Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Int J Mol Sci ; 22(24)2021 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-34948369

RESUMEN

It has been reported that 17ß-estradiol (E2) can exert beneficial effects against the development of obesity, providing women with a healthier metabolic profile and conferring cardiovascular protection. However, a growing body of evidence questions this role in the context of obesity and diabetes. We focus on the adipose tissue-heart axis to address the question of whether E2 can have metabolically detrimental effects in an obese-diabetic rat model. Female Zucker Diabetic Fatty rats were used: LEAN, fa/+; SHAM, sham-operated fa/fa; OVA, ovariectomized fa/fa, and OVA+E2, ovariectomized and E2 treated fa/fa. The secretory expression profile, tissue expansion parameters and composition of visceral adipose tissue, as well as systemic and cardiac parameters related to insulin resistance, fibrosis, and inflammation were analyzed. Ovariectomy induced an attenuation of both diabetic condition and metabolic dysfunction of adipose tissue and cardiac muscle in fa/fa rats, suggesting that E2, in the context of diabetes and obesity, loses its cardioprotective role and could even contribute to greater metabolic alterations. Adipose tissue from OVA rats showed a healthier hyperplastic expansion pattern, which could help maintain tissue function, increase adiponectin expression, and decrease pro-inflammatory adipokines. These findings should be taken into account when considering hormone replacement therapy for obese-diabetic women.


Asunto(s)
Tejido Adiposo/metabolismo , Enfermedades Cardiovasculares/metabolismo , Complicaciones de la Diabetes/metabolismo , Diabetes Mellitus/metabolismo , Estradiol/metabolismo , Obesidad/metabolismo , Tejido Adiposo/patología , Animales , Enfermedades Cardiovasculares/patología , Diabetes Mellitus/patología , Estrógenos/metabolismo , Femenino , Obesidad/patología , Ratas , Ratas Zucker
2.
Biochem Cell Biol ; 94(2): 167-75, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26914441

RESUMEN

Taking into account the sexual dimorphism previously found in white adipose tissue (WAT) regarding mitochondrial function and biogenesis, as well as insulin sensitivity, the aim of this study was to go further into the role of sex hormones in this dimorphism. To achieve this objective, we used ovariectomized rats and performed a screening by means of proteomic analyses of the periovarian WAT, combined with a study of the protein levels of specific factors involved in mitochondrial function. Rats were ovariectomized at 5 weeks of age and subcutaneously injected every 48 h with corn-oil (OVX group) or with 17ß-estradiol (E2, 10 µg/kg body mass; OVX + E2 group) for 4 weeks prior to sacrifice. Beside proteomic analysis, protein levels of Transcription Factor A, Mitochondrial (TFAM), cytochrome oxidase (COX)II, and COXIV were determined by Western blot, and mRNA levels of peroxisome proliferator-activated receptor-γ coactivator (PGC)-1α, ERα, ERß, lipoprotein lipase (LPL), peroxisome proliferator-activated receptor-γ (PPARγ), and adiponectin were quantified by real-time PCR. Our results show that ovariectomy leads to an increase in anabolic processes and inflammatory protein levels as well as to a decrease in some of the markers of mitochondrial function, which are restored, at least in part, by E2 supplementation. Indeed, this E2 supplementation seems to be counteracted by a decline in ERα and in the ERα to ERß ratio values that could be directed to avoid an over-stimulation of the E2 signaling pathway, given the possibility of an activation of extra-gonadal steroid biosynthetic pathways.


Asunto(s)
Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Estradiol/farmacología , Ovariectomía , Proteómica , Animales , Complejo IV de Transporte de Electrones/análisis , Complejo IV de Transporte de Electrones/metabolismo , Estradiol/administración & dosificación , Femenino , Inyecciones Subcutáneas , Ratas , Ratas Wistar , Factores de Transcripción/análisis , Factores de Transcripción/metabolismo
3.
Cell Biochem Funct ; 31(6): 504-10, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23112138

RESUMEN

The aim of the study was to determine, in rats of both sexes, the effect of HF diet feeding on the expression of adipokines involved in inflammatory status and insulin sensitivity and on the levels of proteins involved in lipid handling of retroperitoneal adipose tissue. Eight-week-old Wistar rats of both sexes were fed a control diet (2.9% w/w fat) or an HF diet (30% w/w fat) for 14 weeks. Adiponectin, peroxisome proliferator-activated receptor γ and inflammatory marker mRNA levels were analyzed by real-time polymerase chain reaction. Levels of insulin receptor, glucose transporter 4, carnitine palmitoyltransferase 1, fatty acid synthase, hormone-sensitive lipase and lipoprotein lipase were determined by Western blot. HF diet feeding did not induce hyperphagia or body weight gain but did promote an increase in adiposity although only in male rats. HF diet impaired glucose tolerance and the expression of inflammatory and insulin sensitivity markers in adipose tissue of male rats, but not in female rats. Male rats seem to be more prone to disorders associated with an unbalanced composition of the diet, even in the absence of hyperphagia. In contrast, female rats counteract excessive fat intake by improving their ability to use lipid fuels, which limits adiposity and maintains insulin sensitivity.


Asunto(s)
Dieta Alta en Grasa , Inflamación/metabolismo , Grasa Intraabdominal/metabolismo , Metabolismo de los Lípidos , Caracteres Sexuales , Adiponectina/metabolismo , Animales , Ingestión de Energía/genética , Femenino , Regulación de la Expresión Génica , Inflamación/patología , Insulina/metabolismo , Resistencia a la Insulina/genética , Masculino , PPAR gamma/biosíntesis , Ratas
4.
Biochem Cell Biol ; 90(2): 164-72, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22235846

RESUMEN

Ectopic deposition of lipids in liver and other extrahepatic tissues alters their function and occurs once adipose tissue fat storage capacity is exceeded. We investigated sexual dimorphism in the effects of dietary obesity on the liver insulin signaling pathway, as well as its connection to differences in hepatic fat accumulation. Ten-week-old Wistar rats of both sexes were fed a standard diet or a high-fat diet for 26 weeks. Insulin, adipokine levels, and glucose tolerance were measured. Lipid content, PPARα mRNA expression and protein levels of insulin receptor subunit ß (IRß), IR substrate 2 (IRS-2), Ser/Thr kinase A (Akt), and pyruvate dehydrogenase kinase isozyme 4 (PDK4) were measured in liver. In control rats, serum parameters and hepatic levels of IRß, IRS-2, and Akt proteins pointed to a profile of better insulin sensitivity in females. In response to dietary treatment, female rats exhibited a greater increase in body mass and adiposity and lower liver fat accumulation than males, but maintained better glucose tolerance. The reduced insulin signaling capacity in the liver of obese female rats seems to prevent lipid accumulation and probably lipotoxicity-associated hepatic disorders.


Asunto(s)
Resistencia a la Insulina , Metabolismo de los Lípidos , Hígado/metabolismo , Obesidad/metabolismo , Adiponectina/sangre , Adiposidad , Animales , Glucemia , Peso Corporal , Dieta Alta en Grasa/efectos adversos , Ingestión de Energía , Femenino , Variación Genética , Insulina/sangre , Proteínas Sustrato del Receptor de Insulina/metabolismo , Hígado/patología , Masculino , Obesidad/etiología , Tamaño de los Órganos , PPAR alfa/genética , PPAR alfa/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora , Ratas , Ratas Wistar , Receptor de Insulina/metabolismo , Factores Sexuales , Triglicéridos/sangre
5.
Cell Physiol Biochem ; 27(3-4): 201-6, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21471708

RESUMEN

Aging is responsible for the decline in the function of mitochondria and their increase in size and number--adaptive mechanism to restore mitochondrial function. Estrogens increase mitochondrial function, especially in female rats. The aim of this study was to determine the age-related changes in rat brain mitochondrial function focusing on sex differences. Cellular and mitochondrial protein and DNA content, mitochondrial oxidative and phosphorylative function in male and female rat brain from four different age groups (6, 12, 18 and 24 months old) were analyzed. Mitochondria protein/DNA content decreased with aging shifting toward lesser mitochondrial functional capacity and the mitochondria number increased. A sex dimorphism was determined, with female rat brain showing mitochondria with greater functional capacity than males. These sex differences gradually increased during aging.


Asunto(s)
Envejecimiento , Encéfalo/fisiología , Mitocondrias/fisiología , Animales , Citrato (si)-Sintasa/metabolismo , ADN Mitocondrial/metabolismo , Femenino , Masculino , Mitocondrias/enzimología , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Oxidación-Reducción , Fosforilación , Ratas , Ratas Wistar , Factores Sexuales
6.
Lipids Health Dis ; 10: 52, 2011 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-21486445

RESUMEN

BACKGROUND: High-fat (HF) diet feeding usually leads to hyperphagia and body weight gain, but macronutrient proportions in the diet can modulate energy intake and fat deposition. The mechanisms of fat accumulation and mobilization may differ significantly between depots, and gender can also influence these differences. AIM: To investigate, in rats of both sexes, the effect of an isocaloric intake of a diet with an unbalanced proportion of macronutrients on fatty acid composition of visceral and subcutaneous adipose tissues and how this is influenced by both dietary fatty acids and levels of proteins involved in tissue lipid handling. METHODS: Eight-week-old Wistar rats of both sexes were fed a control diet (3% w/w fat) or high-fat diet (30% w/w fat) for 14 weeks. Fatty acid composition was analyzed by gas-chromatography and levels of LPL, HSL, α2-AR, ß3-AR, PKA and CPT1 were determined by Western blot. RESULTS: The HF diet did not induce hyperphagia or body weight gain, but promoted an increase of adiposity index only in male rats. HF diet produced an increase of the proportion of MUFA and a decrease in that of PUFA in both adipose depots and in both sexes. The levels of proteins involved in the adrenergic control of the lipolytic pathway increased in the gonadal fat of HF females, whereas LPL levels increased in the inguinal fat of HF males and decreased in that of females. CONCLUSION: Sexual dimorphism in adiposity index reflects a differential sex response to dietary fatty acid content and could be related to the levels of the proteins involved in tissue lipid management.


Asunto(s)
Adiposidad/efectos de los fármacos , Grasas de la Dieta/efectos adversos , Ingestión de Energía/efectos de los fármacos , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Animales , Western Blotting , Ingestión de Alimentos/efectos de los fármacos , Femenino , Metabolismo de los Lípidos/efectos de los fármacos , Masculino , Ratas , Factores Sexuales
7.
Antioxid Redox Signal ; 35(9): 753-774, 2021 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-33736456

RESUMEN

Significance: Nonalcoholic fatty liver disease (NAFLD) is a hepatic and systemic disorder with a complex multifactorial pathogenesis. Owing to the rising incidence of obesity and diabetes mellitus, the prevalence of NAFLD and its impact on global health care are expected to increase in the future. Differences in NAFLD exist between males and females, and among females depending on their reproductive status. Clinical and preclinical data show that females in the fertile age are more protected against NAFLD, and studies in postmenopausal women and ovariectomized animal models support a protective role for estrogens. Recent Advances: An efficient crosstalk between the liver and adipose tissue is necessary to regulate lipid and glucose metabolism, protecting the liver from steatosis and insulin resistance contributing to NALFD. New advances in the knowledge of sexual dimorphism in liver and adipose tissue are providing interesting clues about the sex differences in NAFLD pathogenesis that could inspire new therapeutic strategies. Critical Issues: Sex hormones influence key master regulators of lipid metabolism and oxidative stress in liver and adipose tissue. All these sex-biased metabolic adjustments shape the crosstalk between liver and adipose tissue, contributing to the higher protection of females to NAFLD. Future Directions: The development of novel drugs based on the protective action of estrogens, but without its feminizing or undesired side effects, might provide new therapeutic strategies for the management of NAFLD. Antioxid. Redox Signal. 35, 753-774.


Asunto(s)
Enfermedad del Hígado Graso no Alcohólico , Tejido Adiposo/metabolismo , Animales , Estrógenos/metabolismo , Femenino , Humanos , Hígado/metabolismo , Masculino , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Caracteres Sexuales
8.
Cell Physiol Biochem ; 26(3): 291-302, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20798513

RESUMEN

BACKGROUND/AIMS: Mitochondrial biogenesis includes both mitochondrial proliferation and differentiation and its regulation under different physiological conditions is not clear. Given the sexual dimorphism previously found in mitochondrial function, the aim of this study was to investigate the gender-dependent effect of chronic high-fat-diet (HFD) feeding on rat liver mitochondrial function and biogenesis. METHODS: Ten-week old male and female rats were fed a HFD (26% fat) or a control diet (2.9% fat) for 26 weeks. Mitochondrial morphology was studied. Mitochondrial DNA and protein content, hydrogen peroxide production, oxidative capacity, antioxidant defenses, as well as markers of oxidative damage and mitochondrial biogenesis were analyzed. RESULTS: Female rats showed higher levels of mitochondrial protein and an enhanced oxidative capacity per mitochondrion than males. In both genders, HFD feeding increased mtDNA content and decreased mitochondrial differentiation markers. CONCLUSION: In comparison to male rats, females show higher oxidative capacity as a consequence of their greater mitochondrial differentiation under both control and obese status. In response to HFD feeding, the oxidative capacity of the whole mitochondrial population is maintained in both genders. This is obtained by means of an enhancement of mitochondrial proliferation, which counteracts the diet-induced impairment of the function of each mitochondrion.


Asunto(s)
Mitocondrias Hepáticas/metabolismo , Animales , Citrato (si)-Sintasa/metabolismo , Grasas de la Dieta/farmacología , Complejo IV de Transporte de Electrones/metabolismo , Estradiol/sangre , Femenino , Glutatión Peroxidasa/metabolismo , Peróxido de Hidrógeno/metabolismo , Masculino , Estrés Oxidativo , Ratas , Ratas Wistar , Factores Sexuales , Superóxido Dismutasa/metabolismo , Factores de Tiempo
9.
Free Radic Biol Med ; 150: 148-160, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32105829

RESUMEN

The prevalence and severity of nonalcoholic fatty liver disease (NAFLD) is higher in men and postmenopausal women compared to premenopausal women, suggesting a protective role for ovarian hormones. Diet-induced obesity and fatty acids surplus promote mitochondrial dysfunction in liver, triggering oxidative stress and activation of c-Jun N-terminal kinase (JNK) which has been related to the development of insulin resistance and steatosis, the main hallmarks of NAFLD. Considering that estrogen, in particular 17ß-estradiol (E2), have been reported to improve mitochondrial biogenesis and function in liver, our aim was to elucidate the role of E2 in preventing fatty acid-induced insulin resistance in hepatocytes through modulation of mitochondrial function, oxidative stress and JNK activation. An in vivo study was conducted in Wistar rats of both sexes (n = 7) fed control diet and high-fat diet (HFD), and in vitro studies were carried out in HepG2 cells treated with palmitate (PA) and E2 for 24 h. Our HFD-fed male rats showed a prediabetic state characterized by greater systemic and hepatic insulin resistance, as well as higher lipid content in liver, compared to females. JNK activation rose markedly in males in response to HFD feeding, in parallel with mitochondrial dysfunction and oxidative stress. Consistently, in PA-exposed HepG2 cells, E2 treatment prevented JNK activation, insulin resistance and fatty acid accumulation. Altogether, our data highlights the importance of E2 as a mitigating factor of fatty acid-insulin resistance in hepatocytes through downregulation of JNK activation, by means of mitochondrial function improvement.


Asunto(s)
Resistencia a la Insulina , Enfermedad del Hígado Graso no Alcohólico , Animales , Dieta Alta en Grasa/efectos adversos , Estradiol/metabolismo , Estradiol/farmacología , Femenino , Hígado/metabolismo , Masculino , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Enfermedad del Hígado Graso no Alcohólico/etiología , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Estrés Oxidativo , Ratas , Ratas Wistar
10.
J Steroid Biochem Mol Biol ; 185: 256-267, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30253224

RESUMEN

Obesity is associated with inflammation, dysregulated adipokine secretion, and disrupted adipose tissue mitochondrial function. Estradiol (E2) has been previously reported to increase mitochondrial function and biogenesis in several cell lines, but neither the type of oestrogen receptor (ERα, ERß and GPER) involved nor the mechanism whereby such effects are exerted have been fully described. Considering the anti-inflammatory activity of E2 as well as its effects in enhancing mitochondrial biogenesis, the aim of this study was to investigate the contribution of ERα, ERß, and GPER signaling to the E2-mediated enhancement of adipocyte mitochondrial function in a pro-inflammatory situation. 3T3-L1 cells were treated for 24 h with ER agonists (PPT, DPN, and G1) and antagonists (MPP, PHTPP, and G15) in the presence or absence of interleukin 6 (IL6), as a pro-inflammatory stimulus. Inflammation, mitochondrial function and biogenesis markers were analyzed. To confirm the involvement of the PKA pathway, cells were treated with a GPER agonist, a PKA inhibitor, and IL6. Mitochondrial function markers were analyzed. Our results showed that activation of ERα and GPER, but not ERß, was able to counteract the proinflammatory effects of IL6 treatment, as well as mitochondrial biogenesis and function indicators. Inhibition of PKA prevented the E2- and G1-associated increase in mitochondrial function markers. In conclusion E2 prevents IL6 induced inflammation in adipocytes and promotes mitochondrial function through the combined activation of both GPER and ERα. These findings expand our understanding of ER interactions under inflammatory conditions in female rodent white adipose tissue.


Asunto(s)
Adipocitos/patología , Estradiol/metabolismo , Receptor alfa de Estrógeno/metabolismo , Interleucina-6/metabolismo , Mitocondrias/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Células 3T3 , Animales , Línea Celular , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Receptor alfa de Estrógeno/agonistas , Receptor beta de Estrógeno/agonistas , Receptor beta de Estrógeno/metabolismo , Femenino , Inflamación/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/patología , Ratas , Ratas Wistar , Receptores Acoplados a Proteínas G/agonistas
11.
Cell Physiol Biochem ; 22(1-4): 327-36, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18769060

RESUMEN

High fat diets (HFD) usually lead to hyperphagia and body weight gain. However, macronutrient proportions in the diet can modulate energy intake and body fat deposition. The aim of the study was to investigate muscle and liver oxidative metabolism in response to an isocaloric intake of a HFD and to elucidate the possible gender-dependent response. Eight week-old male and female rats were fed either standard chow or HFD for 14 weeks. Energy intake, body weight and whole animal oxygen consumption were determined periodically. Mitochondrial oxygen consumption, hydrogen peroxide production, TBARS levels, Cytochrome-c-oxidase, Citrate synthase and antioxidant enzyme activities were measured in muscle and liver. UCP1 and UCP3 protein levels were analyzed in brown adipose tissue and muscle, respectively. Male rats showed higher energy efficiency, enhanced adiposity, greater hydrogen peroxide production and less effective antioxidant machinery compared to females. HFD feeding increased energy expenditure but did not modify either tissue oxidative metabolism or oxidative damage in either gender. HFD animals over-expressed uncoupling proteins in order to maintain energy balance (brown adipose tissue UCP1) and to avoid oxidative stress (skeletal muscle UCP3), thus counteracting the alterations induced by the modification of the proportion of macronutrients in the diet.


Asunto(s)
Grasas de la Dieta/farmacología , Conducta Alimentaria/efectos de los fármacos , Hígado/metabolismo , Músculo Esquelético/metabolismo , Caracteres Sexuales , Tejido Adiposo Pardo/metabolismo , Animales , Antioxidantes/metabolismo , Metabolismo Energético/efectos de los fármacos , Femenino , Peróxido de Hidrógeno/metabolismo , Canales Iónicos/metabolismo , Hígado/anatomía & histología , Hígado/enzimología , Masculino , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Músculo Esquelético/anatomía & histología , Músculo Esquelético/enzimología , Tamaño de los Órganos , Oxidación-Reducción/efectos de los fármacos , Consumo de Oxígeno/efectos de los fármacos , Ratas , Ratas Wistar , Sustancias Reactivas al Ácido Tiobarbitúrico/metabolismo , Proteína Desacopladora 1
12.
Cell Physiol Biochem ; 22(5-6): 539-48, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-19088436

RESUMEN

Muscle resistance to insulin plays a key role in the metabolic dysregulation associated to obesity. A pro-inflammatory and pro-oxidant status has been proposed to be the link between dietary obesity and insulin resistance. Given the gender differences previously found in mitochondrial function and oxidative stress, the aim of the present study was to investigate whether this gender dimorphism leads to differences in the development of high-fat-diet-induced insulin resistance in rat skeletal muscle. Male and female rats of 15 months of age were fed with a high-fat-diet (32% fat) for 14 weeks. Control male rats showed a more marked insulin resistance status compared to females, as indicated by the glucose tolerance curve profile and the serum insulin, resistin and adiponectin levels. High-fat-diet feeding induced an excess of body weight of 16.2% in males and 38.4% in females, an increase in both muscle mitochondrial hydrogen peroxide production and in oxidative damage, together with a decrease in the Mn-superoxide dismutase activity in both genders. However, high-fat-diet fed female rats showed a less marked insulin resistance profile than males, higher mitochondrial oxygen consumption and cytochrome c oxidase activity, and a better capacity to counteract the oxidative-stress-dependent insulin resistance through an overexpression of both muscle UCP3 and GLUT4 proteins. These results point to a gender dimorphism in the insulin resistance status and in the response of skeletal muscle to high-fat-diet feeding which could be related to a more detrimental effect of age in male rats.


Asunto(s)
Envejecimiento/efectos de los fármacos , Envejecimiento/fisiología , Grasas de la Dieta/farmacología , Resistencia a la Insulina/fisiología , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/fisiología , Caracteres Sexuales , Adipoquinas/sangre , Animales , Antioxidantes/metabolismo , Glucemia/metabolismo , Peso Corporal/efectos de los fármacos , Complejo IV de Transporte de Electrones/metabolismo , Metabolismo Energético/efectos de los fármacos , Femenino , Prueba de Tolerancia a la Glucosa , Transportador de Glucosa de Tipo 4/metabolismo , Peróxido de Hidrógeno/metabolismo , Insulina/sangre , Canales Iónicos/metabolismo , Peróxidos Lipídicos/metabolismo , Masculino , Mitocondrias/efectos de los fármacos , Mitocondrias/enzimología , Proteínas Mitocondriales/metabolismo , Consumo de Oxígeno/efectos de los fármacos , Carbonilación Proteica/efectos de los fármacos , Ratas , Ratas Wistar , Proteína Desacopladora 3 , Aumento de Peso/efectos de los fármacos
13.
J Endocrinol ; 232(2): 297-308, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27885055

RESUMEN

Sexual dimorphism in mitochondrial biogenesis and function has been described in many rat tissues, with females showing larger and more functional mitochondria. The family of the peroxisome proliferator-activated receptor gamma coactivator 1 (PGC1) plays a central role in the regulatory network governing mitochondrial biogenesis and function, but little is known about the different contribution of hepatic PGC1A and PGC1B in these processes. The aim of this study was to elucidate the role of 17ß-estradiol (E2) in mitochondrial biogenesis and function in liver and assess the contribution of both hepatic PGC1A and PGC1B as mediators of these effects. In ovariectomized (OVX) rats (half of which were treated with E2) estrogen deficiency led to impaired mitochondrial biogenesis and function, increased oxidative stress, and defective lipid metabolism, but was counteracted by E2 treatment. In HepG2 hepatocytes, the role of E2 in enhancing mitochondrial biogenesis and function was confirmed. These effects were unaffected by the knockdown of PGC1A, but were impaired when PGC1B expression was knocked down by specific siRNA. Our results reveal a widespread protective role of E2 in hepatocytes, which is explained by enhanced mitochondrial content and oxidative capacity, lower hepatic lipid accumulation, and a reduction of oxidative stress. We also suggest a novel hepatic protective role of PGC1B as a modulator of E2 effects on mitochondrial biogenesis and function supporting activation of PGC1B as a therapeutic target for hepatic mitochondrial disorders.


Asunto(s)
Estradiol/farmacología , Metabolismo de los Lípidos/efectos de los fármacos , Hígado/efectos de los fármacos , Mitocondrias/metabolismo , Coactivadores de Receptor Nuclear/metabolismo , Estrés Oxidativo/efectos de los fármacos , Animales , Femenino , Metabolismo de los Lípidos/fisiología , Hígado/metabolismo , Ratones Transgénicos , Coactivadores de Receptor Nuclear/genética , Biogénesis de Organelos , Ovariectomía , Oxidación-Reducción , Estrés Oxidativo/fisiología , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , ARN Interferente Pequeño , Ratas
14.
J Steroid Biochem Mol Biol ; 172: 9-19, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28529127

RESUMEN

Peroxiredoxin 3 (PRX3) plays a role as a regulator of the adipocyte mitochondrial function due to its antioxidant activity. We have previously reported the existence of a sexual dimorphism in the mitochondrial oxidative stress status of many rat tissues such as white (WAT) and brown (BAT) adipose tissues. The aim was to elucidate whether sex hormones may play a role in PRX3 expression in the adipose tissues of rats. In in vivo experiments, male and female standard diet fed rats, high fat diet (HFD) fed rats and rosiglitazone-supplemented HFD (HDF+Rsg) fed rats, as well as ovariectomized (OVX) and 17beta-estradiol-supplemented OVX (OVX+E2) female rats were used. 3T3-L1 adipocytes and brown adipocyte primary culture were used to study the roles of both E2 and testosterone in in vitro experiments. PRX3 levels were greater in the WAT of female rats than in males. This sexual dimorphism disappeared by HFD feeding but was magnified with Rsg supplementation. PRX3 sexual dimorphism was not observed in BAT, and neither HFD nor ovariectomy modified PRX3 levels. Rsg increased Prx3 expression in the BAT of both sexes. In vitro studies supported the results obtained in vivo and confirmed the contribution of E2 to sex differences in WAT Prx3 expression. Finally, we reported an E2 upregulation of both PRX3 and thioredoxin 2 (TRX2) in WAT but not in BAT that could play a key role in the sex dimorphism reported in the antioxidant defence of WAT in order to palliate the detrimental effect of the oxidative stress.


Asunto(s)
Adipocitos Marrones/metabolismo , Adipocitos Blancos/metabolismo , Estradiol/farmacología , Proteínas de Homeodominio/genética , Caracteres Sexuales , Tiorredoxinas/genética , Células 3T3-L1 , Adipocitos Marrones/efectos de los fármacos , Adipocitos Blancos/efectos de los fármacos , Tejido Adiposo Blanco/efectos de los fármacos , Tejido Adiposo Blanco/metabolismo , Animales , Antioxidantes/metabolismo , Dieta Alta en Grasa , Grasas de la Dieta/efectos adversos , Femenino , Regulación de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Hipoglucemiantes/farmacología , Masculino , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Ovariectomía , Cultivo Primario de Células , Ratas , Ratas Wistar , Rosiglitazona , Transducción de Señal , Testosterona/farmacología , Tiazolidinedionas/farmacología , Tiorredoxinas/metabolismo
15.
J Nutr Biochem ; 17(3): 197-203, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16214326

RESUMEN

Paraoxonase 1 (PON1) associates to specific high-density lipoproteins (HDLs)--those containing apolipoprotein A-I (apoA-I) and apolipoprotein J (apoJ)--and is largely responsible for their antiatherogenic properties. Caloric restriction (CR) has been shown to reduce major atherosclerotic risk factors. The aims of this work were to study PON1 activity response to CR (40% over 14 weeks) and to elucidate whether there are adaptive differences related to gender. Serum and liver paraoxonase and arylesterase activities, serum triglyceride, total and HDL cholesterol concentrations, serum PON1, apoA-I and apoJ contents and liver PON1 mRNA levels were measured. No effects of CR or gender were observed in triglyceride, total cholesterol concentration and PON1 mRNA levels. HDL cholesterol was higher in female rats than in male rats and increased with CR only in the latter animals. Serum PON1 activities tended to be higher in female rats and dropped with CR, with females showing the biggest decrease. Serum PON1 content was higher in female rats and decreased in both genders with CR, whereas apoA-I and apoJ contents, which were higher in female rats too, decreased only in the former animals, accounting for the high PON1 activity decrease observed in these animals. In conclusion, the short-term CR-associated reduction of serum PON1 activity and PON1, apoA-I and apoJ levels points toward a reduced stability of HDL-PON1 complexes and/or HDL particle levels responsible for PON1 transport and function in the blood. Moreover, the variations in PON1 activity and apolipoprotein levels show gender-related differences that are indicative of a different adaptive strategy of male and female rats when faced with a period of food restriction.


Asunto(s)
Arildialquilfosfatasa/sangre , Restricción Calórica , Caracteres Sexuales , Animales , Apolipoproteína A-I/sangre , Arildialquilfosfatasa/genética , Arildialquilfosfatasa/metabolismo , Clusterina/sangre , Femenino , Privación de Alimentos , Lípidos/sangre , Hígado/anatomía & histología , Hígado/enzimología , Masculino , Microsomas Hepáticos/química , Tamaño de los Órganos , Proteínas/análisis , ARN Mensajero/análisis , Ratas
16.
Mol Cell Endocrinol ; 420: 116-24, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26628039

RESUMEN

Considering the sexual dimorphism described in cardiac mitochondrial function and oxidative stress, we aimed to investigate the role of 17ß-estradiol (E2) in these sex differences and the contribution of E2 receptors to these effects. As a model of chronic deprivation of ovarian hormones, we used ovariectomized (OVX) rats, half of which were treated with E2. Ovariectomy decreased markers of cardiac mitochondrial biogenesis and function and also increased oxidative stress, whereas E2 counteracted these effects. In H9c2 cardiomyocytes we observed that G-protein coupled estrogen receptor (GPER) agonist mimicked the effects of E2 in enhancing mitochondrial function and biogenesis, whereas GPER inhibitor neutralized them. These data suggest that E2 enhances mitochondrial function and decreases oxidative stress in cardiac muscle, thus it could be responsible for the sexual dimorphism observed in mitochondrial biogenesis and function in this tissue. These effects seem to be mediated through GPER stimulation.


Asunto(s)
Estradiol/farmacología , Mitocondrias Cardíacas/metabolismo , Biogénesis de Organelos , Receptores Acoplados a Proteínas G/metabolismo , Animales , Biomarcadores/metabolismo , Peso Corporal/efectos de los fármacos , Línea Celular , Estradiol/sangre , Femenino , Mitocondrias Cardíacas/efectos de los fármacos , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Tamaño de los Órganos/efectos de los fármacos , Ovariectomía , Estrés Oxidativo/efectos de los fármacos , Progesterona/sangre , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas Wistar , Receptores de Estrógenos/metabolismo
17.
Mitochondrion ; 5(1): 45-53, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16075478

RESUMEN

Rat brown adipose tissue mitochondrial subpopulations-isolated by differential centrifugation at 1000, 3000 and 8000g, giving the heavy, medium and light mitochondria-were characterized. Thus, contamination by non-mitochondrial subcellular components, morphological features, respiratory chain and antioxidant enzyme activities, both uncoupling protein 1 and mitochondrial protein content, mitochondrial DNA levels and mitochondrial integrity were measured. Results indicate that mitochondrial fractions showed important differences in the morphological, thermogenic and antioxidant properties. All the parameters studied were always higher in heavy mitochondria, which is indicative of a greater mitochondrial differentiation state.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Pardo/ultraestructura , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Termogénesis , Animales , Antioxidantes/metabolismo , Secuencia de Bases , Fraccionamiento Celular , ADN Mitocondrial/genética , ADN Mitocondrial/aislamiento & purificación , Femenino , Microscopía Electrónica , Mitocondrias/clasificación , Proteínas Mitocondriales/aislamiento & purificación , Ratas , Ratas Wistar
18.
Life Sci ; 76(10): 1147-58, 2005 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-15620578

RESUMEN

To investigate the possible existence of a gender dimorphism in the morphology and functionality of brown adipose tissue (BAT) mitochondrial subpopulations, we obtained three mitochondrial fractions - heavy, medium and light - by differential centrifugation. Electron microscopic analysis was carried out and mitochondrial protein content, cytochrome c oxidase and ATP synthase activities, mitochondrial DNA content and UCP1 protein levels were measured in each mitochondrial fraction. Female rats showed a greater mitochondrial size than males, with a different distribution pattern of the subpopulations. These differences were accompanied by higher oxidative and thermogenic capacities and a higher protein content in female rat BAT. This tissue also showed a greater tendency to respiratory chain uncoupling, as well as a close coordination between the oxidative, phosphorylative and thermogenic processes. These differences were found in the heavy subpopulation but not in the light one. Our results demonstrate that female rat BAT shows a highly differentiated mitochondrial pool, with the heavy mitochondrial subpopulation as the main responsible for the greater thermogenic activity of this tissue. In addition, it seems that there is a differential regulation of the mitochondrial growth cycle between genders in BAT, which leads to enhanced thermogenic capacity in female rat mitochondria.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Pardo/ultraestructura , Mitocondrias/ultraestructura , Termogénesis , Animales , Proteínas Portadoras/análisis , ADN Mitocondrial/análisis , Femenino , Canales Iónicos , Masculino , Proteínas de la Membrana/análisis , Mitocondrias/metabolismo , Proteínas Mitocondriales , ATPasas de Translocación de Protón Mitocondriales/análisis , Prostaglandina-Endoperóxido Sintasas/análisis , Ratas , Ratas Wistar , Caracteres Sexuales , Proteína Desacopladora 1
19.
Biochimie ; 106: 75-80, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25128631

RESUMEN

The aim of this study was to investigate the time-course response of retroperitoneal white adipose tissue (WAT) insulin and adiponectin signaling pathway intermediates in relation to the systemic age-associated impairment of insulin sensitivity in male and female rats. The main markers of the insulin and adiponectin signaling pathways of the retroperitoneal WAT, as well as of the systemic insulin sensitivity profile of 3-, 9- and 18-month old Wistar rats of both sexes were determined. Our results indicate that age leads to a decrease in the insulin sensitivity in both sexes that agrees with the decline in the levels of the WAT insulin signaling pathway intermediates, the increase in the adiposity index and the rise in the serum insulin resistance markers. This is accompanied by a sex-dimorphism that involves a gradual insulin signaling pathway decrease in female rats and an earlier and acute decrease in males and suggests a better insulin responsiveness in female rats at any age group. Our results confirm the idea that in rats, the insulin signaling pathway of WAT is altered at earlier ages than that of skeletal muscle and also provides further evidence of the impairment of the WAT adiponectin signaling pathway.


Asunto(s)
Tejido Adiposo Blanco/metabolismo , Envejecimiento/metabolismo , Resistencia a la Insulina , Transducción de Señal , Proteínas Quinasas Activadas por AMP/metabolismo , Adiponectina/sangre , Adiponectina/metabolismo , Tejido Adiposo Blanco/crecimiento & desarrollo , Factores de Edad , Animales , Glucemia/metabolismo , Western Blotting , Peso Corporal , Femenino , Insulina/sangre , Insulina/metabolismo , Masculino , Músculo Esquelético/metabolismo , Tamaño de los Órganos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas Wistar , Receptor de Insulina/metabolismo , Receptores de Adiponectina/metabolismo , Factores Sexuales , Factores de Tiempo
20.
J Mol Endocrinol ; 52(2): 203-14, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24604890

RESUMEN

Sexual dimorphism has been found in both mitochondrial functionality and adiponectin expression in white adipose tissue, with female rats presenting more functional mitochondria than males and greater adiponectin expression. However, little is known about the role of sex hormones in this dimorphism. The aim was to elucidate the role of sex hormones in mitochondrial biogenesis and dynamics and in adiponectin synthesis in white adipocytes, and also to provide new evidence of the link between these processes. 3T3-L1 preadipocytes were differentiated and treated either with 17-ß estradiol (E2; 10  nM), progesterone (Pg), testosterone (1  µM both), or a combination of Pg or testosterone with flutamide (FLT; 10  µM) or E2 (1  µM). The markers of mitochondrial biogenesis and dynamics and adiponectin expression were analyzed. E2 induced mitochondrial proliferation and differentiation in 3T3-L1, although testosterone showed opposite effects. Pg treatment stimulated proliferation but impaired differentiation. In concerns mitochondrial dynamics, these hormones promoted fusion over fission. FLT treatment indicated that Pg elicits its effects on mitochondrial dynamics through the androgen receptor. E2 coadministration with testosterone or Pg reversed its effects. In conclusion, our results show that E2 induces stimulation of mitochondrial biogenesis in white adipocytes in vitro, especially in situations that imply an impairment of mitochondrial function, whereas testosterone would have opposite effects. Moreover, testosterone and Pg alter mitochondrial dynamics by promoting fusion over fission, while E2 stimulates both processes. All these alterations run in parallel with changes in adiponectin expression, thus suggesting the existence of a link between mitochondrial biogenesis and dynamics and adiponectin synthesis in white adipocytes.


Asunto(s)
Adipocitos Blancos/metabolismo , Adiponectina/biosíntesis , Estradiol/farmacología , Recambio Mitocondrial/efectos de los fármacos , Testosterona/farmacología , Células 3T3-L1 , Adipocitos Blancos/efectos de los fármacos , Adiponectina/genética , Animales , Biomarcadores/metabolismo , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Femenino , Flutamida/farmacología , Masculino , Ratones , Dinámicas Mitocondriales/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Receptores de Adiponectina/genética , Receptores de Adiponectina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA