RESUMO
Obesity is a complex medical condition that is linked to various health complications such as infertility, stroke, and osteoarthritis. Understanding the neurobiology of obesity is crucial for responding to the etiology of this disease. The hypothalamus coordinates many integral activities such as hormone regulation and feed intake and numerous studies have observed altered hypothalamic gene regulation in obesity models. Previously, it was reported that the promoter region of the satiety gene, Pomc, has increased DNA methylation in the hypothalamus following short-term exposure to a high fat diet, suggesting that epigenetic-mediated repression of hypothalamic Pomc might contribute to the development of obesity. However, due to technical limitations, this has never been directly tested. Here, we used the CRISPR-dCas9-TET1 and dCas9-DNMT3a systems to test the role of Pomc DNA methylation in the hypothalamus in abnormal weight gain following acute exposure to a high fat diet in male rats. We found that exposure to a high fat diet increases Pomc DNA methylation and reduces gene expression in the hypothalamus. Despite this, we found that CRISPR-dCas9-TET1-mediated demethylation of Pomc was not sufficient to prevent abnormal weight gain following exposure to a high fat diet. Furthermore, CRISPR-dCas9-DNMT3a-mediated methylation of Pomc did not alter weight gain following exposure to standard or high fat diets. Collectively, these results suggest that high fat diet induced changes in Pomc DNA methylation are a consequence of, but do not directly contribute to, abnormal weight gain during the development of obesity.
Assuntos
Metilação de DNA , Pró-Opiomelanocortina , Ratos , Masculino , Animais , Pró-Opiomelanocortina/genética , Pró-Opiomelanocortina/metabolismo , Obesidade/metabolismo , Aumento de Peso/genética , Hipotálamo/metabolismo , Regiões Promotoras Genéticas , Dieta Hiperlipídica/efeitos adversosRESUMO
Genomic imprinting refers to the mono-allelic and parent-specific expression of a subset of genes. While long recognized for their role in embryonic development, imprinted genes have recently emerged as important modulators of postnatal physiology, notably through hypothalamus-driven functions. Here, using mouse models of loss, gain and parental inversion of expression, we report that the paternally expressed Zdbf2 gene controls neonatal growth in mice, in a dose-sensitive but parent-of-origin-independent manner. We further found that Zdbf2-KO neonates failed to fully activate hypothalamic circuits that stimulate appetite, and suffered milk deprivation and diminished circulating Insulin Growth Factor 1 (IGF-1). Consequently, only half of Zdbf2-KO pups survived the first days after birth and those surviving were smaller. This study demonstrates that precise imprinted gene dosage is essential for vital physiological functions at the transition from intra- to extra-uterine life, here the adaptation to oral feeding and optimized body weight gain.
Assuntos
Proteínas de Ligação a DNA/genética , Ingestão de Alimentos/genética , Impressão Genômica/genética , Hipotálamo , Aumento de Peso/genética , Animais , Animais Recém-Nascidos/genética , Animais Recém-Nascidos/fisiologia , Feminino , Hipotálamo/metabolismo , Hipotálamo/fisiologia , Masculino , Camundongos , Camundongos Knockout , GravidezRESUMO
Smoking cessation increases body weight. The underlying mechanisms, however, have not been fully understood. We here report an establishment of a mouse model that exhibits an augmented body weight gain after nicotine withdrawal. High fat diet-fed mice were infused with nicotine for two weeks, and then with vehicle for another two weeks using osmotic minipumps. Body weight increased immediately after nicotine cessation and was significantly higher than that of mice continued on nicotine. Mice switched to vehicle consumed more food than nicotine-continued mice during the first week of cessation, while oxygen consumption was comparable. Elevated expression of orexigenic agouti-related peptide was observed in the hypothalamic appetite center. Pair-feeding experiment revealed that the accelerated weight gain after nicotine withdrawal is explained by enhanced energy intake. As a showcase of an efficacy of pharmacologic intervention, exendin-4 was administered and showed a potent suppression of energy intake and weight gain in mice withdrawn from nicotine. Our current model provides a unique platform for the investigation of the changes of energy regulation after smoking cessation.
Assuntos
Nicotina/efeitos adversos , Síndrome de Abstinência a Substâncias/patologia , Aumento de Peso , Proteína Relacionada com Agouti/metabolismo , Animais , Calorimetria , Respiração Celular/efeitos dos fármacos , Modelos Animais de Doenças , Ingestão de Energia/efeitos dos fármacos , Exenatida/farmacologia , Comportamento Alimentar/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Hipotálamo/metabolismo , Masculino , Camundongos Endogâmicos C57BL , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Síndrome de Abstinência a Substâncias/genética , Aumento de Peso/efeitos dos fármacos , Aumento de Peso/genéticaRESUMO
BACKGROUND: Diet-induced obesity (DIO) is associated with chronic, low-grade inflammation in the hypothalamus. The inflammatory pathway of the hypothalamus is activated during obesity, and inhibition of activation of the inflammatory pathway can partially reverse obesity. Therefore, exploring new targets for inhibiting hypothalamic inflammation will provide new ideas for the prevention and treatment of obesity. Liver kinase B1 (LKB1), a serine/threonine kinase, is a tumor suppressor and metabolic regulator. Recent studies have shown that LKB1 has a certain anti-inflammatory effect. However, a role of LKB1 in the regulation of hypothalamic inflammation remains unclear. Therefore, we examined whether LKB1 overexpression in the hypothalamus could weaken the hypothalamic inflammation and inhibit the development of obesity. METHODS: LKB1 overexpressing adeno-associated virus (AAV) particles were injected stereotactically into the third ventricle (3â¯V) of C57BL/6 mice fed with HFD. We assessed changes in body mass and adiposity, food intake, hypothalamic inflammatory markers, and energy and glucose metabolism. RESULTS: LKB1 up-regulation in hypothalamus attenuated diet-induced hypothalamic inflammation, reduced food intake and body weight gain. In addition, the overexpression of hypothalamic LKB1 increased the insulin sensitivity and improved whole-body lipid metabolism, which attenuated hepatic fat accumulation and serum lipid levels. CONCLUSION: Hypothalamic LKB1 up-regulation attenuates hypothalamic inflammation, and protects against hypothalamic inflammation induced damage to melanocortin system, resulting in lower food intake and lower fat mass accumulation, which consequently protects mice from the development of obesity. Our data suggest LKB1 as a novel negative regulator of hypothalamic inflammation, and also a potentially important target for treating other inflammatory diseases.
Assuntos
Doenças Hipotalâmicas/genética , Inflamação/genética , Obesidade/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas Quinases Ativadas por AMP , Animais , Dieta Hiperlipídica , Doenças Hipotalâmicas/prevenção & controle , Hipotálamo/metabolismo , Hipotálamo/patologia , Inflamação/prevenção & controle , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Obesidade/etiologia , Obesidade/prevenção & controle , Especificidade de Órgãos/genética , Proteínas Serina-Treonina Quinases/metabolismo , Regulação para Cima/genética , Aumento de Peso/genéticaRESUMO
Nutrient excess increases skeletal muscle oxidant production and mitochondrial fragmentation that may result in impaired mitochondrial function, a hallmark of skeletal muscle insulin resistance. This led us to explore whether an endogenous gas molecule, carbon monoxide (CO), which is thought to prevent weight gain and metabolic dysfunction in mice consuming high-fat diets, alters mitochondrial morphology and respiration in C2C12 myoblasts exposed to high glucose (15.6 mM) and high fat (250 µM BSA-palmitate) (HGHF). Also, skeletal muscle mitochondrial morphology, distribution, respiration, and energy expenditure were examined in obese resistant (OR) and obese prone (OP) rats that consumed a high-fat and high-sucrose diet for 10 wk with or without intermittent low-dose inhaled CO and/or exercise training. In cells exposed to HGHF, superoxide production, mitochondrial membrane potential (ΔΨm), mitochondrial fission regulatory protein dynamin-related protein 1 (Drp1) and mitochondrial fragmentation increased, while mitochondrial respiratory capacity was reduced. CO decreased HGHF-induced superoxide production, Drp1 protein levels and mitochondrial fragmentation, maintained ΔΨm, and increased mitochondrial respiratory capacity. In comparison with lean OR rats, OP rats had smaller skeletal muscle mitochondria that contained disorganized cristae, a normal mitochondrial distribution, but reduced citrate synthase protein expression, normal respiratory responses, and a lower energy expenditure. The combination of inhaled CO and exercise produced the greatest effect on mitochondrial morphology, increasing ADP-stimulated respiration in the presence of pyruvate, and preventing a decline in resting energy expenditure. These data support a therapeutic role for CO and exercise in preserving mitochondrial morphology and respiration during metabolic overload.
Assuntos
Monóxido de Carbono/metabolismo , Dinaminas/genética , Obesidade/genética , Aumento de Peso/genética , Animais , Monóxido de Carbono/farmacologia , Dieta Hiperlipídica , Metabolismo Energético/efeitos dos fármacos , Humanos , Camundongos , Mitocôndrias Musculares/metabolismo , Mitocôndrias Musculares/patologia , Dinâmica Mitocondrial/genética , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Mioblastos/metabolismo , Mioblastos/patologia , Obesidade/metabolismo , Obesidade/patologia , Condicionamento Físico Animal , Ratos , Espécies Reativas de Oxigênio/metabolismo , Sacarose/efeitos adversosRESUMO
SCOPE: Previous studies have identified potent anticancer activities of polyphenols in preventing prostate cancer. The aim of the current study is to evaluate the chemopreventive potential of grape powder (GP) supplemented diets in genetically predisposed and obesity-provoked prostate cancer. METHODS AND RESULTS: Prostate-specific Pten heterozygous (Pten+/f ) transgenic mice are fed low- and high-fat diet (LFD and HFD, respectively) supplemented with 10% GP for 33 weeks, ad libitum. Prostate tissues are characterized using immunohistochemistry and western blots, and sera are analyzed by ELISA and qRT-PCR. Pten+/f mice fed LFD and HFD supplemented with 10% GP show favorable histopathology, significant reduction of the proliferative rate of prostate epithelial cells (Ki67), and rescue of PTEN expression. The most potent protective effect of GP supplementation is detected against HFD-induced increase in inflammation (IL-1ß; TGF-ß1), activation of cell survival pathways (Akt, AR), and angiogenesis (CD31) in Pten+/f mice. Moreover, GP supplementation reduces circulating levels of oncogenic microRNAs (miR-34a; miR-22) in Pten+/f mice. There are no significant changes in body weight and food intake in GP supplemented diet groups. CONCLUSIONS: GP diet supplementation can be a beneficial chemopreventive strategy for obesity-related inflammation and prostate cancer progression. Monitoring serum miRNAs can facilitate the non-invasive evaluation of chemoprevention efficacy.
Assuntos
Anticarcinógenos/farmacologia , Neoplasia Prostática Intraepitelial/prevenção & controle , Neoplasias da Próstata/prevenção & controle , Vitis/química , Animais , Linhagem Celular Tumoral , Dieta Hiperlipídica/efeitos adversos , Suplementos Nutricionais , Feminino , Haploinsuficiência/genética , Humanos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , MicroRNAs/metabolismo , PTEN Fosfo-Hidrolase/genética , Pós , Neoplasia Prostática Intraepitelial/etiologia , Neoplasia Prostática Intraepitelial/patologia , Neoplasias da Próstata/etiologia , Neoplasias da Próstata/patologia , Prostatite/etiologia , Prostatite/prevenção & controle , Aumento de Peso/efeitos dos fármacos , Aumento de Peso/genéticaRESUMO
Inclusion of some plants especially spices in the diets of farm animals have been researched upon extensively for parameters like growth, blood chemistry, and feed utilization among others. In contrary, the use of turmeric as feed additive for ruminant production is so low, while its effects on reproduction (especially during pregnancy) in ruminants are not available due to little/no information on its properties, processing, and inclusion rate. Thus, a study was conducted to evaluate pre- and post-weaning performances of kids produced by three breeds of goat fed diets supplemented with graded levels of turmeric powder intensively in southwestern Nigeria. Kids produced by Kalahari Red (KR), West African Dwarf (WAD), and KalaWAD Does fed diets containing turmeric powder (TP) at different levels were monitored for pre-weaning that lasted for 12 weeks, while selected kids were observed for 4 weeks post-weaning. Each Doe breed was fed diet at 5% bodyweight during pregnancy and pre-weaning, including some selected kids during post-weaning. Dietary treatments included concentrate diet (CD) as TP-0, CD + 2 g/kg TP as TP-2g, and CD + 5 g/kg TP as TP-5 g. Brachiaria ruziziensis was used as basal diet. Data obtained were arranged in a 3 × 3 factorial layout in a completely randomized design, while analysis of variance was done using SAS 9.1. Significance level was taken at 5% probability, while means were separated using Duncan's multiple range test of the same statistical package. Results of the study for breed effect revealed that KR kids had the best (p < 0.05) pre-weaning performance from birth (3.2 kg) till weaning (14.5 kg) and for weight gains (11.2 kg and 134.0 g/day). No mortality was recorded for WAD kids. For TP effects, the kids fed TP-2g had the highest (p < 0.05) weight gains (1.6 kg and 58.6 g/day) and reduced value for feed conversion ratio (FCR) of 6.2 for post-weaning performance. Kids fed TP-5g had the highest (p < 0.05) value for protein efficiency ratio (PER) of 1.4 at the end of post-weaning. For interaction effects of breeds and TP, KR kids fed TP-5g performed best (p < 0.05) at the pre-weaning period in terms of weights from birth (3.6 kg) till weaning (15.7 kg), while KR kids fed TP-2g had highest (p < 0.05) pre-weaning weight gains (12.5 kg and 148.9 g/day). KR kids fed TP-5g performed best (p < 0.05) at the post-weaning period in terms of weight gains (1.8 kg and 64.4 g/day). KR kids fed TP-2g and KalaWAD kids fed TP-5g had the lowest FCR (5.2) and the highest PER (1.6) respectively at the post-weaning period. It can be concluded that breeds of kid, TP inclusion, and their interaction influenced pre- and post-weaning performances of the kids.
Assuntos
Ração Animal/análise , Curcuma , Dieta/veterinária , Suplementos Nutricionais , Cabras/crescimento & desenvolvimento , Fenômenos Fisiológicos da Nutrição Animal , Animais , Feminino , Cabras/genética , Masculino , Gravidez , Aumento de Peso/efeitos dos fármacos , Aumento de Peso/genéticaRESUMO
This passive overconsumption of western diet has precipitated a steep rise in obesity and its comorbidities, and obesity has become one of the main threats to health worldwide. Thus, deciphering the molecular mechanisms leading to obesity is therefore of utmost importance to guide the search for novel therapeutic and preventive strategies. Lycopene (LYC), a major carotenoid present in tomato, has been regarded as a nutraceutical that has powerful anti-oxidant and anti-obesity bioactivities. Even though substantial progress has been made in deciphering the mechanism of how LYC affects obesity in recent years, whether thermogenic genes, mitochondrial function and insulin resistance are involved in the anti-obesity effect of LYC is yet to be elucidated. In the current study, we demonstrated that LYC remarkably suppressed HFFD-elevated mice body weight gain. LYC blocked lipid accumulation in adipose tissue by decreasing the expressions of lipogenesis genes and increasing the expressions of lipidolysis related genes, including thermogenic and mitochondrial functional genes. Moreover, LYC improved HFFD-induced insulin resistance in WATs via inhibiting the inflammation responses in WATs, decreasing circulating proinflammatory cytokines, suppressing gut leak and intestinal inflammation. Our study indicating that the supplementation of LYC might be a nutritional preventive strategy to combat obesity.
Assuntos
Tecido Adiposo/efeitos dos fármacos , Dieta Ocidental/efeitos adversos , Resistência à Insulina , Licopeno/farmacologia , Aumento de Peso/efeitos dos fármacos , Adipócitos/efeitos dos fármacos , Adipócitos/patologia , Tecido Adiposo/metabolismo , Animais , Autofagia/efeitos dos fármacos , Suplementos Nutricionais , Frutose/efeitos adversos , Regulação da Expressão Gênica/efeitos dos fármacos , Inflamação/dietoterapia , Inflamação/etiologia , Metabolismo dos Lipídeos/efeitos dos fármacos , Masculino , Camundongos Endogâmicos C57BL , Estresse Oxidativo/efeitos dos fármacos , Termogênese/efeitos dos fármacos , Termogênese/genética , Aumento de Peso/genéticaRESUMO
BACKGROUND: RF-amide-related peptide-3 (RFRP-3), the mammalian ortholog of gonadotropin-inhibiting hormone, operates as inhibitory signal for the reproductive axis. Recently, RFRP-3 has been also suggested to stimulate feeding, and therefore might contribute to the control of body weight and its alterations. Yet, characterization of the metabolic actions of RFRP-3 has been so far superficial and mostly pharmacological. Here, we aim to investigate the physiological roles of RFRP-3 signaling in the control of feeding and metabolic homeostasis using a novel mouse model of genetic ablation of its canonical receptor, NPFF1R. METHODS: Food intake, body weight gain and composition, and key metabolic parameters, including glucose tolerance and insulin sensitivity, were monitored in mice with constitutive inactivation of NPFF1R. RESULTS: Congenital elimination of NPFF1R in male mice resulted in changes in feeding patterns, with a decrease in spontaneous food intake and altered responses to leptin and ghrelin: leptin-induced feeding suppression was exaggerated in NPFF1R null mice, whereas orexigenic responses to ghrelin were partially blunted. Concordant with this pro-anorectic phenotype, hypothalamic expression of Pomc was increased in NPFF1R null mice. In contrast, spontaneous feeding and neuropeptide expression remained unaltered in NPFF1R KO female mice. Despite propensity for reduced feeding, ablation of NPFF1R signaling in male mice did not cause overt alterations in body weight (BW) gain or composition, neither it affected BW responses to high fat diet (HFD), total energy expenditure or RQ ratios. Yet, NPFF1R KO males showed a decrease in locomotor activity. Conversely, NPFF1R null female mice tended to be heavier and displayed exaggerated BW increases in response to obesogenic insults, such as HFD or ovariectomy. These were associated to increased fat mass, decreased total energy expenditure in HFD, and unaltered RQ ratios or spontaneous locomotor activity. Finally, lack of NPFF1R signaling worsened the metabolic impact of HFD on glycemic homeostasis in males, as revealed by impaired glucose tolerance and insulin sensitivity, while female mice remained unaffected. CONCLUSION: Our data support a discernible orexigenic role of NPFF1R signaling selectively in males, which might modulate the effects of leptin and ghrelin on food intake. In addition, our study is the first to disclose the sex-biased, deleterious impact of the lack of NPFF1R signaling on body weight and fat composition, energy expenditure, locomotor activity and glucose balance, which exaggerates some of the metabolic consequences of concurrent obesogenic insults, such as HFD, in a sexually dimorphic manner. SUMMARY OF TRANSLATIONAL RELEVANCE: Our data are the first to document the nature and magnitude of the regulatory actions of RFRP-3/NPFF1R signaling in the control of feeding and metabolic homeostasis in a physiological setting. Our results not only suggest an orexigenic action of endogenous RFRP-3, specifically in males, but reveal also the detrimental impact of ablation of NPFF1R signaling on body composition, energy expenditure, locomotor activity or glucose balance, especially when concurrent with other obesogenic insults, as HFD, thereby providing the first evidence for additional metabolic effects of RFRP-3, other that the mere control of feeding. Interestingly, alterations of such key metabolic parameters occurred in a sex-biased manner, with males being more sensitive to deregulation of locomotor activity and glycemic control, while females displayed clearer obesogenic responses and deregulated energy expenditure. While our study cannot discard the possibility of RFRP-3 actions via alternative pathways, such as NPFF2R, our data pave the way for future analyses addressing the eventual contribution of altered RFRP-3/NPFF1R signaling in the development of metabolic alterations (including obesity and its comorbidities), especially in conditions associated to reproductive dysfunction.
Assuntos
Ingestão de Alimentos/genética , Ingestão de Alimentos/fisiologia , Neuropeptídeos/metabolismo , Receptores de Neuropeptídeos/genética , Receptores de Neuropeptídeos/fisiologia , Animais , Composição Corporal/genética , Dieta Hiperlipídica , Metabolismo Energético/genética , Grelina/farmacologia , Intolerância à Glucose/genética , Homeostase , Hipotálamo/metabolismo , Resistência à Insulina/genética , Leptina/farmacologia , Masculino , Camundongos , Camundongos Knockout , Caracteres Sexuais , Aumento de Peso/genéticaRESUMO
Genetic correlations between 16 meat quality and nutritional value traits and live weight at various ages, live ultrasound fat and muscle depth, carcass measures, and carcass dissection traits were estimated for Merino sheep in the Information Nucleus (IN). Genetic correlations between live weight at various ages and the carcass traits are also reported. The IN comprised 8 genetically linked flocks managed across a range of Australian sheep environments. Meat quality traits included between 1,200 and 1,300 records for progeny from over 170 sires for intramuscular fat (IMF), lean meat yield (LMY), shear force (SF5), pH, meat color, and meat nutritional value traits including iron and zinc levels and long-chain omega-3 and omega-6 polyunsaturated fatty acid levels. The genetic correlations indicated that selection of Merino sheep to either reduce fat or increase muscle using ultrasound assessments will result in little change in IMF and SF5. Myoglobin levels would tend to be reduced following selection for reduced ultrasound fat depth (0.35 ± 0.21, 0.43 ± 0.14), whereas increases in myoglobin levels would occur due to selection for increased ultrasound muscle depth (0.25 ± 0.24, 0.38 ± 0.15). Selection for increased live weight will result in favorable correlated responses in hot carcass weight (0.76 to 0.97), dressing percentage (0.13 to 0.47), and carcass muscle (0.37 to 0.95), but unfavorable responses of increases in carcass fatness (0.13 to 0.65) and possible small reductions in muscle oxidative activity (-0.13 ± 0.14 to -0.73 ± 0.33) and iron content (-0.14 ± 0.15 to -0.38 ± 0.16), and a possible deterioration of shear force from selection at later ages (0.15 ± 0.26, 0.27 ± 0.24). Negligible changes are generally expected for LMY and meat color traits following selection for increased live weight (most genetic correlations less than 0.20 in size). Selection for increased LMY would tend to result in unfavorable changes in several aspects of meat quality, including reduced IMF (-0.27 ± 0.18), meat tenderness (0.53 ± 0.26), and meat redness (-0.69 ± 0.40), as well as reduced iron levels (-0.25 ± 0.22). These genetic correlations are a first step in assisting the development of breeding values for new traits to be incorporated into genetic evaluation programs to improve meat production from Merino sheep and other dual-purpose sheep breeds.
Assuntos
Carne , Ovinos , Animais , Austrália , Composição Corporal/genética , Cruzamento , Cor , Ácidos Graxos Ômega-3 , Feminino , Ferro , Masculino , Carne/análise , Carne/normas , Músculos , Valor Nutritivo , Oxirredução , Fenótipo , Ovinos/crescimento & desenvolvimento , Carneiro Doméstico/genética , Aumento de Peso/genéticaRESUMO
Effectors of the phosphoinositide 3-kinase (PI3K) signal transduction pathway contribute to the hypothalamic regulation of energy and glucose homeostasis in divergent ways. Here we show that central nervous system (CNS) action of the PI3K signaling intermediate atypical protein kinase C (aPKC) constrains food intake, weight gain, and glucose intolerance in both rats and mice. Pharmacological inhibition of CNS aPKC activity acutely increases food intake and worsens glucose tolerance in chow-fed rodents and causes excess weight gain during high-fat diet (HFD) feeding. Similarly, selective deletion of the aPKC isoform Pkc-λ in proopiomelanocortin (POMC) neurons disrupts leptin action, reduces melanocortin content in the paraventricular nucleus, and markedly increases susceptibility to obesity, glucose intolerance, and insulin resistance specifically in HFD-fed male mice. These data implicate aPKC as a novel regulator of energy and glucose homeostasis downstream of the leptin-PI3K pathway in POMC neurons.
Assuntos
Ingestão de Alimentos/genética , Intolerância à Glucose/genética , Glucose/metabolismo , Isoenzimas/genética , Neurônios/metabolismo , Obesidade/genética , Proteína Quinase C/genética , Aumento de Peso/genética , Animais , Dieta Hiperlipídica , Ingestão de Alimentos/efeitos dos fármacos , Metabolismo Energético/efeitos dos fármacos , Metabolismo Energético/genética , Intolerância à Glucose/metabolismo , Hipotálamo/metabolismo , Resistência à Insulina , Leptina/metabolismo , Masculino , Melanocortinas/metabolismo , Camundongos , Obesidade/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Pró-Opiomelanocortina/efeitos dos fármacos , Pró-Opiomelanocortina/metabolismo , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/metabolismo , Ratos , Transdução de Sinais , Aumento de Peso/efeitos dos fármacosRESUMO
Pharmacological activation of the hypothalamic glucagon-like peptide 1 (GLP-1) receptor (GLP-1R) promotes weight loss and improves glucose tolerance. This demonstrates that the hypothalamic GLP-1R is sufficient but does not show whether it is necessary for the effects of exogenous GLP-1R agonists (GLP-1RA) or endogenous GLP-1 on these parameters. To address this, we crossed mice harboring floxed Glp1r alleles to mice expressing Nkx2.1-Cre to knock down Glp1r expression throughout the hypothalamus (GLP-1RKDΔNkx2.1cre). We also generated mice lacking Glp1r expression specifically in two GLP-1RA-responsive hypothalamic feeding nuclei/cell types, the paraventricular nucleus (GLP-1RKDΔSim1cre) and proopiomelanocortin neurons (GLP-1RKDΔPOMCcre). Chow-fed GLP-1RKDΔNkx2.1cre mice exhibited increased food intake and energy expenditure with no net effect on body weight. When fed a high-fat diet, these mice exhibited normal food intake but elevated energy expenditure, yielding reduced weight gain. None of these phenotypes were observed in GLP-1RKDΔSim1cre and GLP-1RKDΔPOMCcre mice. The acute anorectic and glucose tolerance effects of peripherally dosed GLP-1RA exendin-4 and liraglutide were preserved in all mouse lines. Chronic liraglutide treatment reduced body weight in chow-fed GLP-1RKDΔNkx2.1cre mice, but this effect was attenuated with high-fat diet feeding. In sum, classic homeostatic control regions are sufficient but not individually necessary for the effects of GLP-1RA on nutrient homeostasis.
Assuntos
Ingestão de Alimentos/genética , Metabolismo Energético/genética , Receptor do Peptídeo Semelhante ao Glucagon 1/genética , Glucose/metabolismo , Hipotálamo/metabolismo , Animais , Composição Corporal , Dieta Hiperlipídica , Ingestão de Alimentos/efeitos dos fármacos , Exenatida , Técnicas de Silenciamento de Genes , Peptídeo 1 Semelhante ao Glucagon/farmacologia , Receptor do Peptídeo Semelhante ao Glucagon 1/metabolismo , Teste de Tolerância a Glucose , Homeostase/genética , Incretinas/farmacologia , Liraglutida/farmacologia , Masculino , Camundongos , Neurônios/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Peptídeos/farmacologia , Pró-Opiomelanocortina/metabolismo , Peçonhas/farmacologia , Aumento de Peso/efeitos dos fármacos , Aumento de Peso/genéticaRESUMO
BACKGROUND: Constitutional thinness (CT) is a natural state of underweight (13-17.5kg/m2) without the presence of any eating disorders and abnormal hormonal profile, and with preserved menses in women. We previously conducted a four-week fat overfeeding study showing weight gain resistance in CT women and one of our main results was the identification of an energy gap: a positive energy balance (higher energy intake than energy expenditure). OBJECTIVE: This new overfeeding study is designed to confirm the energy gap and propose mechanistic hypothesis. DESIGN: A 2-week overfeeding (daily consumption of one bottle of Renutryl® Booster (600kcal, 30g protein, 72g carbohydrate, 21g fat) on top of the dietary intake) is performed to compare 15 women and men in each CT group (Body Mass Index [BMI]<18.5kg/m2) to their controls (BMI 20-25kg/m2). Bodyweight, food intake, energy expenditure (canopy, calorimetric chamber and Actiheart), body composition (DXA), appetite regulatory hormone profiles after a test meal, proteomics, metabolomics, urinary metabolic profiles, stool microbiome and lipids, fat and muscle transcriptomics are monitored before and after overfeeding. RESULTS AND CONCLUSIONS: Data inter-linking will be able to be established with results of this study. The findings could possibly open to therapeutic approaches to help CT patients to gain weight as well as provide a better understanding of energy regulation with regard to treat obesity (resistance to weight loss), a mirror image of CT (resistance to weight gain).
Assuntos
Terapia Nutricional/métodos , Magreza/etiologia , Magreza/terapia , Aumento de Peso/genética , Adolescente , Adulto , Metabolismo Energético/fisiologia , Feminino , Genômica , Humanos , Masculino , Metabolômica , Hipernutrição , Projetos de Pesquisa , Magreza/genética , Magreza/metabolismo , Adulto JovemRESUMO
C-type natriuretic peptide (CNP) and its receptor, natriuretic peptide receptor-B (NPR-B), are abundantly distributed in the hypothalamus. To explore the role of central CNP/NPR-B signaling in energy regulation, we generated mice with brain-specific NPR-B deletion (BND mice) by crossing Nestin-Cre transgenic mice and mice with a loxP-flanked NPR-B locus. Brain-specific NPR-B deletion prevented body weight gain induced by a high-fat diet (HFD), and the mesenteric fat and liver weights were significantly decreased in BND mice fed an HFD. The decreased liver weight in BND mice was attributed to decreased lipid accumulation in the liver, which was confirmed by histologic findings and lipid content. Gene expression analysis revealed a significant decrease in the mRNA expression levels of CD36, Fsp27, and Mogat1 in the liver of BND mice, and uncoupling protein 2 mRNA expression was significantly lower in the mesenteric fat of BND mice fed an HFD than in that of control mice. This difference was not observed in the epididymal or subcutaneous fat. Although previous studies reported that CNP/NPR-B signaling inhibits SNS activity in rodents, SNS is unlikely to be the underlying mechanism of the metabolic phenotype observed in BND mice. Taken together, CNP/NPR-B signaling in the brain could be a central factor that regulates visceral lipid accumulation and hepatic steatosis under HFD conditions. Further analyses of the precise mechanisms will enhance our understanding of the contribution of the CNP/NPR-B system to energy regulation.
Assuntos
Encéfalo/metabolismo , Fígado Gorduroso/metabolismo , Gordura Intra-Abdominal/metabolismo , Metabolismo dos Lipídeos , Fígado/metabolismo , Peptídeo Natriurético Tipo C/metabolismo , Receptores do Fator Natriurético Atrial/metabolismo , Aciltransferases/genética , Aciltransferases/metabolismo , Animais , Antígenos CD36/genética , Antígenos CD36/metabolismo , Dieta Hiperlipídica/efeitos adversos , Metabolismo Energético/genética , Fígado Gorduroso/genética , Deleção de Genes , Perfilação da Expressão Gênica , Hipotálamo/metabolismo , Gordura Intra-Abdominal/química , Metabolismo dos Lipídeos/genética , Fígado/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Obesidade/genética , Obesidade/metabolismo , Tamanho do Órgão/genética , Proteínas/genética , Proteínas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores do Fator Natriurético Atrial/genética , Transdução de Sinais , Aumento de Peso/genéticaRESUMO
In rodents, food-predictive cues elicit eating in the absence of hunger (Weingarten, 1983). This behavior is disrupted by the disconnection of amygdala pathways to the lateral hypothalamus (Petrovich et al., 2002). Whether this circuit contributes to long-term weight gain is unknown. Using fMRI in 32 healthy individuals, we demonstrate here that the amygdala response to the taste of a milkshake when sated but not hungry positively predicts weight change. This effect is independent of sex, initial BMI, and total circulating ghrelin levels, but it is only present in individuals who do not carry a copy of the A1 allele of the Taq1A polymorphism. In contrast, A1 allele carriers, who have decreased D2 receptor density (Blum et al., 1996), show a positive association between caudate response and weight change. Regardless of genotype, however, dynamic causal modeling supports unidirectional gustatory input from basolateral amygdala (BLA) to hypothalamus in sated subjects. This finding suggests that, as in rodents, external cues gain access to the homeostatic control circuits of the human hypothalamus via the amygdala. In contrast, during hunger, gustatory inputs enter the hypothalamus and drive bidirectional connectivity with the amygdala. These findings implicate the BLA-hypothalamic circuit in long-term weight change related to nonhomeostatic eating and provide compelling evidence that distinct brain mechanisms confer susceptibility to weight gain depending upon individual differences in dopamine signaling.
Assuntos
Tonsila do Cerebelo/fisiologia , Sinais (Psicologia) , Fome , Saciação , Aumento de Peso/fisiologia , Adolescente , Adulto , Alelos , Feminino , Humanos , Hipotálamo/fisiologia , Masculino , Polimorfismo Genético , Receptores de Dopamina D2/genética , Aumento de Peso/genéticaRESUMO
Prader-Willi syndrome (PWS) is a multigene disorder associated with neonatal failure to thrive, developmental delay and endocrine abnormalities suggestive of hypothalamic dysfunction. Children with PWS typically develop overt hyperphagia and obesity â¼8 years of age, later than children with other genetic forms of obesity. This suggests a postnatal developmental or degenerative component to PWS-associated obesity. De novo inactivating mutations in one PWS candidate gene, MAGEL2, have been identified in children with features of PWS. Adult mice lacking Magel2 are insensitive to the anorexic effect of leptin treatment, and their hypothalamic pro-opiomelanocortin (POMC) neurons fail to depolarize in response to leptin. However, it is unclear whether this leptin insensitivity is congenital, or whether normal leptin sensitivity in neonatal Magel2-null mice is lost postnatally. We used in vitro cytosolic calcium imaging to follow the postnatal development of leptin responses in POMC neurons in these mice. Leptin caused an activation of POMC neurons in wild-type acute hypothalamic slice preparations at all ages, reflecting their normal leptin-invoked depolarization. Normal leptin responses were found in Magel2-null mice up to 4 weeks of age, but the proportion of leptin-responsive POMC neurons was reduced in 6-week-old Magel2-null mice. The number of α-melanocyte-stimulating hormone immunoreactive fibers in the paraventricular hypothalamic nucleus was also reduced in mutant mice at 6 weeks of age. A similar progressive loss of leptin sensitivity caused by loss of MAGEL2 in children with PWS could explain the delayed onset of increased appetite and weight gain in this complex disorder.
Assuntos
Antígenos de Neoplasias/genética , Leptina/metabolismo , Neurônios/metabolismo , Síndrome de Prader-Willi/genética , Proteínas/genética , Animais , Núcleo Arqueado do Hipotálamo/crescimento & desenvolvimento , Núcleo Arqueado do Hipotálamo/metabolismo , Núcleo Arqueado do Hipotálamo/patologia , Modelos Animais de Doenças , Humanos , Hipotálamo/crescimento & desenvolvimento , Hipotálamo/metabolismo , Hipotálamo/patologia , Leptina/administração & dosagem , Camundongos , Neurônios/patologia , Síndrome de Prader-Willi/metabolismo , Síndrome de Prader-Willi/patologia , Pró-Opiomelanocortina/metabolismo , Aumento de Peso/genéticaRESUMO
The first epigenome-wide association study of BMI identified DNA methylation at an HIF3A locus associated with BMI. We tested the hypothesis that DNA methylation variants are associated with BMI according to intake of B vitamins. In two large cohorts, we found significant interactions between the DNA methylation-associated HIF3A single nucleotide polymorphism (SNP) rs3826795 and intake of B vitamins on 10-year changes in BMI. The association between rs3826795 and BMI changes consistently increased across the tertiles of total vitamin B2 and B12 intake (all P for interaction <0.01). The differences in the BMI changes per increment of minor allele were -0.10 (SE 0.06), -0.01 (SE 0.06), and 0.12 (SE 0.07) within subgroups defined by increasing tertiles of total vitamin B2 intake and -0.10 (SE 0.06), -0.01 (SE 0.06), and 0.10 (SE 0.07) within subgroups defined by increasing tertiles of total vitamin B12 intake. In two independent cohorts, a DNA methylation variant in HIF3A was associated with BMI changes through interactions with total or supplemental vitamin B2, vitamin B12, and folate. These findings suggest a potential causal relation between DNA methylation and adiposity.
Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Metilação de DNA/genética , Dieta/estatística & dados numéricos , Interação Gene-Ambiente , Obesidade/genética , Complexo Vitamínico B , Aumento de Peso/genética , Adulto , Idoso , Proteínas Reguladoras de Apoptose , Índice de Massa Corporal , Estudos de Coortes , Feminino , Ácido Fólico , Humanos , Estudos Longitudinais , Masculino , Pessoa de Meia-Idade , Obesidade/epidemiologia , Sobrepeso/epidemiologia , Sobrepeso/genética , Polimorfismo de Nucleotídeo Único , Estudos Prospectivos , Proteínas Repressoras , Riboflavina , Reino Unido/epidemiologia , Estados Unidos/epidemiologia , Vitamina B 12 , Vitamina B 6RESUMO
Lead- (Pb-) induced oxidative stress is known to suppress growth performance and feed efficiency in broiler chickens. In an attempt to describe the specific underlying mechanisms of such phenomenon we carried out the current study. Ninety-six one-day-old broiler chicks were randomly assigned to 2 dietary treatment groups of 6 pen replicates, namely, (i) basal diet containing no lead supplement (control) and (ii) basal diet containing 200 mg lead acetate/kg of diet. Following 3 weeks of experimental period, jejunum samples were collected to examine the changes in gene expression of several nutrient transporters, antioxidant enzymes, and heat shock protein 70 (Hsp70) using quantitative real-time PCR. The results showed that addition of lead significantly decreased feed intake, body weight gain, and feed efficiency. Moreover, with the exception of GLUT5, the expression of all sugar, peptide, and amino acid transporters was significantly downregulated in the birds under Pb induced oxidative stress. Exposure to Pb also upregulated the antioxidant enzymes gene expression together with the downregulation of glutathione S-transferase and Hsp70. In conclusion, it appears that Pb-induced oxidative stress adversely suppresses feed efficiency and growth performance in chicken and the possible underlying mechanism for such phenomenon is downregulation of major nutrient transporter genes in small intestine.
Assuntos
Galinhas/genética , Regulação da Expressão Gênica/genética , Intestino Delgado/metabolismo , Chumbo/metabolismo , Proteínas de Membrana Transportadoras/genética , RNA Mensageiro/genética , Ração Animal , Animais , Antioxidantes/metabolismo , Galinhas/metabolismo , Dieta/métodos , Suplementos Nutricionais , Alimentos , Glutationa Transferase/metabolismo , Proteínas de Choque Térmico HSP70/metabolismo , Masculino , Estresse Oxidativo/genética , Aumento de Peso/genéticaRESUMO
Selenium is transferred from the mouse dam to its neonate via milk. Milk contains selenium in selenoprotein form as selenoprotein P (Sepp1) and glutathione peroxidase-3 (Gpx3) as well as in non-specific protein form as selenomethionine. Selenium is also present in milk in uncharacterized small-molecule form. We eliminated selenomethionine from the mice in these experiments by feeding a diet that contained sodium selenite as the source of selenium. Selenium-replete dams with deletion of Sepp1 or Gpx3 were studied to assess the effects of these genes on selenium transfer to the neonate. Sepp1 knockout caused a drop in milk selenium to 27% of the value in wild-type milk and a drop in selenium acquisition by the neonates to 35%. In addition to decreasing milk selenium by eliminating Sepp1, deletion of Sepp1 causes a decline in whole-body selenium, which likely also contributes to the decreased transfer of selenium to the neonate. Deletion of Gpx3 did not decrease milk selenium content or neonate selenium acquisition by measurable amounts. Thus, when the dam is fed selenium-adequate diet (0.25 mg selenium/kg diet), milk Sepp1 transfers a large amount of selenium to neonates but the transfer of selenium by Gpx3 is below detection by our methods.
Assuntos
Glutationa Peroxidase/metabolismo , Leite/metabolismo , Selênio/metabolismo , Selenoproteína P/metabolismo , Animais , Animais Recém-Nascidos , Transporte Biológico , Eletroforese em Gel de Poliacrilamida , Feminino , Glutationa Peroxidase/genética , Hibridização In Situ , Masculino , Glândulas Mamárias Animais/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Selenoproteína P/genética , Desmame , Aumento de Peso/genéticaRESUMO
Common polymorphisms in the first intron of FTO are associated with increased body weight in adults. Previous studies have suggested that a CUX1-regulatory element within the implicated FTO region controls expression of FTO and the nearby ciliary gene, RPGRIP1L. Given the role of ciliary genes in energy homeostasis, we hypothesized that mice hypomorphic for Rpgrip1l would display increased adiposity. We find that Rpgrip1lâº/â» mice are hyperphagic and fatter, and display diminished suppression of food intake in response to leptin administration. In the hypothalamus of Rpgrip1lâº/â» mice, and in human fibroblasts with hypomorphic mutations in RPGRIP1L, the number of AcIII-positive cilia is diminished, accompanied by impaired convening of the leptin receptor to the vicinity of the cilium, and diminished pStat3 in response to leptin. These findings suggest that RPGRIP1L may be partly or exclusively responsible for the obesity susceptibility signal at the FTO locus.