levels or eNOS expression. However, expression of the downstream mediator cGMP-dependent protein kinase 1 (PKG-1) was significantly reduced by both AGE exposure and diabetes. AGEs also augmented hyperglycemia-associated depletion in endothelial nitric oxide production and eNOS protein expression in vitro, and the novel AGE inhibitor, alagebrium chloride, partly restored these parameters. CONCLUSION: We demonstrate that AGEs represent a potentially important cause of vascular dysfunction, linked to the induction of nitric oxide resistance. These findings also emphasize the deleterious and potentially additive effects of AGEs and hyperglycemia in diabetic vasculature.


Asunto(s)
Productos Finales de Glicación Avanzada/farmacología , Óxido Nítrico Sintasa de Tipo III/metabolismo , Óxido Nítrico/metabolismo , Enfermedades Vasculares/inducido químicamente , Animales , Aorta Torácica/citología , Arginina/análogos & derivados , Arginina/sangre , Arginina/metabolismo , Bovinos , Células Cultivadas , Diabetes Mellitus/metabolismo , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Productos Finales de Glicación Avanzada/metabolismo , Hipoglucemiantes/metabolismo , Masculino , Óxido Nítrico/biosíntesis , Óxido Nítrico Sintasa de Tipo III/antagonistas & inhibidores , Ratas , Ratas Sprague-Dawley , Enfermedades Vasculares/complicaciones , Enfermedades Vasculares/metabolismo , Vasodilatación/efectos de los fármacos , Vasodilatadores/metabolismo , Vasodilatadores/farmacología
18.
Int J Biochem Cell Biol ; 41(12): 2522-7, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19695340

RESUMEN

Under conditions of oxidative stress it is well known that the bioavailability of nitric oxide (NO) is known to be significantly reduced. This process is in part due to the combination of NO with superoxide radicals to form peroxynitrite (ONOO(-)). While this process inactivates NO per se, it is not certain to which extent this process may also further impair ongoing NO production. Given the pivotal role of arginine availability for NO synthesis we determined the impact of ONOO(-) on endothelial arginine transport and intracellular arginine metabolism. Peroxynitrite reduced endothelial [(3)H]-L-arginine transport and increased the rate of arginine efflux in a concentration-dependent manner (both p<0.05). In conjunction, exposure to ONOO(-) significantly reduced the intracellular concentration of L-arginine, N(G)-hydroxy-L-arginine (an intermediate of NO biosynthesis) and citrulline by 46%, 45% and 60% respectively (all p<0.05), while asymmetric dimethyl arginine (ADMA) levels rose by 180% (p<0.05). ONOO(-) exposure did not alter the cellular distribution of the principal L-arginine transporter, CAT1, rather the effect on CAT1 activity appeared to be mediated by protein nitrosation. Conclusion Peroxynitrite negatively influences NO production by combined effects on arginine uptake and efflux, most likely due to a nitrosative action of ONOO(-) on CAT-1.


Asunto(s)
Arginina/metabolismo , Citrulina/biosíntesis , Endotelio Vascular/metabolismo , Óxido Nítrico/metabolismo , Ácido Peroxinitroso/farmacología , Animales , Arginina/análogos & derivados , Arginina/química , Bovinos , Células Cultivadas , Endotelio Vascular/efectos de los fármacos , Óxido Nítrico/química , Nitrosación/efectos de los fármacos , Estrés Oxidativo , Ácido Peroxinitroso/química , Transporte de Proteínas/efectos de los fármacos , Canales Catiónicos TRPV/metabolismo
19.
Hypertension ; 48(2): 278-85, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16801489

RESUMEN

Endothelial dysfunction is a hallmark of cardiovascular disease, and the l-arginine:NO pathway plays a critical role in determining endothelial function. Recent studies suggest that smoking, a well-recognized risk factor for vascular disease, may interfere with l-arginine and NO metabolism; however, this remains poorly characterized. Accordingly, we performed a series of complementary in vivo and in vitro studies to elucidate the mechanism by which cigarette smoke adversely affects endothelial function. In current smokers, plasma levels of asymmetrical dimethyl-arginine (ADMA) were 80% higher (P = 0.01) than nonsmokers, whereas citrulline (17%; P < 0.05) and N-hydroxy-l-arginine (34%; P < 0.05) were significantly lower. Exposure to 10% cigarette smoke extract (CSE) significantly affected endothelial arginine metabolism with reductions in the intracellular content of citrulline (81%), N-hydroxy-l-arginine (57%), and arginine (23%), while increasing ADMA (129%). CSE significantly inhibited (38%) arginine uptake in conjunction with a 34% reduction in expression of the arginine transporter, CAT1. In conjunction with these studies, CSE significantly reduced the activity of eNOS and NO production by endothelial cells, while stimulating the production of reactive oxygen species. In conclusion, cigarette smoke adversely affects the endothelial l-arginine NO synthase pathway, resulting in reducing NO production and elevated oxidative stress. In conjunction, exposure to cigarette smoke increases ADMA concentration, the latter being a risk factor for cardiovascular disease.


Asunto(s)
Arginina/metabolismo , Células Endoteliales/metabolismo , Óxido Nítrico/metabolismo , Estrés Oxidativo , Humo/efectos adversos , Fumar , Adulto , Animales , Antioxidantes/farmacología , Arginina/análogos & derivados , Arginina/sangre , Arginina/farmacocinética , Disponibilidad Biológica , Transporte Biológico/efectos de los fármacos , Transportador de Aminoácidos Catiónicos 1/metabolismo , Bovinos , Células Cultivadas , Citrulina/sangre , Cotinina/sangre , Medios de Cultivo/química , Medios de Cultivo/farmacología , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Humanos , Masculino , Óxido Nítrico Sintasa/metabolismo , Oxidantes/farmacología , Especies Reactivas de Oxígeno/metabolismo , Fumar/efectos adversos , Fumar/sangre , Nicotiana
20.
Mol Cell Biochem ; 270(1-2): 131-8, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15792362

RESUMEN

Cardiac ischemia reperfusion leads to oxidative stress and poor physiological recovery. Selenium deficiency down-regulates thioredoxin reductase (Txnrd) and glutathione peroxidase (Gpx) activity, impairing recovery from ischemia-reperfusion. Furthermore, selenium supplementation has been shown to be cardioprotective and lessens oxidative stress in reperfused rat hearts. In this study we have investigated the role of selenium in the mRNA expression of these, and related antioxidant proteins, post ischemia-reperfusion. Male rats were fed varying doses of selenium for five weeks. Hearts were isolated and perfused using the Langendorff method with 22.5 min of global ischemia and 45 min reperfusion. RNA was extracted for quantitative real-time PCR analysis of glutathione peroxidase (Gpx)-1 and 4, glutathione reductase (Gsr), thioredoxin peroxidase-2 (Prdx2), thioredoxin (Txn) and thioredoxin reductase (Txnrd)-1 and 2 gene expression. Selenium deficiency produced significant reductions in Gpx-1, Gpx-4, Prdx2, Txnrd-1 and Txnrd-2 expression. Conversely, selenium supplementation of 1000 microg/kg significantly up-regulated Gpx-1, Gpx-4, Txn, Txnrd-1 and Txnrd-2 transcription. Our results show selenium modulates the cardiac mRNA expression of thioredoxin and glutathione related enzymes post ischemia-reperfusion, and impacts on tolerance to ischemia-reperfusion.


Asunto(s)
Antioxidantes/farmacología , Dieta , Isquemia/patología , ARN Mensajero/metabolismo , Selenio/farmacología , Fenómenos Fisiológicos Nutricionales de los Animales , Animales , Antioxidantes/metabolismo , Peso Corporal , ADN Complementario/metabolismo , Regulación hacia Abajo , Glutatión/metabolismo , Glutatión Peroxidasa/biosíntesis , Glutatión Peroxidasa/metabolismo , Glutatión Reductasa/biosíntesis , Masculino , Miocardio/metabolismo , Estrés Oxidativo , Perfusión , Fosfolípido Hidroperóxido Glutatión Peroxidasa , ARN/metabolismo , Ratas , Ratas Wistar , Daño por Reperfusión , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Selenio/metabolismo , Temperatura , Reductasa de Tiorredoxina-Disulfuro/biosíntesis , Transcripción Genética , Glutatión Peroxidasa GPX1
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA