Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 168
Filtrar
Mais filtros

País/Região como assunto
Intervalo de ano de publicação
1.
J Gastroenterol Hepatol ; 38(11): 1868-1876, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37438882

RESUMO

Obesity is related to several organs, but the liver is particularly affected. Adenosine monophosphate-activated protein kinase (AMPK) is a cellular energy sensor and regulator of liver lipid dysfunction and glucose metabolism. The mechanistic target of rapamycin (mTOR) is a protein kinase regulating cell growth, survival, metabolism, and immunity. Together, these pathways are involved in obesity, insulin resistance, non-alcoholic fatty liver disease (NAFLD) and its progression, and autophagy. During energy demand, liver kinase B (LKB) phosphorylation helps activate the AMPK/mTOR pathways. Likewise, the protein forkhead box O family (FOXO) negatively regulates adipogenesis by binding to the promoter sites of peroxisome proliferator-activated receptor-gamma coactivator 1-alpha, initiating adipogenesis. In addition, acetyl-CoA carboxylase, which regulates de novo lipogenesis, is linked to LKB and FOXO in developing NAFLD. The kinase complex, consisting of Unc-51-like autophagy-activating kinase 1 or 2 (ULK1, ULK2) by stimulating autophagy, and eliminating fat droplets in NAFLD, is regulated by mTORC1 and negatively regulated by AMPK that suppresses liver lipogenesis and increases fatty acid oxidation. Also, ULK1 is essential for initiating phagophore formation, establishing macrophagy, and generating autophagosomes. The selective breakdown of lipid droplets through macroautophagy, or macrolipophagy, occurs on a cellular energy level using free fatty acids. In addition, mTORC1 promotes lipogenesis by activating sterol regulatory element-binding protein. Finding new components and novel regulatory modes in signaling is significant for a better understanding of the AMPK/mTOR pathways, potentially facilitating the development of future diagnostic and therapeutic strategies for NAFLD and its progression to non-alcoholic steatohepatitis, cirrhosis, and hepatocellular carcinoma.


Assuntos
Hepatopatia Gordurosa não Alcoólica , Humanos , Hepatopatia Gordurosa não Alcoólica/metabolismo , Proteínas Quinases Ativadas por AMP/metabolismo , Fígado/patologia , Serina-Treonina Quinases TOR/metabolismo , Obesidade , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo
2.
Int J Obes (Lond) ; 46(1): 21-29, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34465857

RESUMO

BACKGROUND/OBJECTIVES: The weight loss following Semaglutide treatment, a GLP-1 receptor agonist, might be responsible for some effects observed on the nonalcoholic fatty liver disease of obese mice. SUBJECTS/METHODS: Two groups of C57BL/6 male mice (n = 30/group) were fed the diets Control (C) or high-fat (HF) for 16 weeks. Then, separated into six new groups for an additional four weeks (n = 10/group) and treated with Semaglutide (S, 40 µg/kg) or paired feeding (PF) with S groups (C; C-S; C-PF; HF; HF-S; HF-PF). RESULTS: Semaglutide reduced energy consumption leading to weight loss. Simultaneously it improved glucose intolerance, glycated hemoglobin, insulin resistance/sensitivity, plasma lipids, and gastric inhibitory polypeptide. Semaglutide and paired feeding mitigated liver steatosis and adipose differentiation-related protein (Plin2) expression. Semaglutide also improved hormones and adipokines, reduced lipogenesis and inflammation, and increased beta-oxidation. Semaglutide lessened liver glucose uptake and endoplasmic reticulum (ER) stress. Among the 14 genes analyzed, 13 were modified by Semaglutide (93 %, six genes were changed exclusively by Semaglutide, and seven other genes were affected by the combination of Semaglutide and paired feeding). In seven genes, the paired diet showed no effect (50% of the genes tested). No marker was affected exclusively by paired feeding. CONCLUSIONS: Semaglutide and the consequent weight loss reduced obese mice liver inflammation, insulin resistance, and ER stress. However, weight loss alone did show few or no action on some significant study findings, like liver steatosis, leptin, insulin, resistin, and amylin. Furthermore, hepatic inflammation mediated by MCP-1 and partially by TNF-alpha and IL6 were also not reduced by weight loss. Furthermore, weight loss alone did not lessen hepatic lipogenesis as determined by the findings of SREBP-1c, CHREBP, PPAR-alpha, and SIRT1. Semaglutide was implicated in improving glucose uptake and lessening ER stress by reducing GADD45, independent of weight loss.


Assuntos
Retículo Endoplasmático/fisiologia , Receptor do Peptídeo Semelhante ao Glucagon 1/antagonistas & inibidores , Hepatopatia Gordurosa não Alcoólica/etiologia , Obesidade/complicações , Animais , Modelos Animais de Doenças , Retículo Endoplasmático/metabolismo , Receptor do Peptídeo Semelhante ao Glucagon 1/uso terapêutico , Peptídeos Semelhantes ao Glucagon/efeitos adversos , Peptídeos Semelhantes ao Glucagon/antagonistas & inibidores , Peptídeos Semelhantes ao Glucagon/uso terapêutico , Camundongos , Camundongos Endogâmicos C57BL , Hepatopatia Gordurosa não Alcoólica/fisiopatologia , Apoio Nutricional , Obesidade/fisiopatologia , Redução de Peso/fisiologia
3.
Exp Parasitol ; 212: 107889, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32222527

RESUMO

Human and experimental studies have shown that chronic schistosomiasis mansoni protects against metabolic disorders through direct and indirect pathways. This study aims to investigate the co-morbidity between the acute schistosomiasis and nonalcoholic fatty liver. To address this, male C57BL/6 mice fed a high-fat chow (60% fat) or standard chow (10% fat) for 13 weeks and later infected with 80 Schistosoma mansoni cercariae. Mice were assigned into four groups: uninfected fed standard (USC), uninfected fed high-fat chow (UHFC), infected fed standard (ISC), and infected fed high-fat chow (IHFC). Blood sample and tissues were obtained at nine weeks post-infection (acute schistosomiasis) by necropsy. UHFC mice showed higher body mass, visceral adiposity, impaired glucose tolerance, total cholesterol (TC), low-density lipoprotein cholesterol (LDL-c), triglyceride (TG), and liver steatosis compared to USC mice. IHFC mice showed lower blood lipid levels, blood glucose, improved glucose tolerance, body mass, and liver steatosis (macro and microvesicular) compared to UHFC mice. IHFC showed more massive histopathological changes (sinusoidal fibrosis, hepatocellular ballooning, and inflammatory infiltrates) compared to ISC. In conclusion, the co-morbidity results in both beneficial (friend) and detrimental (foe) for the host. While the acute schistosomiasis improves some metabolic features of metabolic syndrome, comorbidity worsens the liver injury.


Assuntos
Síndrome Metabólica/epidemiologia , Esquistossomose mansoni/epidemiologia , Análise de Variância , Animais , Área Sob a Curva , Biomphalaria/parasitologia , Comorbidade , Dieta Hiperlipídica/efeitos adversos , Modelos Animais de Doenças , Fígado Gorduroso/etiologia , Fígado Gorduroso/patologia , Granuloma/etiologia , Granuloma/patologia , Intestinos/parasitologia , Fígado/patologia , Masculino , Síndrome Metabólica/complicações , Síndrome Metabólica/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Esquistossomose mansoni/complicações , Esquistossomose mansoni/metabolismo , Aumento de Peso
4.
Eur J Nutr ; 57(5): 1901-1912, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28534168

RESUMO

PURPOSE: Studies demonstrated the influence of mother's obesity on offspring. However, the father is also related to programming the future generation. The study aimed to evaluate the effects of father's obesity upon white adipose tissue (WAT) remodeling, resulting in activation of signaling pathways and inflammation in male and female offspring. METHODS: Male C57BL/6 mice received control diet (lean father group; 17% energy from lipids) or high-fat diet (obese father group; 49% energy from lipids) for 8 weeks before mating. The mothers received control diet throughout the experiment. Mice were mated: lean mother and lean father, and lean mother and obese father. Offspring received control diet from weaning until 3 months of age when they were studied. RESULTS: In the offspring, father's obesity led to decreased QUICKI with impairment of the insulin signaling pathway in both sexes. In line with these findings, in white adipose tissue, male offspring demonstrated hypertrophied adipocytes, enhanced proinflammatory cytokines, overactivation of components of the local renin-angiotensin system (RAS) and extracellular signal-regulated kinase 1/2 (ERK1/2), and inhibition of peroxisome proliferator-activated receptors (alpha and gamma). CONCLUSIONS: We observed that father's obesity influences the offspring in adult life, with an impairment in insulin homeostasis, adipocyte remodeling, and adipose tissue overexpression of IL-6 and TNF-alpha in male offspring. The activation of local RAS and ERK1/2, a concomitant PPAR diminishing, and impairment in phosphorylation of AKT and IRS-1 could explain at least in part the findings regardless of the increase in body mass in the offspring.


Assuntos
Dieta Hiperlipídica , Obesidade/etiologia , Sistema Renina-Angiotensina/fisiologia , Tecido Adiposo/metabolismo , Animais , Brasil , Dieta Hiperlipídica/efeitos adversos , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/epidemiologia
5.
Eur J Nutr ; 55(1): 159-69, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25612928

RESUMO

PURPOSE: Fish oil (FO) elicits diverse beneficial effects. Reduction in or prevention of body mass (BM) gain in animal models may be associated with modulation of brown adipose tissue (BAT). We aimed to evaluate the effects of different high-fat diets with FO on BAT metabolism and thermogenic markers. METHODS: C57BL/6 male mice (3-month-old) were fed different diets during 8 weeks: standard-chow diet (SC 10% fat), high-fat lard diet (HF-L 50% fat), high-fat lard plus FO diet (HF-L+FO 50% fat), and high-fat FO diet (HF-FO 50% fat). We evaluated BM and performed an oral glucose tolerance test. At euthanasia, plasma was collected for leptin, and triacylglycerol measurement and interscapular BAT was dissected and stored for molecular analyses. RESULTS: HF-L group showed elevated BM; glucose intolerance associated with diminished TC10 and GLUT4 expressions; hypertriglyceridemia associated with increased CD36 and diminished CPT1 expression; elevated expression of pro-inflammatory cytokines; and reduced PPAR expression. Furthermore, these animals showed hyperleptinemia with increased expression of thermogenic markers (beta3-AR, PGC1alpha, and UCP1). Conversely, HF-L+FO and HF-FO groups showed reduced BM gain with regularization of glucose tolerance and triglyceridemia, GLUT4, TC10, CD36, CPT1, and cytokines expressions. Both groups exhibited elevated PPAR and thermogenic markers expression in a dose-dependent way. CONCLUSIONS: FO improves metabolic profile and upregulates thermogenic markers, suggesting an elevated thermogenesis that leads to reduced BM gain.


Assuntos
Tecido Adiposo Marrom/efeitos dos fármacos , Óleos de Peixe/administração & dosagem , Termogênese/efeitos dos fármacos , Regulação para Cima , Tecido Adiposo Marrom/metabolismo , Animais , Biomarcadores/sangue , Biomarcadores/metabolismo , Glicemia/metabolismo , Antígenos CD36/genética , Antígenos CD36/metabolismo , Carnitina O-Palmitoiltransferase/genética , Carnitina O-Palmitoiltransferase/metabolismo , Dieta Hiperlipídica , Gorduras na Dieta , Intolerância à Glucose/metabolismo , Teste de Tolerância a Glucose , Transportador de Glucose Tipo 4/genética , Transportador de Glucose Tipo 4/metabolismo , Insulina/sangue , Interleucina-6/genética , Interleucina-6/metabolismo , Canais Iônicos/genética , Canais Iônicos/metabolismo , Leptina/sangue , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Triglicerídeos/sangue , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo , Proteína Desacopladora 1
6.
Artigo em Inglês | MEDLINE | ID: mdl-26872995

RESUMO

Seasonal plasticity in the small intestine of neonatal tegu lizards was investigated using morphometry and analysis of enzymes involved in supplying energy to the intestinal tissue. In the autumn, the intestinal mass (Mi) was 1.0% of body mass and the scaling exponent b=0.92 indicated that Mi was larger in smaller neonates. During arousal from dormancy Mi was 23% smaller; later in spring, Mi increased 60% in relation to the autumn and the exponent b=0.14 indicated that the recovery was disproportionate in smaller tegus. During the autumn, the intestinal villi were greatly elongated; by midwinter, the Hv, SvEp, and VvEp were smaller than during the autumn (59%, 54%, 29%) and were restored to autumn levels during spring. In the active tegus, the maximum activity (Vmax) of enzymes indicated that the enterocytes can obtain energy from different sources, and possess gluconeogenic capacity. During winter, the Vmax of CS, HOAD, GDH, PEPCK was 40-50% lower in relation to the autumn and spring, while the Vmax of HK, PK, LDH, AST was unchanged. The hypoglycemia and the mucosal atrophy/ischemia during winter would prevent the enterocytes from using glucose, whereas they could slowly oxidize fatty acids released from body stores and amino acids from the tissue proteolysis to satisfy their needs of energy. Contrastingly, starvation during spring caused severe mass loss (50%); the tissue protein and the VvEp and VvLP did not change while the thickness of the muscular layer increased 51%, which suggested different effects along the length of the organ. In addition, the Vmax of the glycolytic enzymes was lower, indicating that a regulatory mechanism would spare blood glucose for vital organs during unanticipated food restriction.


Assuntos
Jejum , Hibernação , Intestino Delgado/metabolismo , Lagartos/fisiologia , Animais , Peso Corporal , Lagartos/crescimento & desenvolvimento , Estações do Ano
7.
Cell Biochem Funct ; 33(4): 249-56, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25959716

RESUMO

Obesity is considered a public health problem worldwide. Fenofibrate, a selective peroxisome proliferator-activated receptor α (PPAR-α) agonist, elicits weight loss in animal models. This study aimed to examine the effects of fenofibrate on energy expenditure, body mass (BM) and gene expression of thermogenic factors in brown adipose tissue of diet-induced obese mice. Male C57BL/6 mice were fed a standard chow (SC; 10% lipids) diet or a high-fat (HF; 50% lipids) diet for 10 weeks. Afterwards, groups were subdivided as SC, SC-F, HF and HF-F (n = 10, each). Treatment with fenofibrate (100 mg kg(-1) BM mixed into the diet) lasted 5 weeks. Treated groups had reduced final BM compared with their counterparts (p < 0·05), explained by the increase in energy expenditure, CO2 production and O2 consumption after treatment with fenofibrate (p < 0·05). Similarly, genes involved in thermogenesis as PPAR-α, PPAR-γ coactivator 1α, nuclear respiratory factor 1, mitochondrial transcription factor A (Tfam), PR domain containing 16 (PRDM16), ß-3 adrenergic receptor (ß3-AR), bone morphogenetic protein 8B and uncoupling protein 1 were significantly expressed in brown adipocytes after the treatment (p < 0·05). All observations ensure that selective PPAR-α agonist can induce thermogenesis by increasing energy expenditure and enhancing the expression of genes involved in the thermogenic pathway. These results suggest fenofibrate as a coadjutant drug for the treatment of obesity.


Assuntos
Adipócitos Marrons/metabolismo , Tecido Adiposo/metabolismo , Dieta/efeitos adversos , Fenofibrato/farmacologia , Obesidade/tratamento farmacológico , PPAR alfa/agonistas , Redução de Peso/efeitos dos fármacos , Adipócitos Marrons/efeitos dos fármacos , Tecido Adiposo/citologia , Tecido Adiposo/efeitos dos fármacos , Animais , Biomarcadores/metabolismo , Metabolismo Energético/efeitos dos fármacos , Hipolipemiantes/farmacologia , Técnicas Imunoenzimáticas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Obesidade/etiologia , Obesidade/metabolismo , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
8.
Exp Physiol ; 99(12): 1663-78, 2014 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-25326526

RESUMO

Telmisartan has previously been used to target obesity, showing peroxisome proliferator-activated receptor (PPAR) ß/δ-related effects in white adipose tissue (WAT). We sought to evaluate whether telmisartan enhances gene and protein expression of all PPAR isoforms in WAT and brown adipose tissue (BAT), as well as their downstream effects upon insulin resistance, adipokine profile and adaptive thermogenesis. Male C57BL/6 mice were fed standard chow (SC; 10% lipids) or high-fat diet (HF; 50% lipids) for 10 weeks. Animals were then randomly allocated into the following four groups: SC, SC-T, HF and HF-T. Telmisartan [10 mg (kg diet)(-1)] was administered for 4 weeks in the diet. Animals in the HF group were overweight and exhibited hypertension, insulin resistance, decreased energy expenditure, a pro-inflammatory adipokine profile and abnormal fat pad mass distribution. Animals in the HF group showed decreased expression of PPARα, ß/δ and γ in WAT and BAT, resulting in impaired glucose uptake and insufficient thermogenesis. Due to the improvement in the adipokine profile and enhanced insulin sensitivity with adequate insulin-stimulated glucose uptake after treatment with telmisartan, the activation of all PPAR isoforms in WAT was beneficial. In BAT, telmisartan induced sustained sympathetic activation, because the ß3-adrenergic receptor was induced by PPARß/δ, while uncoupling protein 1 was induced by PPARα to promote thermogenesis. Telmisartan exerted anti-obesity effects through higher pan-PPAR gene and protein expression. Upon PPARα, ß/δ and γ (pan-PPAR) agonism in adipose tissue of obese mice, telmisartan ameliorates inflammation and insulin resistance, as well as inducing non-shivering thermogenesis. Our results point to new therapeutic targets for the control of obesity and comorbidities through pan-PPAR-related effects.


Assuntos
Tecido Adiposo/metabolismo , Benzimidazóis/farmacologia , Benzoatos/farmacologia , Obesidade/metabolismo , Receptores Ativados por Proliferador de Peroxissomo/metabolismo , Tecido Adiposo/efeitos dos fármacos , Animais , Benzimidazóis/uso terapêutico , Benzoatos/uso terapêutico , Dieta Hiperlipídica , Expressão Gênica/efeitos dos fármacos , Resistência à Insulina/fisiologia , Masculino , Camundongos , Camundongos Obesos , Obesidade/tratamento farmacológico , Obesidade/genética , Receptores Ativados por Proliferador de Peroxissomo/genética , Telmisartan
9.
Mol Nutr Food Res ; 68(4): e2300148, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38085111

RESUMO

SCOPE: To analyze the effects of fexaramine (FEX), as an intestinal FXR agonist, on the modulation of the intestinal microbiota and ileum of mice fed a high-fat (HF) diet. METHODS AND RESULTS: Three-month-old C57Bl/6 male mice are divided into two groups and received a control (C, 10% of energy from lipids) or HF (50% of energy from lipids) diet for 12 weeks. They are subdivided into the C, C + FEX, HF, and HF + FEX groups. FEX is administered (FEX-5 mg kg-1 ) via orogastric gavage for three weeks. Body mass (BM), glucose metabolism, qPCR 16S rRNA gene expression, and ileum gene expression, bile acids (BAs), tight junctions (TJs), and incretin are analyzed. FEX reduces BM and glucose intolerance, reduces plasma lipid concentrations and the Firmicutes/Bacteroidetes ratio, increases the Lactobacillus sp. and Prevotella sp. abundance, and reduces the Escherichia coli abundance. Consequently, the ileal gene expression of Fxr-Fgf15, Tgr5-Glp1, and Cldn-Ocldn-Zo1 is increased, and Tlr4-Il6-Il1beta is decreased. CONCLUSION: FEX stimulates intestinal FXR and improves dysbiosis, intestinal TJs, and the release of incretins, mitigating glucose intolerance and BM increases induced by an HF diet. However, FEX results in glucose intolerance, insulin resistance, and reduces intestinal TJs in a control group, thus demonstrating limitations to this dietary model.


Assuntos
Intolerância à Glucose , Camundongos , Masculino , Animais , Intolerância à Glucose/tratamento farmacológico , Dieta Hiperlipídica/efeitos adversos , Disbiose/tratamento farmacológico , RNA Ribossômico 16S , Junções Íntimas , Inflamação/tratamento farmacológico , Lipídeos , Camundongos Endogâmicos C57BL , Ácidos e Sais Biliares
10.
Nutr Res ; 126: 180-192, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38759501

RESUMO

Metabolic dysfunction-associated steatotic liver disease (MASLD) has attracted increasing attention from the scientific community because of its severe but silent progression and the lack of specific treatment. Glucolipotoxicity triggers endoplasmic reticulum (ER) stress with decreased beta-oxidation and enhanced lipogenesis, promoting the onset of MASLD, whereas regular physical exercise can prevent MASLD by preserving ER and mitochondrial function. Thus, the hypothesis of this study was that high-intensity interval training (HIIT) could prevent the development of MASLD in high-fat (HF)-fed C57BL/6J mice by maintaining insulin sensitivity, preventing ER stress, and promoting beta-oxidation. Forty male C57BL/6J mice (3 months old) comprised 4 experimental groups: the control (C) diet group, the C diet + HIIT (C-HIIT) group, the HF diet group, and the HF diet + HIIT (HF-HIIT) group. HIIT sessions lasted 12 minutes and were performed 3 times weekly by trained mice. The diet and exercise protocols lasted for 10 weeks. The HIIT protocol prevented weight gain and maintained insulin sensitivity in the HF-HIIT group. A chronic HF diet increased ER stress-related gene and protein expression, but HIIT helped to maintain ER homeostasis, preserve mitochondrial ultrastructure, and maximize beta-oxidation. The increased sirtuin-1/peroxisome proliferator-activated receptor-gamma coactivator 1-alpha expression implies that HIIT enhanced mitochondrial biogenesis and yielded adequate mitochondrial dynamics. High hepatic fibronectin type III domain containing 5/irisin agreed with the antilipogenic and anti-inflammatory effects observed in the HF-HIIT group, reinforcing the antisteatotic effects of HIIT. Thus, we confirmed that practicing HIIT 3 times per week maintained insulin sensitivity, prevented ER stress, and enhanced hepatic beta-oxidation, impeding MASLD development in this mouse model even when consuming high energy intake from saturated fatty acids.


Assuntos
Dieta Hiperlipídica , Estresse do Retículo Endoplasmático , Treinamento Intervalado de Alta Intensidade , Resistência à Insulina , Fígado , Camundongos Endogâmicos C57BL , Mitocôndrias Hepáticas , Condicionamento Físico Animal , Animais , Dieta Hiperlipídica/efeitos adversos , Masculino , Fígado/metabolismo , Mitocôndrias Hepáticas/metabolismo , Camundongos , Hepatopatia Gordurosa não Alcoólica/metabolismo , Hepatopatia Gordurosa não Alcoólica/terapia , Fígado Gorduroso/prevenção & controle , Oxirredução
11.
Eur J Nutr ; 52(2): 759-69, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22661265

RESUMO

PURPOSE: To investigate whether a maternal high-fat diet (HF) during pregnancy and/or suckling periods predisposes adult C57BL/6 mice offspring to morphological pancreatic modifications. METHODS: Male pups were divided into 5 groups: SC (standard chow)-from dams fed SC during gestation and lactation, maintaining an SC diet from postweaning to adulthood; G-from dams fed HF diets during gestation; L-from dams fed HF diets during lactation; GL-from dams fed HF diets during gestation and lactation; and GL/HF-from dams fed HF diets during gestation and lactation, maintaining an HF diet from postweaning to adulthood. We analysed body mass (BM), plasma insulin, pancreas and adipose tissue structures. RESULTS: During the entire experiment, the SC group had the lowest BM. However, GL/HF offspring were heavier than the other groups. This weight gain was also accompanied by adipocyte hypertrophy. At 3 months, G offspring showed an increased insulin levels and impairment in carbohydrates metabolism. Furthermore, pancreatic islets were hypertrophied in G, GL and GL/HF offspring in comparison with SC offspring. CONCLUSION: HF diet administration during the gestation period is more harmful than during the lactation period, exerting deleterious effects on pancreatic morphology in addition to larger fat deposits in adult mice offspring.


Assuntos
Dieta Hiperlipídica , Fenômenos Fisiológicos da Nutrição Materna , Pâncreas/fisiopatologia , Adipócitos/citologia , Adipócitos/metabolismo , Tecido Adiposo/metabolismo , Animais , Glicemia/análise , Índice de Massa Corporal , Metabolismo dos Carboidratos , Gorduras na Dieta/administração & dosagem , Feminino , Imunofluorescência , Teste de Tolerância a Glucose , Processamento de Imagem Assistida por Computador , Insulina/sangue , Lactação/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pâncreas/metabolismo , Gravidez , Desmame , Aumento de Peso
12.
Horm Mol Biol Clin Investig ; 44(2): 219-227, 2023 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-36288452

RESUMO

Brown adipose tissue (BAT) remains active in adults, oxidizing fatty acids or glucose and releasing energy in the form of heat. Brown adipocytes and enhanced thermogenesis are targets for treating obesity and its comorbidities. BAT shows high synthesis activity and secretes several signaling molecules. The brown adipokines, or batokines, take action in an autocrine, paracrine, and endocrine manner. Batokines have a role in the homeostasis of the cardiovascular system, central nervous system, white adipose tissue, liver, and skeletal muscle and exert beneficial effects on BAT. The systemic function of batokines gives BAT an endocrine organ profile. Besides, the batokines Fibroblast Growth Factor-21, Vascular Endothelial Growth Factor A, Bone Morphogenetic Protein 8, Neuregulin 4, Myostatin, and Interleukin-6 emerge as targets to treat obesity and its comorbidities, deserving attention. This review outlines the role of six emerging batokines on BAT and their cross-talk with other organs, focusing on their physiological significance and diet-induced changes.


Assuntos
Tecido Adiposo Marrom , Fator A de Crescimento do Endotélio Vascular , Adulto , Humanos , Tecido Adiposo Marrom/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Adipócitos Marrons/metabolismo , Sistema Endócrino , Tecido Adiposo Branco/metabolismo , Obesidade/metabolismo , Termogênese , Metabolismo Energético
13.
Life Sci ; 327: 121858, 2023 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-37315839

RESUMO

Obesity and type 2 diabetes mellitus (T2DM) cause morphofunctional alterations in pancreatic islet alpha and beta cells. Therefore, we hypothesize that the new GLP-1/Glucagon receptor dual agonist cotadutide may benefit islet cell arrangement and function. Twelve-week-old C57BL/6 male mice were fed a control diet (C, 10 % kJ fat) or a high-fat diet (HF, 50 % kJ fat) for ten weeks. Then, the animals were divided into four groups for an additional 30 days and daily treated with subcutaneous cotadutide (30 nmol/kg) or vehicle: C, CC (control+cotadutide), HF, and HFC (high-fat+cotadutide). Cotadutide led to weight loss and reduced insulin resistance in the HFC group, increasing insulin receptor substrate 1 and solute carrier family 2 gene expressions in isolated islets. Also, cotadutide enhanced transcriptional factors related to islet cell transdifferentiation, decreasing aristaless-related homeobox and increasing the paired box 4 and 6, pancreatic and duodenal homeobox 1, v-maf musculoaponeurotic fibrosarcoma oncogene family protein A, neurogenin 3, and neurogenic differentiation 1. In addition, cotadutide improved the proliferating cell nuclear antigen, NK6 homeobox 1, B cell leukemia/lymphoma 2, but lessening caspase 3. Furthermore, cotadutide mitigated the endoplasmic reticulum (ER) stress-responsive genes, reducing transcription factor 4, DNA-damage-inducible transcript 3, and growth arrest and DNA-damage-inducible 45. In conclusion, our data demonstrated significant beneficial actions of cotadutide in DIO mice, such as weight loss, glycemic control, and insulin resistance improvement. In addition, cotadutide counteracted the pathological adaptive cellular arrangement of the pancreatic islet in obese mice, improving the markers of the transdifferentiating pathway, proliferation, apoptosis, and ER stress.


Assuntos
Diabetes Mellitus Tipo 2 , Resistência à Insulina , Células Secretoras de Insulina , Ilhotas Pancreáticas , Masculino , Camundongos , Animais , Camundongos Obesos , Diabetes Mellitus Tipo 2/metabolismo , Insulina/metabolismo , Camundongos Endogâmicos C57BL , Ilhotas Pancreáticas/metabolismo , Células Secretoras de Insulina/metabolismo , Dieta Hiperlipídica/efeitos adversos , Redução de Peso , DNA/metabolismo
14.
J Endocrinol ; 259(1)2023 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-37462522

RESUMO

Gut dysbiosis impairs nonshivering thermogenesis (NST) in obesity. The antiobesogenic effects of exercise training might involve the modulation of gut microbiota and its inflammatory signals to the brown adipose tissue (BAT). This study evaluated whether high-intensity interval training (HIIT) and moderate-intensity continuous training (MICT) prevent overweight through reduced gut-derived inflammatory signals to BAT in high-fat-fed mice. Sixty male C57BL/6 mice (3 months old) comprised six experimental groups: control (C) diet group, C diet + HIIT (C-HIIT) group, C diet + MICT (C-MICT) group, high-fat (HF) diet group, HF diet + HIIT (HF-HIIT) group, and HF diet + MICT (HF-MICT) group. The protocols lasted for 10 weeks. HIIT and MICT restored body mass, mitigated glucose intolerance, and prevented hyperinsulinemia in HF-trained groups. A chronic HF diet caused dysbiosis, but HIIT and MICT prevented gut dysbiosis and preserved tight junction (TJ) gene expression. HF-HIIT and HF-MICT groups exhibited a similar pattern of goblet cell distribution, agreeing with the decreased plasma lipopolysaccharide concentrations and interscapular BAT (iBAT) Lbp-Cd14-Tlr4 expression. The lowered Nlrp3 and Il1ß in the HF-HITT and HF-MICT groups complied with iBAT thermogenic capacity maintenance. This study shows reliable evidence that HIIT and MICT prevented overweight by restoring the diversity of the gut microbiota phyla and TJ gene expression, thereby reducing inflammatory signals to brown adipocytes with preserved thermogenic capacity. Both exercise modalities prevented overweight, but HIIT rescued Zo-1 and Jam-a gene expression, exerting more potent anti-inflammatory effects than MICT (reduced LPS concentrations), providing a sustained increase in thermogenesis with 78% less distance traveled.


Assuntos
Adipócitos Marrons , Sobrepeso , Camundongos , Masculino , Animais , Adipócitos Marrons/metabolismo , Disbiose/prevenção & controle , Camundongos Endogâmicos C57BL , Obesidade/prevenção & controle , Obesidade/metabolismo , Dieta Hiperlipídica/efeitos adversos
15.
J Mol Endocrinol ; 70(3)2023 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-36753306

RESUMO

Obesity, adipose tissue inflammation, and nonalcoholic fatty liver disease (NAFLD) are associated with insulin resistance and type 2 diabetes (T2D). Cotadutide is a dual agonist GLP-1/glucagon, currently in a preclinical study phase 2 that presents an anti-obesity effect. Diet-induced obese (DIO) C57BL/6 mice were treated for 4 weeks with cotadutide (30 nm/kg once a day at 14:00 h). The study focused on epididymal white adipose tissue (eWAT), liver (NAFLD), inflammation, lipid metabolism, AMP-activated protein kinase (AMPK)/mechanistic target of rapamycin (mTOR) pathways, and the endoplasmic reticulum (ER) stress. As a result, cotadutide controlled weight gain, glucose intolerance, and insulin resistance and showed beneficial effects on plasma markers in DIO mice (triacylglycerol, total cholesterol, alanine aminotransferase, and aspartate aminotransferase, leptin, adiponectin, monocyte chemoattractant protein-1, resistin, interleukin-6, tumor necrosis factor-alpha). Also, cotadutide lessened liver fat accumulation, eWAT proinflammatory markers, and ER stress. In addition, cotadutide improved lipid metabolism genes in eWAT, fatty acid synthase, peroxisome proliferator-activated receptor gamma and mitigates adipocyte hypertrophy and apoptosis. Furthermore, the effects of cotadutide were related to liver AMPK/mTOR pathway and ER stress. In conclusion, cotadutide induces weight loss and treats glucose intolerance and insulin resistance in DIO mice. In addition, cotadutide shows beneficial effects on liver lipid metabolism, mitigating steatosis, inflammation, and ER stress. Besides, in adipocytes, cotadutide decreases hypertrophy and reduces apoptosis. These actions rescuing the AMPK and mTOR pathway, improving lipid metabolism, and lessening NAFLD, inflammation, and ER stress in both eWAT and liver of DIO mice indicate cotadutide as a potentially new pharmacological treatment for T2D and associated obesity.


Assuntos
Diabetes Mellitus Tipo 2 , Intolerância à Glucose , Resistência à Insulina , Hepatopatia Gordurosa não Alcoólica , Animais , Camundongos , Hepatopatia Gordurosa não Alcoólica/tratamento farmacológico , Hepatopatia Gordurosa não Alcoólica/etiologia , Hepatopatia Gordurosa não Alcoólica/metabolismo , Camundongos Obesos , Intolerância à Glucose/metabolismo , Intolerância à Glucose/patologia , Diabetes Mellitus Tipo 2/metabolismo , Proteínas Quinases Ativadas por AMP/metabolismo , Camundongos Endogâmicos C57BL , Fígado/metabolismo , Obesidade/metabolismo , Tecido Adiposo/metabolismo , Inflamação/metabolismo , Hipertrofia/metabolismo , Hipertrofia/patologia , Serina-Treonina Quinases TOR/metabolismo , Dieta Hiperlipídica
16.
Clin Sci (Lond) ; 123(4): 259-70, 2012 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-22420611

RESUMO

The aim of the present study was to evaluate the effects of ST (rosuvastatin) and GZ (rosiglitazone) on IR (insulin resistance) and on liver as well as adipose tissue in mice fed on an HF (high-fat) diet. Our data show that treatment with ST resulted in a marked improvement in insulin sensitivity characterized by enhanced glucose clearance during the insulin tolerance test and a 70% decrease in the HOMA-IR (homoeostasis model assessment of insulin resistance) index level (P=0.0008). The ST-treated mice exhibited lower gains in BM (body mass; -8%; P<0.01) and visceral fat pad thickness (-60%; P<0.01) compared with the untreated HF group. In comparison with HF-diet-fed mice, HF+ST-treated mice showed a significant reduction in hepatomegaly and liver steatosis (-6%, P<0.05; and -21%, P<0.01 respectively). In HF+ST-treated mice, the hepatic TAG (triacylglycerol) levels were reduced by 58% compared with the HF group (P<0.01). In addition, the expression of SREBP-1c (sterol-regulatory-element-binding protein-1c) was decreased by 50% in the livers of HF+ST-treated mice (P<0.01) relative to the HF-diet-fed mice. The levels of resistin were lower in the HF+ST-treated group compared with the HF group (44% less, P< 0.01). In conclusion, we demonstrated that ST treatment improved insulin sensitivity and decreased liver steatosis in mice fed on an HF diet. Furthermore, ST reduced BM gains, improved the circulating levels of plasma cholesterol and TAG, and reduced hepatic TAG, which was concomitant with lower resistin levels.


Assuntos
Adiposidade/efeitos dos fármacos , Fígado Gorduroso/prevenção & controle , Fluorbenzenos/uso terapêutico , Resistência à Insulina/fisiologia , Pirimidinas/uso terapêutico , Sulfonamidas/uso terapêutico , Adipocinas/metabolismo , Tecido Adiposo/efeitos dos fármacos , Animais , Glicemia/metabolismo , Composição Corporal/efeitos dos fármacos , Dieta Hiperlipídica , Homeostase , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Camundongos , Hepatopatia Gordurosa não Alcoólica , Resistina/sangue , Rosiglitazona , Rosuvastatina Cálcica , Proteína de Ligação a Elemento Regulador de Esterol 1/biossíntese , Tiazolidinedionas/uso terapêutico , Triglicerídeos/sangue , Triglicerídeos/metabolismo
17.
Neuroendocrinology ; 96(4): 272-84, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22456428

RESUMO

A fatty diet during pregnancy in mouse dams causes metabolic abnormalities (similar to metabolic syndrome in humans) in the rodents' offspring. We tested the hypothesis that the offspring of dams fed a high-fat diet during pregnancy and lactation develop metabolic abnormalities and leptin resistance. Pregnant C57BL/6 mice (n = 20) were fed either standard chow (SC; 19% fat) or a high-fat diet (HF; 49% fat). After weaning, male offspring were divided into four groups, according to the diet of dams and offspring: SC(dams)/SC(offspring), SC/HF, HF/SC and HF/HF (n = 30/group). For a metabolic analysis, we evaluated body mass, fat mass depots, blood plasma and adipocyte structure at 12 weeks of age. To analyse leptin sensitivity, each group was divided into two groups (vehicle or leptin) to identify the feeding response and pSTAT3 expression after acute intracerebroventricular (ICV) treatment. The offspring of mothers fed a high-fat diet presented increased body mass and visceral fat, adipocyte hypertrophy and insulin resistance. This phenotype was not associated with central leptin resistance. Thus, maternal programming by HF predisposes offspring to metabolic abnormalities despite leptin sensitivity.


Assuntos
Adiposidade/fisiologia , Dieta Hiperlipídica/efeitos adversos , Resistência à Insulina/fisiologia , Leptina/sangue , Fenômenos Fisiológicos da Nutrição Materna/fisiologia , Efeitos Tardios da Exposição Pré-Natal/sangue , Animais , Glicemia/metabolismo , Dieta Hiperlipídica/métodos , Gorduras na Dieta/administração & dosagem , Gorduras na Dieta/efeitos adversos , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fenótipo , Gravidez
18.
Eur J Nutr ; 51(1): 89-96, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21499941

RESUMO

PURPOSE: To study the effects of a diet rich in salt and/or saturated fat on atrial natriuretic peptide (ANP)-granules, hypertension, renin expression, and cardiac structure in C57Bl/6 mice. METHODS: Young adult male mice were separated into four groups (n = 12) and fed one of the following for 9 weeks: standard chow/normal salt (SC-NS), high-fat chow/normal salt (HF-NS), standard chow/high salt (SC-HS) and high-fat chow/high salt (HF-HS). Alterations in the serum ANP, ultrastructural analysis of cardiomyocytes that produce ANP, structural analysis of the left ventricle, blood pressure, renin expression, glomerular filtration rate (GFR), feed efficiency, and lipid and glucose parameters were examined. RESULTS: The HF-NS diet showed a small increase in ANP production and left ventricular hypertrophy, increased food efficiency, and abnormal lipid and glucose parameters. The SC-HS diet showed a large increase in ANP granules in myocytes and corresponding elevation in ANP serum levels, left ventricular hypertrophy, hypertension, decrease in renin levels, and increase in GFR. The combination of the two diets (HF-HS) had an additive effect. CONCLUSION: The incorporation of a high-fat high-salt diet induced ultrastructural changes in cardiomyocytes, increased the production of ANP and increased its serum level, and reduced the amount of renin in the kidney.


Assuntos
Fator Natriurético Atrial/metabolismo , Dieta Hiperlipídica/efeitos adversos , Rim/metabolismo , Síndrome Metabólica/metabolismo , Miócitos Cardíacos/metabolismo , Renina/metabolismo , Cloreto de Sódio na Dieta/efeitos adversos , Animais , Fator Natriurético Atrial/sangue , Gorduras na Dieta/efeitos adversos , Regulação para Baixo , Taxa de Filtração Glomerular , Hipertensão/etiologia , Hipertrofia Ventricular Esquerda/etiologia , Rim/patologia , Rim/fisiopatologia , Masculino , Síndrome Metabólica/sangue , Síndrome Metabólica/patologia , Síndrome Metabólica/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Miócitos Cardíacos/ultraestrutura , Distribuição Aleatória , Vesículas Secretórias/metabolismo , Vesículas Secretórias/ultraestrutura , Óleo de Soja/efeitos adversos , Regulação para Cima
19.
Am J Orthod Dentofacial Orthop ; 142(3): 333-8, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22920699

RESUMO

INTRODUCTION: Asthma is a common systemic disease occurring in infancy and adolescence, time periods that could encompass orthodontic treatment. Asthma is an inflammatory disease; therefore, it might interfere with orthodontic tooth movement. The purpose of the study was to analyze the histomorphologic aspects of the periodontal ligament of asthmatic Wistar rats in the initial period of orthodontic movement. METHODS: Thirty-two Wistar rats were divided into 4 groups: 2 control groups consisting of rats without induced allergic asthma, and 2 experimental groups consisting of rats with induced allergic asthma. The animals of the first control and experimental groups did not receive orthodontic forces, whereas those in the second control and experimental groups were subjected to mesial movement of the maxillary left first molar for 3 days. The samples were prepared for histomorphometric analysis of the periodontal ligament. The area of the periodontal ligament was calculated as a function of root length in the cervical and apical regions of the distal face of the maxillary first molar mesial root. The Student t test and the Welch correlation test were applied to the data obtained. RESULTS: There was a statistically significant difference (P <0.05) between the control and experimental groups. An enhanced response to orthodontic force was observed in the asthmatic animals: the periodontal ligament was more compressed at the pressure area and more stretched in the traction area. CONCLUSIONS: Our findings indicate that experimental allergic asthma seems to exacerbate orthodontic movement in rats.


Assuntos
Asma/patologia , Análise do Estresse Dentário , Ligamento Periodontal/anatomia & histologia , Técnicas de Movimentação Dentária , Animais , Asma/imunologia , Fenômenos Biomecânicos , Brônquios/patologia , Força Compressiva , Contagem de Leucócitos , Dente Molar , Ratos , Ratos Wistar , Resistência à Tração , Raiz Dentária/anatomia & histologia
20.
Life Sci ; 288: 120188, 2022 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-34861287

RESUMO

The therapeutic arsenal for treating type 2 diabetes mellitus (T2DM) has been enriched recently with the inclusion of type 1 glucagon-like peptide (GLP-1). GLP-1 receptor agonists (RA) secondarily reduce appetite, decrease gastric emptying, and reduce body weight. This effect has been used to treat overweight/obesity, especially with comorbidities associated with T2DM. However, the first formulations and adverse effects gradually gave way to new formulations with fewer unpleasant effects and a more extended period of action (weekly subcutaneous administration and even oral administration), which improved the acceptance and adherence to the treatment. Therefore, titration of GLP-1RA should be done gradually. Furthermore, when side effects are consistent and intolerable after weeks/months of titration, a lower dose or a combination of antidiabetic therapies should be implemented, avoiding treatment interruption. The effort to produce increasingly powerful molecules with fewer side effects is the driving force behind the pharmaceutical industry. The unimolecular dual agonism GLP-1RA plus glucagon receptor agonism (GRA) represents an updated pharmacological indication for controlling blood glucose levels in treating T2DM and its comorbidities, showing better effects with less adverse impact than mono GLP-1RA. There are currently different proposals in this way by different laboratories. Nevertheless, the experimental results are promising and show that soon, we will have the contribution of new drugs for the treatment of T2DM.


Assuntos
Diabetes Mellitus Tipo 2/tratamento farmacológico , Peptídeo 1 Semelhante ao Glucagon/agonistas , Receptor do Peptídeo Semelhante ao Glucagon 1/agonistas , Glucagon/metabolismo , Hipoglicemiantes/uso terapêutico , Incretinas/uso terapêutico , Obesidade/tratamento farmacológico , Animais , Diabetes Mellitus Tipo 2/patologia , Humanos , Obesidade/fisiopatologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA