Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Int J Mol Sci ; 22(24)2021 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-34948369

RESUMO

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.


Assuntos
Tecido Adiposo/metabolismo , Doenças Cardiovasculares/metabolismo , Complicações do Diabetes/metabolismo , Diabetes Mellitus/metabolismo , Estradiol/metabolismo , Obesidade/metabolismo , Tecido Adiposo/patologia , Animais , Doenças Cardiovasculares/patologia , Diabetes Mellitus/patologia , Estrogênios/metabolismo , Feminino , Obesidade/patologia , Ratos , Ratos Zucker
2.
Biochem Cell Biol ; 94(2): 167-75, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26914441

RESUMO

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.


Assuntos
Tecido Adiposo/efeitos dos fármacos , Tecido Adiposo/metabolismo , Estradiol/farmacologia , Ovariectomia , Proteômica , Animais , Complexo IV da Cadeia de Transporte de Elétrons/análise , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Estradiol/administração & dosagem , Feminino , Injeções Subcutâneas , Ratos , Ratos Wistar , Fatores de Transcrição/análise , Fatores de Transcrição/metabolismo
3.
Nutrients ; 16(18)2024 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-39339665

RESUMO

Obesity and type 2 diabetes mellitus are global public health issues. Although males show higher obesity and insulin resistance prevalence, current treatments often neglect sex-specific differences. White adipose tissue (WAT) is crucial in preventing lipotoxicity and inflammation and has become a key therapeutic target. Rosiglitazone (RSG), a potent PPARγ agonist, promotes healthy WAT growth and mitochondrial function through MitoNEET modulation. Recent RSG-based strategies specifically target white adipocytes, avoiding side effects. Our aim was to investigate whether sex-specific differences in the insulin-sensitizing effects of RSG exist on WAT during obesity and inflammation. We used Wistar rats of both sexes fed a high-fat diet (HFD, 22.5% fat content) for 16 weeks. Two weeks before sacrifice, a group of HFD-fed rats received RSG treatment (4 mg/kg of body weight per day) within the diet. HFD male rats showed greater insulin resistance, inflammation, mitochondrial dysfunction, and dyslipidemia than females. RSG had more pronounced effects in males, significantly improving insulin sensitivity, fat storage, mitochondrial function, and lipid handling in WAT while reducing ectopic fat deposition and enhancing adiponectin signaling in the liver. Our study suggests a significant sexual dimorphism in the anti-diabetic effects of RSG on WAT, correlating with the severity of metabolic dysfunction.


Assuntos
Tecido Adiposo Branco , Dieta Hiperlipídica , Resistência à Insulina , Rosiglitazona , Animais , Feminino , Masculino , Ratos , Adiponectina/metabolismo , Tecido Adiposo Branco/metabolismo , Tecido Adiposo Branco/efeitos dos fármacos , Dieta Hiperlipídica/efeitos adversos , Hipoglicemiantes/farmacologia , Inflamação/tratamento farmacológico , Fígado/metabolismo , Fígado/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Obesidade/tratamento farmacológico , Obesidade/metabolismo , PPAR gama/metabolismo , PPAR gama/agonistas , Ratos Wistar , Rosiglitazona/farmacologia , Caracteres Sexuais , Fatores Sexuais
4.
Free Radic Biol Med ; 224: 574-587, 2024 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-39241985

RESUMO

Estrogens have a well-known protective role in the development of the metabolic syndrome. Nevertheless, recent epidemiological data question the cardioprotective effect of estrogens in obese and diabetic women. In this context, white adipose tissue (WAT) becomes dysfunctional, which has an impact on the cardiovascular system. The aim of the study was to elucidate the role of 17ß-estradiol (E2) in the interplay between adipose tissue and endothelial function in an animal model of diabesity. We used ZDF (fa/fa) female rats subjected to ovariectomy (OVA), OVA + E2 or sham operated, as well as non-obese non-diabetic ZDF (fa/+) rats. Endothelial function and vascular remodeling markers were assessed in the aorta, while mitochondrial function, oxidative stress, and adiponectin production were analyzed in gonadal WAT. Conditioned media from gonadal WAT explants were used to assess the effects of WAT secretome on HUVEC. Additionally, the adiponectin receptor agonist AdipoRON and E2 were utilized to examine potential interactions. Ovariectomy ameliorated the WAT dysfunction associated to the obese and diabetic state and promoted adiponectin secretion, effects that were linked to a reduction of endothelial dysfunction and inflammatory markers in the aorta of OVA rats and in HUVEC treated with OVA-conditioned media. Our findings provide evidence supporting the idea that in the context of obesity and diabetes, ovariectomy improves WAT secretome and positively impacts endothelial function, suggesting a detrimental role for E2. Additionally, our results point to adiponectin as the primary driver of the effects exerted by ovariectomy on the adipovascular axis.

5.
Cell Biochem Funct ; 31(6): 504-10, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23112138

RESUMO

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.


Assuntos
Dieta Hiperlipídica , Inflamação/metabolismo , Gordura Intra-Abdominal/metabolismo , Metabolismo dos Lipídeos , Caracteres Sexuais , Adiponectina/metabolismo , Animais , Ingestão de Energia/genética , Feminino , Regulação da Expressão Gênica , Inflamação/patologia , Insulina/metabolismo , Resistência à Insulina/genética , Masculino , PPAR gama/biossíntese , Ratos
6.
Biochem Cell Biol ; 90(2): 164-72, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22235846

RESUMO

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.


Assuntos
Resistência à Insulina , Metabolismo dos Lipídeos , Fígado/metabolismo , Obesidade/metabolismo , Adiponectina/sangue , Adiposidade , Animais , Glicemia , Peso Corporal , Dieta Hiperlipídica/efeitos adversos , Ingestão de Energia , Feminino , Variação Genética , Insulina/sangue , Proteínas Substratos do Receptor de Insulina/metabolismo , Fígado/patologia , Masculino , Obesidade/etiologia , Tamanho do Órgão , PPAR alfa/genética , PPAR alfa/metabolismo , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Piruvato Desidrogenase Quinase de Transferência de Acetil , Ratos , Ratos Wistar , Receptor de Insulina/metabolismo , Fatores Sexuais , Triglicerídeos/sangue
7.
Cell Physiol Biochem ; 27(3-4): 201-6, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21471708

RESUMO

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.


Assuntos
Envelhecimento , Encéfalo/fisiologia , Mitocôndrias/fisiologia , Animais , Citrato (si)-Sintase/metabolismo , DNA Mitocondrial/metabolismo , Feminino , Masculino , Mitocôndrias/enzimologia , Mitocôndrias/metabolismo , Proteínas Mitocondriais/metabolismo , Oxirredução , Fosforilação , Ratos , Ratos Wistar , Fatores Sexuais
8.
Lipids Health Dis ; 10: 52, 2011 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-21486445

RESUMO

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.


Assuntos
Adiposidade/efeitos dos fármacos , Gorduras na Dieta/efeitos adversos , Ingestão de Energia/efeitos dos fármacos , Tecido Adiposo/efeitos dos fármacos , Tecido Adiposo/metabolismo , Animais , Western Blotting , Ingestão de Alimentos/efeitos dos fármacos , Feminino , Metabolismo dos Lipídeos/efeitos dos fármacos , Masculino , Ratos , Fatores Sexuais
9.
Antioxid Redox Signal ; 35(9): 753-774, 2021 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-33736456

RESUMO

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.


Assuntos
Hepatopatia Gordurosa não Alcoólica , Tecido Adiposo/metabolismo , Animais , Estrogênios/metabolismo , Feminino , Humanos , Fígado/metabolismo , Masculino , Hepatopatia Gordurosa não Alcoólica/tratamento farmacológico , Hepatopatia Gordurosa não Alcoólica/metabolismo , Caracteres Sexuais
10.
Cell Physiol Biochem ; 26(3): 291-302, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20798513

RESUMO

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.


Assuntos
Mitocôndrias Hepáticas/metabolismo , Animais , Citrato (si)-Sintase/metabolismo , Gorduras na Dieta/farmacologia , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Estradiol/sangue , Feminino , Glutationa Peroxidase/metabolismo , Peróxido de Hidrogênio/metabolismo , Masculino , Estresse Oxidativo , Ratos , Ratos Wistar , Fatores Sexuais , Superóxido Dismutase/metabolismo , Fatores de Tempo
11.
Free Radic Biol Med ; 150: 148-160, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32105829

RESUMO

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.


Assuntos
Resistência à Insulina , Hepatopatia Gordurosa não Alcoólica , Animais , Dieta Hiperlipídica/efeitos adversos , Estradiol/metabolismo , Estradiol/farmacologia , Feminino , Fígado/metabolismo , Masculino , Hepatopatia Gordurosa não Alcoólica/tratamento farmacológico , Hepatopatia Gordurosa não Alcoólica/etiologia , Hepatopatia Gordurosa não Alcoólica/metabolismo , Estresse Oxidativo , Ratos , Ratos Wistar
12.
J Steroid Biochem Mol Biol ; 185: 256-267, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30253224

RESUMO

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.


Assuntos
Adipócitos/patologia , Estradiol/metabolismo , Receptor alfa de Estrogênio/metabolismo , Interleucina-6/metabolismo , Mitocôndrias/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Células 3T3 , Animais , Linhagem Celular , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Receptor alfa de Estrogênio/agonistas , Receptor beta de Estrogênio/agonistas , Receptor beta de Estrogênio/metabolismo , Feminino , Inflamação/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/patologia , Ratos , Ratos Wistar , Receptores Acoplados a Proteínas G/agonistas
13.
Cell Physiol Biochem ; 22(1-4): 327-36, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18769060

RESUMO

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.


Assuntos
Gorduras na Dieta/farmacologia , Comportamento Alimentar/efeitos dos fármacos , Fígado/metabolismo , Músculo Esquelético/metabolismo , Caracteres Sexuais , Tecido Adiposo Marrom/metabolismo , Animais , Antioxidantes/metabolismo , Metabolismo Energético/efeitos dos fármacos , Feminino , Peróxido de Hidrogênio/metabolismo , Canais Iônicos/metabolismo , Fígado/anatomia & histologia , Fígado/enzimologia , Masculino , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Proteínas Mitocondriais/metabolismo , Músculo Esquelético/anatomia & histologia , Músculo Esquelético/enzimologia , Tamanho do Órgão , Oxirredução/efeitos dos fármacos , Consumo de Oxigênio/efeitos dos fármacos , Ratos , Ratos Wistar , Substâncias Reativas com Ácido Tiobarbitúrico/metabolismo , Proteína Desacopladora 1
14.
Cell Physiol Biochem ; 22(5-6): 539-48, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19088436

RESUMO

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.


Assuntos
Envelhecimento/efeitos dos fármacos , Envelhecimento/fisiologia , Gorduras na Dieta/farmacologia , Resistência à Insulina/fisiologia , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/fisiologia , Caracteres Sexuais , Adipocinas/sangue , Animais , Antioxidantes/metabolismo , Glicemia/metabolismo , Peso Corporal/efeitos dos fármacos , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Metabolismo Energético/efeitos dos fármacos , Feminino , Teste de Tolerância a Glucose , Transportador de Glucose Tipo 4/metabolismo , Peróxido de Hidrogênio/metabolismo , Insulina/sangue , Canais Iônicos/metabolismo , Peróxidos Lipídicos/metabolismo , Masculino , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/enzimologia , Proteínas Mitocondriais/metabolismo , Consumo de Oxigênio/efeitos dos fármacos , Carbonilação Proteica/efeitos dos fármacos , Ratos , Ratos Wistar , Proteína Desacopladora 3 , Aumento de Peso/efeitos dos fármacos
15.
J Steroid Biochem Mol Biol ; 172: 9-19, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28529127

RESUMO

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.


Assuntos
Adipócitos Marrons/metabolismo , Adipócitos Brancos/metabolismo , Estradiol/farmacologia , Proteínas de Homeodomínio/genética , Caracteres Sexuais , Tiorredoxinas/genética , Células 3T3-L1 , Adipócitos Marrons/efeitos dos fármacos , Adipócitos Brancos/efeitos dos fármacos , Tecido Adiposo Branco/efeitos dos fármacos , Tecido Adiposo Branco/metabolismo , Animais , Antioxidantes/metabolismo , Dieta Hiperlipídica , Gorduras na Dieta/efeitos adversos , Feminino , Regulação da Expressão Gênica , Proteínas de Homeodomínio/metabolismo , Hipoglicemiantes/farmacologia , Masculino , Camundongos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Ovariectomia , Cultura Primária de Células , Ratos , Ratos Wistar , Rosiglitazona , Transdução de Sinais , Testosterona/farmacologia , Tiazolidinedionas/farmacologia , Tiorredoxinas/metabolismo
16.
J Endocrinol ; 232(2): 297-308, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27885055

RESUMO

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.


Assuntos
Estradiol/farmacologia , Metabolismo dos Lipídeos/efeitos dos fármacos , Fígado/efeitos dos fármacos , Mitocôndrias/metabolismo , Coativadores de Receptor Nuclear/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Animais , Feminino , Metabolismo dos Lipídeos/fisiologia , Fígado/metabolismo , Camundongos Transgênicos , Coativadores de Receptor Nuclear/genética , Biogênese de Organelas , Ovariectomia , Oxirredução , Estresse Oxidativo/fisiologia , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/genética , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , RNA Interferente Pequeno , Ratos
17.
J Nutr Biochem ; 17(3): 197-203, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16214326

RESUMO

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.


Assuntos
Arildialquilfosfatase/sangue , Restrição Calórica , Caracteres Sexuais , Animais , Apolipoproteína A-I/sangue , Arildialquilfosfatase/genética , Arildialquilfosfatase/metabolismo , Clusterina/sangue , Feminino , Privação de Alimentos , Lipídeos/sangue , Fígado/anatomia & histologia , Fígado/enzimologia , Masculino , Microssomos Hepáticos/química , Tamanho do Órgão , Proteínas/análise , RNA Mensageiro/análise , Ratos
18.
Mol Cell Endocrinol ; 420: 116-24, 2016 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-26628039

RESUMO

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.


Assuntos
Estradiol/farmacologia , Mitocôndrias Cardíacas/metabolismo , Biogênese de Organelas , Receptores Acoplados a Proteínas G/metabolismo , Animais , Biomarcadores/metabolismo , Peso Corporal/efeitos dos fármacos , Linhagem Celular , Estradiol/sangue , Feminino , Mitocôndrias Cardíacas/efeitos dos fármacos , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Tamanho do Órgão/efeitos dos fármacos , Ovariectomia , Estresse Oxidativo/efeitos dos fármacos , Progesterona/sangue , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos Wistar , Receptores de Estrogênio/metabolismo
19.
Mitochondrion ; 5(1): 45-53, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16075478

RESUMO

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.


Assuntos
Tecido Adiposo Marrom/metabolismo , Tecido Adiposo Marrom/ultraestrutura , Mitocôndrias/metabolismo , Mitocôndrias/ultraestrutura , Termogênese , Animais , Antioxidantes/metabolismo , Sequência de Bases , Fracionamento Celular , DNA Mitocondrial/genética , DNA Mitocondrial/isolamento & purificação , Feminino , Microscopia Eletrônica , Mitocôndrias/classificação , Proteínas Mitocondriais/isolamento & purificação , Ratos , Ratos Wistar
20.
Life Sci ; 76(10): 1147-58, 2005 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-15620578

RESUMO

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.


Assuntos
Tecido Adiposo Marrom/metabolismo , Tecido Adiposo Marrom/ultraestrutura , Mitocôndrias/ultraestrutura , Termogênese , Animais , Proteínas de Transporte/análise , DNA Mitocondrial/análise , Feminino , Canais Iônicos , Masculino , Proteínas de Membrana/análise , Mitocôndrias/metabolismo , Proteínas Mitocondriais , ATPases Mitocondriais Próton-Translocadoras/análise , Prostaglandina-Endoperóxido Sintases/análise , Ratos , Ratos Wistar , Caracteres Sexuais , Proteína Desacopladora 1
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA