Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Antioxidants (Basel) ; 10(8)2021 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-34439522

RESUMEN

We have evaluated the role of mitochondrial oxidative stress and its association with endoplasmic reticulum (ER) stress activation in the progression of obesity-related cardiovascular fibrosis. MitoQ (200 µM) was orally administered for 7 weeks to male Wistar rats that were fed a high-fat diet (HFD, 35% fat) or a control diet (CT, 3.5% fat). Obese animals presented cardiovascular fibrosis accompanied by increased levels of extracellular matrix proteins and profibrotic mediators. These alterations were associated with ER stress activation characterized by enhanced levels (in heart and aorta vs. CT group, respectively) of immunoglobulin binding protein (BiP; 2.1-and 2.6-fold, respectively), protein disulfide-isomerase A6 (PDIA6; 1.9-fold) and CCAAT-enhancer-binding homologous protein (CHOP; 1.5- and 1.8-fold, respectively). MitoQ treatment was able to prevent (p < 0.05) these modifications at cardiac and aortic levels. MitoQ (5 nM) and the ER stress inhibitor, 4-phenyl butyric acid (4 µM), were able to block the prooxidant and profibrotic effects of angiotensin II (Ang II, 10-6 M) in cardiac and vascular cells. Therefore, the data show a crosstalk between mitochondrial oxidative stress and ER stress activation, which mediates the development of cardiovascular fibrosis in the context of obesity and in which Ang II can play a relevant role.

2.
Cells ; 9(2)2020 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-32079154

RESUMEN

The impact of the mitochondria-targeted antioxidant MitoQ was evaluated in the cardiac alterations associated with obesity. Male Wistar rats were fed either a high fat diet (HFD, 35% fat) or a standard diet (CT, 3.5% fat) for 7 weeks and treated with MitoQ (200 µM). The effect of MitoQ (5 nM) in rat cardiac myoblasts treated for 24 h with palmitic acid (PA, 200 µM) was evaluated. MitoQ reduced cardiac oxidative stress and prevented the development of cardiac fibrosis, hypertrophy, myocardial 18-FDG uptake reduction, and mitochondrial lipid remodeling in HFD rats. It also ameliorated cardiac mitochondrial protein level changes observed in HFD: reductions in fumarate hydratase, complex I and II, as well as increases in mitofusin 1 (MFN1), peroxisome proliferator-activated receptor gamma coactivator 1-alpha, and cyclophilin F (cycloF). In vitro, MitoQ prevented oxidative stress and ameliorated alterations in mitochondrial proteins observed in palmitic acid (PA)-stimulated cardiac myoblasts: increases in carnitine palmitoyltransferase 1A, cycloF, and cytochrome C. PA induced phosphorylation of extracellular signal-regulated kinases and nuclear factor-κB p65. Therefore, the data show the beneficial effects of MitoQ in the cardiac damage induced by obesity and suggests a crosstalk between lipotoxicity and mitochondrial oxidative stress in this damage.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Mitocondrias/metabolismo , Miocardio/metabolismo , Obesidad/complicaciones , Compuestos Organofosforados/uso terapéutico , Ubiquinona/análogos & derivados , Animales , Modelos Animales de Enfermedad , Humanos , Masculino , Compuestos Organofosforados/farmacología , Estrés Oxidativo , Ratas , Ratas Wistar , Ubiquinona/farmacología , Ubiquinona/uso terapéutico
3.
FASEB J ; 33(11): 12060-12072, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31370681

RESUMEN

The impact of the mitochondria-targeted antioxidant MitoQ was evaluated in the metabolic alterations and the adipose tissue remodeling associated with obesity. Male Wistar rats were fed either a high-fat diet (HFD; 35% fat) or a standard diet (3.5% fat) for 7 wk and treated with MitoQ (200 µM). A proteomic analysis of visceral adipose tissue from patients with obesity and patients without obesity was performed. MitoQ partially prevented the increase in body weight, adiposity, homeostasis model assessment index, and adipose tissue remodeling in HFD rats. It also ameliorated protein level changes of factors involved in insulin signaling observed in adipose tissue of obese rats: reductions in adiponectin and glucose transporter 4 (GLUT 4) and increases in dipeptidylpeptidase 4, suppressor of cytokine signaling 3 (SOCS3), and insulin receptor substrate 1 phosphorylation. MitoQ prevented down-regulation of adiponectin and GLUT 4 and increases in SOCS3 levels in a TNF-α-induced insulin-resistant 3T3-L1 adipocyte model. MitoQ also ameliorated alterations in mitochondrial proteins observed in obese rats: increases in cyclophylin F and carnitine palmitoyl transferase 1A and reductions in mitofusin1, peroxiredoxin 4, and fumarate hydratase. The proteomic analysis of the visceral adipose tissue from patients with obesity show alterations in mitochondrial proteins similar to those observed in obese rats. Therefore, the data show the beneficial effect of MitoQ in the metabolic dysfunction induced by obesity.-Marín-Royo, G., Rodríguez, C., Le Pape, A., Jurado-López, R., Luaces, M., Antequera, A., Martínez-González, J., Souza-Neto, F. V., Nieto, M. L., Martínez-Martínez, E., Cachofeiro, V. The role of mitochondrial oxidative stress in the metabolic alterations in diet-induced obesity in rats.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Mitocondrias/metabolismo , Obesidad/metabolismo , Estrés Oxidativo , Células 3T3-L1 , Adiposidad/efectos de los fármacos , Adiposidad/genética , Adulto , Animales , Femenino , Expresión Génica/efectos de los fármacos , Humanos , Masculino , Ratones , Persona de Mediana Edad , Obesidad/etiología , Compuestos Organofosforados/administración & dosificación , Proteómica/métodos , Ratas Wistar , Ubiquinona/administración & dosificación , Ubiquinona/análogos & derivados , Proteína Desacopladora 1/genética , Proteína Desacopladora 1/metabolismo
4.
Sci Rep ; 9(1): 444, 2019 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-30679580

RESUMEN

Cardiac lipotoxicity is involved in the cardiac functional consequences associated with obesity. Therefore, the aim of this study was to explore whether changes in the mitochondrial lipid cardiac profile could reflect differences in cardiac function and structure in obese and non-obese rats with myocardial infarction (MI). Whether these changes can also be reflected in a specific plasma miRNA signature as markers of cardiac damage was also evaluated. Rats were fed with either standard (3.5% fat) or high fat diet (35% fat) for 6 weeks before the induction of MI and sacrificed 4 weeks later. MI showed cardiac lipotoxicity independently of the presence of obesity, although obese and non-obese rats did not present the same cardiac lipid profile at mitochondrial level. Several cardiac lipid species in mitochondria, including cardiolipins and triglycerides, were associated with myocardial fibrosis, with mitochondrial triglyceride levels being independently associated with it; this supports that lipotoxicity can affect cardiac function. MI down-regulated plasma levels of miRNA 15b-5p and 194-5p in obese and non-obese animals, which were associated with cardiac function, mitochondrial lipids and myocardial fibrosis, with miRNA 15b-5p levels being independently associated with cardiac fibrosis. This could support that lipotoxicity could affect heart function by modulating plasma miRNAs.


Asunto(s)
Cardiomiopatías/genética , Lípidos/análisis , MicroARNs/genética , Obesidad/genética , Transducción de Señal/genética , Animales , Cardiolipinas/análisis , Cardiomiopatías/metabolismo , Cardiomiopatías/fisiopatología , Dieta Alta en Grasa/efectos adversos , Fibrosis , Perfilación de la Expresión Génica/métodos , Masculino , MicroARNs/sangre , Infarto del Miocardio/etiología , Infarto del Miocardio/genética , Infarto del Miocardio/metabolismo , Miocardio/metabolismo , Miocardio/patología , Obesidad/fisiopatología , Ratas Wistar , Triglicéridos/análisis
5.
Dis Model Mech ; 11(2)2018 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-29361517

RESUMEN

Obesity is accompanied by metabolic alterations characterized by insulin resistance and cardiac lipotoxicity. Galectin-3 (Gal-3) induces cardiac inflammation and fibrosis in the context of obesity; however, its role in the metabolic consequences of obesity is not totally established. We have investigated the potential role of Gal-3 in the cardiac metabolic disturbances associated with obesity. In addition, we have explored whether this participation is, at least partially, acting on mitochondrial damage. Gal-3 inhibition in rats that were fed a high-fat diet (HFD) for 6 weeks with modified citrus pectin (MCP; 100 mg/kg/day) attenuated the increase in cardiac levels of total triglyceride (TG). MCP treatment also prevented the increase in cardiac protein levels of carnitine palmitoyl transferase IA, mitofusin 1, and mitochondrial complexes I and II, reactive oxygen species accumulation and decrease in those of complex V but did not affect the reduction in 18F-fluorodeoxyglucose uptake observed in HFD rats. The exposure of cardiac myoblasts (H9c2) to palmitic acid increased the rate of respiration, mainly due to an increase in the proton leak, glycolysis, oxidative stress, ß-oxidation and reduced mitochondrial membrane potential. Inhibition of Gal-3 activity was unable to affect these changes. Our findings indicate that Gal-3 inhibition attenuates some of the consequences of cardiac lipotoxicity induced by a HFD since it reduced TG and lysophosphatidyl choline (LPC) levels. These reductions were accompanied by amelioration of the mitochondrial damage observed in HFD rats, although no improvement was observed regarding insulin resistance. These findings increase the interest for Gal-3 as a potential new target for therapeutic intervention to prevent obesity-associated cardiac lipotoxicity and subsequent mitochondrial dysfunction.


Asunto(s)
Galectina 3/antagonistas & inhibidores , Corazón/efectos de los fármacos , Lípidos/toxicidad , Obesidad/patología , Animales , Presión Sanguínea/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Dieta Alta en Grasa , Fibrosis , Fluorodesoxiglucosa F18/química , Galectina 3/metabolismo , Glucosa/metabolismo , Glucólisis/efectos de los fármacos , Corazón/diagnóstico por imagen , Corazón/fisiopatología , Resistencia a la Insulina , Masculino , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Dinámicas Mitocondriales , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Oxidación-Reducción , Ratas Wistar , Superóxidos/metabolismo
6.
Clin Investig Arterioscler ; 30(1): 10-20, 2018.
Artículo en Inglés, Español | MEDLINE | ID: mdl-28869040

RESUMEN

AIMS: To explore the impact of obesity on the cardiac lipid profile in rats with diet-induced obesity, as well as to evaluate whether or not the specific changes in lipid species are associated with cardiac fibrosis. METHODS: Male Wistar rats were fed either a high-fat diet (HFD, 35% fat) or standard diet (3.5% fat) for 6 weeks. Cardiac lipids were analyzed using by liquid chromatography-tandem mass spectrometry. RESULTS: HFD rats showed cardiac fibrosis and enhanced levels of cardiac superoxide anion (O2), HOMA index, adiposity, and plasma leptin, as well as a reduction in those of cardiac glucose transporter (GLUT 4), compared with control animals. Cardiac lipid profile analysis showed a significant increase in triglycerides, especially those enriched with palmitic, stearic, and arachidonic acid. An increase in levels of diacylglycerol (DAG) was also observed. No changes in cardiac levels of diacyl phosphatidylcholine, or even a reduction in total levels of diacyl phosphatidylethanolamine, diacyl phosphatidylinositol, and sphingomyelins (SM) was observed in HFD, as compared with control animals. After adjustment for other variables (oxidative stress, HOMA, cardiac hypertrophy), total levels of DAG were independent predictors of cardiac fibrosis while the levels of total SM were independent predictors of the cardiac levels of GLUT 4. CONCLUSIONS: These data suggest that obesity has a significant impact on cardiac lipid composition, although it does not modulate the different species in a similar manner. Nonetheless, these changes are likely to participate in the cardiac damage in the context of obesity, since total DAG levels can facilitate the development of cardiac fibrosis, and SM levels predict GLUT4 levels.


Asunto(s)
Transportador de Glucosa de Tipo 4/metabolismo , Cardiopatías/patología , Metabolismo de los Lípidos , Obesidad/complicaciones , Animales , Cromatografía Liquida , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Fibrosis , Cardiopatías/etiología , Leptina/metabolismo , Masculino , Ratas , Ratas Wistar , Espectrometría de Masas en Tándem
7.
Sci Rep ; 7(1): 16802, 2017 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-29196758

RESUMEN

We have investigated whether mineralocorticoid receptor activation can participate in the profibrotic effects of leptin in cardiac myofibroblasts, as well as the potential mechanisms involved. The presence of eplerenone reduced the leptin-induced increase in protein levels of collagen I, transforming growth factor ß, connective tissue growth factor and galectin-3 and the levels of both total and mitochondrial of superoxide anion (O2.-) in cardiac myofibroblasts. Likewise, the MEK/ERK inhibitor, PD98059, and the PI3/Akt inhibitor, LY294002, showed a similar pattern. Mitochondrial reactive oxygen species (ROS) scavenger (MitoTempo) attenuated the increase in body weight observed in rats fed a high fat diet (HFD). No differences were found in cardiac function or blood pressure among any group. However, the cardiac fibrosis and enhanced O2.-levels observed in HFD rats were attenuated by MitoTempo, which also prevented the increased circulating leptin and aldosterone levels in HFD fed animals. This study supports a role of mineralocorticoid receptor in the cardiac fibrosis induced by leptin in the context of obesity and highlights the role of the mitochondrial ROS in this process.


Asunto(s)
Fibrosis Endomiocárdica/metabolismo , Leptina/metabolismo , Miocardio/citología , Obesidad/complicaciones , Especies Reactivas de Oxígeno/metabolismo , Receptores de Mineralocorticoides/metabolismo , Animales , Colágeno Tipo I/metabolismo , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Fibrosis Endomiocárdica/etiología , Eplerenona/farmacología , Fibroblastos/citología , Galectina 3/metabolismo , Masculino , Mitocondrias/metabolismo , Obesidad/inducido químicamente , Obesidad/metabolismo , Estrés Oxidativo , Ratas , Ratas Wistar , Factor de Crecimiento Transformador beta/metabolismo
8.
J Mol Cell Cardiol ; 92: 96-104, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26780438

RESUMEN

Lysyl oxidase (LOX) is an extracellular matrix (ECM)-modifying enzyme that has been involved in cardiovascular remodeling. We explore the impact of LOX inhibition in ECM alterations induced by obesity in the cardiovascular system. LOX is overexpressed in the heart and aorta from rats fed a high-fat diet (HFD). ß-Aminopropionitrile (BAPN), an inhibitor of LOX activity, significantly attenuated the increase in body weight and cardiac hypertrophy observed in HFD rats. No significant differences were found in cardiac function or blood pressure among any group. However, HFD rats showed cardiac and vascular fibrosis and enhanced levels of superoxide anion (O2(-)), collagen I and transforming growth factor ß (TGF-ß) in heart and aorta and connective tissue growth factor (CTGF) in aorta, effects that were attenuated by LOX inhibition. Interestingly, BAPN also prevented the increase in circulating leptin levels detected in HFD fed animals. Leptin increased protein levels of collagen I, TGF-ß and CTGF, Akt phosphorylation and O2(-) production in both cardiac myofibroblasts and vascular smooth muscle cells in culture, while LOX inhibition ameliorated these alterations. LOX knockdown also attenuated leptin-induced collagen I production in cardiovascular cells. Our findings indicate that LOX inhibition attenuates the fibrosis and the oxidative stress induced by a HFD on the cardiovascular system. The reduction of leptin levels by BAPN in vivo and the ability of this compound to inhibit leptin-induced profibrotic mediators and ROS production in cardiac and vascular cells suggest that interactions between leptin and LOX regulate downstream events responsible for myocardial and vascular fibrosis in obesity.


Asunto(s)
Fibrosis/tratamiento farmacológico , Leptina/metabolismo , Miocardio/metabolismo , Obesidad/tratamiento farmacológico , Proteína-Lisina 6-Oxidasa/biosíntesis , Aminopropionitrilo/administración & dosificación , Animales , Aorta/metabolismo , Aorta/patología , Presión Sanguínea/efectos de los fármacos , Peso Corporal , Dieta Alta en Grasa , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/genética , Fibrosis/genética , Fibrosis/patología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Miocardio/patología , Obesidad/genética , Obesidad/patología , Proteína-Lisina 6-Oxidasa/antagonistas & inhibidores , Proteína-Lisina 6-Oxidasa/genética , Ratas , Especies Reactivas de Oxígeno/metabolismo
9.
Hypertension ; 66(5): 961-9, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26351031

RESUMEN

Remodeling, diastolic dysfunction, and arterial stiffness are some of the alterations through which obesity affects the cardiovascular system. Fibrosis and inflammation are important mechanisms underlying cardiovascular remodeling, although the precise promoters involved in these processes are still unclear. Galectin-3 (Gal-3) induces inflammation and fibrosis in the cardiovascular system. We have investigated the potential role of Gal-3 in cardiac damage in morbidly obese patients, and we have evaluated the protective effect of the Gal-3 inhibition in the occurrence of cardiovascular fibrosis and inflammation in an experimental model of obesity. Morbid obesity is associated with alterations in cardiac remodeling, mainly left ventricular hypertrophy and diastolic dysfunction. Obesity and hypertension are the main determinants of left ventricular hypertrophy. Insulin resistance, left ventricular hypertrophy, and circulating levels of C-reactive protein and Gal-3 are associated with a worsening of diastolic function in morbidly obese patients. Obesity upregulates Gal-3 production in the cardiovascular system in a normotensive animal model of diet-induced obesity by feeding for 6 weeks a high-fat diet (33.5% fat). Gal-3 inhibition with modified citrus pectin (100 mg/kg per day) reduced cardiovascular levels of Gal-3, total collagen, collagen I, transforming and connective growth factors, osteopontin, and monocyte chemoattractant protein-1 in the heart and aorta of obese animals without changes in body weight or blood pressure. In morbidly obese patients, Gal-3 levels are associated with diastolic dysfunction. In obese animals, Gal-3 blockade decreases cardiovascular fibrosis and inflammation. These data suggest that Gal-3 could be a novel therapeutic target in cardiac fibrosis and inflammation associated with obesity.


Asunto(s)
Sistema Cardiovascular/fisiopatología , Galectina 3/fisiología , Hipertrofia Ventricular Izquierda/etiología , Hipertrofia Ventricular Izquierda/fisiopatología , Obesidad/complicaciones , Obesidad/fisiopatología , Remodelación Ventricular/fisiología , Adulto , Animales , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Femenino , Fibrosis/epidemiología , Galectina 3/antagonistas & inhibidores , Galectina 3/efectos de los fármacos , Ventrículos Cardíacos/diagnóstico por imagen , Humanos , Incidencia , Inflamación/epidemiología , Modelos Lineales , Masculino , Persona de Mediana Edad , Miocardio/patología , Obesidad/etiología , Pectinas/farmacología , Ratas , Ratas Wistar , Análisis de Regresión , Ultrasonografía
10.
Hypertension ; 66(4): 767-75, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26238446

RESUMEN

Hypertensive cardiac remodeling is accompanied by molecular inflammation and fibrosis, 2 mechanisms that finally affect cardiac function. At cardiac level, aldosterone promotes inflammation and fibrosis, although the precise mechanisms are still unclear. Galectin-3 (Gal-3), a ß-galactoside-binding lectin, is associated with inflammation and fibrosis in the cardiovascular system. We herein investigated whether Gal-3 inhibition could block aldosterone-induced cardiac inflammation and fibrosis and its potential role in cardiac damage associated with hypertension. Aldosterone-salt-treated rats presented hypertension, cardiac inflammation, and fibrosis that were prevented by the pharmacological inhibition of Gal-3 with modified citrus pectin. Cardiac inflammation and fibrosis presented in spontaneously hypertensive rats were prevented by modified citrus pectin treatment, whereas Gal-3 blockade did not modify blood pressure levels. In the absence of blood pressure modifications, Gal-3 knockout mice were resistant to aldosterone-induced cardiac inflammation. In human cardiac fibroblasts, aldosterone increased Gal-3 expression via its mineralocorticoid receptor. Gal-3 and aldosterone enhanced proinflammatory and profibrotic markers, as well as metalloproteinase activities in human cardiac fibroblasts, effects that were not observed in Gal-3-silenced cells treated with aldosterone. In experimental hyperaldosteronism, the increase in Gal-3 expression was associated with cardiac inflammation and fibrosis, alterations that were prevented by Gal-3 blockade independently of blood pressure levels. These data suggest that Gal-3 could be a new molecular mechanism linking cardiac inflammation and fibrosis in situations with high-aldosterone levels, such as hypertension.


Asunto(s)
Galectina 3/antagonistas & inhibidores , Hiperaldosteronismo/complicaciones , Hipertensión/complicaciones , Miocarditis/prevención & control , Espironolactona/uso terapéutico , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Fibroblastos/efectos de los fármacos , Fibroblastos/patología , Fibrosis/etiología , Fibrosis/patología , Galectina 3/biosíntesis , Humanos , Hiperaldosteronismo/tratamiento farmacológico , Hiperaldosteronismo/metabolismo , Hipertensión/tratamiento farmacológico , Hipertensión/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Antagonistas de Receptores de Mineralocorticoides/uso terapéutico , Miocarditis/etiología , Miocarditis/patología , Ratas , Ratas Endogámicas WKY , Ratas Wistar
11.
Dis Model Mech ; 8(6): 543-51, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26035864

RESUMEN

Extracellular matrix (ECM) remodelling of the adipose tissue plays a pivotal role in the pathophysiology of obesity. The lysyl oxidase (LOX) family of amine oxidases, including LOX and LOX-like (LOXL) isoenzymes, controls ECM maturation, and upregulation of LOX activity is essential in fibrosis; however, its involvement in adipose tissue dysfunction in obesity is unclear. In this study, we observed that LOX is the main isoenzyme expressed in human adipose tissue and that its expression is strongly upregulated in samples from obese individuals that had been referred to bariatric surgery. LOX expression was also induced in the adipose tissue from male Wistar rats fed a high-fat diet (HFD). Interestingly, treatment with ß-aminopropionitrile (BAPN), a specific and irreversible inhibitor of LOX activity, attenuated the increase in body weight and fat mass that was observed in obese animals and shifted adipocyte size toward smaller adipocytes. BAPN also ameliorated the increase in collagen content that was observed in adipose tissue from obese animals and improved several metabolic parameters - it ameliorated glucose and insulin levels, decreased homeostasis model assessment (HOMA) index and reduced plasma triglyceride levels. Furthermore, in white adipose tissue from obese animals, BAPN prevented the downregulation of adiponectin and glucose transporter 4 (GLUT4), as well as the increase in suppressor of cytokine signaling 3 (SOCS3) and dipeptidyl peptidase 4 (DPP4) levels, triggered by the HFD. Likewise, in the TNFα-induced insulin-resistant 3T3-L1 adipocyte model, BAPN prevented the downregulation of adiponectin and GLUT4 and the increase in SOCS3 levels, and consequently normalised insulin-stimulated glucose uptake. Therefore, our data provide evidence that LOX plays a pathologically relevant role in the metabolic dysfunction induced by obesity and emphasise the interest of novel pharmacological interventions that target adipose tissue fibrosis and LOX activity for the clinical management of this disease.


Asunto(s)
Aminopropionitrilo/farmacología , Aminopropionitrilo/uso terapéutico , Metaboloma/efectos de los fármacos , Obesidad/tratamiento farmacológico , Obesidad/metabolismo , Proteína-Lisina 6-Oxidasa/antagonistas & inhibidores , Aumento de Peso/efectos de los fármacos , Células 3T3-L1 , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Adiponectina/metabolismo , Adiposidad/efectos de los fármacos , Animales , Tamaño de la Célula/efectos de los fármacos , Colágeno/metabolismo , Dieta Alta en Grasa , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Femenino , Fibrosis , Glucosa/metabolismo , Transportador de Glucosa de Tipo 4/metabolismo , Humanos , Insulina/metabolismo , Resistencia a la Insulina , Grasa Intraabdominal/efectos de los fármacos , Grasa Intraabdominal/metabolismo , Masculino , Ratones , Modelos Biológicos , Proteína-Lisina 6-Oxidasa/metabolismo , Ratas Wistar , Transducción de Señal/efectos de los fármacos
12.
Mol Cell Endocrinol ; 411: 20-7, 2015 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-25896545

RESUMEN

Interleukin-33 (IL-33) but not soluble ST2 (sST2) exerts anti-inflammatory and protective effects in several tissues. Aldosterone, a proinflammatory mediator which promotes adipogenesis, is elevated in obese patients. The aim of this study was to investigate the interactions between IL-33/ST2 system and Aldosterone in adipose tissue. Rats fed a high fat diet presented increased sST2 expression, diminished IL-33/sST2 ratio and enhanced levels of differentiation and inflammation in adipose tissue as compared to controls. A similar pattern was observed in adipose tissue from C57BL/6 Aldosterone-treated mice. In both animal models, Aldosterone was correlated with sST2. Treatment of 3T3-L1 adipocytes with IL-33 delayed adipocyte differentiation diminished lipid accumulation and decreased inflammation. Aldosterone decreased IL-33 and increased sST2 expressions in differentiated adipocytes. Aldosterone-induced adipocyte differentiation and inflammation were blocked by IL-33 treatment, but sST2 did not exert any effects. The crosstalk between IL-33/ST2 and Aldosterone could be relevant in the metabolic consequences of obesity.


Asunto(s)
Adipogénesis/fisiología , Tejido Adiposo/metabolismo , Aldosterona/farmacología , Interleucina-33/metabolismo , Receptores de Interleucina/metabolismo , Células 3T3-L1 , Adipogénesis/efectos de los fármacos , Tejido Adiposo/efectos de los fármacos , Animales , Dieta Alta en Grasa , Inflamación/metabolismo , Proteína 1 Similar al Receptor de Interleucina-1 , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/metabolismo , Ratas , Ratas Wistar
13.
J Hypertens ; 32(5): 1104-14; discussion 1114, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24695395

RESUMEN

OBJECTIVE: Leptin acts as a cardiac profibrotic factor. However, the mechanisms underlying this effect are unclear. Therefore, we sought to elucidate the mediators involved in this process and the potential role of leptin in cardiac fibrosis associated with obesity. METHODS: Male Wistar rats were fed either a high-fat diet (HFD; 33.5% fat), or a standard diet (3.5% fat) for 6 weeks. RESULTS: HFD animals show cardiac hypertrophy, fibrosis and an increase in O2- production as evaluated by dihydroethidium. Echocardiographic parameters of cardiac structure and systolic function were similar in both groups. Cardiac levels of leptin, collagen I, galectin-3 and transforming growth factor ß (TGF-ß) were higher in HFD than in controls. In cardiac myofibroblasts, leptin (10-100 ng/ml) increased O2-, collagen I, galectin-3, TGF-ß and connective tissue growth factor production (CTGF). These effects were prevented by the presence of either melatonin (10 mmol/l) or the inhibitor of mTOR, rapamycin (10 mmol/l). Blockage of galectin-3 activity by N-acetyllactosamine (LacNac 10 mmol/l) reduced both collagen I and O2(*-) production induced by leptin. The p70S6 kinase activation/phosphorylation, the downstream mediator of mTOR, induced by leptin was not modified by melatonin. Leptin reduced the metalloproteinase (MMP) 2 activity and the presence of melatonin, rapamycin or LacNac were unable to prevent it. CONCLUSION: The data suggest that leptin locally produced in the heart could participate in the fibrosis observed in HFD by affecting collagen turnover. Collagen synthesis induced by leptin seems to be mediated by the production of galectin-3, TGF-ß and CTGF through oxidative stress increased by activation of mTOR pathway.


Asunto(s)
Cardiomiopatías/metabolismo , Fibrosis/metabolismo , Galectina 3/metabolismo , Leptina/fisiología , Obesidad/metabolismo , Estrés Oxidativo , Serina-Treonina Quinasas TOR/metabolismo , Animales , Células Cultivadas , Grasas de la Dieta/administración & dosificación , Masculino , Fosforilación , Ratas , Ratas Wistar , Superóxidos/metabolismo
14.
PLoS One ; 8(11): e79176, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24265755

RESUMEN

BACKGROUND: The function of the Interleukin-33 (IL-33)/ST2 system has been mainly investigated on immunological aspects, but recent data suggest that this pathway plays also an important role in cardiovascular system and adipose tissue. Whereas IL-33 has been demonstrated to exert anti-inflammatory and protective effects, circulating soluble ST2 (sST2) has emerged as a prognostic biomarker in patients with myocardial infarction and heart failure. Furthermore, sST2 is increased in severe obesity, although its role in the pathogenesis of vascular remodeling associated with obesity is still not well defined. METHODOLOGY/PRINCIPAL FINDINGS: Male Wistar rats fed standard diet (Control) or high fat diet (HFD) for 6 weeks. Aortic tunica media from diet-induced obese animals showed hypertrophy and fibrosis. The IL-33/ST2 system was spontaneously expressed in the aorta from Wistar rats. Administration of HFD in animals did not modify IL-33 expression at the transcriptional level. By contrast, HFD group showed an increase in aortic soluble sST2 and a decrease in the transmembrane isoform (ST2L) levels, resulting in decreased protective pathway activity. Aortic sST2 mRNA levels were associated with parameters showing vascular hypertrophy and fibrosis. In vitro experiments showed that primary cultured vascular smooth muscle cells (VSMCs) spontaneously expressed the IL-33/ST2 system. VSMCs stimulated with sST2 showed an increase in collagen type I, fibronectin and profibrotic factors. CONCLUSIONS: This is the first study demonstrating a deleterious role for sST2 in the vascular remodeling associated with obesity. In addition, we demonstrated that sST2 may act not only as a decoy receptor by binding IL-33 and preventing ST2L, but also modulating ECM remodeling and turnover. Thus, sST2 could be a new therapeutic target to reduce vascular remodeling in the context of obesity.


Asunto(s)
Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/patología , Obesidad/metabolismo , Obesidad/patología , Receptores de Interleucina-1/metabolismo , Animales , Aorta/metabolismo , Aorta/patología , Biomarcadores/metabolismo , Presión Sanguínea , Vasos Sanguíneos/efectos de los fármacos , Peso Corporal , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Fibrosis , Inflamación , Interleucina-33 , Interleucinas/metabolismo , Masculino , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Ratas , Transducción de Señal/efectos de los fármacos
15.
PLoS One ; 7(7): e41545, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22844495

RESUMEN

BACKGROUND: The natural triterpenes, erythrodiol and uvaol, exert anti-inflammatory, vasorelaxing and anti-proliferative effects. Angiotensin II is a well-known profibrotic and proliferative agent that participates in the cardiac remodeling associated with different pathological situations through the stimulation and proliferation of cardiac fibroblasts. Therefore, the aim of the study was to investigate the preventive effects of the natural triterpenes erythrodiol and uvaol on the proliferation and collagen production induced by angiotensin II in cardiac myofibroblasts. Their actions on cardiac hypertrophy triggered by angiotensin II were also studied. METHODOLOGY/PRINCIPAL FINDINGS: The effect of erythrodiol and uvaol on angiotensin II-induced proliferation was evaluated in cardiac myofibroblasts from adult rats in the presence or the absence of the inhibitors of PPAR-γ, GW9662 or JNK, SP600125. The effect on collagen levels induced by angiotensin II was evaluated in cardiac myofibroblasts and mouse heart. The presence of low doses of both triterpenes reduced the proliferation of cardiac myofibroblasts induced by angiotensin II. Pretreatment with GW9662 reversed the effect elicited by both triterpenes while SP600125 did not modify it. Both triterpenes at high doses produced an increase in annexing-V binding in the presence or absence of angiotensin II, which was reduced by either SP600125 or GW9662. Erythrodiol and uvaol decreased collagen I and galectin 3 levels induced by angiotensin II in cardiac myofribroblasts. Finally, cardiac hypertrophy, ventricular remodeling, fibrosis, and increases in myocyte area and brain natriuretic peptide levels observed in angiotensin II-infused mice were reduced in triterpene-treated animals. CONCLUSIONS/SIGNIFICANCE: Erythrodiol and uvaol reduce cardiac hypertrophy and left ventricle remodeling induced by angiotensin II in mice by diminishing fibrosis and myocyte area. They also modulate growth and survival of cardiac myofibroblasts. They inhibit the angiotensin II-induced proliferation in a PPAR-γ-dependent manner, while at high doses they activate pathways of programmed cell death that are dependent on JNK and PPAR-γ.


Asunto(s)
Angiotensina II/farmacología , Cardiomegalia/inducido químicamente , Cardiomegalia/prevención & control , Ácido Oleanólico/análogos & derivados , Triterpenos/farmacología , Animales , Apoptosis/efectos de los fármacos , Cardiomegalia/metabolismo , Cardiomegalia/patología , Proliferación Celular/efectos de los fármacos , Colágeno/metabolismo , Relación Dosis-Respuesta a Droga , Fibrosis , Masculino , Ratones , Miofibroblastos/efectos de los fármacos , Miofibroblastos/metabolismo , Miofibroblastos/patología , Ácido Oleanólico/farmacología , Ratas
16.
J Vasc Res ; 49(2): 111-22, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22249107

RESUMEN

The effect of adiponectin and leptin on the proliferation of the human microvascular endothelial cell line (HMEC-1) was studied in the absence or presence of fetal bovine serum (FBS). The participation of extracellular signal-regulated kinase (ERK) and phosphatidylinositol 3-kinase/Akt (PI-3K/Akt) pathways in this effect were evaluated. We studied the effect of both adipokines on the motility, mitosis, proliferation and cell death processes of HMEC-1 cells using live-cell imaging techniques. Adiponectin but not leptin further increased the proliferative effect induced by FBS on HMEC-1. This effect seems to be the consequence of an increase in the mitotic index in adiponectin-treated cells when compared to untreated ones. The presence of either the mitogen-activated protein kinase (MAPK) inhibitor (PD98059), or PI-3K inhibitor (LY294002), reduced the effect of adiponectin in a dose-dependent manner. Neither adipokine was able to affect HMEC-1 proliferation in FBS-free conditions. Duration of mitosis, cell motility and the cell death process were similar in all conditions. These data suggest that adiponectin and leptin exert different effects on endothelial cell function. Adiponectin was able to potentiate proliferation of HMEC-1. This effect involves the activation of both PI3-K/Akt and ERK/MAPK pathways. However, it seems to exert minimal effects on HMEC-1 function in the case of leptin.


Asunto(s)
Adiponectina/farmacología , Proliferación Celular/efectos de los fármacos , Leptina/farmacología , Línea Celular , Movimiento Celular/efectos de los fármacos , Cromonas/farmacología , Endotelio Vascular/citología , Quinasas MAP Reguladas por Señal Extracelular/efectos de los fármacos , Flavonoides/farmacología , Humanos , Microcirculación , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Morfolinas/farmacología , Inhibidores de las Quinasa Fosfoinosítidos-3
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...