Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
JACC Basic Transl Sci ; 7(10): 973-981, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-36337921

RESUMEN

Interstitial collagen loss and cardiomyocyte ultrastructural damage accounts for left ventricular (LV) sphericity and decrease in LV twist and circumferential strain. Normal LV diastolic function belies significantly abnormal left atrial (LA) function and early LV diastolic untwist rate. This underscores the complex interplay of LV and LA myocardial remodeling and function in the pathophysiology of primary mitral regurgitation. In this study, we connect LA function with LV systolic and diastolic myocardial remodeling and function using cardiac magnetic resonance tissue tagging in primary mitral regurgitation.

2.
Redox Biol ; 1: 381-6, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24024175

RESUMEN

Hypercholesterolemia is a risk factor for the development of hypertrophic cardiomyopathy. Nevertheless, there are few studies aimed at determining the effects of dietary compounds on early or mild cardiac hypertrophy associated with dyslipidemia. Here we describe left ventricular (LV) hypertrophy in 12 week-old Apo E(-/-) hypercholesterolemic mice. The LV end diastolic posterior wall thickness and overall LV mass were significantly increased in Apo E(-/-) mice compared with wild type (WT) controls. Fractional shortening, LV end diastolic diameter, and hemodynamic parameters were unchanged from WT mice. Oral low dose quercetin (QCN; 0.1 µmol QCN/kg body weight for 6 weeks) significantly reduced total cholesterol and very low density lipoprotein in the plasma of Apo E(-/-) mice. QCN treatment also significantly decreased LV posterior wall thickness and LV mass in Apo E(-/-) mice. Myocardial geometry and function were unaffected in WT mice by QCN treatment. These data suggest that dietary polyphenolic compounds such as QCN may be effective modulators of plasma cholesterol and could prevent maladaptive myocardial remodeling.


Asunto(s)
Antioxidantes/administración & dosificación , Apolipoproteínas E/genética , Hipercolesterolemia/dietoterapia , Hipertrofia Ventricular Izquierda/dietoterapia , Quercetina/administración & dosificación , Animales , Antioxidantes/uso terapéutico , Colesterol/sangre , Hipercolesterolemia/genética , Hipercolesterolemia/patología , Hipertrofia Ventricular Izquierda/complicaciones , Hipertrofia Ventricular Izquierda/fisiopatología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Quercetina/uso terapéutico , Remodelación Ventricular/efectos de los fármacos
3.
Free Radic Biol Med ; 51(11): 1975-84, 2011 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-21925594

RESUMEN

Xanthine oxidoreductase (XOR) is increased in the left ventricle (LV) of humans with volume overload (VO), and mitochondrial inhibition of the respiratory chain occurs in animal models of VO. Because mitochondria are both a source and a target of reactive oxygen and nitrogen species, we hypothesized that activation of XOR and mitochondrial dysfunction are interdependent. To test this we used the aortocaval fistula (ACF) rat model of VO and a simulation of the stretch response in isolated adult cardiomyocytes with and without the inhibitor of XOR, allopurinol, or the mitochondrially targeted antioxidant MitoQ. Xanthine oxidase (XO) activity was increased in cardiomyocytes from ACF vs sham rats (24h) without an increase in XO protein. A twofold increase in LV end-diastolic pressure/wall stress and a decrease in LV systolic elastance with ACF were improved when allopurinol treatment (100mg/kg) was started at ACF induction. Subsarcolemmal State 3 mitochondrial respiration was significantly decreased in ACF and normalized by allopurinol. Cardiomyocytes subjected to 3h cyclical stretch resulted in an increase in XO activity and mitochondrial swelling, which was prevented by allopurinol or MitoQ pretreatment. These studies establish an early interplay between cardiomyocyte XO activation and bioenergetic dysfunction that may provide a new target that prevents progression to heart failure in VO.


Asunto(s)
Fístula Arteriovenosa/metabolismo , Volumen Cardíaco , Mitocondrias/metabolismo , Xantina Oxidasa/metabolismo , Alopurinol/farmacología , Animales , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Insuficiencia Cardíaca/metabolismo , Mitocondrias/efectos de los fármacos , Miocitos Cardíacos/enzimología , Miocitos Cardíacos/metabolismo , Ratas , Ratas Sprague-Dawley , Xantina Oxidasa/antagonistas & inhibidores
4.
J Mol Cell Cardiol ; 50(1): 147-56, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21059354

RESUMEN

Volume overload (VO) caused by aortocaval fistula (ACF) is associated with oxidative/inflammatory stress. The resulting inflammation, matrix metalloproteinase (MMP) activation, and collagen degradation is thought to play a pivotal role in left ventricular (LV) dilatation and failure. Since mitochondria are also targets for inflammation and oxidative stress, we hypothesized that there would be bioenergetic dysfunction with acute VO. In Sprague-Dawley rats subjected to 24 hrs of ACF, there was a two-fold increase in LV pressure-volume area in vivo, consistent with increased LV myocardial oxygen usage and increased bioenergetic demand in cardiomyocytes. Isolated cardiomyocytes from ACF LVs demonstrated increased hydrogen peroxide and superoxide formation and increased MMP activity. Subsarcolemmal mitochondria (SSM) showed a 40% decrease in state 3 respiration and proteomic analysis of SSM demonstrated decreased levels of complexes I-V in ACF. Immunohistochemical analysis revealed disruption of the subsarcolemmal location of the SSM network in ACF. To test for a potential link between SSM dysfunction and loss of interstitial collagen, rats were treated with the MMP-inhibitor PD166793 prior to ACF. MMP-inhibitor preserved interstitial collagen, integrin-α5 and the SSM structural arrangement. In addition, the decrease in state 3 mitochondrial respiration with ACF was prevented by PD166793. These studies established an important interaction between degradation of interstitial collagen in acute VO and the disruption of SSM structure and function which could contribute to progression to heart failure.


Asunto(s)
Colágeno/metabolismo , Mitocondrias Cardíacas/metabolismo , Miocitos Cardíacos/metabolismo , Animales , Western Blotting , Ecoencefalografía , Inmunohistoquímica , Microscopía Confocal , Microscopía Electrónica de Transmisión , Mitocondrias Cardíacas/ultraestructura , Miocitos Cardíacos/ultraestructura , Estrés Oxidativo/fisiología , Ratas , Ratas Sprague-Dawley , Función Ventricular Izquierda/fisiología
5.
Biochem J ; 432(1): 9-19, 2010 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-20825366

RESUMEN

Mitochondrial production of ROS (reactive oxygen species) is thought to be associated with the cellular damage resulting from chronic exposure to high glucose in long-term diabetic patients. We hypothesized that a mitochondria-targeted antioxidant would prevent kidney damage in the Ins2(+/)⁻(AkitaJ) mouse model (Akita mice) of Type 1 diabetes. To test this we orally administered a mitochondria-targeted ubiquinone (MitoQ) over a 12-week period and assessed tubular and glomerular function. Fibrosis and pro-fibrotic signalling pathways were determined by immunohistochemical analysis, and mitochondria were isolated from the kidney for functional assessment. MitoQ treatment improved tubular and glomerular function in the Ins2(+/)⁻(AkitaJ) mice. MitoQ did not have a significant effect on plasma creatinine levels, but decreased urinary albumin levels to the same level as non-diabetic controls. Consistent with previous studies, renal mitochondrial function showed no significant change between any of the diabetic or wild-type groups. Importantly, interstitial fibrosis and glomerular damage were significantly reduced in the treated animals. The pro-fibrotic transcription factors phospho-Smad2/3 and ß-catenin showed a nuclear accumulation in the Ins2(+/)⁻(AkitaJ) mice, which was prevented by MitoQ treatment. These results support the hypothesis that mitochondrially targeted therapies may be beneficial in the treatment of diabetic nephropathy. They also highlight a relatively unexplored aspect of mitochondrial ROS signalling in the control of fibrosis.


Asunto(s)
Nefropatías Diabéticas/prevención & control , Insulina/deficiencia , Mitocondrias/efectos de los fármacos , Ubiquinona/farmacología , Animales , Glucemia/metabolismo , Núcleo Celular/metabolismo , Diabetes Mellitus Tipo 1/sangre , Diabetes Mellitus Tipo 1/complicaciones , Nefropatías Diabéticas/complicaciones , Nefropatías Diabéticas/genética , Fibrosis/prevención & control , Mesangio Glomerular/efectos de los fármacos , Mesangio Glomerular/fisiopatología , Inmunohistoquímica , Insulina/genética , Riñón/efectos de los fármacos , Riñón/patología , Riñón/ultraestructura , Túbulos Renales/efectos de los fármacos , Túbulos Renales/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Microscopía Electrónica , Mitocondrias/metabolismo , Fosforilación/efectos de los fármacos , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , beta Catenina/metabolismo
6.
Am J Physiol Gastrointest Liver Physiol ; 291(5): G857-67, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16825707

RESUMEN

An early event that occurs in response to alcohol consumption is mitochondrial dysfunction, which is evident in changes to the mitochondrial proteome, respiration defects, and mitochondrial DNA (mtDNA) damage. S-adenosylmethionine (SAM) has emerged as a potential therapeutic for treating alcoholic liver disease through mechanisms that appear to involve decreases in oxidative stress and proinflammatory cytokine production as well as the alleviation of steatosis. Because mitochondria are a source of reactive oxygen/nitrogen species and a target for oxidative damage, we tested the hypothesis that SAM treatment during alcohol exposure preserves organelle function. Mitochondria were isolated from livers of rats fed control and ethanol diets with and without SAM for 5 wk. Alcohol feeding caused a significant decrease in state 3 respiration and the respiratory control ratio, whereas SAM administration prevented these alcohol-mediated defects and preserved hepatic SAM levels. SAM treatment prevented alcohol-associated increases in mitochondrial superoxide production, mtDNA damage, and inducible nitric oxide synthase induction, without a significant lessening of steatosis. Accompanying these indexes of oxidant damage, SAM prevented alcohol-mediated losses in cytochrome c oxidase subunits as shown using blue native PAGE proteomics and immunoblot analysis, which resulted in partial preservation of complex IV activity. SAM treatment attenuated the upregulation of the mitochondrial stress chaperone prohibitin. Although SAM supplementation did not alleviate steatosis by itself, SAM prevented several key alcohol-mediated defects to the mitochondria genome and proteome that contribute to the bioenergetic defect in the liver after alcohol consumption. These findings reveal new molecular targets through which SAM may work to alleviate one critical component of alcohol-induced liver injury: mitochondria dysfunction.


Asunto(s)
Hepatopatías Alcohólicas/prevención & control , Mitocondrias Hepáticas/efectos de los fármacos , Mitocondrias Hepáticas/metabolismo , S-Adenosilmetionina/farmacología , Animales , Western Blotting , Citocromo P-450 CYP2E1/metabolismo , ADN Mitocondrial/metabolismo , Complejo IV de Transporte de Electrones/metabolismo , Electroforesis en Gel de Poliacrilamida , Hígado/patología , Hepatopatías Alcohólicas/metabolismo , Hepatopatías Alcohólicas/patología , Pruebas de Función Hepática , Masculino , Chaperonas Moleculares/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Fosforilación Oxidativa/efectos de los fármacos , Prohibitinas , Ratas , Ratas Sprague-Dawley , Proteínas Represoras/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
7.
Free Radic Biol Med ; 38(3): 297-306, 2005 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-15629859

RESUMEN

It is now recognized that mitochondria play an integral role in orchestrating the response of the cell to a wide variety of metabolic and environmental stressors. Of particular interest are the interactions of reactive oxygen and nitrogen species with the organelle and their potential regulatory function. The best understood example is the O(2) sensitive binding of NO (nitric oxide) to the heme group in cytochrome c oxidase. We have proposed that this reversible process serves the function of both regulating the formation of hydrogen peroxide from the respiratory chain for the purposes of signal transduction and controlling O(2) gradients in complex organs such as the liver or heart. It now appears that maladaptation in this pathway leads to a mitochondrial dysfunction which has some of the characteristics of hypoxia, such as a deficit in ATP, but occurs in the presence of normal or enhanced levels of O(2). These are the optimal conditions for the formation of reactive nitrogen species (RNS), such as peroxynitrite which lead to the irreversible modification of proteins. We term this unique pathological condition Nitroxia and describe how it may contribute to the pathology of chronic inflammatory diseases using ethanol-dependent hepatotoxicity as an example.


Asunto(s)
Complejo IV de Transporte de Electrones/fisiología , Hepatitis Alcohólica/metabolismo , Óxido Nítrico/fisiología , Transducción de Señal/fisiología , Animales , Respiración de la Célula/fisiología , Complejo IV de Transporte de Electrones/metabolismo , Etanol/farmacología , Hígado/efectos de los fármacos , Hígado/metabolismo , Mitocondrias/fisiología , Óxido Nítrico/metabolismo , Oxidación-Reducción , Especies Reactivas de Oxígeno/metabolismo
8.
Hepatology ; 40(3): 565-73, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15349894

RESUMEN

Nitric oxide (NO) is now known to control both mitochondrial respiration and organelle biogenesis. Under conditions of ethanol-dependent hepatic dysfunction, steatosis is increased, and this is associated with increased expression of inducible nitric oxide synthase (iNOS). We have previously shown that after chronic exposure to ethanol, the sensitivity of mitochondrial respiration to inhibition by NO is enhanced, and we have proposed that this contributes to ethanol-dependent hypoxia. This study examines the role of iNOS in controlling the NO-dependent modification of mitochondrial function. Mitochondria were isolated from the livers of both wild-type (WT) and iNOS knockout (iNOS-/-) mice that were fed an isocaloric ethanol-containing diet for a period of 5 weeks. All animals that consumed ethanol showed some evidence of fatty liver; however, this was to a lesser extent in the iNOS-/- mice compared to controls. At this early stage in ethanol-dependent hepatic dysfunction, infiltration of inflammatory cells and the formation of nitrated proteins was also decreased in response to ethanol feeding in the iNOS-/- animals. Mitochondria isolated from wild-type ethanol-fed mice showed a significant decrease in respiratory control ratio and an increased sensitivity to NO-dependent inhibition of respiration relative to their pair-fed controls. In contrast, liver mitochondria isolated from iNOS-/- mice fed ethanol showed no change in the sensitivity to NO-dependent inhibition of respiration. In conclusion, the hepatic response to chronic alcohol-dependent cytotoxicity involves a change in mitochondrial function dependent on the induction of iNOS.


Asunto(s)
Etanol/toxicidad , Hígado/efectos de los fármacos , Mitocondrias Hepáticas/efectos de los fármacos , Óxido Nítrico Sintasa/fisiología , Animales , Hígado/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias Hepáticas/fisiología , Nitratos/metabolismo , Óxido Nítrico/fisiología , Óxido Nítrico Sintasa de Tipo II , Tirosina/metabolismo
9.
Am J Physiol Gastrointest Liver Physiol ; 286(4): G521-7, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-14670822

RESUMEN

Redox modification of mitochondrial proteins is thought to play a key role in regulating cellular function, although direct evidence to support this hypothesis is limited. Using an in vivo model of mitochondrial redox stress, ethanol hepatotoxicity, the modification of mitochondrial protein thiols was examined using a proteomics approach. Specific labeling of reduced thiols in the mitochondrion from the livers of control and ethanol-fed rats was achieved by using the thiol reactive compound (4-iodobutyl)triphenylphosphonium (IBTP). This molecule selectively accumulates in the organelle and can be used to identify thiol-containing proteins. Mitochondrial proteins that have been modified are identified by decreased labeling with IBTP using two-dimensional SDS-PAGE followed by immunoblotting with an antibody directed against the triphenylphosphonium moiety of the IBTP molecule. Analyses of these data showed a significant decrease in IBTP labeling of thiols present in specific mitochondria matrix proteins from ethanol-fed rats compared with their corresponding controls. These proteins were identified as the low-K(m) aldehyde dehydrogenase and glucose-regulated protein 78. The decrease in IBTP labeling in aldehyde dehydrogenase was accompanied by a decrease in specific activity of the enzyme. These data demonstrate that mitochondrial protein thiol modification is associated with chronic alcohol intake and might contribute to the pathophysiology associated with hepatic injury. Taken together, we have developed a protocol to chemically tag and select thiol-modified proteins that will greatly enhance efforts to establish posttranslational redox modification of mitochondrial protein in in vivo models of oxidative or nitrosative stress.


Asunto(s)
Depresores del Sistema Nervioso Central/toxicidad , Etanol/toxicidad , Mitocondrias Hepáticas/metabolismo , Proteínas/metabolismo , Compuestos de Sulfhidrilo/metabolismo , Aldehído Deshidrogenasa/metabolismo , Animales , Enfermedad Crónica , Electroforesis en Gel de Poliacrilamida , Metabolismo Energético/fisiología , Procesamiento de Imagen Asistido por Computador , Immunoblotting , Técnicas In Vitro , Masculino , Peso Molecular , Oxidación-Reducción , Ratas , Ratas Sprague-Dawley , Especies de Nitrógeno Reactivo/metabolismo , Especies Reactivas de Oxígeno/metabolismo
10.
Biochem Soc Symp ; (71): 107-20, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15777016

RESUMEN

Cellular redox signalling is mediated by the post-translational modification of proteins in signal-transduction pathways by ROS/RNS (reactive oxygen species/reactive nitrogen species) or the products derived from their reactions. NO is perhaps the best understood in this regard with two important modifications of proteins known to induce conformational changes leading to modulation of function. The first is the addition of NO to haem groups as shown for soluble guanylate cyclase and the newly discovered NO/cytochrome c oxidase signalling pathway in mitochondria. The second mechanism is through the modification of thiols by NO to form an S-nitrosated species. Other ROS/RNS can also modify signalling proteins although the mechanisms are not as clearly defined. For example, electrophilic lipids, formed as the reaction products of oxidation reactions, orchestrate adaptive responses in the vasculature by reacting with nucleophilic cysteine residues. In modifying signalling proteins ROS/RNS appear to change the overall activity of signalling pathways in a process that we have termed 'redox tone'. In this review, we discuss these different mechanisms of redox cell signalling, and give specific examples of ROS/RNS participation in signal transduction.


Asunto(s)
Metabolismo de los Lípidos , Óxido Nítrico/metabolismo , Oxidación-Reducción , Especies de Nitrógeno Reactivo/metabolismo , Transducción de Señal/fisiología , Procesamiento Postranscripcional del ARN
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...