Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Br J Nutr ; 121(7): 748-755, 2019 04.
Article in English | MEDLINE | ID: mdl-30806344

ABSTRACT

Treatment of liver fibrosis is very limited as there is currently no effective anti-fibrotic therapy. Spirulina platensis (SP) is a blue-green alga that is widely supplemented in healthy foods. The objective of this study was to determine whether SP supplementation can prevent obesity-induced liver fibrosis in vivo. Male C57BL/6J mice were randomly assigned to a low-fat or a high-fat (HF)/high-sucrose/high-cholesterol diet or an HF diet supplemented with 2·5 % SP (w/w) (HF/SP) for 16 or 20 weeks. There were no significant differences in body weight, activity, energy expenditure, serum lipids or glucose tolerance between mice on HF and HF/SP diets. However, plasma alanine aminotransferase level was significantly reduced by SP at 16 weeks. Expression of fibrotic markers and trichrome stains showed no differences between HF and HF/SP. Splenocytes isolated from HF/SP fed mice had lower inflammatory gene expression and cytokine secretion compared with splenocytes from HF-fed mice. SP supplementation did not attenuate HF-induced liver fibrosis. However, the expression and secretion of inflammatory genes in splenocytes were significantly reduced by SP supplementation, demonstrating the anti-inflammatory effects of SP in vivo. Although SP did not show appreciable effect on the prevention of liver fibrosis in this mouse model, it may be beneficial for other inflammatory conditions.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Dietary Supplements , Liver Cirrhosis/prevention & control , Spirulina , Spleen/cytology , Animals , Diet, High-Fat/adverse effects , Disease Models, Animal , Liver Cirrhosis/etiology , Male , Mice , Mice, Inbred C57BL , Obesity/complications
2.
Br J Nutr ; 121(2): 146-154, 2019 01.
Article in English | MEDLINE | ID: mdl-30400998

ABSTRACT

We have recently reported that soluble dietary fibre, glucomannan, increased colonic alkaline phosphatase (ALP) activity and the gene expression without affecting the small-intestinal activity and that colonic ALP was correlated with gut mucins (index of intestinal barrier function). We speculated that dietary fermentable carbohydrates including oligosaccharides commonly elevate colonic ALP and gene expression as well as increase mucin secretion and microbial fermentation. To test this hypothesis, male Sprague-Dawley rats were fed a diet containing 30 % lard with or without 4 % fructo-oligosaccharides (FOS), galacto-oligosaccharides (GOS), raffinose (RAF) and lactulose (LAC), which are non-digestible oligosaccharides or isomalto-oligosaccharides (IMOS; some digestible oligosaccharides) for 2 weeks. Colon ALP activity, the gene expression and gut luminal variables including mucins, organic acids and microbiota were measured. Colonic ALP was significantly elevated in the FOS, RAF and LAC groups, and a similar trend was observed in the GOS group. Colonic expression of intestinal alkaline phosphatase (IAP -I), an ALP gene, was significantly elevated in the FOS, GOS and RAF groups and tended to be increased in the LAC group. Dietary FOS, GOS, RAF and LAC significantly elevated faecal mucins, caecal n-butyrate and faecal ratio of Bifidobacterium spp. Dietary IMOS had no effect on colonic ALP, mucins, organic acids and microbiota. Colon ALP was correlated with mucins, caecal n-butyrate and faecal Bifidobacterium spp. This study demonstrated that non-digestible and fermentable oligosaccharides commonly elevate colonic ALP activity and the expression of IAP-I, with increasing mucins and microbial fermentation, which might be important for protection of gut epithelial homoeostasis.


Subject(s)
Alkaline Phosphatase/metabolism , Colon/enzymology , Gastrointestinal Microbiome/physiology , Isoenzymes/genetics , Mucins/metabolism , Oligosaccharides/administration & dosage , Alkaline Phosphatase/genetics , Animals , Bacteria/metabolism , Bifidobacterium/isolation & purification , Butyrates/analysis , Cecum/chemistry , Dietary Carbohydrates , Dietary Fats/administration & dosage , Digestion , Feces/microbiology , Fermentation/drug effects , Male , Oligosaccharides/metabolism , Rats , Rats, Sprague-Dawley , Up-Regulation/drug effects
3.
Br J Nutr ; 120(1): 13-22, 2018 07.
Article in English | MEDLINE | ID: mdl-29936922

ABSTRACT

Dietary fatty acid (FA) composition may influence metabolism, possibly affecting weight management. The purpose of this study was to compare the effects of a 5-d diet rich in PUFA v. MUFA. A total of fifteen normal-weight men participated in a randomised cross-over design with two feeding trials (3 d lead-in diet, pre-diet visit, 5-d PUFA- or MUFA-rich diet, post-diet visit). The 5-d diets (50 % fat) were rich in either PUFA (25 % of energy) or MUFA (25 % of energy). At pre- and post-diet visits, subjects consumed breakfast and lunch test meals, rich in the FA for that 5-d diet. Indirect calorimetry was used for 4 h after each meal. There were no treatment differences in fasting metabolism acutely or after the 5-d diet. For acute meal responses before diet, RER was higher for PUFA v. MUFA (0·86 (sem 0·01) v. 0·84 (sem 0·01), P<0·05), whereas diet-induced thermogenesis (DIT) was lower for PUFA v. MUFA (18·91 (SEM 1·46) v. 21·46 (SEM 1·34) kJ, P<0·05). After the 5-d diets, the change in RER was different for PUFA v. MUFA (-0·02 (sem 0·01) v. 0·00 (sem 0·01), P<0·05). Similarly, the change in fat oxidation was greater for PUFA v. MUFA (0·18 (sem 0·07) v. 0·04 (sem 0·06) g, P<0·05). In conclusion, acutely, a MUFA-rich meal results in lower RER and greater DIT. However, after a 5-d high-fat diet, the change in metabolic responses was greater in the PUFA diet, showing the metabolic adaptability of a PUFA-rich diet.


Subject(s)
Diet, High-Fat , Fatty Acids, Monounsaturated/metabolism , Fatty Acids, Unsaturated/metabolism , Adolescent , Adult , Body Weight , Calorimetry , Calorimetry, Indirect , Cross-Over Studies , Dietary Fats/administration & dosage , Energy Metabolism , Fasting , Fatty Acids/metabolism , Humans , Male , Meals , Obesity/metabolism , Oxygen/chemistry , Postprandial Period , Single-Blind Method , Thermogenesis , Young Adult
4.
Br J Nutr ; 119(9): 981-991, 2018 05.
Article in English | MEDLINE | ID: mdl-29502540

ABSTRACT

Little is known about how diet-induced obesity and insulin resistance affect protein and amino acid (AA) metabolism in tissues. The natural relative abundances of the heavy stable isotopes of C (δ 13C) and N (δ 15N) in tissue proteins offer novel and promising biomarkers of AA metabolism. They, respectively, reflect the use of dietary macronutrients for tissue AA synthesis and the relative metabolic use of tissue AA for oxidation v. protein synthesis. In this study, δ 13C and δ 15N were measured in the proteins of various tissues in young adult rats exposed perinatally and/or fed after weaning with a normal- or a high-fat (HF) diet, the aim being to characterise HF-induced tissue-specific changes in AA metabolism. HF feeding was shown to increase the routing of dietary fat to all tissue proteins via non-indispensable AA synthesis, but did not affect AA allocation between catabolic and anabolic processes in most tissues. However, the proportion of AA directed towards oxidation rather than protein synthesis was increased in the small intestine and decreased in the tibialis anterior muscle and adipose tissue. In adipose tissue, the AA reallocation was observed in the case of perinatal or post-weaning exposure to HF, whereas in the small intestine and tibialis anterior muscle the AA reallocation was only observed after HF exposure that covered both the perinatal and post-weaning periods. In conclusion, HF exposure induced an early reorganisation of AA metabolism involving tissue-specific effects, and in particular a decrease in the relative allocation of AA to oxidation in several peripheral tissues.


Subject(s)
Amino Acids/metabolism , Carbon/metabolism , Diet, High-Fat/adverse effects , Nitrogen/metabolism , Animal Feed/analysis , Animal Nutritional Physiological Phenomena , Animals , Carbon/chemistry , Carbon Isotopes , Diet/veterinary , Nitrogen/chemistry , Nitrogen Isotopes , Rats , Rats, Sprague-Dawley
5.
Br J Nutr ; 119(11): 1220-1232, 2018 06.
Article in English | MEDLINE | ID: mdl-29770757

ABSTRACT

Diets high in fat can result in obesity and non-alcoholic fatty liver disease (NAFLD). The improvement of obesity and NAFLD is an important issue. ß-Conglycinin, one of the soya proteins, is known to prevent hyperlipidaemia, obesity and NAFLD. Therefore, we aimed to investigate the effects of ß-conglycinin on the improvement of obesity and NAFLD in high-fat (HF) diet-induced obese (DIO) mice and clarify the mechanism underlying these effects in liver and white adipose tissue (WAT). DIO male ddY mice were divided into six groups: HF, medium-fat (MF) and low-fat (LF) groups fed casein, and HF, MF and LF groups in all of which the casein was replaced by ß-conglycinin. A period of 5 weeks later, the ß-conglycinin-supplemented group resulted in lower body weight, relative weight of subcutaneous WAT, and hepatic TAG content (P=0·001). Furthermore, ß-conglycinin suppressed the hepatic expression of Pparγ2 in the HF dietary group, sterol regulatory element-binding protein-1c and the target genes. The expressions of inflammation-related genes were significantly low in the epididymal and subcutaneous WAT from the mice fed ß-conglycinin compared with those fed casein in the HF dietary group. Moreover, the expressions of Pparγ1 and Pparγ2 mRNA were suppressed in subcutaneous WAT in the HF dietary group but not in epididymal WAT. The concentrations of insulin and leptin were low in the serum of the mice fed ß-conglycinin. In conclusion, ß-conglycinin effectively improved obesity and NAFLD in DIO mice, and it appears to be a promising dietary protein for the amelioration of NAFLD and obesity.


Subject(s)
Antigens, Plant/pharmacology , Down-Regulation/drug effects , Fatty Liver/prevention & control , Globulins/pharmacology , Obesity/prevention & control , PPAR gamma/metabolism , Seed Storage Proteins/pharmacology , Soybean Proteins/pharmacology , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Animal Feed/analysis , Animals , Antigens, Plant/administration & dosage , Carbon Dioxide , Diet , Epididymis , Gene Expression Regulation/drug effects , Globulins/administration & dosage , Male , Mice , Obesity/etiology , Oxygen Consumption , PPAR gamma/genetics , Seed Storage Proteins/administration & dosage , Soybean Proteins/administration & dosage
6.
Br J Nutr ; 117(3): 377-385, 2017 02.
Article in English | MEDLINE | ID: mdl-28222826

ABSTRACT

Nutrition during pregnancy can impact on the susceptibility of the offspring to CVD. Postnatal consumption of trans-fatty acids (TFA), associated with partially hydrogenated vegetable oil (PHVO), increases the risk of atherosclerosis, whereas evidence for those TFA associated with ruminant-derived dairy products and meat remain equivocal. In this study, we investigate the impact of maternal consumption of dietary PHVO (P) and ruminant milk fat (R) on the development of atherosclerosis in their offspring, using the transgenic apoE*3 Leiden mouse. Dams were fed either chow (C) or one of three high-fat diets: a diet reflecting the SFA content of a 'Western' diet (W) or one enriched with either P or R. Diets were fed during either pregnancy alone or pregnancy and lactation. Weaned offspring were then transferred to an atherogenic diet for 12 weeks. Atherosclerosis was assessed as lipid staining in cross-sections of the aorta. There was a significant effect of maternal diet during pregnancy on development of atherosclerosis (P=0·013) in the offspring with those born of mothers fed R or P during pregnancy displaying smaller lesions that those fed C or W. This was not associated with changes in total or lipoprotein cholesterol. Continuing to feed P during lactation increased atherosclerosis compared with that seen in offspring of dams fed P only during pregnancy (P<0·001). No such effect was seen in those from mothers fed R (P=0·596) or W (P=901). We conclude that dietary TFA have differing effects on cardiovascular risk at different stages of the lifecycle.


Subject(s)
Apolipoproteins E/metabolism , Atherosclerosis/etiology , Dietary Fats/adverse effects , Maternal Nutritional Physiological Phenomena , Milk/chemistry , Plant Oils/chemistry , Trans Fatty Acids/adverse effects , Animals , Animals, Genetically Modified , Apolipoproteins E/genetics , Atherosclerosis/blood , Atherosclerosis/pathology , Cholesterol/blood , Diet, High-Fat , Disease Susceptibility , Female , Lactation , Lipoproteins/blood , Male , Mice , Pregnancy , Prenatal Nutritional Physiological Phenomena , Risk Factors , Ruminants
7.
Br J Nutr ; 117(6): 796-803, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28412994

ABSTRACT

This study evaluated the effects of a post-weaning high-fat (HF) diet on somatic growth, food consumption, metabolic parameters, phagocytic rate and nitric oxide (NO) production of peritoneal macrophages in young rats submitted to a maternal low-protein (LP) diet. Male Wistar rats (aged 60 d) were divided in two groups (n 22/each) according to their maternal diet during gestation and lactation: control (C, dams fed 17 % casein) and LP (dams fed 8 % casein). At weaning, half of the groups were fed HF diet and two more groups were formed (HF and low protein-high fat (LP-HF)). Somatic growth, food and energy intake, fat depots, serum glucose, cholesterol and leptin concentrations were evaluated. Phagocytic rate and NO production were analysed in peritoneal macrophages under stimulation of zymosan and lipopolysaccharide (LPS)+interferon γ (IFN-γ), respectively. The maternal LP diet altered the somatic parameters of growth and development of pups. LP and LP-HF pups showed a higher body weight gain and food intake than C pups. HF and LP-HF pups showed increased retroperitoneal and epididymal fat depots, serum level of TAG and total cholesterol compared with C and LP pups. After LPS+IFN-γ stimulation, LP and LP-HF pups showed reduced NO production when compared with their pairs. Increased phagocytic activity and NO production were seen in LP but not LP-HF peritoneal macrophages. However, peritoneal macrophages of LP pups were hyporesponsive to LPS+IFN-γ induced NO release, even after a post-weaning HF diet. Our data demonstrated that there was an immunomodulation related to dietary fatty acids after the maternal LP diet-induced metabolic programming.


Subject(s)
Diet, High-Fat , Diet, Protein-Restricted , Dietary Fats/pharmacology , Macrophages, Peritoneal/drug effects , Malnutrition/complications , Maternal Nutritional Physiological Phenomena , Weight Gain , Animals , Dietary Fats/administration & dosage , Energy Intake/drug effects , Female , Interferon-gamma/blood , Lactation , Lipopolysaccharides , Macrophages, Peritoneal/metabolism , Male , Nitric Oxide/metabolism , Obesity/etiology , Obesity/immunology , Obesity/metabolism , Pregnancy , Pregnancy Complications , Prenatal Exposure Delayed Effects , Rats, Wistar , Weaning , Zymosan
8.
Br J Nutr ; 118(10): 788-803, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29110748

ABSTRACT

Perinatal maternal high-fat (HF) diet programmes offspring obesity. Obesity is associated with overactivation of the endocannabinoid system (ECS) in adult subjects, but the role of the ECS in the developmental origins of obesity is mostly unknown. The ECS consists of endocannabinoids, cannabinoid receptors (cannabinoid type-1 receptor (CB1) and cannabinoid type-2 receptor (CB2)) and metabolising enzymes. We hypothesised that perinatal maternal HF diet would alter the ECS in a sex-dependent manner in white and brown adipose tissue of rat offspring at weaning in parallel to obesity development. Female rats received standard diet (9 % energy content from fat) or HF diet (29 % energy content from fat) before mating, during pregnancy and lactation. At weaning, male and female offspring were killed for tissue harvest. Maternal HF diet induced early obesity, white adipocyte hypertrophy and increased lipid accumulation in brown adipose tissue associated with sex-specific changes of the ECS's components in weanling rats. In male pups, maternal HF diet decreased CB1 and CB2 protein in subcutaneous adipose tissue. In female pups, maternal HF diet increased visceral and decreased subcutaneous CB1. In brown adipose tissue, maternal HF diet increased CB1 regardless of pup sex. In addition, maternal HF diet differentially changed oestrogen receptor across the adipose depots in male and female pups. The ECS and oestrogen signalling play an important role in lipogenesis, adipogenesis and thermogenesis, and we observed early changes in their targets in adipose depots of the offspring. The present findings provide insights into the involvement of the ECS in the developmental origins of metabolic disease induced by inadequate maternal nutrition in early life.


Subject(s)
Adipose Tissue/metabolism , Diet, High-Fat/adverse effects , Endocannabinoids/metabolism , Maternal Nutritional Physiological Phenomena , Obesity/etiology , Receptors, Cannabinoid/metabolism , Weaning , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Adiposity , Animal Nutritional Physiological Phenomena , Animals , Dietary Fats/administration & dosage , Dietary Fats/adverse effects , Female , Lactation , Lipid Metabolism , Male , Obesity/metabolism , Pregnancy , Prenatal Nutritional Physiological Phenomena , Rats, Wistar , Receptors, Estrogen/metabolism , Sex Factors , Thermogenesis
9.
Br J Nutr ; 115(11): 1887-95, 2016 06.
Article in English | MEDLINE | ID: mdl-27080153

ABSTRACT

Peripheral blood mononuclear cells (PBMC) are increasingly used for nutrigenomic studies. In this study, we aimed to identify whether these cells could reflect the development of an obesogenic profile associated with the intake of high-fat (HF) diets. We analysed, by real-time RT-PCR, the dietary response of key genes related to lipid metabolism, obesity and inflammation in PBMC of control rats, rats fed a cafeteria or a commercial HF diet and rats fed a control diet after the intake of a cafeteria diet (post-cafeteria model). Cafeteria diet intake, which resulted in important overweight and related complications, altered the expressions of most of the studied genes in PBMC, evidencing the development of an obesogenic profile. Commercial HF diet, which produced metabolic alterations but in the absence of noticeably increased body weight, also altered PBMC gene expression, inducing a similar regulatory pattern as that observed for the cafeteria diet. Regulation of carnitine palmitoyltransferase I (Cpt1a) mRNA expression was of special interest; its expression reflected metabolic alterations related to the intake of both obesogenic diets (independently of increased body weight) even at an early stage as well as metabolic recovery in post-cafeteria animals. Thus, PBMC constitute an important source of biomarkers that reflect the increased adiposity and metabolic deregulation associated with the intake of HF diets. In particular, we propose an analysis of Cpt1a expression as a good biomarker to detect the early metabolic alterations caused by the consumption of hyperlipidic diets, and also as a marker of metabolic recovery associated to weight loss.


Subject(s)
Carnitine O-Palmitoyltransferase/metabolism , Diet, High-Fat/adverse effects , Dietary Fats/metabolism , Leukocytes, Mononuclear/metabolism , Obesity/metabolism , Transcriptome , Adipogenesis/genetics , Adiposity , Animals , Biomarkers/metabolism , Carnitine O-Palmitoyltransferase/genetics , Diet , Dietary Fats/administration & dosage , Dietary Fats/adverse effects , Epigenesis, Genetic , Male , Obesity/etiology , Obesity/genetics , RNA, Messenger/metabolism , Rats, Wistar
10.
Nutr Res Rev ; 29(2): 180-193, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27364352

ABSTRACT

As obesity and metabolic diseases rise, there is need to investigate physiological and behavioural aspects associated with their development. Circadian rhythms have a profound influence on metabolic processes, as they prepare the body to optimise energy use and storage. Moreover, food-related signals confer temporal order to organs involved in metabolic regulation. Therefore food intake should be synchronised with the suprachiasmatic nucleus (SCN) to elaborate efficient responses to environmental challenges. Human studies suggest that a loss of synchrony between mealtime and the SCN promotes obesity and metabolic disturbances. Animal research using different paradigms has been performed to characterise the effects of timing of food intake on metabolic profiles. Therefore the purpose of the present review is to critically examine the evidence of animal studies, to provide a state of the art on metabolic findings and to assess whether the paradigms used in rodent models give the evidence to support a 'best time' for food intake. First we analyse and compare the current findings of studies where mealtime has been shifted out of phase from the light-dark cycle. Then, we analyse studies restricting meal times to different moments within the active period. So far animal studies correlate well with human studies, demonstrating that restricting food intake to the active phase limits metabolic disturbances produced by high-energy diets and that eating during the inactive/sleep phase leads to a worse metabolic outcome. Based on the latter we discuss the missing elements and possible mechanisms leading to the metabolic consequences, as these are still lacking.


Subject(s)
Circadian Rhythm , Eating , Obesity , Suprachiasmatic Nucleus/physiology , Animals , Humans , Motor Activity
11.
Br J Nutr ; 114(11): 1774-83, 2015 Dec 14.
Article in English | MEDLINE | ID: mdl-26395755

ABSTRACT

Obesity is one of the major health problems throughout the world. The present study investigated the preventive effect of epilactose--a rare non-digestible disaccharide--on obesity and metabolic disorders in mice fed high-fat (HF) diets. Feeding with HF diets increased body weight gain, fat pad weight and adipocyte size in mice (P<0·01), and these increases were effectively prevented by the use of supplemental epilactose without influencing food intake (P<0·01). Caecal pools of SCFA such as acetic and propionic acids in mice fed epilactose were higher compared with mice not receiving epilactose. Supplemental epilactose increased the expression of uncoupling protein (UCP)-1, which enhances energy expenditure, to 2-fold in the gastrocnemius muscle (P=0·04) and to 1·3-fold in the brown adipose tissue (P=0·02) in mice fed HF diets. Feeding HF diets induced pro-inflammatory macrophage infiltration into white adipose tissue, as indicated by the increased expression of monocyte chemotactic protein-1, TNF-α and F4/80, and these increases were attenuated by supplemental epilactose. In differentiated myogenic-like C2C12 cells, propionic acid, but not acetic or n-butyric acids, directly enhanced UCP-1 expression by approximately 2-fold (P<0·01). Taken together, these findings indicate that the epilactose-mediated increase in UCP-1 in the skeletal muscle and brown adipose tissue can enhance whole-body energy expenditure, leading to effective prevention of obesity and metabolic disorders in mice fed HF diets. It is suggested that propionic acid--a bacterial metabolite--acts as a mediator to induce UCP-1 expression in skeletal muscles.


Subject(s)
Anti-Obesity Agents/therapeutic use , Disaccharides/therapeutic use , Ion Channels/agonists , Mitochondrial Proteins/agonists , Muscle, Skeletal/metabolism , Obesity/prevention & control , Prebiotics , Up-Regulation , Adipose Tissue, Brown/immunology , Adipose Tissue, Brown/metabolism , Adipose Tissue, Brown/pathology , Adipose Tissue, White/immunology , Adipose Tissue, White/metabolism , Adipose Tissue, White/pathology , Animals , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Anti-Obesity Agents/metabolism , Cell Line , Diet, High-Fat/adverse effects , Disaccharides/metabolism , Energy Metabolism , Fermentation , Gastrointestinal Microbiome , Ion Channels/genetics , Ion Channels/metabolism , Macrophage Activation , Male , Mice, Inbred C57BL , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Muscle, Skeletal/immunology , Obesity/immunology , Obesity/metabolism , Obesity/microbiology , Propionates/metabolism , Random Allocation , Uncoupling Protein 1
12.
Br J Nutr ; 114(6): 866-72, 2015 Sep 28.
Article in English | MEDLINE | ID: mdl-26268732

ABSTRACT

n-3 Long-chain PUFA up-regulate intestinal lipid metabolism. However, whether these metabolic effects of PUFA on intestine are mediated by AMP-activated protein kinase (AMPK) remains to be elucidated. To determine the effects of α-linolenic acid (ALA) on intestinal fatty acid (FA) metabolism and whether these effects were affected by AMPK deletion, mice deficient in the catalytic subunit of AMPKα1 or AMPKα2 and wild-type (WT) mice were fed either a high-fat diet (HF) or HF supplemented with ALA (HF-A). The results showed that ALA supplementation decreased serum TAG content in WT mice. ALA also increased mRNA expression of genes (carnitine palmitoyltransferase 1a, acyl-CoA oxidase 1, medium-chain acyl-CoA dehydrogenase, cytochrome P450 4A10 and pyruvate dehydrogenase kinase isoenzyme 4a) involved in intestinal lipid oxidation and mRNA expression of TAG synthesis-related genes (monoacylglycerol O-acyltransferase 2, diacylglycerol O-acyltransferases 1 and 2) in WT mice. Consistent with these, expression levels of phosphorylated AMPKα1 and AMPKα2 were also increased in WT mice after ALA addition. However, in the absence of either AMPKα1 or AMPKα2, ALA supplementation failed to increase intestinal lipid oxidation. In addition, no significant effects of either diet (HF and HF-A) or genotype (WT, AMPKα1(-/-) and AMPKα2(-/-)) on FA uptake in the intestine and faecal TAG output were observed. Our results suggest that AMPK is indispensable for the effects of ALA on intestinal lipid oxidation.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Enzyme Induction , Intestinal Mucosa/metabolism , Lipid Metabolism , Up-Regulation , alpha-Linolenic Acid/therapeutic use , AMP-Activated Protein Kinases/genetics , Animals , Diet, High-Fat/adverse effects , Dietary Supplements , Feces/chemistry , Hypertriglyceridemia/blood , Hypertriglyceridemia/etiology , Hypertriglyceridemia/metabolism , Hypertriglyceridemia/prevention & control , Ileum/enzymology , Ileum/metabolism , Intestinal Mucosa/enzymology , Jejunum/enzymology , Jejunum/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Protein Processing, Post-Translational , Triglycerides/adverse effects , Triglycerides/analysis , Triglycerides/blood , Triglycerides/metabolism
13.
Br J Nutr ; 114(8): 1132-42, 2015 Oct 28.
Article in English | MEDLINE | ID: mdl-26285832

ABSTRACT

High-protein diets are known to reduce adiposity in the context of high carbohydrate and Western diets. However, few studies have investigated the specific high-protein effect on lipogenesis induced by a high-sucrose (HS) diet or fat deposition induced by high-fat feeding. We aimed to determine the effects of high protein intake on the development of fat deposition and partitioning in response to high-fat and/or HS feeding. A total of thirty adult male Wistar rats were assigned to one of the six dietary regimens with low and high protein, sucrose and fat contents for 5 weeks. Body weight (BW) and food intake were measured weekly. Oral glucose tolerance tests and meal tolerance tests were performed after 4th and 5th weeks of the regimen, respectively. At the end of the study, the rats were killed 2 h after ingestion of a calibrated meal. Blood, tissues and organs were collected for analysis of circulating metabolites and hormones, body composition and mRNA expression in the liver and adipose tissues. No changes were observed in cumulative energy intake and BW gain after 5 weeks of dietary treatment. However, high-protein diets reduced by 20 % the adiposity gain induced by HS and high-sucrose high-fat (HS-HF) diets. Gene expression and transcriptomic analysis suggested that high protein intake reduced liver capacity for lipogenesis by reducing mRNA expressions of fatty acid synthase (fasn), acetyl-CoA carboxylase a and b (Acaca and Acacb) and sterol regulatory element binding transcription factor 1c (Srebf-1c). Moreover, ketogenesis, as indicated by plasma ß-hydroxybutyrate levels, was higher in HS-HF-fed mice that were also fed high protein levels. Taken together, these results suggest that high-protein diets may reduce adiposity by inhibiting lipogenesis and stimulating ketogenesis in the liver.


Subject(s)
Adipose Tissue/metabolism , Diet, High-Fat/adverse effects , Dietary Proteins/administration & dosage , Dietary Sucrose/adverse effects , Lipogenesis , 3-Hydroxybutyric Acid/blood , Acetyl-CoA Carboxylase/genetics , Acetyl-CoA Carboxylase/metabolism , Adiposity , Animals , Blood Glucose/metabolism , Body Composition , Body Weight , Cholesterol, HDL/blood , Cholesterol, LDL/blood , Dietary Fats/administration & dosage , Dietary Sucrose/administration & dosage , Energy Intake , Fatty Acid Synthases/genetics , Fatty Acid Synthases/metabolism , Ghrelin/blood , Glucose Tolerance Test , Hypothalamus/metabolism , Leptin/blood , Liver/metabolism , Male , Muscle, Skeletal/metabolism , Rats , Rats, Wistar , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 1/metabolism , Triglycerides/blood
14.
Biochem Biophys Rep ; 28: 101168, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34825068

ABSTRACT

PURPOSE: This current study investigated the effect of metformin treatment on hepatic oxidative stress and inflammation associated with nonalcoholic fatty liver disease (NADLD) in high fat diet (HFD) fed rats. METHOD: Wistar rats were fed with a HFD or laboratory chow diet for 8 weeks. Metformin was administered orally at a dose of 200 mg/kg. Body weight, food and water intake were recorded on daily basis. Oral glucose tolerance test (OGTT), biochemical analysis and histological examinations were conducted on plasma and tissue samples. Antioxidant and anti-inflammatory mRNA expression was analyzed using reverse transcription polymeric chain reaction (RT-PCR). RESULTS: Metformin treatment for 8 weeks prevented HFD-induced weight gain and decreased fat deposition in HFD fed rats. Biochemical analysis revealed that metformin treatment significantly attenuated nitro-oxidative stress markers malondialdehyde (MDA), advanced protein oxidation product (APOP), and excessive nitric oxide (NO) levels in the liver of HFD fed rats. Gene expression analysis demonestrated that metformin treatment was associated with an enhanced expression of antioxidant genes such as Nrf-2, HO-1, SOD and catalase in liver of HFD fed rats. Metformin treatment also found to modulate the expression of fat metabolizing and anti-inflammatory genes including PPAR--γ, C/EBP-α, SREBP1c, FAS, AMPK and GLUT-4. Consistent with the biochemical and gene expression data, the histopathological examination unveiled that metformin treatment attenuated inflammatory cells infiltration, steatosis, hepatocyte necrosis, collagen deposition, and fibrosis in the liver of HFD fed rats. CONCLUSION: In conclusion, this study suggests that metformin might be effective in the prevention and treatment of HFD-induced steatosis by reducing hepatic oxidative stress and inflammation in the liver.

15.
Toxicol Rep ; 8: 846-862, 2021.
Article in English | MEDLINE | ID: mdl-33948438

ABSTRACT

Exposure to air pollution from traffic-generated sources is known to contribute to the etiology of inflammatory diseases, including cardiovascular disease (CVD) and obesity; however, the signaling pathways involved are still under investigation. Dysregulation of the renin-angiotensin system (RAS) can contribute to CVD and alter lipid storage and inflammation in adipose tissue. Our previous exposure studies revealed that traffic-generated emissions increase RAS signaling, further exacerbated by a high-fat diet. Thus, we investigated the hypothesis that exposure to engine emissions increases systemic and local adipocyte RAS signaling, promoting the expression of factors involved in CVD and obesity. Male C57BL/6 mice (6-8 wk old) were fed either a high-fat (HF, n = 16) or low-fat (LF, n = 16) diet, beginning 30d prior to exposures, and then exposed via inhalation to either filtered air (FA, controls) or a mixture of diesel engine + gasoline engine vehicle emissions (MVE: 100 µg PM/m3) via whole-body inhalation for 6 h/d, 7 d/wk, 30d. Endpoints were assessed via immunofluorescence and RT-qPCR. MVE-exposure promoted vascular adhesion factors (VCAM-1, ICAM-1) expression, monocyte/macrophage sequestration, and oxidative stress in the vasculature, associated with increased angiotensin II receptor type 1 (AT1) expression. In the kidney, MVE-exposure promoted the expression of renin, AT1, and AT2 receptors. In adipose tissue, both HF-diet and MVE-exposure mediated increased epididymal fat pad weight and adipocyte hypertrophy, associated with increased angiotensinogen and AT1 receptor expression; however, these outcomes were further exacerbated in the MVE + HF group. MVE-exposure also induced inflammation, monocyte chemoattractant protein (MCP)-1, and leptin, while reducing insulin receptor and glucose transporter, GLUT4, expression in adipose tissue. Our results indicate that MVE-exposure promotes systemic and local adipose RAS signaling, associated with increased expression of factors contributing to CVD and obesity, further exacerbated by HF diet consumption.

16.
Comput Struct Biotechnol J ; 18: 1914-1924, 2020.
Article in English | MEDLINE | ID: mdl-32774786

ABSTRACT

Circadian rhythms are 24-hour oscillations affecting an organism at multiple levels from gene expression all the way to tissues and organs. They have been observed in organisms across the kingdom of life, spanning from cyanobacteria to humans. In mammals, the master circadian pacemaker is located in the hypothalamic suprachiasmatic nuclei (SCN) in the brain where it synchronizes the peripheral oscillators that exist in other tissues. This system regulates the circadian activity of a large part of the transcriptome and recent findings indicate that almost every cell in the body has this clock at the molecular level. In this review, we briefly summarize the different factors that can influence the circadian transcriptome, including light, temperature, and food intake. We then summarize recently identified general principles governing genome-scale circadian regulation, as well as future lines of research. Genome-scale circadian activity represents a fascinating study model for computational biology. For this purpose, systems biology methods are promising exploratory tools to decode the global regulatory principles of circadian regulation.

17.
Cell Mol Gastroenterol Hepatol ; 5(3): 367-398, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29552625

ABSTRACT

BACKGROUND & AIMS: Nonalcoholic steatohepatitis (NASH) is associated with oxidative stress. We surmised that pharmacologic activation of NF-E2 p45-related factor 2 (Nrf2) using the acetylenic tricyclic bis(cyano enone) TBE-31 would suppress NASH because Nrf2 is a transcriptional master regulator of intracellular redox homeostasis. METHODS: Nrf2+/+ and Nrf2-/- C57BL/6 mice were fed a high-fat plus fructose (HFFr) or regular chow diet for 16 weeks or 30 weeks, and then treated for the final 6 weeks, while still being fed the same HFFr or regular chow diets, with either TBE-31 or dimethyl sulfoxide vehicle control. Measures of whole-body glucose homeostasis, histologic assessment of liver, and biochemical and molecular measurements of steatosis, endoplasmic reticulum (ER) stress, inflammation, apoptosis, fibrosis, and oxidative stress were performed in livers from these animals. RESULTS: TBE-31 treatment reversed insulin resistance in HFFr-fed wild-type mice, but not in HFFr-fed Nrf2-null mice. TBE-31 treatment of HFFr-fed wild-type mice substantially decreased liver steatosis and expression of lipid synthesis genes, while increasing hepatic expression of fatty acid oxidation and lipoprotein assembly genes. Also, TBE-31 treatment decreased ER stress, expression of inflammation genes, and markers of apoptosis, fibrosis, and oxidative stress in the livers of HFFr-fed wild-type mice. By comparison, TBE-31 did not decrease steatosis, ER stress, lipogenesis, inflammation, fibrosis, or oxidative stress in livers of HFFr-fed Nrf2-null mice. CONCLUSIONS: Pharmacologic activation of Nrf2 in mice that had already been rendered obese and insulin resistant reversed insulin resistance, suppressed hepatic steatosis, and mitigated against NASH and liver fibrosis, effects that we principally attribute to inhibition of ER, inflammatory, and oxidative stress.

18.
Proc Nutr Soc ; 76(4): 455-465, 2017 11.
Article in English | MEDLINE | ID: mdl-28514983

ABSTRACT

There is increasing evidence for important roles of key cognitive processes, including attention, memory and learning, in the short-term decision making about eating. There is parallel evidence that people who are overweight or obese tend to perform worse on a variety of cognitive tasks. In this review, the evidence for these two ideas is summarised and then the idea that overconsumption of Western-style high-fat (HF)-high-sugar diets may underlie the association between obesity and poorer cognitive performance is explored. In particular, evidence in animals and human subjects that repeated consumption of HF or HF and sugar (HFS) diets leads to specific impairments in the functioning of the hippocampus, which underpin the consequent changes in cognition is summarised. These findings lead into the vicious cycle model (VCM), which suggests that these cognitive changes have knock-on negative effects for future appetite control, and evidence that altered hippocampal function is also associated with impaired appetite control is explored. The review concludes that there is consistent evidence in the animal literature and emerging evidence from human studies that supports this VCM. It is also noted, however, that to date studies lack the nutritional specificity needed to be able to translate these basic research findings into clear nutritional effects, and concludes that there is an urgent need for additional research to clarify the precise nature of the apparent effects of consuming HFS diets on cognition.


Subject(s)
Cognitive Dysfunction/etiology , Dietary Fats/adverse effects , Dietary Sugars/adverse effects , Eating/psychology , Obesity/psychology , Animals , Cognition , Diet, High-Fat/adverse effects , Diet, High-Fat/psychology , Diet, Western/adverse effects , Diet, Western/psychology , Hippocampus/physiopathology , Humans , Obesity/etiology , Obesity/physiopathology
19.
Mol Metab ; 6(8): 873-881, 2017 08.
Article in English | MEDLINE | ID: mdl-28752051

ABSTRACT

OBJECTIVE: Dietary protein dilution (PD) has been associated with metabolic advantages such as improved glucose homeostasis and increased energy expenditure. This phenotype involves liver-induced release of FGF21 in response to amino acid insufficiency; however, it has remained unclear whether dietary dilution of specific amino acids (AAs) is also required. Circulating branched chain amino acids (BCAAs) are sensitive to protein intake, elevated in the serum of obese humans and mice and thought to promote insulin resistance. We tested whether replenishment of dietary BCAAs to an AA-diluted (AAD) diet is sufficient to reverse the glucoregulatory benefits of dietary PD. METHODS: We conducted AA profiling of serum from healthy humans and lean and high fat-fed or New Zealand obese (NZO) mice following dietary PD. We fed wildtype and NZO mice one of three amino acid defined diets: control, total AAD, or the same diet with complete levels of BCAAs (AAD + BCAA). We quantified serum AAs and characterized mice in terms of metabolic efficiency, body composition, glucose homeostasis, serum FGF21, and tissue markers of the integrated stress response (ISR) and mTORC1 signaling. RESULTS: Serum BCAAs, while elevated in serum from hyperphagic NZO, were consistently reduced by dietary PD in humans and murine models. Repletion of dietary BCAAs modestly attenuated insulin sensitivity and metabolic efficiency in wildtype mice but did not restore hyperglycemia in NZO mice. While hepatic markers of the ISR such as P-eIF2α and FGF21 were unabated by dietary BCAA repletion, hepatic and peripheral mTORC1 signaling were fully or partially restored, independent of changes in circulating glucose or insulin. CONCLUSIONS: Repletion of BCAAs in dietary PD is sufficient to oppose changes in somatic mTORC1 signaling but does not reverse the hepatic ISR nor induce insulin resistance in type 2 diabetes during dietary PD.


Subject(s)
Amino Acids, Branched-Chain/metabolism , Dietary Proteins/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Adult , Amino Acids, Branched-Chain/blood , Animals , Fibroblast Growth Factors/blood , Fibroblast Growth Factors/metabolism , Humans , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Signal Transduction
20.
Proc Nutr Soc ; 75(1): 90-95, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26621202

ABSTRACT

The importance of folate during pregnancy was established more than 80 years ago by Lucy Wills' ground-breaking studies of tropical macrocytic anaemia. More recently, it has become apparent that the adverse consequences of inadequate nutrient supply during early developmental may be exacerbated by over-nutrition postnatally. The present paper aims to review recent evidence that maternal methyl donor (notably folate) supply peri-conceptually and during pregnancy has long-term effects on offspring (metabolic) health. In addition, we propose the hypothesis that epigenetic mechanisms, especially DNA methylation, may mediate the effects of these early life nutritional insults. We discuss evidence from a natural experiment in human subjects which provides proof of principle for the hypothesis. We describe an attempt to test this hypothesis using a mouse model in which female C57Bl/6 mice were randomised to low or normal folate diets prior to, and during, pregnancy and lactation. Low maternal folate supply resulted in offspring that were more susceptible to detrimental metabolic effects of a high-fat diet fed from weaning, manifested as increased circulating TAG concentration. Interestingly, this metabolic phenotype in adult offspring occurred without any detectable change in adiposity, suggesting a different aetiological origin from the more commonly reported observation that maternal undernutrition leads to increased offspring adiposity and to symptoms of the Metabolic Syndrome. The widespread prevalence of overweight and obesity and of folate deficiency among women of child-bearing age highlights the possibility that this double nutritional insult may exacerbate the risk of metabolic disease in their offspring.

SELECTION OF CITATIONS
SEARCH DETAIL