Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 139
Filtrar
1.
Nat Commun ; 13(1): 246, 2022 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-35017472

RESUMO

The molecular targets and mechanisms of propolis ameliorating metabolic syndrome are not fully understood. Here, we report that Brazilian green propolis reduces fasting blood glucose levels in obese mice by disrupting the formation of CREB/CRTC2 transcriptional complex, a key regulator of hepatic gluconeogenesis. Using a mammalian two-hybrid system based on CREB-CRTC2, we identify artepillin C (APC) from propolis as an inhibitor of CREB-CRTC2 interaction. Without apparent toxicity, APC protects mice from high fat diet-induced obesity, decreases fasting glucose levels, enhances insulin sensitivity and reduces lipid levels in the serum and liver by suppressing CREB/CRTC2-mediated both gluconeogenic and SREBP transcriptions. To develop more potential drugs from APC, we designed and found a novel compound, A57 that exhibits higher inhibitory activity on CREB-CRTC2 association and better capability of improving insulin sensitivity in obese animals, as compared with APC. In this work, our results indicate that CREB/CRTC2 is a suitable target for developing anti-metabolic syndrome drugs.


Assuntos
Proteína de Ligação a CREB/metabolismo , Sistemas de Liberação de Medicamentos , Síndrome Metabólica/metabolismo , Camundongos Obesos/metabolismo , Própole/metabolismo , Fatores de Transcrição/metabolismo , Animais , Glicemia , Brasil , Proteína de Ligação a CREB/genética , Desenvolvimento de Medicamentos , Descoberta de Drogas , Gluconeogênese , Resistência à Insulina , Fígado/metabolismo , Síndrome Metabólica/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Camundongos Obesos/genética , Obesidade/metabolismo , Própole/genética , Fatores de Transcrição/genética
2.
Artigo em Inglês | MEDLINE | ID: mdl-34619367

RESUMO

Eicosapentaenoic acid (EPA) ethyl esters are of interest given their clinical approval for lowering circulating triglycerides and cardiometabolic disease risk. EPA ethyl esters prevent metabolic complications driven by a high fat diet in male mice; however, their impact on female mice is less studied. Herein, we first investigated how EPA influences the metabolic profile of female C57BL/6J mice consuming a high fat diet. EPA lowered murine fat mass accumulation, potentially through increased biosynthesis of 8-hydroxyeicosapentaenoic acid (HEPE), as revealed by mass spectrometry and cell culture studies. EPA also reversed the effects of a high fat diet on circulating levels of insulin, glucose, and select inflammatory/metabolic markers. Next, we studied if the improved metabolic profile of obese mice consuming EPA was associated with a reduction in the abundance of key gut Gram-negative bacteria that contribute toward impaired glucose metabolism. Using fecal 16S-ribosomal RNA gene sequencing, we found EPA restructured the gut microbiota in a time-dependent manner but did not lower the levels of key Gram-negative bacteria. Interestingly, EPA robustly increased the abundance of the Gram-negative Akkermansia muciniphila, which controls glucose homeostasis. Finally, predictive functional profiling of microbial communities revealed EPA-mediated reversal of high fat diet-associated changes in a wide range of genes related to pathways such as Th-17 cell differentiation and PI3K-Akt signaling. Collectively, these results show that EPA ethyl esters prevent some of the deleterious effects of a high fat diet in female mice, which may be mediated mechanistically through 8-HEPE and the upregulation of intestinal Akkermansia muciniphila.


Assuntos
Ácido Eicosapentaenoico/farmacologia , Microbioma Gastrointestinal/genética , Ácidos Hidroxieicosatetraenoicos/biossíntese , Akkermansia/genética , Akkermansia/crescimento & desenvolvimento , Animais , Fatores de Risco Cardiometabólico , Diferenciação Celular/efeitos dos fármacos , Dieta Hiperlipídica/efeitos adversos , Ácido Eicosapentaenoico/metabolismo , Feminino , Microbioma Gastrointestinal/efeitos dos fármacos , Glucose/metabolismo , Humanos , Ácidos Hidroxieicosatetraenoicos/sangue , Masculino , Espectrometria de Massas , Camundongos , Camundongos Obesos/genética , Camundongos Obesos/microbiologia , Fosfatidilinositol 3-Quinases/genética , Proteínas Proto-Oncogênicas c-akt/genética , RNA Ribossômico 16S/genética , Caracteres Sexuais , Células Th17/metabolismo , Triglicerídeos/sangue
3.
Artigo em Inglês | MEDLINE | ID: mdl-34606993

RESUMO

BACKGROUND: Obesity and type 2 diabetes are two interrelated metabolic disorders characterized by insulin resistance and a mild chronic inflammatory state. We previously observed that leptin (ob/ob) and leptin receptor (db/db) knockout mice display a distinct inflammatory tone in the liver and adipose tissue. The present study aimed at investigating whether alterations in these tissues of the molecules belonging to the endocannabinoidome (eCBome), an extension of the endocannabinoid (eCB) signaling system, whose functions are important in the context of metabolic disorders and inflammation, could reflect their different inflammatory phenotypes. RESULTS: The basal eCBome lipid and gene expression profiles, measured by targeted lipidomics and qPCR transcriptomics, respectively, in the liver and subcutaneous or visceral adipose tissues, highlighted a differentially altered eCBome tone, which may explain the impaired hepatic function and more pronounced liver inflammation remarked in the ob/ob mice, as well as the more pronounced inflammatory state observed in the subcutaneous adipose tissue of db/db mice. In particular, the levels of linoleic acid-derived endocannabinoid-like molecules, of one of their 12-lipoxygenase metabolites and of Trpv2 expression, were always altered in tissues exhibiting the highest inflammation. Correlation studies suggested the possible interactions with some gut microbiota bacterial taxa, whose respective absolute abundances were significantly different between ob/ob and the db/db mice. CONCLUSIONS: The present findings emphasize the possibility that bioactive lipids and the respective receptors and enzymes belonging to the eCBome may sustain the tissue-dependent inflammatory state that characterizes obesity and diabetes, possibly in relation with gut microbiome alterations.


Assuntos
Canais de Cálcio/genética , Diabetes Mellitus Tipo 2/genética , Leptina/genética , Obesidade/genética , Receptores para Leptina/genética , Canais de Cátion TRPV/genética , Tecido Adiposo/metabolismo , Animais , Araquidonato 12-Lipoxigenase/genética , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patologia , Modelos Animais de Doenças , Endocanabinoides/genética , Microbioma Gastrointestinal/genética , Regulação da Expressão Gênica/genética , Humanos , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , Camundongos , Camundongos Endogâmicos NOD/genética , Camundongos Endogâmicos NOD/microbiologia , Camundongos Obesos/genética , Camundongos Obesos/microbiologia , Obesidade/metabolismo , Obesidade/patologia , Transcriptoma/genética
4.
Cells ; 10(12)2021 12 09.
Artigo em Inglês | MEDLINE | ID: mdl-34943988

RESUMO

Obesity is associated with chronic low-grade inflammation. The benefits of exercise are partly attributed to its anti-inflammatory effect, but whether exercise can regulate NLRP3 inflammasome activation in obese adipose tissue remains unknown. Meteorin-like (METRNL), a recently discovered myokine, has been implicated in mediating the effect of exercise on metabolism. Herein, we examined the effect of exercise and METRNL on NLRP3 inflammasome activation. High-fat diet (HFD)-induced obese mice were subjected to treadmill exercise for 8 weeks. A subgroup of HFD mice was switched to normal chow with the exercise intervention. Exercise and diet attenuated weight gain, fat accumulation, and insulin resistance in obese mice. In addition, exercise downregulated gene and protein levels of inflammasome markers, including NLRP3 and caspase-1, in adipose tissue. In isolated bone marrow-derived macrophages, activation of NLRP3 inflammasome was suppressed in the exercise group, as confirmed by the downregulation of IL-1ß and IL-18. Exercise significantly enhanced the expression of METRNL in various muscle depots, and further in vitro analysis revealed that recombinant METRNL treatment inhibited IL-1ß secretion in macrophages. In conclusion, exercise exerts its anti-inflammatory action by suppressing adipose tissue NLRP3 inflammasome, and this is, in part, associated with METRNL induction in muscle and its anti-inflammatory effects in macrophages.


Assuntos
Inflamação/terapia , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Fatores de Crescimento Neural/genética , Obesidade/terapia , Animais , Dieta Hiperlipídica/efeitos adversos , Terapia por Exercício , Humanos , Inflamassomos/genética , Inflamassomos/metabolismo , Inflamação/genética , Inflamação/patologia , Resistência à Insulina/genética , Macrófagos/metabolismo , Macrófagos/patologia , Camundongos , Camundongos Obesos/genética , Obesidade/genética , Obesidade/patologia , Condicionamento Físico Animal
5.
J Cell Physiol ; 236(1): 132-145, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32468615

RESUMO

Pericardial adipose tissue (PAT), a visceral fat depot enveloping the heart, is an active endocrine organ and a source of free fatty acids and inflammatory cytokines. As in other fat adult tissues, PAT contains a population of adipose stem cells; however, whether these cells and/or their environment play a role in physiopathology is unknown. We analyzed several stem cell-related properties of pericardial adipose stem cells (PSCs) isolated from obese and ex-obese mice. We also performed RNA-sequencing to profile the transcriptional landscape of PSCs isolated from the different diet regimens. Finally, we tested whether these alterations impacted on the properties of cardiac mesoangioblasts isolated from the same mice. We found functional differences between PSCs depending on their source: specifically, PSCs from obese PSC (oPSC) and ex-obese PSC (dPSC) mice showed alterations in apoptosis and migratory capacity when compared with lean, control PSCs, with increased apoptosis in oPSCs and blunted migratory capacity in oPSCs and dPSCs. This was accompanied by different gene expression profiles across the cell types, where we identified some genes altered in obese conditions, such as BMP endothelial cell precursor-derived regulator (BMPER), an important regulator of BMP-related signaling pathways for endothelial cell function. The importance of BMPER in PSCs was confirmed by loss- and gain-of-function studies. Finally, we found an altered production of BMPER and some important chemokines in cardiac mesoangioblasts in obese conditions. Our findings point to BMPER as a potential new regulator of PSC function and suggest that its dysregulation could be associated with obesity and may impact on cardiac cells.


Assuntos
Adipócitos/metabolismo , Proteínas de Transporte/metabolismo , Obesidade/genética , Obesidade/metabolismo , Pericárdio/metabolismo , Células-Tronco/metabolismo , Regulação para Cima/genética , Tecido Adiposo/metabolismo , Animais , Apoptose/genética , Diferenciação Celular/genética , Células Cultivadas , Dieta Hiperlipídica/efeitos adversos , Células Endoteliais/metabolismo , Ácidos Graxos não Esterificados/metabolismo , Feminino , Gordura Intra-Abdominal/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos/genética , Camundongos Obesos/metabolismo , Transdução de Sinais/genética
6.
Proc Natl Acad Sci U S A ; 117(46): 29025-29034, 2020 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-33144503

RESUMO

As a physiological regulator of bile acid homeostasis, FGF19 is also a potent insulin sensitizer capable of normalizing plasma glucose concentration, improving lipid profile, ameliorating fatty liver disease, and causing weight loss in both diabetic and diet-induced obesity mice. There is therefore a major interest in developing FGF19 as a therapeutic agent for treating type 2 diabetes and cholestatic liver disease. However, the known tumorigenic risk associated with prolonged FGF19 administration is a major hurdle in realizing its clinical potential. Here, we show that nonmitogenic FGF19 variants that retain the full beneficial glucose-lowering and bile acid regulatory activities of WT FGF19 (FGF19WT) can be engineered by diminishing FGF19's ability to induce dimerization of its cognate FGF receptors (FGFR). As proof of principle, we generated three such variants, each with a partial defect in binding affinity to FGFR (FGF19ΔFGFR) and its coreceptors, i.e., ßklotho (FGF19ΔKLB) or heparan sulfate (FGF19ΔHBS). Pharmacological assays in WT and db/db mice confirmed that these variants incur a dramatic loss in mitogenic activity, yet are indistinguishable from FGF19WT in eliciting glycemic control and regulating bile acid synthesis. This approach provides a robust framework for the development of safer and more efficacious FGF19 analogs.


Assuntos
Fatores de Crescimento de Fibroblastos/genética , Fatores de Crescimento de Fibroblastos/metabolismo , Mitógenos/metabolismo , Animais , Ácidos e Sais Biliares/metabolismo , Diabetes Mellitus Tipo 2 , Dimerização , Modelos Animais de Doenças , Fatores de Crescimento de Fibroblastos/química , Fatores de Crescimento de Fibroblastos/farmacologia , Engenharia Genética , Glucose/metabolismo , Células Hep G2 , Homeostase , Humanos , Proteínas Klotho , Masculino , Proteínas de Membrana/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Obesos/genética , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo
7.
Int J Mol Sci ; 21(15)2020 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-32752112

RESUMO

The expansion of adipose tissue mass is the primary characteristic of the process of becoming obesity, which causes chronic adipose inflammation and is closely associated with type 2 diabetes mellitus (T2DM). Adipocyte hypertrophy restricts oxygen availability, leading to microenvironmental hypoxia and adipose dysfunction. This study aimed at investigating the effects of oxygenated water (OW) on adipocyte differentiation (adipogenesis) and the metabolic function of mature adipocytes. The effects of OW on adipogenesis and the metabolic function of mature adipocytes were examined. Meanwhile, the in vivo metabolic effects of long-term OW consumption on diet-induced obesity (DIO) mice were investigated. OW inhibited adipogenesis and lipid accumulation through down-regulating critical adipogenic transcription factors and lipogenic enzymes. While body weight, blood and adipose parameters were not significantly improved by long-term OW consumption, transient circulatory triglyceride-lowering and glucose tolerance-improving effects were identified. Notably, hepatic lipid contents were significantly reduced, indicating that the DIO-induced hepatic steatosis was attenuated, despite no improvements in fibrosis and lipid contents in adipose tissue being observed in the OW-drinking DIO mice. The study provides evidence regarding OW's effects on adipogenesis and mature adipocytes, and the corresponding molecular mechanisms. OW exhibits transient triglyceride-lowering and glucose tolerance-improving activity as well as hepatic steatosis-attenuating functions.


Assuntos
Adipogenia/efeitos dos fármacos , Fígado Gorduroso/tratamento farmacológico , Lipogênese/efeitos dos fármacos , Água/metabolismo , Células 3T3-L1 , Adipócitos/efeitos dos fármacos , Tecido Adiposo/efeitos dos fármacos , Tecido Adiposo/metabolismo , Animais , Diferenciação Celular/efeitos dos fármacos , Dieta Hiperlipídica/efeitos adversos , Fígado Gorduroso/metabolismo , Fígado Gorduroso/patologia , Humanos , Camundongos , Camundongos Obesos/genética , Camundongos Obesos/metabolismo , Obesidade/genética , Obesidade/metabolismo , Obesidade/patologia , Obesidade/prevenção & controle , Oxigênio/metabolismo , Água/farmacologia
8.
Nutrients ; 12(6)2020 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-32545529

RESUMO

: The molecular adaptations that underpin body composition changes and health benefits of intermittent fasting (IF) and high-intensity interval training (HIIT) are unclear. The present study investigated these adaptations within the hypothalamus, white adipose and skeletal muscle tissue following 12 weeks of IF and/or HIIT in diet-induced obese mice. Mice (C57BL/6, 8-week-old, males/females) were fed high-fat (59%) and sugar (30%) water (HF/S) for 12 weeks followed by an additional 12 weeks of HF/S plus either IF, HIIT, combination (IF+HIIT) or HF/S only control (CON). Tissues were harvested at 12 and 24 weeks and analysed for various molecular markers. Hypothalamic NPY expression was significantly lower following IF+HIIT compared to CON in females. In adipose tissue, leptin expression was significantly lower following IF and IF+HIIT compared to CON in males and females. Males demonstrated increased markers of fat oxidation (HADH, FABP4) following IF+HIIT, whereas females demonstrated reduced markers of adipocyte differentiation/storage (CIDEC and FOXO1) following IF and/or IF+HIIT. In muscle, SIRT1, UCP3, PGC1α, and AS160 expression was significantly lower following IF compared to CON in males and/or females. This investigation suggests that males and females undertaking IF and HIIT may prevent weight gain via different mechanisms within the same tissue.


Assuntos
Tecido Adiposo Branco/metabolismo , Jejum , Treinamento Intervalado de Alta Intensidade/métodos , Hipotálamo/metabolismo , Músculo Esquelético/metabolismo , Adaptação Fisiológica/genética , Animais , Composição Corporal , Dieta Hiperlipídica/efeitos adversos , Feminino , Leptina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos/genética , Camundongos Obesos/metabolismo , Condicionamento Físico Animal/métodos , RNA Mensageiro/genética , Caracteres Sexuais , Aumento de Peso
9.
Obesity (Silver Spring) ; 28(6): 1075-1085, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32348021

RESUMO

OBJECTIVE: Identifying novel approaches to combat obesity is important to improve health span. It was hypothesized that methionine restriction (MR) will induce weight loss in obese mice by reducing adipose tissue mass caused by increased energy expenditure and reprogramming of adipose tissue homeostasis. The roles of adiponectin (ADIPOQ) and fibroblast growth factor 21 (FGF21) during weight loss in MR mice were also tested. METHODS: Diet-induced obese (DIO) male C57BL/6J (wild type), Adipoq-deficient (Adipoq knockout [KO]), Fgf21-KO, and Adipoq-Fgf21 double-KO mice were used. Following a switch to high-fat control (DIO-CF, 60% fat/0.86% methionine) or MR (DIO-MR, 60% fat/0.12% methionine) diet, physiological parameters were measured, and inguinal and perigonadal adipose tissues were examined. RESULTS: Obese mice subjected to MR showed loss of body weight and adiposity, increased energy expenditure, and improved glucose tolerance that were independent of the actions of ADIPOQ and FGF21. MR induced reduction of circulating lipids, glucose, insulin, leptin, and insulin like growth factor 1 and increased ß-hydroxybutyrate, ADIPOQ, and FGF21 concentrations. In fat, MR upregulated protein levels of adipose triglyceride lipase, apoptosis-inducing factor, lysosomal-associated membrane proteins 1 and 2, autophagy-related protein 5, beclin-1, and light chain 3B I and II. CONCLUSIONS: MR reduction of adipose tissue mass in obese mice is associated with elevated lipolysis, apoptosis, and autophagy and occurs independently of the actions of ADIPOQ and FGF21.


Assuntos
Adiponectina/metabolismo , Adiposidade/fisiologia , Fatores de Crescimento de Fibroblastos/metabolismo , Metionina/metabolismo , Camundongos Obesos/genética , Redução de Peso/fisiologia , Animais , Masculino , Camundongos
10.
Oncol Rep ; 42(6): 2797-2805, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31638239

RESUMO

Sodium­glucose cotransporter 2 inhibitors were developed for the treatment of diabetes mellitus. Although recent studies have indicated that sodium­glucose cotransporter 2 inhibitors have suppressive effects on several types of cancer, their effects against colorectal cancer remain unknown. The purpose of the present study was to investigate the effects of tofogliflozin, a sodium­glucose cotransporter 2 inhibitor, on the development of colorectal cancer in diabetic and obese mice. The direct effects of tofogliflozin on the proliferation of colorectal cancer cells were also evaluated. C57BL/KsJ­db/db mice were injected with azoxymethane to induce colorectal pre­malignancy and they received drinking water with or without tofogliflozin. At the end of the study, administration of tofogliflozin was revealed to significantly suppress the development of colorectal neoplastic lesions and ß­catenin accumulated crypts. In the tofogliflozin­treated mice, the levels of blood glucose and serum TNF­α, as well as mRNA expression of the pro­inflammatory markers in the white adipose tissue, were reduced. Furthermore, macrophage infiltrations in the white adipose tissues were also reduced significantly. The proliferation of the sodium­glucose cotransporter 2­expressing human colorectal cancer cells was not altered by tofogliflozin. These results indicated that tofogliflozin ameliorated chronic inflammation and hyperglycemic condition leading to prevention of colorectal tumorigenesis in a diabetes­ and obesity­related colorectal cancer model.


Assuntos
Compostos Benzidrílicos/farmacologia , Carcinogênese/efeitos dos fármacos , Neoplasias Colorretais/tratamento farmacológico , Glucosídeos/farmacologia , Transportador 2 de Glucose-Sódio/genética , Animais , Azoximetano/toxicidade , Glicemia/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Neoplasias Colorretais/induzido quimicamente , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Diabetes Mellitus Tipo 2/tratamento farmacológico , Diabetes Mellitus Tipo 2/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Hipoglicemiantes/farmacologia , Camundongos , Camundongos Endogâmicos NOD/genética , Camundongos Obesos/genética , Obesidade/tratamento farmacológico , Obesidade/genética , Inibidores do Transportador 2 de Sódio-Glicose/farmacologia , Fator de Necrose Tumoral alfa/genética
11.
Sci Rep ; 9(1): 13427, 2019 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-31530849

RESUMO

Obesity and associated metabolic comorbidities represent a growing public health problem. In this study, we demonstrate the use of a newly created fusion gene of exendin-4 and α1-antitrypsin to control obesity and obesity-associated metabolic disorders including insulin resistance, fatty liver and hyperglycemia. The fusion gene encodes a protein with exendin-4 peptide placed at the N-terminus of human α-1 antitrypsin, and is named EAT. Hydrodynamic transfer of the EAT gene to mice prevents high-fat diet-induced obesity, insulin resistance and fatty liver development. In diet-induced obese mice, expression of EAT gene induces weight loss, improves glucose homeostasis, and attenuates hepatic steatosis. In ob/ob mice, EAT gene transfer suppresses body weight gain, maintains metabolic homeostasis, and completely blocks fatty liver development. Six-month overexpression of the EAT fusion gene in healthy mice does not lead to any detectable toxicity. Mechanistic study reveals that the resulting metabolic benefits are achieved by a reduced food take and down-regulation of transcription of pivotal genes responsible for lipogenesis and lipid droplet formation in the liver and chronic inflammation in visceral fat. These results validate the feasibility of gene therapy in preventing and restoring metabolic homeostasis under diverse pathologic conditions, and provide evidence in support of a new strategy to control obesity and related metabolic diseases.


Assuntos
Exenatida/genética , Obesidade/terapia , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/genética , alfa 1-Antitripsina/genética , Adiposidade/genética , Animais , Fármacos Antiobesidade/administração & dosagem , Fármacos Antiobesidade/farmacologia , Dieta Hiperlipídica/efeitos adversos , Regulação da Expressão Gênica , Vetores Genéticos , Glucose/metabolismo , Resistência à Insulina/genética , Leptina/genética , Masculino , Camundongos Endogâmicos C57BL , Camundongos Obesos/genética , Hepatopatia Gordurosa não Alcoólica/etiologia , Hepatopatia Gordurosa não Alcoólica/genética , Hepatopatia Gordurosa não Alcoólica/prevenção & controle , Engenharia de Proteínas/métodos , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/farmacologia , Aumento de Peso/genética
12.
PLoS One ; 13(7): e0200779, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30036374

RESUMO

BACKGROUND: Achieving weight loss is the cornerstone of the treatment of the metabolic consequences of obesity, in particular of glucose intolerance. OBJECTIVE: To determine whether improvement in glucose control depends on dietary macronutrient composition of the diet at identical weight loss. MATERIALS AND METHODS: Twenty-two weeks old diet-induced obese C57BL/6 mice lost weight through caloric restriction on normal chow (R-NC) or high fat diet (R-HF). Control mice were fed normal chow (LEAN) or high fat diet (OBESE) ad libitum. Body weight and composition were assessed after 8 weeks of dietary intervention. Glucose homeostasis was evaluated by intraperitoneal glucose tolerance tests (IPGTT). Epididymal white adipose (eWAT) and hepatic tissues were analyzed by immunohistochemistry and RT-qPCR. RESULTS: By 30 weeks of age, the body weight of the mice on R-NC (31.6±1.7g, mean±SEM) and R-HF (32.3±0.9g) was similar to LEAN mice (31.9±1.4g), while OBESE mice weighed 51.7±2.4g. Glucose tolerance in R-NC was better than in LEAN mice (69% AUC IPGTT, P 0.0168) whereas R-HF mice remained significantly less glucose tolerant (125% AUC IPGTT, P 0.0279 vs LEAN), despite identical weight loss. The eWAT pads and adipocyte size were similar in LEAN and R-NC mice, while the eWAT pad size of R-HF was 180% of R-NC (P < 0.0001) and the average adipocyte size of R-HF mice was 134% of R-NC fed mice (P 0.0285). No LEAN or R-NC mice had hepatic steatosis, in contrast to 28.6% of R-HF mice. Compared to OBESE mice, inflammatory markers were lower in eWAT and liver tissue of R-NC, but not in R-HF mice. Measures of visceral adiposity correlated well with glucose tolerance parameters. CONCLUSIONS: In mice, caloric restriction on a normal chow diet improved glucose tolerance significantly more when identical weight loss was achieved on a high fat diet.


Assuntos
Glicemia/metabolismo , Restrição Calórica , Dieta Hiperlipídica , Camundongos Obesos/genética , Nutrientes/química , Adipócitos/metabolismo , Tecido Adiposo/metabolismo , Tecido Adiposo Branco/metabolismo , Adiposidade , Animais , Composição Corporal , Peso Corporal , Calorimetria , Gorduras na Dieta/metabolismo , Ingestão de Alimentos , Fígado Gorduroso/metabolismo , Glucose/química , Intolerância à Glucose/metabolismo , Homeostase , Inflamação , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/metabolismo , Redução de Peso
13.
In Vivo ; 32(4): 813-817, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29936463

RESUMO

BACKGROUND/AIM: Adipocyte gene expression is altered in obese individuals through multiple metabolic and biochemical pathways. In this study, we aimed to examine the expression of resistin (Retn), amylin (Iapp), and dopamine receptor domain 5 (Drd5) genes previously suggested to contribute to the pathogenesis of obesity, albeit controversially. We also aimed to determine the effects on short and long-term mRNA levels of these genes in obese mice, induced with high-fat diet (HFD). MATERIALS AND METHODS: Two obesity models were created in our study: group T1 (20 mice) was fed with HFD (60% fat) for 3 months, and group T2 (20 mice) was fed with HFD (60% fat) for 6 months. The control group T0 (20 mice) was fed with a diet of 10% kcal fat supplement for 6 months. At the end of the experiment, their adipose tissues were dissected surgically. Tissue samples of each group were pooled for RNA isolation, cDNA synthesis was carried out and the mRNA levels were examined by quantitative real-time polymerase chain reaction. Serum resistin levels were measured using multiplex bead (luminex) technology for validation. RESULTS: In T2 mice, the mRNA expression of Retn showed a moderate up-regulation (fold change=8.32; p=0.0019) in the adipose tissues. Iapp expression was also significantly up-regulated (fold change=9.78; p=0.012). Moreover, a 6.36-fold up-regulation for Drd5 was observed in the adipose tissues of T2 mice (p<0.001). At the same time, serum levels of resistin were found to be high in T1 and T2 mice compared to the control group (p<0.001 and p=0.024, respectively). CONCLUSION: Our study demonstrated that the mRNA levels of the genetic markers considered to play a role in adipogenesis were different in short- and long-term obesity models formed in C57BL/6J mice using HFD.


Assuntos
Polipeptídeo Amiloide das Ilhotas Pancreáticas/genética , Camundongos Obesos/genética , Obesidade/genética , Receptores de Dopamina D5/genética , Resistina/genética , Adipócitos/metabolismo , Tecido Adiposo/metabolismo , Tecido Adiposo/patologia , Animais , Peso Corporal/genética , Dieta Hiperlipídica/efeitos adversos , Regulação da Expressão Gênica/genética , Fígado/metabolismo , Camundongos , Obesidade/patologia , RNA Mensageiro/genética
14.
Cell Physiol Biochem ; 42(2): 603-614, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28595178

RESUMO

AIMS: To determine whether irisin could improve endothelial dysfunction by regulating heme oxygenase-1(HO-1)/adiponectin axis in perivascular adipose tissue (PVAT) in obesity. METHODS: Male C57BL/6 mice were fed with a high-fat diet (HFD) with or without irisin treatment. Endothelium-dependent vasorelaxation of the thoracic aorta with or without PVAT (PVAT+ or PVAT-) was determined. Western blot was employed to determine the levels of HO-1 and adiponectin in PVAT. UCP-1, Cidea, and TNF-α gene expression in PVAT were tested by real-time PCR. RESULTS: The presence of PVAT significantly impaired endothelial function in the HFD mice. Treatment of HFD mice with irisin significantly restored this impairment and improved endothelial function in vivo or ex vivo. Incubated aortic rings (PVAT-) with PVAT-derived conditioned medium (CM) from HFD mice impaired endothelial function in control mice. This impairment was prevented by incubating the aortic rings (PVAT-) from HFD mice with PVAT-derived CM from irisin. However, the beneficial effects were partly attenuated in the presence of HO-1 inhibitor and adiponectin receptor blocking peptide. Treatment of HFD mice with irisin significantly increased NO production, protein levels of HO-1 and adiponectin, mRNA expressions of UCP-1 and Cidea, and decreased superoxide production and TNF-α expression in PVAT. CONCLUSION: Irisin improved endothelial function by modulating HO-1/ adiponectin axis in PVAT in HFD-induced obese mice. These findings suggest that regulating PVAT function may be a potential mechanism by which irisin improves endothelial function in obesity.


Assuntos
Adiponectina/genética , Endotélio Vascular/metabolismo , Fibronectinas/genética , Heme Oxigenase-1/genética , Obesidade/genética , Tecido Adiposo/metabolismo , Tecido Adiposo/patologia , Animais , Aorta Torácica/metabolismo , Dieta Hiperlipídica , Endotélio Vascular/patologia , Regulação da Expressão Gênica , Humanos , Camundongos , Camundongos Obesos/genética , Obesidade/metabolismo , Obesidade/patologia , Receptores de Adiponectina/metabolismo , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
15.
Physiol Rep ; 5(9)2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28483861

RESUMO

C57Bl/6J mice are the gold standard animal model of diet-induced obesity. These animals become obese with higher adiposity, blood fasting glucose, triglycerides, and total cholesterol when fed a high-fat diet (HFD). Conversely, the FVB/N mouse line is thought to be resistant to diet-induced obesity, with low or no weight gain and adiposity in response to a HFD In this study, we investigated whether FVB/N mice are resistant or susceptible to metabolic disorder that is promoted by a HFD Biometric parameters and blood chemistry were analyzed in C57Bl/6J and FVB/N mice that were fed a chow diet or HFD Glucose and insulin sensitivity were assessed by performing the glucose tolerance test and measuring serum insulin/glucose and homeostasis model assessment-insulin resistance. Metabolism-related gene expression was investigated by real-time reverse transcription polymerase chain reaction. Adipocyte morphology and liver steatosis were evaluated using standard histology. FVB/N mice had higher adiposity than C57Bl/6J mice that were fed a chow diet and were glucose intolerant. FVB/N mice that were fed a HFD presented higher insulin resistance and greater liver steatosis. Epididymal white adipose tissue exhibited severe inflammation in FVB/N mice that were fed a HFD The FVB/N mouse strain is suitable for studies of diet-induced obesity, and the apparent lack of a HFD-induced response may reveal several strain-specific events that are triggered by a HFD Further studies of the FVB/N background may shed light on the complex multifactorial symptoms of obesity and metabolic syndrome.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Camundongos Obesos/metabolismo , Obesidade/etiologia , Adiposidade , Animais , Glicemia/metabolismo , Modelos Animais de Doenças , Patrimônio Genético , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos/genética , Obesidade/genética
16.
Diabetes ; 66(6): 1535-1547, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28325853

RESUMO

Although the importance of macrophages in nonalcoholic fatty liver disease (NAFLD) and type 2 diabetes mellitus (T2DM) has been recognized, how macrophages affect hepatocytes remains elusive. Mineralocorticoid receptor (MR) has been implicated to play important roles in NAFLD and T2DM. However, cellular and molecular mechanisms are largely unknown. We report that myeloid MR knockout (MRKO) improves glucose intolerance, insulin resistance, and hepatic steatosis in obese mice. Estrogen signaling is sufficient and necessary for such improvements. Hepatic gene and protein expression suggests that MRKO reduces hepatic lipogenesis and lipid storage. In the presence of estrogen, MRKO in macrophages decreases lipid accumulation and increases insulin sensitivity of hepatocytes through hepatocyte growth factor (HGF)/Met signaling. MR directly regulates estrogen receptor 1 (Esr1 [encoding ERα]) in macrophages. Knockdown of hepatic Met eliminates the beneficial effects of MRKO in female obese mice. These findings identify a novel MR/ERα/HGF/Met pathway that conveys metabolic signaling from macrophages to hepatocytes in hepatic steatosis and insulin resistance and provide potential new therapeutic strategies for NAFLD and T2DM.


Assuntos
Receptor alfa de Estrogênio/genética , Fator de Crescimento de Hepatócito/metabolismo , Hepatócitos/metabolismo , Resistência à Insulina/genética , Macrófagos/metabolismo , Camundongos Obesos/genética , Hepatopatia Gordurosa não Alcoólica/genética , Proteínas Proto-Oncogênicas c-met/genética , Receptores de Mineralocorticoides/genética , Animais , Células Cultivadas , Imunoprecipitação da Cromatina , Diabetes Mellitus Tipo 2/metabolismo , Ensaio de Imunoadsorção Enzimática , Receptor alfa de Estrogênio/metabolismo , Ácidos Graxos não Esterificados/metabolismo , Feminino , Técnicas de Silenciamento de Genes , Teste de Tolerância a Glucose , Immunoblotting , Insulina/metabolismo , Lipogênese , Masculino , Camundongos , Camundongos Knockout , Camundongos Obesos/metabolismo , Hepatopatia Gordurosa não Alcoólica/metabolismo , Proteínas Proto-Oncogênicas c-met/metabolismo , Células RAW 264.7 , Transdução de Sinais , Triglicerídeos/metabolismo
17.
Pharmacol Rep ; 69(2): 229-234, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28095353

RESUMO

BACKGROUND: Obesity is a major health problem worldwide, for which preventive and therapeutic means are still needed. Alpha-amylase is a digestive enzyme whose inhibition has been targeted as a potential anti-obesity strategy. However, alpha-amylase gene expression has not been particularly attended to, and in contrast with pancreatic and salivary amylases, fewer studies have focused on liver alpha-amylase. The present study aimed at investigating the expression of alpha-amylase gene in obese and normal mice at RNA and protein level as well as acarbose effect on this gene expression in hepatocyte cell culture. METHODS: Control and case groups were fed by normal mouse pellet and high-fat diet respectively, during 8 weeks. After this period, serum biochemical parameters including glucose, cholesterol, triglycerides, AST, ALT and alpha-amylase were assayed. Liver alpha-amylase gene was analyzed by real time PCR, and liver enzyme was assayed with Bernfeld and ELISA methods Hepatocyte cell culture derived from both group were also treated by acarbose and alpha-amylase activity and gene expression was analyzed by above mentioned methods. RESULTS: All biochemical factors showed an increase in obese mice, but the increase in ALT and AST were not statistically significant. Alpha-amylase levels were also increased in obese mice, both at RNA and protein level, while a decrease was seen in obese mice derived hepatocytes after acarbose treatment. CONCLUSIONS: Elevated liver alpha-amylase levels may be indicative of initial stages of obesity and the use of acarbose could be considered as a treatment of obesity which could be potentially effective at multiple levels.


Assuntos
Biomarcadores/sangue , Expressão Gênica/genética , Fígado/metabolismo , Camundongos Obesos/genética , Obesidade/genética , alfa-Amilases/genética , Acarbose/farmacologia , Animais , Células Cultivadas , Colesterol/sangue , Expressão Gênica/efeitos dos fármacos , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Fígado/efeitos dos fármacos , Masculino , Camundongos , Camundongos Obesos/sangue , Obesidade/sangue , Triglicerídeos/sangue
18.
Breast Cancer Res ; 18(1): 29, 2016 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-26956474

RESUMO

BACKGROUND: Somatostatin (SST) and cortistatin (CORT), two structurally and functionally related peptides, share a family of widespread receptors (sst1-5) to exert apparently similar biological actions, including endocrine/metabolic regulation and suppression of tumor cell proliferation. However, despite their therapeutic potential, attempts to apply SST-analogs to treat breast cancer have yielded unsatisfactory results. Actually, the specific roles of SST and CORT in mammary gland tumorigenesis (MGT), particularly in relation to metabolic dysregulation (i.e. obesity), remain unknown. METHODS: The role of endogenous SST and CORT in carcinogen-induced MGT was investigated under normal (lean) and obesity conditions. To that end, SST- and CORT-knockout (KO) mice and their respective littermate-controls, fed low-fat (LF) or high-fat (HF) diets, were treated with 7,12-dimethyl-benza-anthracene (DMBA) once a week (wk) for 3 wk, and MGT was monitored for 25 wk. Additionally, we examined the effect of SST or CORT removal in the development of the mammary gland. RESULTS: Lack of SST did not alter DMBA-induced MGT incidence under lean conditions; conversely, lack of endogenous CORT severely aggravated DMBA-induced MGT in LF-fed mice. These differences were not attributable to altered mammary gland development. HF-diet modestly increased the sensitivity to DMBA-induced carcinogenesis in control mice, whereas, as observed in LF-fed CORT-KO, HF-fed CORT-KO mice exhibited aggravated tumor incidence, discarding a major influence of obesity on these CORT actions. In marked contrast, HF-fed SST-KO mice exhibited much higher tumor incidence than LF-fed SST-KO mice, which could be associated with higher mammary complexity. CONCLUSIONS: Endogenous SST and CORT distinctly impact on DMBA-induced MGT, in a manner that is strongly dependent on the metabolic/endocrine milieu (lean vs. obese status). Importantly, CORT, rather than SST, could represent a major inhibitor of MGT under normal/lean-conditions, whereas both neuropeptides would similarly influence MGT under obesity conditions. The mechanisms mediating these different effects likely involve mammary development and hormones, but the precise underlying factors are still to be fully elucidated. However, our findings comprise suggestive evidence that CORT-like molecules, rather than classic SST-analogs, may help to identify novel tools for the medical treatment of breast cancer.


Assuntos
Carcinogênese/genética , Neoplasias Mamárias Animais/genética , Neuropeptídeos/genética , Somatostatina/genética , 9,10-Dimetil-1,2-benzantraceno/toxicidade , Animais , Feminino , Humanos , Glândulas Mamárias Animais/efeitos dos fármacos , Glândulas Mamárias Animais/patologia , Neoplasias Mamárias Animais/induzido quimicamente , Neoplasias Mamárias Animais/patologia , Camundongos , Camundongos Knockout , Camundongos Obesos/genética
19.
Sci Rep ; 5: 16262, 2015 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-26548909

RESUMO

Mitochondria-related microRNAs (miRNAs) have recently emerged as key regulators of cell metabolism and can modulate mitochondrial fusion and division. In order to investigate the roles of mitochondria-related miRNAs played in obesity, we conducted comprehensive molecular analysis in vitro and in vivo. Based on high-fat-diet (HFD) induced obese mice, we found that hepatic mitochondrial function was markedly altered. Subsequently, we evaluated the expression levels of selected mitochondria-related miRNAs and found that miR-141-3p was up-regulated strikingly in HFD mice. To further verify the role of miR-141-3p in obesity, we carried out gain-and-loss-of-function study in human HepG2 cells. We found that miR-141-3p could modulate ATP production and induce oxidative stress. Through luciferase report gene assay, we identified that phosphatase and tensin homolog (PTEN) was a target of miR-141-3p. Inhibiting PTEN could alter the mitochondrial function, too. Our study suggested that mitochondria-related miR-141-3p induced mitochondrial dysfunction by inhibiting PTEN.


Assuntos
MicroRNAs/genética , Obesidade/genética , PTEN Fosfo-Hidrolase/biossíntese , Trifosfato de Adenosina/genética , Animais , Dieta Hiperlipídica , Regulação da Expressão Gênica , Células Hep G2 , Humanos , Camundongos , Camundongos Obesos/genética , Mitocôndrias/genética , Mitocôndrias/patologia , Obesidade/patologia , Estresse Oxidativo/genética , PTEN Fosfo-Hidrolase/genética
20.
Physiol Behav ; 152(Pt A): 315-22, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-26440318

RESUMO

Despite the increase in obesity prevalence over the last decades, humans show large inter-individual variability for susceptibility to diet-induced obesity. Understanding the biological basis of this susceptibility could identify new therapeutic alternatives against obesity. We characterized behavioral changes associated with propensity to obesity induced by cafeteria (CAF) diet consumption in mice. We show that Balb/c mice fed a CAF diet display a large inter-individual variability in susceptibility to diet-induced obesity, such that based on changes in adiposity we can classify mice as obesity prone (OP) or obesity resistant (OR). Both OP and OR were hyperphagic relative to control-fed mice but caloric intake was similar between OP and OR mice. In contrast, OR had a larger increase in locomotor activity following CAF diet compared to OP mice. Obesity resistant and prone mice showed similar intake of sweet snacks, but OR ate more savory snacks than OP mice. Two bottle sucrose preference tests showed that OP decreased their sucrose preference compared to OR mice after CAF diet feeding. Finally, to test the robustness of the OR phenotype in response to further increases in caloric intake, we fed OR mice with a personalized CAF (CAF-P) diet based on individual snack preferences. When fed a CAF-P diet, OR increased their calorie intake compared to OP mice fed the standard CAF diet, but did not reach adiposity levels observed in OP mice. Together, our data show the contribution of hedonic intake, individual snack preference and physical activity to individual susceptibility to obesity in Balb/c mice fed a standard and personalized cafeteria-style diet.


Assuntos
Dieta , Comportamento Alimentar/fisiologia , Preferências Alimentares/fisiologia , Camundongos Endogâmicos BALB C/fisiologia , Camundongos Obesos/fisiologia , Atividade Motora/fisiologia , Ração Animal/efeitos adversos , Animais , Comportamento de Escolha/fisiologia , Dieta/efeitos adversos , Dieta/psicologia , Sacarose Alimentar/administração & dosagem , Modelos Animais de Doenças , Ingestão de Alimentos/fisiologia , Ingestão de Alimentos/psicologia , Comportamento Alimentar/psicologia , Preferências Alimentares/psicologia , Predisposição Genética para Doença , Hiperfagia/etiologia , Hiperfagia/fisiopatologia , Hiperfagia/psicologia , Masculino , Camundongos Endogâmicos BALB C/genética , Camundongos Endogâmicos BALB C/psicologia , Camundongos Obesos/genética , Camundongos Obesos/psicologia , Especificidade da Espécie
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA