Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Life Sci ; 304: 120718, 2022 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-35714704

RESUMO

Previously, we have shown that Maternal Separation and Early Weaning (MSEW) exacerbates high fat diet (HF)-induced visceral obesity in female offspring compared to normally reared female mice. Stress hormones such as glucocorticoids and mineralocorticoids are critical mediators in the process of fat expansion, and both can activate the mineralocorticoid receptor (MR) in the adipocyte. Therefore, this study aimed to, comprehend the specific effects of MSEW on adipose tissue basic homeostatic function, and investigate whether female MSEW mice show an exacerbated obesogenic response mediated by MR. Gonadal white adipose tissue (gWAT), a type of visceral fat, was collected to assess lipidomics, transcriptomics, and in vitro lipolysis assay. Obese female MSEW mice showed increased adiposity, elevated 44:2/FA 18:2 + NH4 lipid class and reduced mitochondrial DNA density compared to obese control counterparts. In addition, single-cell RNA sequencing in isolated pre- and mature adipocytes showed a ~9-fold downregulation of aquaglycerolporin 3 (Aqp3), a channel responsible for glycerol efflux in adipocytes. Obese MSEW mice showed high levels of circulating aldosterone and gWAT-derived corticosterone compared to controls. Further, the MR blocker spironolactone (Spiro, 100 mg/kg/day, 2 weeks) normalized the elevated intracellular glycerol levels, the greater in vitro lipolysis response, and the number of large size adipocytes in MSEW mice compared to the controls. Our data suggests that MR plays a role promoting adipocyte hypertrophy in female MSEW mice by preventing lipolysis via glycerol release in favor of triglyceride formation and storage.


Assuntos
Obesidade , Receptores de Mineralocorticoides , Estresse Psicológico , Animais , Feminino , Camundongos , Adipócitos , Glicerol/farmacologia , Lipólise , Privação Materna , Camundongos Endogâmicos C57BL , Camundongos Obesos , Receptores de Mineralocorticoides/genética , Triglicerídeos
2.
Biochem Biophys Res Commun ; 568: 158-166, 2021 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-34217973

RESUMO

The lactate dehydrogenase isoform A (LDHA) is a key metabolic enzyme that preferentially catalyzes the conversion of pyruvate to lactate. Whereas LDHA is highly expressed in many tissues, its expression is turned off in the differentiated adult ß-cell within the pancreatic islets. The repression of LDHA under normal physiological condition and its inappropriate upregulation under a diabetogenic environment is well-documented in rodent islets/ß-cells but little is known about LDHA expression in human islet cells and whether its abundance is altered under diabetic conditions. Analysis of public single-cell RNA-seq (sc-RNA seq) data as well as cell type-specific immunolabeling of human pancreatic islets showed that LDHA was mainly localized in human α-cells while it is expressed at a very low level in ß-cells. Furthermore, LDHA, both at mRNA and protein, as well as lactate production is upregulated in human pancreatic islets exposed to chronic high glucose treatment. Microscopic analysis of stressed human islets and autopsy pancreases from individuals with type 2 diabetes (T2D) showed LDHA upregulation mainly in human α-cells. Pharmacological inhibition of LDHA in isolated human islets enhanced insulin secretion under physiological conditions but did not significantly correct the deregulated secretion of insulin or glucagon under diabetic conditions.


Assuntos
Diabetes Mellitus Tipo 2/genética , Células Secretoras de Glucagon/metabolismo , L-Lactato Desidrogenase/genética , Células Cultivadas , Diabetes Mellitus Tipo 2/metabolismo , Células Secretoras de Glucagon/citologia , Glucose/metabolismo , Humanos , Secreção de Insulina , L-Lactato Desidrogenase/análise , L-Lactato Desidrogenase/metabolismo , RNA Mensageiro/análise , RNA Mensageiro/genética , Regulação para Cima
3.
Am J Physiol Endocrinol Metab ; 320(3): E609-E618, 2021 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-33459178

RESUMO

Obesity is associated with alterations in hepatic lipid metabolism. We previously identified the prorenin receptor (PRR) as a potential contributor to liver steatosis. Therefore, we aimed to determine the relative contribution of PRR and its soluble form, sPRR, to lipid homeostasis. PRR-floxed male mice were treated with an adeno-associated virus with thyroxine-binding globulin promoter-driven Cre to delete PRR in the liver [liver PRR knockout (KO) mice]. Hepatic PRR deletion did not change the body weight but increased liver weights. The deletion of PRR in the liver decreased peroxisome proliferator-activated receptor gamma (PPARγ) and triglyceride levels, but liver PRR KO mice exhibited higher plasma cholesterol levels and lower hepatic low-density lipoprotein receptor (LDLR) and Sortilin 1 (SORT1) proteins than control (CTL) mice. Surprisingly, hepatic PRR deletion elevated hepatic cholesterol, and up-regulated hepatic sterol regulatory element-binding protein 2 (SREBP2) and 3-hydroxy-3-methylglutaryl coenzyme A reductase (HMG CoA-R) genes. In addition, the plasma levels of sPRR were significantly higher in liver PRR KO mice than in controls. In vitro studies in HepG2 cells demonstrated that sPRR treatment upregulated SREBP2, suggesting that sPRR could contribute to hepatic cholesterol biosynthesis. Interestingly, PRR, total cleaved and noncleaved sPRR contents, furin, and Site-1 protease (S1P) were elevated in the adipose tissue of liver PRR KO mice, suggesting that adipose tissue could contribute to the circulating pool of sPRR. Overall, this work supports previous works and opens a new area of investigation concerning the function of sPRR in lipid metabolism and adipose tissue-liver cross talk.NEW & NOTEWORTHY Hepatic PRR and its soluble form, sPRR, contribute to triglyceride and cholesterol homeostasis and hepatic inflammation. Deletion of hepatic PRR decreased triglyceride levels through a PRR-PPARγ-dependent mechanism but increased hepatic cholesterol synthesis through sPRR-medicated upregulation of SREBP-2. Our study highlighted a new paradigm of cross talk between the liver and the adipose tissue involving cholesterol and sPRR.


Assuntos
Homeostase/genética , Metabolismo dos Lipídeos/genética , Receptores de Superfície Celular/fisiologia , Tecido Adiposo/metabolismo , Animais , Fígado Gorduroso/genética , Fígado Gorduroso/metabolismo , Fígado Gorduroso/patologia , Células Hep G2 , Humanos , Fígado/metabolismo , Fígado/patologia , Masculino , Camundongos , Camundongos Knockout , Obesidade/genética , Obesidade/metabolismo , Obesidade/patologia , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiologia , Receptores de Superfície Celular/química , Receptores de Superfície Celular/genética , Solubilidade , Triglicerídeos/metabolismo , Receptor de Pró-Renina
4.
Mol Metab ; 42: 101071, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32896668

RESUMO

OBJECTIVE: Sodium-glucose cotransporter 2 (SGLT2) inhibitors (SGLT2i), or gliflozins, are anti-diabetic drugs that lower glycemia by promoting glucosuria, but they also stimulate endogenous glucose and ketone body production. The likely causes of these metabolic responses are increased blood glucagon levels, and decreased blood insulin levels, but the mechanisms involved are hotly debated. This study verified whether or not SGLT2i affect glucagon and insulin secretion by a direct action on islet cells in three species, using multiple approaches. METHODS: We tested the in vivo effects of two selective SGLT2i (dapagliflozin, empagliflozin) and a SGLT1/2i (sotagliflozin) on various biological parameters (glucosuria, glycemia, glucagonemia, insulinemia) in mice. mRNA expression of SGLT2 and other glucose transporters was assessed in rat, mouse, and human FACS-purified α- and ß-cells, and by analysis of two human islet cell transcriptomic datasets. Immunodetection of SGLT2 in pancreatic tissues was performed with a validated antibody. The effects of dapagliflozin, empagliflozin, and sotagliflozin on glucagon and insulin secretion were assessed using isolated rat, mouse and human islets and the in situ perfused mouse pancreas. Finally, we tested the long-term effect of SGLT2i on glucagon gene expression. RESULTS: SGLT2 inhibition in mice increased the plasma glucagon/insulin ratio in the fasted state, an effect correlated with a decline in glycemia. Gene expression analyses and immunodetections showed no SGLT2 mRNA or protein expression in rodent and human islet cells, but moderate SGLT1 mRNA expression in human α-cells. However, functional experiments on rat, mouse, and human (29 donors) islets and the in situ perfused mouse pancreas did not identify any direct effect of dapagliflozin, empagliflozin or sotagliflozin on glucagon and insulin secretion. SGLT2i did not affect glucagon gene expression in rat and human islets. CONCLUSIONS: The data indicate that the SGLT2i-induced increase of the plasma glucagon/insulin ratio in vivo does not result from a direct action of the gliflozins on islet cells.


Assuntos
Glucagon/metabolismo , Secreção de Insulina/fisiologia , Transportador 2 de Glucose-Sódio/metabolismo , Animais , Compostos Benzidrílicos/farmacologia , Glicemia/metabolismo , Glucagon/efeitos dos fármacos , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Células Secretoras de Glucagon/metabolismo , Glucose/metabolismo , Glucosídeos/farmacologia , Humanos , Insulina/metabolismo , Secreção de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Camundongos , Pâncreas/metabolismo , Ratos , Transportador 2 de Glucose-Sódio/fisiologia , Inibidores do Transportador 2 de Sódio-Glicose/farmacologia
5.
Hypertension ; 74(4): 1014-1020, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31378099

RESUMO

Obesity-related hypertension is a major public health concern. We recently demonstrated that plasma levels of the soluble form of the prorenin receptor (sPRR) were elevated in obesity-associated hypertension. Therefore, in the present study, we investigated the contribution of sPRR to blood pressure (BP) elevation in the context of obesity. High fat-fed C57BL/6 male mice were infused with vehicle or sPRR (30 µg/kg per day) via subcutaneously implanted osmotic minipump for 4 weeks. BP parameters were recorded using radiotelemetry devices. Male mice infused with sPRR exhibited higher systolic BP and mean arterial pressure and lower spontaneous baroreflex sensitivity than mice infused with vehicle. To define mechanisms involved in systolic BP elevation, mice were injected with an AT1R (Ang II [angiotensin II] type 1 receptor) antagonist (losartan), a muscarinic receptor antagonist (atropine), a ß-adrenergic antagonist (propranolol), and a ganglionic blocker (chlorisondamine). Losartan did not blunt sPRR-induced elevation in systolic BP. Chlorisondamine treatment exacerbated the decrease in mean arterial pressure in male mice infused with sPRR. These results demonstrated that sPRR induced autonomic nervous dysfunction. Interestingly, plasma leptin levels were increased in high fat-fed C57BL/6 male mice infused with sPRR. Overall, our results indicated that sPRR increased systolic BP through an impairment of the baroreflex sensitivity and an increase in the sympathetic tone potentially mediated by leptin in high fat-fed C57BL/6 male mice.


Assuntos
Pressão Sanguínea/efeitos dos fármacos , Dieta Hiperlipídica , Receptores de Superfície Celular/administração & dosagem , Antagonistas Adrenérgicos beta/farmacologia , Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Animais , Atropina/farmacologia , Barorreflexo/efeitos dos fármacos , Clorisondamina/farmacologia , Bloqueadores Ganglionares/farmacologia , Infusões Subcutâneas , Leptina/sangue , Losartan/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Antagonistas Muscarínicos/farmacologia , Propranolol/farmacologia , Receptor de Pró-Renina
6.
Am J Physiol Heart Circ Physiol ; 316(3): H506-H515, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30550352

RESUMO

Deletion of the prorenin receptor (PRR) in adipose tissue elevates systolic blood pressure (SBP) and the circulating soluble form of PRR (sPRR) in male mice fed a high-fat (HF) diet. However, sex differences in the contribution of adipose-PRR and sPRR to the regulation of the renin-angiotensin system (RAS) in key organs for blood pressure control are undefined. Therefore, we assessed blood pressure and the systemic and intrarenal RAS status in adipose-PRR knockout (KO) female mice. Blockade of RAS with losartan blunted SBP elevation in HF diet-fed adipose-PRR KO mice. ANG II levels were significantly increased in the renal cortex of HF diet-fed adipose-PRR KO female mice, but not systemically. HF diet-fed adipose-PRR KO mice exhibited higher vasopressin levels, water retention, and lower urine output than wild-type (WT) mice. The results also showed that deletion of adipose-PRR increased circulating sPRR and total hepatic sPRR contents, suggesting the liver as a major source of elevated plasma sPRR in adipose-PRR KO mice. To mimic the elevation of circulating sPRR and define the direct contribution of systemic sPRR to the regulation of the RAS and vasopressin, C57BL/6 female mice fed a standard diet were infused with recombinant sPRR. sPRR infusion increased plasma renin levels, renal and hepatic angiotensinogen expression, and vasopressin. Together, these results demonstrate that the deletion of adipose-PRR induced an elevation of SBP likely mediated by an intrarenal ANG II-dependent mechanism and that sPRR participates in RAS regulation and body fluid homeostasis via its capacity to activate the RAS and increase vasopressin levels. NEW & NOTEWORTHY The elevation of systolic blood pressure appears to be primarily mediated by cortical ANG II in high-fat diet-fed adipose-prorenin receptor knockout female mice. In addition, our data support a role for soluble prorenin receptor in renin-angiotensin system activation and vasopressin regulation.


Assuntos
Tecido Adiposo/efeitos dos fármacos , Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Anti-Hipertensivos/farmacologia , Pressão Sanguínea , Losartan/farmacologia , Receptores de Superfície Celular/sangue , Sistema Renina-Angiotensina , Tecido Adiposo/metabolismo , Angiotensina II/sangue , Angiotensinogênio/genética , Angiotensinogênio/metabolismo , Animais , Feminino , Rim/efeitos dos fármacos , Rim/metabolismo , Fígado/efeitos dos fármacos , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Superfície Celular/deficiência , Receptores de Superfície Celular/genética , Vasopressinas/farmacologia , Receptor de Pró-Renina
7.
Food Funct ; 9(5): 2922-2930, 2018 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-29741190

RESUMO

Little is still known about brain protein synthesis. In order to increase our knowledge of it, we aimed to modulate brain protein synthesis rates through aging, variations in nutritional state (fed state vs. fasted state), high sucrose diet and micronutrient supplementation. Four groups of 16 month-old male rats were fed for five months with a diet containing either 13% or 62% sucrose (wheat starch was replaced with sucrose), supplemented or not with rutin (5 g kg-1 diet), vitamin E (4×), A (2×), D (5×), selenium (10×) and zinc (+44%) and compared with an adult control group. We measured cerebellum protein synthesis and hippocampus gene expression of antioxidant enzymes, inflammatory cytokines and transcription factors. We showed that cerebellum protein synthesis was unchanged by the nutritional state, decreased during aging (-8%), and restored to the adult level by micronutrient supplementation. Sucrose diet did not change protein synthesis but reduced the protein content. Micronutrient supplementation had no effect in sucrose fed rats. Hippocampus gene expressions were affected by age (an increase of TNF-α), sucrose treatment (an increase of IL-1ß and IL-6), and micronutrient supplementation (a decrease of heme oxygenase, catalase, glutathione peroxidase, TNF-α, and Nrf2). We noted that cerebellum protein synthesis and hippocampus TNF-α gene expression were modulated by the same factors: they were affected by aging and micronutrient supplementation and unchanged by feeding and by high sucrose diet.


Assuntos
Envelhecimento/metabolismo , Encéfalo/metabolismo , Sacarose Alimentar/metabolismo , Micronutrientes/metabolismo , Biossíntese de Proteínas , Rutina/metabolismo , Envelhecimento/efeitos dos fármacos , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/crescimento & desenvolvimento , Sacarose Alimentar/efeitos adversos , Suplementos Nutricionais/análise , Hipocampo/efeitos dos fármacos , Hipocampo/crescimento & desenvolvimento , Hipocampo/metabolismo , Humanos , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Masculino , Micronutrientes/farmacologia , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , Estado Nutricional , Biossíntese de Proteínas/efeitos dos fármacos , Ratos , Ratos Wistar , Rutina/farmacologia , Selênio/metabolismo , Selênio/farmacologia , Vitamina A/metabolismo , Vitamina A/farmacologia , Vitamina D/metabolismo , Vitamina D/farmacologia , Vitamina E/metabolismo , Vitamina E/farmacologia , Zinco/metabolismo , Zinco/farmacologia
8.
J Physiol Biochem ; 74(4): 569-577, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29637446

RESUMO

High-sugar intake and senescence share common deleterious effects, in particular in liver, but combination of these two factors was little studied. Our aims were to examine the effect of a high-sucrose diet in liver of old rats and also the potential benefices of a polyphenol/micronutrient supplementation. Four groups of 22-month-old male rats fed during 5 months with a diet containing either 13 or 62% sucrose, supplemented or not with rutin, vitamin E, A, D, selenium, and zinc were compared. We measured liver macronutrient composition, glycation/oxidative stress, enzyme activities (lipogenesis, ß-oxidation, fructokinase), gene expression (enzymes and transcription factors), in vivo protein synthesis rates and plasma parameters. Sucrose induced an increase in plasma and liver lipid content, and a stimulation of liver protein synthesis rates. Gene expression was little changed by sucrose, with lower levels for LXR-α and LXR-ß. Polyphenol/micronutrient supplementation tended to limit liver triglyceride infiltration through variations in fatty acid synthase, acyl coA oxidase, and possibly ATP-citrate lyase activities. In conclusion, despite differences in enzymatic regulations, and blunted responses of gene expression, high-sucrose diet was still able to induce a marked increase in liver lipid content in old animals. However, it probably attenuated the positive impact of polyphenol/micronutrients.


Assuntos
Envelhecimento , Antioxidantes/uso terapêutico , Sacarose Alimentar/efeitos adversos , Suplementos Nutricionais , Fígado/metabolismo , Hepatopatia Gordurosa não Alcoólica/prevenção & controle , Rutina/uso terapêutico , Animais , Antioxidantes/metabolismo , Dieta da Carga de Carboidratos/efeitos adversos , Regulação da Expressão Gênica no Desenvolvimento , Glicosilação , Metabolismo dos Lipídeos , Fígado/crescimento & desenvolvimento , Receptores X do Fígado/genética , Receptores X do Fígado/metabolismo , Masculino , Micronutrientes/administração & dosagem , Micronutrientes/metabolismo , Micronutrientes/uso terapêutico , Hepatopatia Gordurosa não Alcoólica/etiologia , Hepatopatia Gordurosa não Alcoólica/metabolismo , Estresse Oxidativo , Processamento de Proteína Pós-Traducional , Distribuição Aleatória , Ratos Wistar
9.
Nutr Res Rev ; 30(2): 191-207, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28511733

RESUMO

In our societies, the proportions of elderly people and of obese individuals are increasing. Both factors are associated with high health-related costs. During obesity, many authors suggest that it is a high chronic intake of added sugars (HCIAS) that triggers the shift towards pathology. However, the majority of studies were performed in young subjects and only a few were interested in the interaction with the ageing process. Our purpose was to discuss the metabolic effects of HCIAS, compare with the effects of ageing, and evaluate how deleterious the combined action of HCIAS and ageing could be. This effect of HCIAS seems mediated by fructose, targeting the liver first, which may lead to all subsequent metabolic alterations. The first basic alterations induced by fructose are increased oxidative stress, protein glycation, inflammation, dyslipidaemia and insulin resistance. These alterations are also present during the ageing process, and are closely related to each other, one leading to the other. These basic alterations are also involved in more complex syndromes, which are also favoured by HCIAS, and present during ageing. These include non-alcoholic fatty liver disease, hypertension, neurodegenerative diseases, sarcopenia and osteoporosis. Cumulative effects of ageing and HCIAS have been seldom tested and may not always be strictly additive. Data also suggest that some of the metabolic alterations that are more prevalent during ageing could be related more with nutritional habits than to intrinsic ageing. In conclusion, it is clear that HCIAS interacts with the ageing process, accelerates the accumulation of metabolic alterations, and that it should be avoided.


Assuntos
Envelhecimento/fisiologia , Açúcares da Dieta/administração & dosagem , Açúcares da Dieta/efeitos adversos , Animais , Dislipidemias/epidemiologia , Dislipidemias/etiologia , Frutose/administração & dosagem , Frutose/efeitos adversos , Frutose/metabolismo , Glicosilação/efeitos dos fármacos , Humanos , Inflamação/epidemiologia , Inflamação/etiologia , Resistência à Insulina , Fígado/efeitos dos fármacos , Fígado/fisiopatologia , Hepatopatia Gordurosa não Alcoólica/epidemiologia , Hepatopatia Gordurosa não Alcoólica/etiologia , Obesidade/epidemiologia , Obesidade/fisiopatologia , Estresse Oxidativo/efeitos dos fármacos
10.
J Proteome Res ; 15(6): 1862-74, 2016 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-27115730

RESUMO

We aimed to determine the time-course of metabolic changes related to the early onset of insulin resistance (IR), trying to evidence breaking points preceding the appearance of the clinical IR phenotype. The model chosen was the fructose (FRU)-fed rat compared to controls fed with starch. We focused on the hepatic metabolism after 0, 5, 12, 30, or 45 days of FRU intake. The hepatic molecular metabolic changes followed indeed a multistep trajectory rather than a continuous progression. After 5 d of FRU feeding, we observed deep modifications in the hepatic metabolism, driven by the induction of lipogenic genes and important glycogen depletion. Thereafter, a steady-state period between days 12 and 30 was observed, characterized by a switch from carbohydrate to lipid utilization at the hepatic level and increased insulin levels aiming at alleviating lipid accumulation and hyperglycemia, respectively. The FRU-fed animals were only clinically IR at day 45 (altered homeostasis model assessment-estimated insulin resistance and muscle glucose transport). Furthermore, the urine metabolome revealed even earlier metabolic trajectory changes that precede the hepatic alterations. We identified several candidate metabolites linked to the tryptophan-nicotinamide metabolism and the installation of fasting hyperglycemia that suggest a role of this metabolic pathway on the development of the IR phenotype in the FRU-fed rats.


Assuntos
Frutose/farmacologia , Resistência à Insulina , Metabolismo , Animais , Metabolismo dos Carboidratos , Frutose/administração & dosagem , Hiperglicemia/metabolismo , Metabolismo dos Lipídeos , Fígado/metabolismo , Metabolômica , Niacinamida/metabolismo , Ratos , Fatores de Tempo , Triptofano/metabolismo
11.
J Nutr ; 145(5): 923-30, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25809681

RESUMO

BACKGROUND: Today, high chronic intake of added sugars is frequent, which leads to inflammation, oxidative stress, and insulin resistance. These 3 factors could reduce meal-induced stimulation of muscle protein synthesis and thus aggravate the age-related loss of muscle mass (sarcopenia). OBJECTIVES: Our aims were to determine if added sugars could accelerate sarcopenia and to assess the capacity of antioxidants and anti-inflammatory agents to prevent this. METHODS: For 5 mo, 16-mo-old male rats were starch fed (13% sucrose and 49% wheat starch diet) or sucrose fed (62% sucrose and 0% wheat starch diet) with or without rutin (5 g/kg diet), vitamin E (4 times), vitamin A (2 times), vitamin D (5 times), selenium (10 times), and zinc (+44%) (R) supplementation. We measured the evolution of body composition and inflammation, plasma insulin-like growth factor 1 (IGF-I) concentration and total antioxidant status, insulin sensitivity (oral-glucose-tolerance test), muscle weight, superoxide dismutase activity, glutathione concentration, and in vivo protein synthesis rates. RESULTS: Sucrose-fed rats lost significantly more lean body mass (-8.1% vs. -5.4%, respectively) and retained more fat mass (+0.2% vs. -33%, respectively) than starch-fed rats. Final muscle mass was 11% higher in starch-fed rats than in sucrose-fed rats. Sucrose had little effect on inflammation, oxidative stress, and plasma IGF-I concentration but reduced the insulin sensitivity index (divided by 2). Meal-induced stimulation of muscle protein synthesis was significantly lower in sucrose-fed rats (+7.3%) than in starch-fed rats (+22%). R supplementation slightly but significantly reduced oxidative stress and increased muscle protein concentration (+4%) but did not restore postprandial stimulation of muscle protein synthesis. CONCLUSIONS: High chronic sucrose intake accelerates sarcopenia in older male rats through an alteration of postprandial stimulation of muscle protein synthesis. This effect could be explained by a decrease of insulin sensitivity rather than by changes in plasma IGF-I, inflammation, and/or oxidative stress.


Assuntos
Envelhecimento , Sacarose Alimentar/efeitos adversos , Regulação da Expressão Gênica no Desenvolvimento , Resistência à Insulina , Proteínas Musculares/biossíntese , Músculo Esquelético/metabolismo , Sarcopenia/etiologia , Adiposidade , Animais , Anti-Inflamatórios não Esteroides/uso terapêutico , Antioxidantes/uso terapêutico , Composição Corporal , Sacarose Alimentar/antagonistas & inibidores , Suplementos Nutricionais , Glutationa/metabolismo , Fator de Crescimento Insulin-Like I/análise , Masculino , Músculo Esquelético/imunologia , Músculo Esquelético/patologia , Estresse Oxidativo , Período Pós-Prandial , Distribuição Aleatória , Ratos Wistar , Sarcopenia/imunologia , Sarcopenia/metabolismo , Sarcopenia/prevenção & controle
12.
PLoS One ; 9(9): e109098, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25268515

RESUMO

Our aim was to compare and combine 3 nutritional strategies to slow down the age-related loss of muscle mass in healthy old rats: 1) increase protein intake, which is likely to stimulate muscle protein anabolism; 2) use leucine rich, rapidly digested whey proteins as protein source (whey proteins are recognized as the most effective proteins to stimulate muscle protein anabolism). 3) Supplement animals with a mixture of chamomile extract, vitamin E, vitamin D (reducing inflammation and oxidative stress is also effective to improve muscle anabolism). Such comparisons and combinations were never tested before. Nutritional groups were: casein 12% protein, whey 12% protein, whey 18% protein and each of these groups were supplemented or not with polyphenols/antioxidants. During 6 months, we followed changes of weight, food intake, inflammation (plasma fibrinogen and alpha-2-macroglobulin) and body composition (DXA). After 6 months, we measured muscle mass, in vivo and ex-vivo fed and post-absorptive muscle protein synthesis, ex-vivo muscle proteolysis, and oxidative stress parameters (liver and muscle glutathione, SOD and total antioxidant activities, muscle carbonyls and TBARS). We showed that although micronutrient supplementation reduced inflammation and oxidative stress, the only factor that significantly reduced the loss of lean body mass was the increase in whey protein intake, with no detectable effect on muscle protein synthesis, and a tendency to reduce muscle proteolysis. We conclude that in healthy rats, increasing protein intake is an effective way to delay sarcopenia.


Assuntos
Envelhecimento/efeitos dos fármacos , Suplementos Nutricionais , Proteínas Musculares/metabolismo , Músculo Esquelético/efeitos dos fármacos , Sarcopenia/prevenção & controle , Proteínas do Soro do Leite/administração & dosagem , Envelhecimento/metabolismo , Animais , Peso Corporal/efeitos dos fármacos , Camomila/química , Dieta/métodos , Ingestão de Alimentos/efeitos dos fármacos , Fibrinogênio/metabolismo , Glutationa/metabolismo , Masculino , Proteínas Musculares/agonistas , Músculo Esquelético/metabolismo , Tamanho do Órgão/efeitos dos fármacos , Extratos Vegetais/administração & dosagem , Ratos , Ratos Wistar , Superóxido Dismutase/metabolismo , Substâncias Reativas com Ácido Tiobarbitúrico/metabolismo , Vitamina D/administração & dosagem , Vitamina E/administração & dosagem , alfa-Macroglobulinas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA