Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Heliyon ; 10(7): e27709, 2024 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-38590904

RESUMO

Diets high in fat and sugar lead to metabolic syndrome (MetS) and related chronic diseases. We investigated the effects of commercially available, cold-pressed polyphenol-rich black currant (BC) and cornelian cherry (CC) juices on the prevention of MetS in Wistar rats induced by a 10-weeks high-fat high-fructose (HFF) diet. Juice consumption, either BC or CC, with a HFF diet resulted in lower serum triglycerides compared to only the HFF consumption. Both juices also mitigated the effects of HFF on the liver, pancreas, and adipose tissue, by preserving liver and pancreas histomorphology and reducing visceral fat and adipocyte size. Furthermore, supplementation with both juices reduced glucagon and up-regulated insulin expression in the pancreas of the rats on the HFF diet, whereas the BC also showed improved glucose regulation. BC juice also reduced the expression of IL-6 and hepatic inflammation compared to the group only on HFF diet. Both juices, especially BC, could be a convenient solution for the prevention of MetS in humans.

2.
Biomedicines ; 11(6)2023 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-37371678

RESUMO

Polycystic ovary syndrome (PCOS) is a common endocrinopathy in women of reproductive age, often associated with obesity and insulin resistance. Childhood obesity is an important predisposing factor for the development of PCOS later in life. Being particularly interested in the interplay between prepubertal obesity and hyperandrogenemia, we investigated the effects of early postnatal overfeeding, accomplished by reducing litter size during the period of suckling, on energy sensing and insulin signaling pathways in the gastrocnemius muscle of a rat model of PCOS-induced by 5α-dihydrotestosterone (DHT). The combination of overfeeding and DHT treatment caused hyperinsulinemia and decreased systemic insulin sensitivity. Early postnatal overfeeding induced defects at critical nodes of the insulin signaling pathway in skeletal muscle, which was associated with reduced glucose uptake in the presence of hyperandrogenemia. In this setting, under a combination of overfeeding and DHT treatment, skeletal muscle switched to mitochondrial ß-oxidation of fatty acids, resulting in oxidative stress and inflammation that stimulated AMP-activated protein kinase (AMPK) activity and its downstream targets involved in mitochondrial biogenesis and antioxidant protection. Overall, a combination of overfeeding and hyperandrogenemia resulted in a prooxidative and insulin-resistant state in skeletal muscle. This was accompanied by the activation of AMPK, which could represent a potential therapeutic target in insulin-resistant PCOS patients.

3.
Biofactors ; 49(1): 90-107, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34767656

RESUMO

Macrophage migration inhibitory factor (MIF) is a pro-inflammatory cytokine that represents a link between diet-induced inflammation and insulin resistance. Our aim was to examine whether fructose diet affects inflammation and insulin signaling in the prefrontal cortex (PFC) of Mif knockout mice (MIF-KO), and their possible link to neural plasticity and behavior. We analyzed nuclear factor κB (NF-κB) and glucocorticoid signaling, expression of F4/80, IL-1ß, TNF-α, TLR-4, MyD88, arginase 1 (Arg-1), mannose receptor (Mrc-1), and leukemia inhibitory factor (Lif) to assess inflammation in the PFC of C57/BL6J and MIF-KO mice consuming 20% fructose solution for 9 weeks. Insulin receptor (IR), IRS-1 serine phosphorylations (307 and 1101) and activity of PKCα, Akt, GSK-3ß and AMPKα were used to analyze insulin signaling. Brain-derived neurotrophic factor (BDNF) and insulin-like growth factor 1 (IGF-1) mRNA levels, together with synapthophysin and PSD-95 protein level and calcium calmodulin-dependent kinase 2 (CaMKII) activity, were used as plasticity markers. Behavior was examined in elevated plus maze, light dark box and novel object recognition test. The results showed concomitant increase of Tnf-α, Tlr-4, MyD88 and M2 microglia markers (Arg-1, Mrc-1, Lif) in the PFC of MIF-KO, paralleled with unchanged glucocorticoid and insulin signaling. Increase of BDNF and IGF-1 was paralleled with increased CaMKII activity, decreased PSD-95 protein level, anxiogenic behavior, and impaired memory in MIF-KO mice. Fructose feeding restored these parameters in the PFC to the control level and mitigated behavioral changes, suggesting that ameliorating effects of fructose on neuroinflammation and behavior depend on the presence of MIF.


Assuntos
Fatores Inibidores da Migração de Macrófagos , Camundongos , Masculino , Animais , Fator de Crescimento Insulin-Like I/metabolismo , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Glucocorticoides , Fator de Necrose Tumoral alfa/metabolismo , Frutose , Proteína 4 Homóloga a Disks-Large/metabolismo , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Glicogênio Sintase Quinase 3 beta/metabolismo , Fator 88 de Diferenciação Mieloide/metabolismo , Receptor 4 Toll-Like/metabolismo , Inflamação/metabolismo , Dieta , Insulina/metabolismo , Córtex Pré-Frontal/metabolismo , Plasticidade Neuronal , Camundongos Endogâmicos C57BL , Camundongos Knockout
4.
Int J Mol Sci ; 23(16)2022 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-36012206

RESUMO

Polycystic ovary syndrome (PCOS) is a well-known reproductive syndrome usually associated with obesity, insulin resistance, and hyperinsulinemia. Although the first signs of PCOS begin early in adolescence, it is underexplored whether peripubertal obesity predisposes women to PCOS metabolic disturbances. To highlight that, we examined the impact of postnatal overfeeding-induced obesity, achieved by litter size reduction during the suckling period, on metabolic disturbances associated with visceral and subcutaneous adipose tissue (VAT and SAT) function in the 5α-dihydrotestosterone (5α-DHT)-induced animal model of PCOS. We analyzed markers of insulin signaling, lipid metabolism, and energy sensing in the VAT and SAT. Our results showed that postnatally overfed DHT-treated Wistar rats had increased VAT mass with hypertrophic adipocytes, together with hyperinsulinemia and increased HOMA index. In the VAT of these animals, insulin signaling remained unchanged while lipogenic markers decreased, which was accompanied by increased AMPK activation. In the SAT of the same animals, markers of lipogenesis and lipolysis increased, while the activity of AMPK decreased. Taken together, obtained results showed that postnatal overfeeding predisposes development of PCOS systemic insulin resistance, most likely as a result of worsened metabolic function of SAT, while VAT preserved its tissue insulin sensitivity through increased activity of AMPK.


Assuntos
Resistência à Insulina , Síndrome do Ovário Policístico , Proteínas Quinases Ativadas por AMP/metabolismo , Tecido Adiposo/metabolismo , Animais , Feminino , Humanos , Insulina/metabolismo , Resistência à Insulina/fisiologia , Obesidade/metabolismo , Síndrome do Ovário Policístico/metabolismo , Ratos , Ratos Wistar , Gordura Subcutânea/metabolismo
5.
Acta Biochim Pol ; 69(3): 647-655, 2022 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-35877942

RESUMO

Appetite regulation in the hypothalamus is dependent on hormonal signals from the periphery, such as insulin and leptin, and can be modulated by both sugar-rich diet and stress. Our aim was to explore the effects of 9-week feeding with 20% fructose solution combined with 4-week chronic unpredictable stress, on appetite-regulating neuropeptides and modulatory role of leptin and insulin signalling in the hypothalamus of male Wistar rats. Energy intake, body mass and adiposity, as well as circulatory leptin and insulin concentrations were assessed. Hypothalamic insulin signalling was analysed at the level of glucose transporters, as well as at the protein level and phosphorylation of insulin receptor, insulin receptor supstrate-1, Akt and ERK. Phosphorylation of AMP-activated protein kinase (AMPK), level of protein tyrosine phosphatase 1B (PTP1B) and expression of leptin receptor (ObRb) and suppressor of cytokine signalling 3 (SOCS3) were also analysed, together with the expression of orexigenic agouti-related protein (AgRP) and anorexigenic proopiomelanocortin (POMC) neuropeptides. The results revealed that stress decreased body mass and adiposity, blood leptin level and expression of ObRb, SOCS3 and POMC, while combination with fructose diet led to marked increase of AgRP, associated with AMPK phosphorylation despite increased plasma insulin. Reduced Akt, enhanced ERK activity and elevated PTP1B were also observed in the hypothalamus of these animals. In conclusion, our results showed that joint effects of fructose diet and stress are more deleterious than the separate ones, since inappropriate appetite control in the hypothalamus may provide a setting for the disturbed energy homeostasis in the long run.


Assuntos
Neuropeptídeos , Pró-Opiomelanocortina , Proteínas Quinases Ativadas por AMP/metabolismo , Proteína Relacionada com Agouti/metabolismo , Proteína Relacionada com Agouti/farmacologia , Animais , Citocinas/metabolismo , Dieta , Frutose/efeitos adversos , Frutose/metabolismo , Glucose/metabolismo , Hipotálamo/metabolismo , Insulina , Leptina , Masculino , Neuropeptídeos/metabolismo , Neuropeptídeos/farmacologia , Fosforilação , Pró-Opiomelanocortina/metabolismo , Pró-Opiomelanocortina/farmacologia , Proteína Tirosina Fosfatase não Receptora Tipo 1/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 1/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Wistar , Receptor de Insulina/metabolismo , Receptores para Leptina/metabolismo
6.
Front Nutr ; 9: 899255, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35677539

RESUMO

Aging is a progressive process that could disturb metabolic homeostasis in the liver via ectopic lipid accumulation, oxidative stress, and deterioration of inflammatory response. Although calorie restriction (CR) is recognized as beneficial for life span and health span prolongation, it is still unclear how late-onset CR, characterized by late beginning and short duration, affects age-related processes. The aim of this study was to examine how late-onset CR-induced metabolic adjustments impact lipid status and inflammation in the liver of old rats. The experiments were conducted on aging male Wistar rats fed ad libitum (AL) or exposed to late-onset CR (60% of AL daily intake) from 21st to 24th month. The results showed that late-onset CR reduces body weight, visceral adipose tissue and liver mass, and triglyceride levels when compared to old animals on AL diet. The ameliorating effects of CR on lipid metabolism include increased activity of AMP-activated protein kinase, suppressed de novo fatty acid synthesis, stimulated ß-oxidation, decreased lipotoxicity, and limited triglyceride synthesis and packaging in the liver. Restricted diet regime, however, does not improve expression of antioxidant enzymes, although it leads to progression of age-related inflammation in the liver, partially through lower corticosterone concentration and decreased activation of glucocorticoid receptor. In conclusion, late-onset CR is able to restore age-related imbalance of lipid metabolism in the liver, but has a negative impact on hepatic inflammatory status, implying that the type of diet for older individuals must be balanced and chosen carefully with appropriate duration and start point.

7.
Biofactors ; 47(3): 363-375, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33522030

RESUMO

Dietary fructose can disturb hepatic lipid metabolism in a way that leads to lipid accumulation and steatosis, which is often accompanied with low-grade inflammation. The macrophage migration inhibitory factor (MIF) is a proinflammatory cytokine with important role not only in the regulation of inflammation, but also in the modulation of energy metabolism in the liver. Thus, the aim of this study was to investigate the role of Mif deficiency in fructose-induced disturbances of hepatic lipid metabolism and ectopic lipid accumulation. Wild type (WT) and Mif deficient (MIF-/- ) C57Bl/6J mice were used to analyze the effects of 9-week 20% fructose-enriched diet on hepatic lipid metabolism (both lipogenesis and ß-oxidation) and histology, inflammatory status and glucocorticoid receptor (GR) signaling. The results showed fructose-induced elevation of lipogenic genes (fatty acid synthase (Fas) and stearoyl-CoA desaturase-1 (Scd1) and transcriptional lipogenic regulators (liver X receptor (LXR), sterol regulatory element binding protein 1c (SREBP1c), and carbohydrate response element-binding protein (ChREBP)). However, microvesicular fatty changes, accompanied with enhanced inflammation, were observable only in fructose-fed Mif deficient animals, and were most likely result of GR activation and facilitated uptake and decreased ß-oxidation of FFA, as evidenced by elevated protein level of fatty acid translocase (FAT/CD36) and decreased carnitine palmitoyl transferase 1 (CPT1) level. In conclusion, the results show that Mif deficiency aggravates the effects of energy-rich fructose diet on hepatic lipid accumulation, most likely through enhanced inflammation and activation of GR signaling pathway.


Assuntos
Dieta/métodos , Frutose/metabolismo , Frutose/farmacologia , Metabolismo dos Lipídeos/efeitos dos fármacos , Fígado/efeitos dos fármacos , Fatores Inibidores da Migração de Macrófagos/deficiência , Animais , Lipogênese , Fígado/metabolismo , Fatores Inibidores da Migração de Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais
8.
Endocrine ; 72(2): 562-572, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33449293

RESUMO

PURPOSE: Polycystic ovary syndrome (PCOS) is a complex reproductive disorder often associated with obesity, insulin resistance, and dyslipidemia. Hormonal changes in PCOS may also include altered glucocorticoid signaling. Our aim was to examine whether alterations in hepatic glucocorticoid signaling are associated with disturbances of glucose and lipid metabolism in animal model of PCOS. METHODS: Female rats, 3 weeks old, were subcutaneously implanted with 5α-dihydrotestosterone (DHT) or placebo pellets for 90 days to induce PCOS. Expression of 11ß-hydroxysteroid dehydrogenase 1 (11ßHSD1) and A-ring reductases (5α and 5ß), as well as intracellular distribution of glucocorticoid receptor (GR) and expression of its regulated genes were examined in the liver. Proteins of hepatic lipid and carbohydrate metabolism and markers of inflammation were also assessed. RESULTS: DHT treatment induced increase in body and liver mass, as well as in triglycerides and free fatty acids levels in plasma. Elevation of 11ßHSD1 and reduction of 5α-reductase expression was observed together with increased hepatic corticosterone concentration and nuclear GR activation. Induced expression of Krüppel-like factor 15 and decreased expression of genes for proinflammatory cytokines and de novo lipogenesis (DNL) were detected in the liver of DHT-treated rats, while DNL regulators and proinflammatory markers were not changed. However, increased mRNA levels of stearoyl-CoA desaturase and apolipoprotein B were observed in DHT animals. CONCLUSIONS: DHT treatment stimulated hepatic glucocorticoid prereceptor metabolism through increased corticosterone availability which is associated with enhanced GR activation. This does not affect gluconeogenesis and DNL, but could be linked to stimulated triglyceride synthesis and hypertriglyceridemia.


Assuntos
Síndrome do Ovário Policístico , Animais , Di-Hidrotestosterona , Modelos Animais de Doenças , Feminino , Glucocorticoides/metabolismo , Humanos , Metabolismo dos Lipídeos , Fígado/metabolismo , Síndrome do Ovário Policístico/induzido quimicamente , Síndrome do Ovário Policístico/metabolismo , Ratos , Ratos Wistar
9.
Mol Nutr Food Res ; 64(13): e1901141, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32379936

RESUMO

SCOPE: Intake of fructose-sweetened beverages and chronic stress (CS) both increase risk of cardiometabolic diseases. The aim is to investigate whether these factors synergistically perturb lipid metabolism in rat liver and kidney. METHODS AND RESULTS: Fractional de novo lipogenesis (fDNL), intrahepatic- and intrarenal-triglycerides (IHTG and IRTG), de novo palmitate (DNPalm) content, FA composition, VLDL-TGs kinetics, and key metabolic gene expression at the end of the feeding and non-feeding phases in rats exposed to standard chow diet, chow diet + CS, 20% liquid high-fructose supplementation (HFr), or HFr+CS are measured. HFr induces hypertriglyceridemia, up-regulates fructose-metabolism and gluconeogenic enzymes, increases IHTG and DNPalm content in IHTG and IRTG, and augments fDNL at the end of the feeding phase. These changes are diminished after the non-feeding phase. CS does not exert such effects, but when combined with HFr, it reduces IHTG and visceral adiposity, enhances lipogenic gene expression and fDNL, and increases VLDL-DNPalm secretion. CONCLUSION: Liquid high-fructose supplementation increases IHTG and VLDL-TG secretion after the feeding phase, the latter being the result of stimulated hepatic and renal DNL. Chronic stress potentiates the effects of high fructose on fDNL and export of newly synthesized VLDL-TGs, and decreases fructose-induced intrahepatic TG accumulation after the feeding phase.


Assuntos
Frutose/efeitos adversos , Rim/efeitos dos fármacos , Lipogênese , Fígado/metabolismo , Estresse Psicológico/fisiopatologia , Animais , Composição Corporal , Ingestão de Alimentos , Ingestão de Energia , Enzimas/genética , Enzimas/metabolismo , Regulação da Expressão Gênica , Gluconeogênese/efeitos dos fármacos , Gluconeogênese/fisiologia , Rim/metabolismo , Metabolismo dos Lipídeos/efeitos dos fármacos , Metabolismo dos Lipídeos/fisiologia , Lipogênese/efeitos dos fármacos , Lipoproteínas VLDL/metabolismo , Fígado/efeitos dos fármacos , Masculino , Palmitatos/metabolismo , Ratos Wistar , Triglicerídeos/metabolismo
10.
Int J Food Sci Nutr ; 71(7): 815-825, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32070154

RESUMO

Overconsumption of fructose-enriched beverages and everyday stress are involved in the pathogenesis of metabolic disorders through modulation of hepatic glucose metabolism. The aim of the study was to investigate whether interaction of high-fructose diet and chronic stress alter insulin and glucocorticoid signalling thus affecting hepatic glucose homeostasis. High-fructose diet led to hyperinsulinemia, increased glucose transporter 2 level, elevated protein kinase B (Akt) phosphorylation, increased glucokinase mRNA and phospho-to-total glycogen synthase kinase 3 ratio and decreased expression of gluconeogenic genes. Fructose diet also led to stimulated glucocorticoid prereceptor metabolism, but downstream signalling remained unchanged due to increased glucocorticoid clearance. Stress did not affect hepatic insulin and glucocorticoid signalling nor glucose metabolism, while the interaction of the factors was observed only for glucokinase expression. The results suggest that, under conditions of fructose-induced hyperinsulinemia, suppression of gluconeogenesis and glycogen synthase activation contribute to the maintenance of glucose homeostasis. The increased glucocorticoid inactivation may represent an adaptive mechanism to prevent hyperglycaemia.


Assuntos
Açúcares da Dieta/administração & dosagem , Frutose/administração & dosagem , Glucose/metabolismo , Homeostase/efeitos dos fármacos , Fígado/efeitos dos fármacos , Animais , Relação Dose-Resposta a Droga , Regulação da Expressão Gênica/efeitos dos fármacos , Glucocorticoides/metabolismo , Insulina/genética , Insulina/metabolismo , Fígado/metabolismo , Masculino , Ratos , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos , Estresse Fisiológico
11.
Eur J Nutr ; 58(5): 1829-1845, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29845385

RESUMO

PURPOSE: High-fructose consumption and chronic stress are both associated with metabolic inflammation and insulin resistance. Recently, disturbed activity of energy sensor AMP-activated protein kinase (AMPK) was recognized as mediator between nutrient-induced stress and inflammation. Thus, we analyzed the effects of high-fructose diet, alone or in combination with chronic stress, on glucose homeostasis, inflammation and expression of energy sensing proteins in the rat liver. METHODS: In male Wistar rats exposed to 9-week 20% fructose diet and/or 4-week chronic unpredictable stress we measured plasma and hepatic corticosterone level, indicators of glucose homeostasis and lipid metabolism, hepatic inflammation (pro- and anti-inflammatory cytokine levels, Toll-like receptor 4, NLRP3, activation of NFκB, JNK and ERK pathways) and levels of energy-sensing proteins AMPK, SIRT1 and peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC-1α). RESULTS: High-fructose diet led to glucose intolerance, activation of NFκB and JNK pathways and increased intrahepatic IL-1ß, TNFα and inhibitory phosphorylation of insulin receptor substrate 1 on Ser307. It also decreased phospho-AMPK/AMPK ratio and increased SIRT1 expression. Stress alone increased plasma and hepatic corticosterone but did not influence glucose tolerance, nor hepatic inflammatory or energy-sensing proteins. After the combined treatment, hepatic corticosterone was increased, glucose tolerance remained preserved, while hepatic inflammation was partially prevented despite decreased AMPK activity. CONCLUSION: High-fructose diet resulted in glucose intolerance, hepatic inflammation, decreased AMPK activity and reduced insulin sensitivity. Chronic stress alone did not exert such effects, but when applied together with high-fructose diet it could partially prevent fructose-induced inflammation, presumably due to increased hepatic glucocorticoids.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Dieta/métodos , Frutose/administração & dosagem , Inflamação/fisiopatologia , Estresse Fisiológico/fisiologia , Animais , Doença Crônica , Modelos Animais de Doenças , Fígado , Masculino , Ratos , Ratos Wistar
12.
J Endocrinol ; 240(2): 133-145, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30400058

RESUMO

The macrophage migration inhibitory factor (MIF) is a proinflammatory cytokine involved in inflammation, regulation of energy metabolism and glucocorticoid action. Chronic low-grade inflammation may be caused by fructose intake, contributing to visceral adipose tissue (VAT) dysfunction. Since MIF is a known antagonist of glucocorticoid signaling, and deregulated glucocorticoid signaling can contribute to lipid metabolism disturbances, we hypothesized that altered MIF signaling might underlie fructose-induced adiposity through glucocorticoid action. We analyzed physiological and biochemical parameters, adipose tissue histology, insulin sensitivity and lipid metabolism in WT and MIF-/- C57Bl/6J mice consuming 20% fructose solution for 9 weeks. Glucocorticoid prereceptor metabolism and glucocorticoid receptor (GR) protein level were examined in VAT, together with the expression of glucocorticoid-target genes involved in lipid metabolism. The expression of adipogenic and lipogenic transcriptional regulators peroxisome proliferator-activated receptor gamma (PPARG) and sterol regulatory element-binding protein 1c (SREBP1c) was also assessed. Results showed disturbed insulin sensitivity in all MIF-/- mice, regardless of the diet. Mice on fructose diet had increased energy intake, but increased visceral adiposity and enlarged adipocytes were observed only in fructose-fed MIF-/- mice. Increased VAT corticosterone level and 11 beta-hydroxysteroid dehydrogenase type 1, hexose-6-phosphate dehydrogenase and GR protein levels were observed in the same animals, together with induced expression of examined lipogenic genes and accumulation of PPARG and SREBP1c. In conclusion, the results showed that dietary fructose was associated with increased visceral adiposity through activation of GR-regulated lipogenic genes, but only in the absence of MIF, which set the state of hyperinsulinemia and insulin resistance.


Assuntos
Tecido Adiposo/metabolismo , Adiposidade/genética , Corticosterona/metabolismo , Fatores Inibidores da Migração de Macrófagos/genética , Adipócitos/metabolismo , Tecido Adiposo/efeitos dos fármacos , Adiposidade/efeitos dos fármacos , Animais , Frutose/administração & dosagem , Frutose/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Glucocorticoides/metabolismo , Resistência à Insulina/genética , Metabolismo dos Lipídeos/efeitos dos fármacos , Metabolismo dos Lipídeos/genética , Lipogênese/efeitos dos fármacos , Lipogênese/genética , Fatores Inibidores da Migração de Macrófagos/deficiência , Camundongos Endogâmicos C57BL , Camundongos Knockout , Obesidade/etiologia , Obesidade/genética , Obesidade/metabolismo , Receptores de Glucocorticoides/metabolismo
13.
Mol Cell Endocrinol ; 476: 110-118, 2018 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-29729371

RESUMO

Both fructose overconsumption and increased glucocorticoids secondary to chronic stress may contribute to overall dyslipidemia. In this study we specifically assessed the effects and interactions of dietary fructose and chronic stress on lipid metabolism in the visceral adipose tissue (VAT) of male Wistar rats. We analyzed the effects of 9-week 20% high fructose diet and 4-week chronic unpredictable stress, separately and in combination, on VAT histology, glucocorticoid prereceptor metabolism, glucocorticoid receptor subcellular redistribution and expression of major metabolic genes. Blood triglycerides and fatty acid composition were also measured to assess hepatic Δ9 desaturase activity. The results showed that fructose diet increased blood triglycerides and Δ9 desaturase activity. On the other hand, stress led to corticosterone elevation, glucocorticoid receptor activation and decrease in adipocyte size, while phosphoenolpyruvate carboxykinase, adipose tissue triglyceride lipase, FAT/CD36 and sterol regulatory element binding protein-1c (SREBP-1c) were increased, pointing to VAT lipolysis and glyceroneogenesis. The combination of stress and fructose diet was associated with marked stimulation of fatty acid synthase and acetyl-CoA carboxylase mRNA level and with increased 11ß-hydroxysteroid dehydrogenase type 1 and hexose-6-phosphate dehydrogenase protein levels, suggesting a coordinated increase in hexose monophosphate shunt and de novo lipogenesis. It however did not influence the level of peroxisome proliferator-activated receptor-gamma, SREBP-1c and carbohydrate responsive element-binding protein. In conclusion, our results showed that only combination of dietary fructose and stress increase glucocorticoid prereceptor metabolism and stimulates lipogenic enzyme expression suggesting that interaction between stress and fructose may be instrumental in promoting VAT expansion and dysfunction.


Assuntos
Dieta , Gordura Intra-Abdominal/metabolismo , Metabolismo dos Lipídeos , Receptores de Glucocorticoides/metabolismo , Transdução de Sinais , Estresse Psicológico/metabolismo , Animais , Corticosterona/sangue , Ácidos Graxos/sangue , Frutose , Regulação da Expressão Gênica , Insulina/sangue , Metabolismo dos Lipídeos/genética , Masculino , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos Wistar , Estearoil-CoA Dessaturase/metabolismo , Estresse Psicológico/sangue , Transcrição Gênica , Triglicerídeos/sangue
14.
Horm Behav ; 96: 95-103, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28919555

RESUMO

Macrophage migration inhibitory factor (MIF) is a multifunctional cytokine well known for its role in inflammation enhancement. However, a growing body of evidence is emerging on its role in energy metabolism in insulin sensitive tissues such as hippocampus, a brain region implicated in cognition, learning and memory. We hypothesized that genetic deletion of MIF may result in the specific behavioral changes, which may be linked tо impairments in brain or systemic insulin sensitivity by possible changes of the hippocampal synaptic plasticity. To assess memory, exploratory behavior and anxiety, three behavioral tests were applied on Mif gene-deficient (MIF-/-) and "wild type" C57BL/6J mice (WT). The parameters of systemic and hippocampal insulin sensitivity were also determined. The impact of MIF deficiency on hippocampal plasticity was evaluated by analyzing the level of synaptosomal polysialylated-neural cell adhesion molecule (PSA-NCAM) plasticity marker and mRNA levels of different neurotrophic factors. The results showed that MIF-/- mice exhibit emphasized anxiety-like behaviors, as well as impaired recognition memory, which may be hippocampus-dependent. This behavioral phenotype was associated with impaired systemic insulin sensitivity and attenuated hippocampal insulin sensitivity, characterized by increased inhibitory Ser307 phosphorylation of insulin receptor substrate 1 (IRS1). Finally, MIF-/- mice displayed a decreased hippocampal PSA-NCAM level and unchanged Bdnf, NT-3, NT-4 and Igf-1 mRNA levels. The results suggest that the lack of MIF leads to disturbances of systemic and hippocampal insulin sensitivity, which are possibly responsible for memory deficits and anxiety, most likely through decreased PSA-NCAM-mediated neuroplasticity rather than through neurotrophic factors.


Assuntos
Comportamento Animal/fisiologia , Hipocampo/metabolismo , Resistência à Insulina/genética , Oxirredutases Intramoleculares/genética , Fatores Inibidores da Migração de Macrófagos/genética , Molécula L1 de Adesão de Célula Nervosa/metabolismo , Ácidos Siálicos/metabolismo , Animais , Ansiedade/genética , Ansiedade/metabolismo , Comportamento Exploratório/fisiologia , Masculino , Aprendizagem em Labirinto/fisiologia , Transtornos da Memória/genética , Transtornos da Memória/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Plasticidade Neuronal/genética
15.
Exp Clin Endocrinol Diabetes ; 125(8): 522-529, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28407665

RESUMO

Polycystic ovary syndrome is a heterogeneous endocrine and metabolic disorder associated with abdominal obesity, dyslipidemia and insulin resistance. Since abdominal obesity is characterized by low-grade inflammation, the aim of the study was to investigate whether visceral adipose tissue inflammation linked to abdominal obesity and dyslipidemia could lead to impaired insulin sensitivity in the animal model of polycystic ovary syndrome.Female Wistar rats were treated with nonaromatizable 5α-dihydrotestosterone pellets in order to induce reproductive and metabolic characteristics of polycystic ovary syndrome. Glucose, triglycerides, non-esterified fatty acids and insulin were determined in blood plasma. Visceral adipose tissue inflammation was evaluated by the nuclear factor kappa B intracellular distribution, macrophage migration inhibitory factor protein level, as well as TNFα, IL6 and IL1ß mRNA levels. Insulin sensitivity was assessed by intraperitoneal glucose tolerance test and homeostasis model assessment index, and through analysis of insulin signaling pathway in the visceral adipose tissue.Dihydrotestosterone treatment led to increased body weight, abdominal obesity and elevated triglycerides and non-esterified fatty acids, which were accompanied by the activation of nuclear factor kappa B and increase in macrophage migration inhibitory factor, IL6 and IL1ß levels in the visceral adipose tissue. In parallel, insulin sensitivity was affected in 5α-dihydrotestosterone-treated animals only at the systemic and not at the level of visceral adipose tissue.The results showed that abdominal obesity and dyslipidemia in the animal model of polycystic ovary syndrome were accompanied with low-grade inflammation in the visceral adipose tissue. However, these metabolic disturbances did not result in decreased tissue insulin sensitivity.


Assuntos
Di-Hidrotestosterona/efeitos adversos , Insulina/metabolismo , Gordura Intra-Abdominal/metabolismo , Obesidade Abdominal/metabolismo , Síndrome do Ovário Policístico/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Di-Hidrotestosterona/farmacologia , Modelos Animais de Doenças , Feminino , Inflamação/induzido quimicamente , Inflamação/metabolismo , Inflamação/patologia , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Gordura Intra-Abdominal/patologia , Obesidade Abdominal/induzido quimicamente , Obesidade Abdominal/patologia , Síndrome do Ovário Policístico/induzido quimicamente , Síndrome do Ovário Policístico/patologia , Ratos , Ratos Wistar
16.
J Sci Food Agric ; 95(11): 2319-24, 2015 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-25307280

RESUMO

BACKGROUND: Increased fructose consumption correlates with rising prevalence of various metabolic disorders, some of which were linked to oxidative stress. The relationship between fructose consumption and oxidative stress is complex and effects of a fructose-rich diet on the young population have not been fully elucidated. The aim of this study was to investigate whether high-fructose diet applied in the period from weaning to adulthood induces oxidative stress in the liver, thus contributing to induction or aggravation of metabolic disturbances in later adulthood. To that end we examined the effects of high-fructose diet on expression and activity of antioxidant enzymes, markers of lipid peroxidation and protein damage in the liver as the main fructose metabolizing tissue. RESULTS: High-fructose diet increased only SOD2 (mitochondrial manganese superoxide dismutase) activity, with no effect on other antioxidant enzymes, lipid peroxidation or accumulation of damaged proteins in the liver. CONCLUSION: The results show that fructose-induced metabolic disturbances could not be attributed to oxidative stress, at least not at young age. The absence of oxidative stress in the liver observed herein implies that young organisms are capable of maintaining redox homeostasis when challenged by fructose-derived energy overload.


Assuntos
Antioxidantes/metabolismo , Dieta , Frutose/farmacologia , Fígado/efeitos dos fármacos , Doenças Metabólicas , Estresse Oxidativo , Desmame , Animais , Frutose/efeitos adversos , Peroxidação de Lipídeos/efeitos dos fármacos , Fígado/enzimologia , Fígado/metabolismo , Masculino , Doenças Metabólicas/etiologia , Carbonilação Proteica/efeitos dos fármacos , Ratos Wistar , Superóxido Dismutase/metabolismo , Substâncias Reativas com Ácido Tiobarbitúrico
17.
Mol Cell Endocrinol ; 399: 22-31, 2015 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-25179821

RESUMO

Polycystic ovary syndrome (PCOS) is a reproductive and metabolic disorder characterized by hyperandrogenism, ovulatory dysfunction, visceral obesity and insulin resistance. We hypothesized that changes in glucocorticoid metabolism and signaling in the visceral adipose tissue may contribute to disturbances of lipid metabolism in the rat model of PCOS obtained by 5α-dihydrotestosterone (DHT) treatment of prepubertal female Wistar rats. The results confirmed that DHT treatment caused anovulation, obesity and dyslipidemia. Enhanced glucocorticoid prereceptor metabolism, assessed by elevated intracellular corticosterone and increased 11 beta-hydroxysteroid dehydrogenase type 1 mRNA and protein levels, was accompanied by glucocorticoid receptor (GR) nuclear accumulation. In concert with the increased expression of GR-regulated prolipogenic genes (lipin-1, sterol regulatory element binding protein 1, fatty acid synthase, phosphoenolpyruvate carboxykinase), histological analyses revealed hypertrophic adipocytes. The results suggest that glucocorticoids influence lipid metabolism in the visceral adipose tissue in the way that may contribute to pathogenesis of metabolic disturbances associated with PCOS.


Assuntos
Adipócitos/metabolismo , Androgênios/efeitos adversos , Di-Hidrotestosterona/efeitos adversos , Glucocorticoides/metabolismo , Gordura Intra-Abdominal/metabolismo , Síndrome do Ovário Policístico/metabolismo , 11-beta-Hidroxiesteroide Desidrogenase Tipo 1/biossíntese , Adipócitos/patologia , Androgênios/farmacologia , Animais , Di-Hidrotestosterona/farmacologia , Ácido Graxo Sintase Tipo I/biossíntese , Feminino , Gordura Intra-Abdominal/patologia , Proteínas Nucleares/biossíntese , Obesidade/etiologia , Obesidade/metabolismo , Obesidade/patologia , Fosfoenolpiruvato Carboxiquinase (ATP)/biossíntese , Síndrome do Ovário Policístico/induzido quimicamente , Síndrome do Ovário Policístico/complicações , Síndrome do Ovário Policístico/patologia , Ratos , Ratos Wistar , Receptores de Glucocorticoides/biossíntese , Proteína de Ligação a Elemento Regulador de Esterol 1/biossíntese
18.
J Mol Neurosci ; 55(4): 959-67, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25367797

RESUMO

Although dexamethasone (DEX), a synthetic glucocorticoid receptor (GR) analog with profound effects on energy metabolism, immune system, and hypothalamic-pituitary-adrenal axis, is widely used therapeutically, its impact on the brain is poorly understood. The aim of the present study was to explore the effect of repeated low-dose DEX administration on the activity and expression of the ectonucleotidase enzymes which hydrolyze and therefore control extracellular ATP and adenosine concentrations in the synaptic cleft. Ectonucleotidases tested were ectonucleoside triphosphate diphosphohydrolase 1-3 (NTPDase1-3) and ecto-5'-nucleotidase (eN), whereas the effects were evaluated in two brain areas that show different sensitivity to glucocorticoid action, hippocampus, and cerebral cortex. In the hippocampus, but not in cerebral cortex, modest level of neurodegenerative changes as well as increase in ATP, ADP, and AMP hydrolysis and upregulation of NTPDase1 and eN mRNA expression ensued under the influence of DEX. The observed pattern of ectonucleotidase activation, which creates tissue volume with enhanced capacity for adenosine formation, is the hallmark of the response after different insults to the brain.


Assuntos
5'-Nucleotidase/metabolismo , Antígenos CD/metabolismo , Apirase/metabolismo , Dexametasona/farmacologia , Glucocorticoides/farmacologia , Hipocampo/enzimologia , Regulação para Cima , 5'-Nucleotidase/genética , Animais , Antígenos CD/genética , Apirase/genética , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/enzimologia , Córtex Cerebral/metabolismo , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Masculino , Especificidade de Órgãos , Ratos , Ratos Wistar
19.
Nutr Neurosci ; 18(2): 66-75, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24257416

RESUMO

OBJECTIVES: High fructose diet has been shown to have damaging effects on the hippocampus, a brain region critical for learning and memory. Fructose-induced hippocampal dysfunction may arise from insulin resistance and inflammation, and from concomitant changes in plasticity-related presynaptic proteins. We hypothesized that long-term access to fructose (10% and 60% solutions over a period of 9 weeks) affects insulin sensitivity, hippocampal inflammation, and synaptic plasticity in male Wistar rats. METHODS: We used the area under curve (AUC) glucose value and inhibitory Ser³°7 phosphorylation of hippocampal insulin receptor substrate 1 (IRS-1) as hallmarks of insulin resistance. To examine inflammatory state, we analysed protein levels and intracellular redistribution of glucocorticoid receptor and nuclear factor-κB (NFκB), as well as mRNA levels of tumour necrosis factor-α (TNF-α), interleukin-6 (IL-6), and interleukin-1ß (IL-1ß). Polysialylated neural cell adhesion molecule (PSA-NCAM) protein was used as a synaptic plasticity marker. RESULTS: The results indicate different impacts of diverse fructose-enriched diets on insulin sensitivity and hippocampal inflammation and plasticity. Long-term ingestion of 10% fructose solution led to increase in AUC glucose value, as well as to elevation in hippocampal IRS-1 Ser³°7 phosphorylation and increase in IL-6 mRNA. In rats consuming 60% fructose, the level of PSA-NCAM was reduced, in parallel with augmented glucocorticoid signalization. DISCUSSION: The results showed that long-term consumption of 10% fructose solution induces hippocampal insulin resistance and inflammation, with no concomitant plasticity changes. Interestingly, rats fed with higher concentrations of fructose displayed impaired plastic response of the hippocampus, coinciding with augmented glucocorticoid signalling, which may provide a basis for cognitive deficits associated with metabolic syndrome.


Assuntos
Carboidratos da Dieta/efeitos adversos , Frutose/efeitos adversos , Hipocampo/metabolismo , Resistência à Insulina , Molécula L1 de Adesão de Célula Nervosa/metabolismo , Plasticidade Neuronal , Neurônios/metabolismo , Ácidos Siálicos/metabolismo , Animais , Biomarcadores/metabolismo , Citocinas/genética , Citocinas/metabolismo , Carboidratos da Dieta/administração & dosagem , Frutose/administração & dosagem , Hipocampo/imunologia , Proteínas Substratos do Receptor de Insulina/metabolismo , Masculino , NF-kappa B/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurônios/imunologia , Fosforilação , Processamento de Proteína Pós-Traducional , Transporte Proteico , Distribuição Aleatória , Ratos Wistar , Receptores de Glucocorticoides/metabolismo
20.
Nutr Res ; 34(7): 646-52, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25150124

RESUMO

Increased fructose consumption is correlated with the rising prevalence of obesity, metabolic syndrome, and type 2 diabetes. It is believed that reactive oxygen species contribute to the development and progression of metabolic disturbances, especially those associated with insulin resistance. Dietary fructose produces both pro-oxidative and antioxidative effects, depending upon the experimental conditions, dosage, duration of treatment, and pathophysiological milieu. The effects of fructose overconsumption on young populations, which have an increased risk of developing metabolic disorders in adulthood, have not been fully elucidated. We have previously shown that rats subjected to a long-term fructose-enriched diet immediately after weaning display impaired hepatic insulin sensitivity. In this study, we tested the hypothesis that long-term fructose consumption induces alterations in the redox setting of the liver. Starting from the 21st day after birth, male Wistar rats were maintained for 9 weeks on a standard diet (control) or a fructose-enriched diet that consisted of standard food and 10% fructose solution instead of drinking water. The expression and activity of antioxidant enzymes as well as lipid peroxidation and protein damage markers were measured. The results showed that a fructose-enriched diet led to an increased expression of mitochondrial manganese superoxide dismutase but did not affect antioxidant enzymes activity, lipid peroxidation, thiol content, and the level of protein oxidation. Therefore, our results suggest that the decrease in hepatic insulin sensitivity that was previously observed in rats that were kept on the same diet regime might be attributed to molecular mechanisms other than redox disbalance. A possible fructose-related micronutrient deficiency should be examined.


Assuntos
Dieta , Carboidratos da Dieta/farmacologia , Frutose/farmacologia , Resistência à Insulina , Fígado/efeitos dos fármacos , Doenças Metabólicas/etiologia , Estresse Oxidativo/efeitos dos fármacos , Animais , Antioxidantes/metabolismo , Carboidratos da Dieta/efeitos adversos , Frutose/efeitos adversos , Peroxidação de Lipídeos/efeitos dos fármacos , Fígado/metabolismo , Masculino , Doenças Metabólicas/metabolismo , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Oxirredução , Carbonilação Proteica/efeitos dos fármacos , Ratos Wistar , Superóxido Dismutase/metabolismo , Substâncias Reativas com Ácido Tiobarbitúrico/metabolismo , Desmame
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...