Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Sci Rep ; 10(1): 2217, 2020 02 10.
Article in English | MEDLINE | ID: mdl-32041991

ABSTRACT

Blueberries are a rich source of polyphenols, widely studied for the prevention or attenuation of metabolic diseases. However, the health contribution and mechanisms of action of polyphenols depend on their type and structure. Here, we evaluated the effects of a wild blueberry polyphenolic extract (WBE) (Vaccinium angustifolium Aiton) on cardiometabolic parameters, gut microbiota composition and gut epithelium histology of high-fat high-sucrose (HFHS) diet-induced obese mice and determined which constitutive polyphenolic fractions (BPF) was responsible for the observed effects. To do so, the whole extract was separated in three fractions, F1) Anthocyanins and phenolic acids, F2) oligomeric proanthocyanidins (PACs), phenolic acids and flavonols (PACs degree of polymerization DP < 4), and F3) PACs polymers (PACs DP > 4) and supplied at their respective concentration in the whole extract. After 8 weeks, WBE reduced OGTT AUC by 18.3% compared to the HFHS treated rodents and the F3 fraction  contributed the most to this effect. The anthocyanin rich F1 fraction did not reproduce this response. WBE and the BPF restored the colonic mucus layer. Particularly, the polymeric PACs-rich F3 fraction increased the mucin-secreting goblet cells number. WBE caused a significant 2-fold higher proportion of Adlercreutzia equolifaciens whereas oligomeric PACs-rich F2 fraction increased by 2.5-fold the proportion of Akkermansia muciniphila. This study reveals the key role of WBE PACs in modulating the gut microbiota and restoring colonic epithelial mucus layer, providing a suitable ecological niche for mucosa-associated symbiotic bacteria, which may be crucial in triggering health effects of blueberry polyphenols.


Subject(s)
Blueberry Plants/chemistry , Gastrointestinal Microbiome/drug effects , Glucose Intolerance/drug therapy , Intestinal Mucosa/drug effects , Plant Extracts/administration & dosage , Proanthocyanidins/administration & dosage , Administration, Oral , Animals , Blood Glucose/analysis , Colon/drug effects , Colon/microbiology , Colon/pathology , Diet, High-Fat/adverse effects , Dietary Sucrose/adverse effects , Disease Models, Animal , Glucose/metabolism , Glucose Intolerance/etiology , Glucose Intolerance/metabolism , Glucose Intolerance/pathology , Humans , Insulin Resistance , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Male , Mice , Plant Extracts/chemistry
2.
PLoS One ; 14(3): e0213267, 2019.
Article in English | MEDLINE | ID: mdl-30845245

ABSTRACT

Resistin promotes hypothalamic neuroinflammation and insulin resistance through Toll like receptor 4 (TLR4), this hormone is thought to be a link between obesity and insulin-resistance. Indeed, resistin plasma levels are higher in obese and insulin resistant subjects. However, the impact of maternal resistin on the predisposition of offspring to hypothalamic neuroinflammation is unknown. Here, female mice were treated with resistin during gestation/lactation periods, then hypothalamic neuroinflammation was investigated in male offspring at p28 and p90. At p28, resistin increased the expression of inflammation markers (IL6, TNFα and NFκB) and TLR4 in the hypothalamus and decreased both hypothalamic insulin and leptin receptors' expression. The hypothalamic up-regulation IL6, TNFα and TLR4 was sustained until p90 promoting most likely hypothalamic inflammation. Maternal resistin also increased IL6 and TNFα in the adipose tissue of offspring at p90 associated with a higher body weight gain. In contrast, liver and muscle were not affected. These findings reveal that the augmentation of maternal resistin during gestation and lactation promotes hypothalamic and adipose tissue inflammation of offspring as evidenced by sustained increase of inflammation markers from weaning to adulthood. Thus, maternal resistin programs offspring hypothalamic and adipose tissue inflammation predisposing then offspring to body weight gain.


Subject(s)
Glucose Intolerance/etiology , Hypothalamus/immunology , Inflammation/etiology , Insulin Resistance , Insulinoma/etiology , Resistin/adverse effects , Weight Gain/drug effects , Animals , Animals, Newborn , Body Weight , Female , Glucose Intolerance/metabolism , Glucose Intolerance/pathology , Hypothalamus/drug effects , Hypothalamus/metabolism , Hypothalamus/pathology , Inflammation/metabolism , Inflammation/pathology , Inflammation Mediators/metabolism , Insulinoma/metabolism , Insulinoma/pathology , Lactation , Leptin/metabolism , Male , Maternal Nutritional Physiological Phenomena , Mice , Pregnancy , Resistin/administration & dosage , Weaning
3.
Amino Acids ; 51(4): 727-738, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30830312

ABSTRACT

Obesity in fathers leads to DNA damage and epigenetic changes in sperm that may carry potential risk factors for metabolic diseases to the next generation. Taurine (TAU) supplementation has demonstrated benefits against testicular dysfunction and pancreatic islet impairments induced by obesity, but it is not known if these protective actions prevent the propagation of metabolic disruptions to the next generation; as such, we hypothesized that paternal obesity may increase the probability of endocrine pancreatic dysfunction in offspring, and that this could be prevented by TAU supplementation in male progenitors. To test this, male C57Bl/6 mice were fed on a control diet (CTL) or a high-fat diet (HFD) without or with 5% TAU in their drinking water (CTAU and HTAU) for 4 months. Subsequently, all groups of mice were mated with CTL females, and the F1 offspring were identified as: CTL-F1, CTAU-F1, HFD-F1, and HTAU-F1. HFD-fed mice were normoglycemic, but glucose intolerant and their islets hypersecreted insulin. However, at 90 days of age, HFD-F1 offspring displayed normal glucose homeostasis and adiposity, but reduced glucose-induced insulin release. HFD-F1 islets also exhibited ß- and α-cell hypotrophy, and lower δ-cell number per islet. Paternal TAU supplementation prevented the decrease in glucose-induced insulin secretion and normalized ß-cell size and δ-cell number, and increased α-cell size/islet in HTAU-F1 mice. In conclusion, HFD consumption by male founders decreases ß-cell secretion and islet-cell distribution in their offspring. TAU attenuates the deleterious effects of paternal obesity on insulin secretion and islet-cell morphology in F1 offspring.


Subject(s)
Diet, High-Fat/adverse effects , Dietary Supplements , Endocrine System/drug effects , Glucose Intolerance/drug therapy , Islets of Langerhans/drug effects , Pancreatic Diseases/drug therapy , Taurine/administration & dosage , Animals , Endocrine System/physiopathology , Glucose Intolerance/etiology , Glucose Intolerance/pathology , Homeostasis , Insulin Secretion , Islets of Langerhans/physiopathology , Male , Mice , Mice, Inbred C57BL , Obesity/physiopathology , Pancreatic Diseases/etiology , Pancreatic Diseases/pathology
4.
J Biol Chem ; 294(13): 4946-4955, 2019 03 29.
Article in English | MEDLINE | ID: mdl-30709906

ABSTRACT

Hypoxia-inducible factor-2α (HIF2α) is a nuclear transcription factor that plays a critical role in cell survival including metabolic adaptation under hypoxia as well as normoxia, but whether HIF2α contributes to the control of whole-body metabolic balance is unclear. In this study, we found that the hypothalamic HIF2α protein level rapidly increases in young mice that are centrally stimulated with insulin. However, this insulin-induced HIF2α up-regulation is substantially attenuated in mice of advanced age. This attenuation is comparable with the effect of high-calorie feeding in young mice. Of note, unlike high-calorie feeding conditions, age-dependent HIF2α attenuation occurs without impaired activation of the hypothalamic IR/IRS-2/AKT/FOXO1 pathway in response to insulin. Molecular and physiological analyses revealed that hypothalamic HIF2α contributes to the action of central insulin in regulation of proopiomelanocortin (Pomc) gene expression and food intake. HIF2α knockout in POMC neurons led to age-dependent excess weight gain and fat increase, a phenotype that was associated with a mild degree of glucose intolerance and insulin resistance. In conclusion, hypothalamic HIF2α responds to insulin, and the up-regulation is involved in adaptive metabolic regulation as age increases, whereas impairment of HIF2α in the hypothalamus contributes to weight gain and glucose disorders in age-dependent manners.


Subject(s)
Aging/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Glucose Intolerance/metabolism , Hypothalamus/metabolism , Insulin/metabolism , Signal Transduction , Aging/genetics , Aging/pathology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Gene Expression Regulation , Glucose Intolerance/genetics , Glucose Intolerance/pathology , Hypothalamus/pathology , Insulin/genetics , Mice , Mice, Transgenic , Pro-Opiomelanocortin/biosynthesis , Pro-Opiomelanocortin/genetics
5.
Diabetes ; 67(8): 1512-1523, 2018 08.
Article in English | MEDLINE | ID: mdl-29793999

ABSTRACT

Altering the gut microbiome may be beneficial to the host and recently arose as a promising strategy to manage obesity. Here, we investigated the relative contribution of ω3 polyunsaturated fatty acid (PUFA)-mediated alterations in the microbiota to metabolic parameter changes in mice. Four groups were compared: male fat-1 transgenic mice (with constitutive production of ω3 PUFAs) and male wild-type (WT) littermates fed an obesogenic (high fat/high sucrose [HFHS]) or a control diet. Unlike WT mice, HFHS-fed fat-1 mice were protected against obesity, glucose intolerance, and hepatic steatosis. Unlike WT mice, fat-1 mice maintained a normal barrier function, resulting in a significantly lower metabolic endotoxemia. The fat-1 mice displayed greater phylogenic diversity in the cecum, and fecal microbiota transplantation from fat-1 to WT mice was able to reverse weight gain and to normalize glucose tolerance and intestinal permeability. We concluded that the ω3 PUFA-mediated alteration of gut microbiota contributed to the prevention of metabolic syndrome in fat-1 mice. It occurred independently of changes in the PUFA content of host tissues and may represent a promising strategy to prevent metabolic disease and preserve a lean phenotype.


Subject(s)
Fatty Acids, Omega-3/metabolism , Fecal Microbiota Transplantation , Gastrointestinal Microbiome , Glucose Intolerance/prevention & control , Insulin Resistance , Non-alcoholic Fatty Liver Disease/prevention & control , Obesity/prevention & control , Animals , Cadherins/genetics , Cadherins/metabolism , Diet, Carbohydrate Loading/adverse effects , Diet, High-Fat/adverse effects , Dietary Sucrose/adverse effects , Dysbiosis/microbiology , Dysbiosis/physiopathology , Dysbiosis/therapy , Endotoxemia/etiology , Endotoxemia/prevention & control , Fecal Microbiota Transplantation/adverse effects , Glucose Intolerance/microbiology , Glucose Intolerance/pathology , Glucose Intolerance/physiopathology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Intestinal Mucosa/physiopathology , Intestines/microbiology , Intestines/pathology , Intestines/physiopathology , Liver/metabolism , Liver/pathology , Male , Mice, Transgenic , Muscle, Skeletal/metabolism , Non-alcoholic Fatty Liver Disease/microbiology , Non-alcoholic Fatty Liver Disease/pathology , Non-alcoholic Fatty Liver Disease/physiopathology , Obesity/microbiology , Obesity/pathology , Obesity/physiopathology , Permeability , Phylogeny
6.
J Biol Chem ; 293(11): 4159-4166, 2018 03 16.
Article in English | MEDLINE | ID: mdl-29374061

ABSTRACT

The responsiveness of glucose sensing per se to regulate whole-body glucose homeostasis is dependent on the ability of a rise in glucose to lower hepatic glucose production and increase peripheral glucose uptake in vivo In both rodents and humans, glucose sensing is lost in diabetes and obesity, but the site(s) of impairment remains elusive. Here, we first report that short-term high-fat feeding disrupts hypothalamic glucose sensing to lower glucose production in rats. Second, leptin administration into the hypothalamus of high-fat-fed rats restored hypothalamic glucose sensing to lower glucose production during a pancreatic (basal insulin)-euglycemic clamp and increased whole-body glucose tolerance during an intravenous glucose tolerance test. Finally, both chemical inhibition of hypothalamic lactate dehydrogenase (LDH) (achieved via hypothalamic LDH inhibitor oxamate infusion) and molecular knockdown of LDHA (achieved via hypothalamic lentiviral LDHA shRNA injection) negated the ability of hypothalamic leptin infusion to enhance glucose sensing to lower glucose production in high fat-fed rats. In summary, our findings illustrate that leptin enhances LDHA-dependent glucose sensing in the hypothalamus to lower glucose production in high-fat-fed rodents in vivo.


Subject(s)
Diet, High-Fat/adverse effects , Gene Expression Regulation, Enzymologic/drug effects , Glucose Intolerance/prevention & control , Glucose/metabolism , Hypothalamus/enzymology , L-Lactate Dehydrogenase/metabolism , Leptin/pharmacology , Animals , Glucose Intolerance/etiology , Glucose Intolerance/pathology , Glucose Tolerance Test , Homeostasis , Insulin Resistance , Male , Rats , Rats, Sprague-Dawley
7.
Sci Rep ; 7(1): 12770, 2017 10 06.
Article in English | MEDLINE | ID: mdl-28986580

ABSTRACT

To assess the metabolically beneficial effects of fenugreek (Trigonella foenum-graecum), C57BL/6J mice were fed a low- or high-fat diet for 16 weeks with or without 2% (w/w) fenugreek supplementation. Body weight, body composition, energy expenditure, food intake, and insulin/glucose tolerance were measured regularly, and tissues were collected for histological and biochemical analysis after 16 weeks of diet exposure. Fenugreek did not alter body weight, fat mass, or food intake in either group, but did transiently improve glucose tolerance in high fat-fed mice. Fenugreek also significantly improved high-density lipoprotein to low-density lipoprotein ratios in high fat-fed mice without affecting circulating total cholesterol, triglycerides, or glycerol levels. Fenugreek decreased hepatic expression of fatty acid-binding protein 4 and increased subcutaneous inguinal adipose tissue expression of adiponectin, but did not prevent hepatic steatosis. Notably, fenugreek was not as effective at improving glucose tolerance as was four days of voluntary wheel running. Overall, our results demonstrate that fenugreek promotes metabolic resiliency via significant and selected effects on glucose regulation, hyperlipidemia, and adipose pathology; but may not be as effective as behavioral modifications at preventing the adverse metabolic consequences of a high fat diet.


Subject(s)
Biomarkers/metabolism , Diet, High-Fat , Dietary Supplements , Feeding Behavior , Health , Metabolism , Trigonella/chemistry , Adiponectin/metabolism , Adipose Tissue/metabolism , Adiposity , Animals , Blood Glucose/metabolism , Body Weight , Epididymis/metabolism , Fatty Acid Synthases/metabolism , Fatty Acid-Binding Proteins/metabolism , Fatty Acids/metabolism , Glucose Intolerance/blood , Glucose Intolerance/pathology , Inflammation/pathology , Insulin/blood , Insulin Resistance , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Lipoproteins, HDL/blood , Lipoproteins, LDL/blood , Male , Mice, Inbred C57BL , Physical Conditioning, Animal , Triglycerides/blood
8.
Diabetes ; 66(10): 2555-2563, 2017 10.
Article in English | MEDLINE | ID: mdl-28710138

ABSTRACT

Neuronal circuits in the brain help to control feeding behavior and systemic metabolism in response to afferent nutrient and hormonal signals. Although astrocytes have historically been assumed to have little relevance for such neuroendocrine control, we investigated whether lipid uptake via lipoprotein lipase (LPL) in astrocytes is required to centrally regulate energy homeostasis. Ex vivo studies with hypothalamus-derived astrocytes showed that LPL expression is upregulated by oleic acid, whereas it is decreased in response to palmitic acid or triglycerides. Likewise, astrocytic LPL deletion reduced the accumulation of lipid droplets in those glial cells. Consecutive in vivo studies showed that postnatal ablation of LPL in glial fibrillary acidic protein-expressing astrocytes induced exaggerated body weight gain and glucose intolerance in mice exposed to a high-fat diet. Intriguingly, astrocytic LPL deficiency also triggered increased ceramide content in the hypothalamus, which may contribute to hypothalamic insulin resistance. We conclude that hypothalamic LPL functions in astrocytes to ensure appropriately balanced nutrient sensing, ceramide distribution, body weight regulation, and glucose metabolism.


Subject(s)
Astrocytes/metabolism , Diet, High-Fat/adverse effects , Obesity/etiology , Obesity/metabolism , Animals , Astrocytes/cytology , Body Weight/physiology , Ceramides/metabolism , Flow Cytometry , Glucose/metabolism , Glucose Intolerance/metabolism , Glucose Intolerance/pathology , Glucose Tolerance Test , Humans , Hypothalamus/cytology , Immunohistochemistry , In Situ Hybridization , Lipoprotein Lipase/genetics , Lipoprotein Lipase/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/pathology , Real-Time Polymerase Chain Reaction
9.
Biol Trace Elem Res ; 180(2): 223-232, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28378114

ABSTRACT

The objective of this study was to investigate the effects of peroral administration of chromium-enriched yeast on glucose tolerance in Holstein calves, assessed by insulin signaling pathway molecule determination and intravenous glucose tolerance test (IVGTT). Twenty-four Holstein calves, aged 1 month, were chosen for the study and divided into two groups: the PoCr group (n = 12) that perorally received 0.04 mg of Cr/kg of body mass daily, for 70 days, and the NCr group (n = 12) that received no chromium supplementation. Skeletal tissue samples from each calf were obtained on day 0 and day 70 of the experiment. Chromium supplementation increased protein content of the insulin ß-subunit receptor, phosphorylation of insulin receptor substrate 1 at Tyrosine 632, phosphorylation of Akt at Serine 473, glucose transporter-4, and AMP-activated protein kinase in skeletal muscle tissue, while phosphorylation of insulin receptor substrate 1 at Serine 307 was not affected by chromium treatment. Results obtained during IVGTT, which was conducted on days 0, 30, 50, and 70, suggested an increased insulin sensitivity and, consequently, a better utilization of glucose in the PoCr group. Lower basal concentrations of glucose and insulin in the PoCr group on days 30 and 70 were also obtained. Our results indicate that chromium supplementation improves glucose utilization in calves by enhancing insulin intracellular signaling in the skeletal muscle tissue.


Subject(s)
Animal Nutritional Physiological Phenomena , Chromium/therapeutic use , Glucose Intolerance/veterinary , Insulin Resistance , Muscle, Skeletal/metabolism , Signal Transduction , Yeast, Dried/therapeutic use , Animals , Animals, Inbred Strains , Biopsy/veterinary , Cattle , Cyclic AMP-Dependent Protein Kinases/chemistry , Cyclic AMP-Dependent Protein Kinases/metabolism , Dairying , Female , Glucose Intolerance/metabolism , Glucose Intolerance/pathology , Glucose Intolerance/prevention & control , Glucose Transporter Type 4/agonists , Glucose Transporter Type 4/metabolism , Hamstring Muscles , Insulin Receptor Substrate Proteins/agonists , Insulin Receptor Substrate Proteins/metabolism , Muscle, Skeletal/growth & development , Muscle, Skeletal/pathology , Phosphorylation , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-akt/agonists , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Insulin/agonists , Receptor, Insulin/metabolism , Weaning
10.
Clin Sci (Lond) ; 131(8): 673-687, 2017 Apr 25.
Article in English | MEDLINE | ID: mdl-28188238

ABSTRACT

The aim of the present study was to demonstrate the role of autophagy and incretins in the fructose-induced alteration of ß-cell mass and function. Normal Wistar rats were fed (3 weeks) with a commercial diet without (C) or with 10% fructose in drinking water (F) alone or plus sitagliptin (CS and FS) or exendin-4 (CE and FE). Serum levels of metabolic/endocrine parameters, ß-cell mass, morphology/ultrastructure and apoptosis, vacuole membrane protein 1 (VMP1) expression and glucose-stimulated insulin secretion (GSIS) were studied. Complementary to this, islets isolated from normal rats were cultured (3 days) without (C) or with F and F + exendin-4 or chloroquine. Expression of autophagy-related proteins [VMP1 and microtubule-associated protein light chain 3 (LC3)], apoptotic/antiapoptotic markers (caspase-3 and Bcl-2), GSIS and insulin mRNA levels were measured. F rats developed impaired glucose tolerance (IGT) and a significant increase in plasma triacylglycerols, thiobarbituric acid-reactive substances, insulin levels, homoeostasis model assessment (HOMA) for insulin resistance (HOMA-IR) and ß-cell function (HOMA-ß) indices. A significant reduction in ß-cell mass was associated with an increased apoptotic rate and morphological/ultrastructural changes indicative of autophagic activity. All these changes were prevented by either sitagliptin or exendin-4. In cultured islets, F significantly enhanced insulin mRNA and GSIS, decreased Bcl-2 mRNA levels and increased caspase-3 expression. Chloroquine reduced these changes, suggesting the participation of autophagy in this process. Indeed, F induced the increase of both VMP1 expression and LC3-II, suggesting that VMP1-related autophagy is activated in injured ß-cells. Exendin-4 prevented islet-cell damage and autophagy development. VMP1-related autophagy is a reactive process against F-induced islet dysfunction, being prevented by exendin-4 treatment. This knowledge could help in the use of autophagy as a potential target for preventing progression from IGT to type 2 diabetes mellitus.


Subject(s)
Autophagy/drug effects , Diet/adverse effects , Fructose/pharmacology , Incretins/pharmacology , Insulin-Secreting Cells/drug effects , Membrane Proteins/physiology , Animals , Autophagy/physiology , Body Weight , Cells, Cultured , Drug Evaluation, Preclinical/methods , Energy Intake , Exenatide , Fructose/administration & dosage , Glucose Intolerance/etiology , Glucose Intolerance/pathology , Glucose Intolerance/prevention & control , Glucose Tolerance Test , Hypoglycemic Agents/pharmacology , Insulin/biosynthesis , Insulin/genetics , Insulin-Secreting Cells/ultrastructure , Male , Microscopy, Electron , Peptides/pharmacology , RNA, Messenger/genetics , Rats, Wistar , Sitagliptin Phosphate/pharmacology , Venoms/pharmacology
11.
Bone ; 97: 175-183, 2017 04.
Article in English | MEDLINE | ID: mdl-28126632

ABSTRACT

Obesity, diabetes and osteoporosis have become a major public heath burden, and understanding the underlying mechanisms of these pathophysiological process will benefit their treatment. Osteoblast lineage cells in charge of the bone formation have been showed to participate in the whole-body energy metabolism. In this study, we identify that wnt/ß-catenin signaling in osteoblasts could regulate global energy metabolism, including glucose homeostasis, fat accumulation and energy expenditure. Mice lacking ß-catenin specifically in osteoblasts postnatally exhibit decreased bone mass, increased glucose level, decreased insulin production, decreased fat accumulation and increased energy expenditure. Osteocalcin supplement can rescue the impaired glucose balance by improving insulin production but cannot influence the abnormal fat accumulation and energy expenditure. Osteoprotegerin (OPG) overexpression exclusively in osteoblasts in ß-catenin deletion mice can normalize not only the decreased bone mass but also the decreased fat accumulation and increased energy expenditure. The effect of ß-catenin deletion and OPG overexpression in osteoblasts on global energy metabolism had no relation with inguinal fat browning. These results suggest that the regulation of bone on energy metabolism and fat accumulation is not mediated exclusively by osteocalcin. Our findings may provide a new insight into the regulation of bone on fat accumulation and energy metabolism.


Subject(s)
Energy Metabolism , Osteoblasts/metabolism , Wnt Signaling Pathway , Adipose Tissue, Brown/pathology , Adiposity , Animals , Bone and Bones/pathology , Glucose Intolerance/metabolism , Glucose Intolerance/pathology , Homeostasis , Insulin/metabolism , Mice, Inbred C57BL , Mice, Knockout , Organ Size , Osteocalcin/metabolism , Osteoprotegerin/metabolism , beta Catenin/metabolism
12.
Biochim Biophys Acta ; 1862(8): 1375-82, 2016 08.
Article in English | MEDLINE | ID: mdl-27112275

ABSTRACT

AIM: Acylcarnitines are fatty acid oxidation (FAO) intermediates, which have been implicated in diet-induced insulin resistance. Elevated acylcarnitine levels are found in obese, insulin resistant humans and rodents, and coincide with lower free carnitine. We hypothesized that increasing free carnitine levels by administration of the carnitine precursor γ-butyrobetaine (γBB) could facilitate FAO, thereby improving insulin sensitivity. METHODS: C57BL/6N mice were fed with a high fat or chow diet with or without γBB supplementation (n=10 per group). After 8weeks of diet, indirect calorimetry, glucose tolerance and insulin sensitivity tests were performed. AC profiles and carnitine biosynthesis intermediates were analyzed in plasma and tissues by tandem mass spectrometry (MS) and liquid chromatography tandem MS. RESULTS: γBB supplementation did not facilitate FAO, was unable to curb bodyweight and did not prevent impaired glucose homeostasis in the HFD fed mice in spite of marked alterations in the acylcarnitine profiles in plasma and liver. Remarkably, γBB did not affect the acylcarnitine profile in other tissues, most notably muscle. Administration of a bolus acetylcarnitine also caused significant changes in plasma and liver, but not in muscle acylcarnitine profiles, again without effect on glucose tolerance. CONCLUSION: Altogether, increasing carnitine availability affects acylcarnitine profiles in plasma and liver but does not modulate glucose tolerance or insulin sensitivity. This may be due to the lack of an effect on muscle acylcarnitine profiles, as muscle tissue is an important contributor to whole body insulin sensitivity. These results warrant caution on making associations between plasma acylcarnitine levels and insulin resistance.


Subject(s)
Carnitine/analogs & derivatives , Energy Metabolism , Glucose Intolerance/blood , Insulin Resistance , Obesity/blood , Animals , Betaine/analogs & derivatives , Betaine/pharmacology , Carnitine/blood , Carnitine/pharmacology , Dietary Fats/adverse effects , Dietary Fats/pharmacology , Glucose Intolerance/chemically induced , Glucose Intolerance/pathology , Liver/metabolism , Liver/pathology , Mice , Mice, Obese , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Obesity/chemically induced , Obesity/pathology
13.
Can J Physiol Pharmacol ; 94(5): 488-97, 2016 May.
Article in English | MEDLINE | ID: mdl-26871756

ABSTRACT

This study investigates the effect of the ergogenic supplement ß-hydroxy-ß-methylbutyrate (HMB) on insulin resistance induced by high-fructose diet (HFD) in rats. Male Sprague Dawley rats were fed 60% HFD for 12 weeks and HMB (320 mg·kg(-1)·day(-1), orally) for 4 weeks. HFD significantly increased fasting insulin, fasting glucose, glycosylated hemoglobin (HBA1C), liver glycogen content, and homeostasis model assessment of insulin resistance (HOMA-IR) index, while it decreased glucose and insulin tolerance. Furthermore, HFD significantly increased serum triglycerides (TG), low density lipoprotein cholesterol (LDL-C), and very low density lipoprotein cholesterol (VLDL-C) levels, while it significantly decreased high density lipoprotein cholesterol (HDL-C). Moreover, HFD significantly increased mRNA expression of glucose transporter type-2 (GLUT-2), the mammalian target of rapamycin (mTOR), and sterol regulatory element-binding protein-1c (SREBP-1c) but decreased peroxisome proliferator-activated receptor-alpha (PPAR-α) in liver. Aortic relaxation to acetylcholine (ACh) was impaired and histopathology showed severe hepatic steatosis. HMB significantly increased insulin tolerance and decreased fasting insulin, HOMA-IR, HBA1C, hepatic glycogen content, serum TG, LDL-C, and VLDL-C. Additionally, HMB enhanced ACh-induced relaxation, ameliorated hepatic steatosis, and decreased mRNA expression of GLUT-2. In conclusion, HMB may attenuate insulin resistance and hepatic steatosis through inhibiting GLUT-2 in liver.


Subject(s)
Dietary Supplements , Glucose Transporter Type 2/antagonists & inhibitors , Insulin Resistance , Liver/metabolism , Non-alcoholic Fatty Liver Disease/prevention & control , Performance-Enhancing Substances/therapeutic use , Valerates/therapeutic use , Animals , Dietary Carbohydrates/adverse effects , Endothelium, Vascular/physiopathology , Fructose/adverse effects , Gene Expression Regulation , Glucose Intolerance/metabolism , Glucose Intolerance/pathology , Glucose Intolerance/physiopathology , Glucose Intolerance/prevention & control , Glucose Transporter Type 2/genetics , Glucose Transporter Type 2/metabolism , Hyperinsulinism/metabolism , Hyperinsulinism/pathology , Hyperinsulinism/physiopathology , Hyperinsulinism/prevention & control , Hyperlipidemias/metabolism , Hyperlipidemias/pathology , Hyperlipidemias/physiopathology , Hyperlipidemias/prevention & control , Liver/pathology , Male , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Non-alcoholic Fatty Liver Disease/physiopathology , PPAR alpha/metabolism , Random Allocation , Rats, Sprague-Dawley , Sterol Regulatory Element Binding Protein 1/metabolism , TOR Serine-Threonine Kinases/metabolism , Vascular Resistance
14.
Endocr J ; 62(6): 531-42, 2015.
Article in English | MEDLINE | ID: mdl-25877295

ABSTRACT

Tissue-selective estrogen complex (TSEC), which combines a selective estrogen receptor modulator (SERM) with one or more estrogens, is a novel approach to menopausal therapy. It has been demonstrated that the phytoestrogen genistein (GEN) exhibits mixed estrogen receptor agonist and antagonist activity, suggesting that GEN may have potential for use as a natural SERM. We evaluated, for the first time, the effects of GEN, conjugated estrogens (CE), and their pairing effects as a TSEC treatment on estrogen-induced endometrial hyperplasia and metabolic dysfunction in ovariectomized (OVX) mice fed a high-fat diet. CE replacement prevented fat accumulation in the adipose tissue and liver, improved glucose homeostasis, and induced endometrial hyperplasia in OVX mice. GEN at 100 mg/kg showed CE mimetic effects in preventing ovariectomy-induced metabolic dysfunctions without endometrial stimulation. Combination treatments with CE and GEN prevented metabolic dysfunctions more strongly than CE alone, but at both low and high doses, GEN did not reverse CE-induced endometrial hyperplasia. In addition, we found that in a TSEC regimen, a typical SERM raloxifene maintains the metabolic benefits of CE while simultaneously protecting the endometrium in OVX mice. These findings indicate that GEN acts as an estrogen agonist in metabolic regulation, but has no SERM function in the uteri of OVX mice.


Subject(s)
Dietary Supplements , Endometrial Hyperplasia/prevention & control , Estrogen Replacement Therapy , Estrogens, Conjugated (USP)/therapeutic use , Genistein/therapeutic use , Glucose Intolerance/prevention & control , Phytoestrogens/therapeutic use , Adiposity/drug effects , Animals , Diet, High-Fat/adverse effects , Dietary Supplements/adverse effects , Endometrial Hyperplasia/chemically induced , Endometrial Hyperplasia/metabolism , Endometrial Hyperplasia/pathology , Endometrium/drug effects , Endometrium/metabolism , Endometrium/pathology , Estrogen Replacement Therapy/adverse effects , Estrogens/adverse effects , Estrogens/therapeutic use , Estrogens, Conjugated (USP)/adverse effects , Female , Genistein/administration & dosage , Genistein/adverse effects , Glucose Intolerance/chemically induced , Glucose Intolerance/metabolism , Glucose Intolerance/pathology , Liver/drug effects , Liver/metabolism , Liver/pathology , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/chemically induced , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Non-alcoholic Fatty Liver Disease/prevention & control , Ovariectomy/adverse effects , Overweight/etiology , Overweight/metabolism , Overweight/pathology , Overweight/prevention & control , Phytoestrogens/administration & dosage , Phytoestrogens/adverse effects , Raloxifene Hydrochloride/adverse effects , Raloxifene Hydrochloride/therapeutic use , Random Allocation , Selective Estrogen Receptor Modulators/adverse effects , Selective Estrogen Receptor Modulators/therapeutic use
15.
PLoS One ; 10(3): e0119843, 2015.
Article in English | MEDLINE | ID: mdl-25774877

ABSTRACT

Besides regulating calcium homeostasis, the effects of vitamin D on vascular tone and metabolic disturbances remain scarce in the literature despite an increase intake with high-fructose corn syrup worldwide. We investigated the effects of calcitriol, an active form of vitamin D, on vascular relaxation, glucose tolerance, and visceral fat pads in fructose-fed rats. Male Wistar-Kyoto rats were divided into 4 groups (n = 6 per group). Group Con: standard chow diet for 8 weeks; Group Fru: high-fructose diet (60% fructose) for 8 weeks; Group Fru-HVD: high-fructose diet as Group Fru, high-dose calcitriol treatment (20 ng / 100 g body weight per day) 4 weeks after the beginning of fructose feeding; and Group Fru-LVD: high-fructose diet as Group Fru, low-dose calcitriol treatment (10 ng / 100 g body weight per day) 4 weeks after the beginning of fructose feeding. Systolic blood pressure was measured twice a week by the tail-cuff method. Blood was examined for serum ionized calcium, phosphate, creatinine, glucose, triglycerides, and total cholesterol. Intra-peritoneal glucose intolerance test, aortic vascular reactivity, the weight of visceral fat pads, adipose size, and adipose angiotensin II levels were analyzed at the end of the study. The results showed that the fructose-fed rats significantly developed hypertension, impaired glucose tolerance, heavier weight and larger adipose size of visceral fat pads, and raised adipose angiotensin II expressions compared with the control rats. High- and low-dose calcitriol reduced modestly systolic blood pressure, increased endothelium-dependent aortic relaxation, ameliorated glucose intolerance, reduced the weight and adipose size of visceral fat pads, and lowered adipose angiotensin II expressions in the fructose-fed rats. However, high-dose calcitriol treatment mildly increased serum ionized calcium levels (1.44 ± 0.05 mmol/L). These results suggest a protective role of calcitriol treatment on endothelial function, glucose tolerance, and visceral adiposity in fructose-fed rats.


Subject(s)
Calcitriol/administration & dosage , Fructose/adverse effects , Glucose Intolerance/drug therapy , Hypertension/drug therapy , Obesity, Abdominal/drug therapy , Vitamins/administration & dosage , Adiposity/drug effects , Angiotensin II/metabolism , Animals , Blood Pressure/drug effects , Body Weight/drug effects , Calcitriol/pharmacology , Dose-Response Relationship, Drug , Glucose Intolerance/chemically induced , Glucose Intolerance/pathology , Glucose Tolerance Test , Hypertension/chemically induced , Hypertension/pathology , Male , Obesity, Abdominal/chemically induced , Obesity, Abdominal/pathology , Rats , Rats, Inbred WKY , Vasodilation/drug effects , Vitamins/pharmacology
16.
J Sci Food Agric ; 95(2): 401-8, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24817112

ABSTRACT

BACKGROUND: Rhizoma dioscorea, Lycium barbarum, Prunella vulgaris and hawthorn are well known in both traditional food and folk medicine. Each of these plants reportedly possesses beneficial effects in the treatment of diabetes. In this study an anti-diabetic health-promoting diet was formulated by mixing the herbs in a ratio of 6:4:2:3, and the anti-diabetic effect and underlying mechanism were elucidated in vivo. RESULTS: Compared with the model control group, the formula, especially its ethanol extract (EF), could improve glucose intolerance and normalize the lipid profile. The mechanisms responsible for the amelioration of glucose and lipid metabolism in mice were an increase in peripheral and hepatic insulin sensitivity, a decrease in serum free fatty acid level, enhanced hepatic glucokinase activity and glycogen content and improved serum antioxidant activity. Hepatic histopathological examination also showed that EF administration markedly decreased fatty deposits in the liver of mice. CONCLUSION: The results of the present study demonstrated that the prepared functional formula diet is a potent alternative as an anti-diabetic health-promoting diet.


Subject(s)
Blood Glucose/metabolism , Diabetes Mellitus, Experimental/drug therapy , Hypoglycemic Agents/therapeutic use , Lipids/blood , Magnoliopsida , Phytotherapy , Plant Extracts/therapeutic use , Adipose Tissue/metabolism , Animals , Antioxidants/metabolism , Crataegus , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diet, High-Fat , Dioscorea , Fatty Liver/metabolism , Fatty Liver/prevention & control , Fructose/adverse effects , Glucose Intolerance/drug therapy , Glucose Intolerance/metabolism , Glucose Intolerance/pathology , Hypoglycemic Agents/pharmacology , Insulin/metabolism , Insulin Resistance , Liver/drug effects , Liver/metabolism , Liver/pathology , Lycium , Male , Mice, Inbred ICR , Plant Extracts/pharmacology , Prunella
17.
Eur J Nutr ; 53(3): 761-8, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24005871

ABSTRACT

PURPOSE: There is an increased interest in the benefits of conjugated α-linolenic acid (CLNA) on obesity-related complications such as insulin resistance and diabetes. The aim of the study was to investigate whether a 1% dietary supplementation of mono-CLNA isomers (c9-t11-c15-18:3 + c9-t13-c15-18:3) improved glucose and lipid metabolism in neonatal pigs. METHODS: Since mono-CLNA isomers combine one conjugated two-double-bond system with an n-3 polyunsaturated fatty acid (PUFA) structure, the experimental protocol was designed to isolate the dietary structural characteristics of the molecules by comparing a CLNA diet with three other dietary fats: (1) conjugated linoleic acid (c9-t11-18:2 + t10-c12-18:2; CLA), (2) non-conjugated n-3 PUFA, and (3) n-6 PUFA. Thirty-two piglets weaned at 3 weeks of age were distributed among the four dietary groups. Diets were isoenergetic and food intake was controlled by a gastric tube. After 2 weeks of supplementation, gastro-enteral (OGTT) and parenteral (IVGTT) glucose tolerance tests were conducted. RESULTS: Dietary supplementation with mono-CLNA did not modify body weight/fat or blood lipid profiles (p > 0.82 and p > 0.57, respectively) compared with other dietary groups. Plasma glucose, insulin, and C-peptide responses to OGTT and IVGTT in the CLNA group were not different from the three other dietary groups (p > 0.18 and p > 0.15, respectively). Compared to the non-conjugated n-3 PUFA diet, CLNA-fed animals had decreased liver composition in three n-3 fatty acids (18:3n-3; 20:3n-3; 22:5n-3; p < 0.001). CONCLUSIONS: These results suggest that providing 1% mono-CLNA is not effective in improving insulin sensitivity in neonatal pigs.


Subject(s)
Dietary Supplements , Disease Models, Animal , Glucose Intolerance/prevention & control , Insulin Resistance , Lipid Metabolism , Liver/metabolism , alpha-Linolenic Acid/therapeutic use , Animals , Canada , Crosses, Genetic , Dietary Supplements/analysis , Emulsions , Fatty Acids, Unsaturated/analysis , Fatty Acids, Unsaturated/chemistry , Fatty Acids, Unsaturated/metabolism , Fatty Acids, Unsaturated/therapeutic use , Female , Glucose Intolerance/blood , Glucose Intolerance/metabolism , Glucose Intolerance/pathology , Liver/pathology , Male , Orchiectomy/veterinary , Random Allocation , Stereoisomerism , Sus scrofa , Weaning , Weight Gain , alpha-Linolenic Acid/analysis , alpha-Linolenic Acid/chemistry , alpha-Linolenic Acid/metabolism
18.
Naunyn Schmiedebergs Arch Pharmacol ; 386(8): 733-45, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23620335

ABSTRACT

Green tea extract (GTE) is regarded to be effective against obesity and type 2 diabetes, but definitive evidences have not been proven. Based on the assumption that the gallated catechins (GCs) in GTE attenuate intestinal glucose and lipid absorption, while enhancing insulin resistance when GCs are present in the circulation through inhibiting cellular glucose uptake in various tissues, this study attempted to block the intestinal absorption of GCs and prolong their residence time in the lumen. We then observed whether GTE containing the nonabsorbable GCs could ameliorate body weight (BW) gain and glucose intolerance in db/db and high-fat diet mice. Inhibition of the intestinal absorption of GCs was accomplished by co-administering the nontoxic polymer polyethylene glycol-3350 (PEG). C57BLKS/J db/db and high-fat diet C57BL/6 mice were treated for 4 weeks with drugs as follows: GTE, PEG, GTE+PEG, voglibose, or pioglitazone. GTE mixed with meals did not have any ameliorating effects on BW gain and glucose intolerance. However, the administration of GTE plus PEG significantly reduced BW gain, insulin resistance, and glucose intolerance, without affecting food intake and appetite. The effect was comparable to the effects of an α-glucosidase inhibitor and a peroxisome proliferator-activated receptor-γ/α agonist. These results indicate that prolonging the action of GCs of GTE in the intestinal lumen and blocking their entry into the circulation may allow GTE to be used as a prevention and treatment for both obesity and obesity-induced type 2 diabetes.


Subject(s)
Anti-Obesity Agents/administration & dosage , Camellia sinensis , Diabetes Mellitus, Type 2/drug therapy , Obesity/drug therapy , Plant Extracts/administration & dosage , Polyethylene Glycols/administration & dosage , Adiponectin/blood , Adipose Tissue/drug effects , Adipose Tissue/pathology , Animals , Caco-2 Cells , Catechin/analogs & derivatives , Catechin/metabolism , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/pathology , Diet, High-Fat , Glucose/metabolism , Glucose Intolerance/blood , Glucose Intolerance/drug therapy , Glucose Intolerance/pathology , Humans , Insulin Resistance , Male , Mice , Mice, Inbred C57BL , Obesity/blood , Obesity/pathology , Organ Size/drug effects , Retinol-Binding Proteins, Plasma/metabolism
19.
J Clin Invest ; 122(11): 4172-89, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23093779

ABSTRACT

Long-term glucocorticoid treatment is associated with numerous adverse outcomes, including weight gain, insulin resistance, and diabetes; however, the pathogenesis of these side effects remains obscure. Glucocorticoids also suppress osteoblast function, including osteocalcin synthesis. Osteocalcin is an osteoblast-specific peptide that is reported to be involved in normal murine fuel metabolism. We now demonstrate that osteoblasts play a pivotal role in the pathogenesis of glucocorticoid-induced dysmetabolism. Osteoblast-targeted disruption of glucocorticoid signaling significantly attenuated the suppression of osteocalcin synthesis and prevented the development of insulin resistance, glucose intolerance, and abnormal weight gain in corticosterone-treated mice. Nearly identical effects were observed in glucocorticoid-treated animals following heterotopic (hepatic) expression of both carboxylated and uncarboxylated osteocalcin through gene therapy, which additionally led to a reduction in hepatic lipid deposition and improved phosphorylation of the insulin receptor. These data suggest that the effects of exogenous high-dose glucocorticoids on insulin target tissues and systemic energy metabolism are mediated, at least in part, through the skeleton.


Subject(s)
Anti-Inflammatory Agents/adverse effects , Corticosterone/adverse effects , Energy Metabolism/drug effects , Glucocorticoids/adverse effects , Osteoblasts/metabolism , Signal Transduction/drug effects , Animals , Anti-Inflammatory Agents/pharmacology , Corticosterone/pharmacology , Energy Metabolism/genetics , Glucocorticoids/pharmacology , Glucose Intolerance/genetics , Glucose Intolerance/metabolism , Glucose Intolerance/pathology , Insulin Resistance/genetics , Mice , Mice, Transgenic , Osteoblasts/pathology , Osteocalcin/biosynthesis , Osteocalcin/genetics , Rats , Signal Transduction/genetics
20.
J Endocrinol ; 215(1): 51-8, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22836489

ABSTRACT

Liver X receptor (LXR) α and ß are nuclear receptors that are crucial for the regulation of carbohydrate and lipid metabolism. Activation of LXRs in the brain facilitates cholesterol clearance and improves cognitive deficits, thus they are considered as promising drug targets to treat diseases such as atherosclerosis and Alzheimer's disease. Nevertheless, little is known about the function and localization of LXRs in the brain. Here, we studied the expression of LXR in the brains of rats that received free access to 10% (w/v) fructose group (FG) in their beverages or water control drinks (control group (CG)). After 6 weeks rats in the FG presented with hypertriglyceridemia, hyperinsulinemia, and became glucose intolerant, suggesting a progression toward type 2 diabetes. We found that hypothalamic LXR expression was altered in fructose-fed rats. Rats in the FG presented with a decrease in LXRß levels while showing an increase in LXRα expression in the hypothalamus but not in the hippocampus, cerebellum, or neocortex. Moreover, both LXRα and ß expression correlated negatively with insulin and triglyceride levels. Interestingly, LXRß showed a negative correlation with the area under the curve during the glucose tolerance test in the CG and a positive correlation in the FG. Immunocytochemistry revealed that the paraventricular and ventromedial nuclei express mainly LXRα whereas the arcuate nucleus expresses LXRß. Both LXR immunosignals were found in the median preoptic area. This is the first study showing a relationship between glucose and lipid homeostasis and the expression of LXRs in the hypothalamus, suggesting that LXRs may trigger neurochemical and neurophysiological responses for the control of food intake and energy expenditure through these receptors.


Subject(s)
Glucose Intolerance/metabolism , Hypothalamus/metabolism , Orphan Nuclear Receptors/metabolism , Animals , Blood Glucose/metabolism , Glucose Intolerance/blood , Glucose Intolerance/pathology , Glucose Tolerance Test , Hypothalamus/pathology , Immunohistochemistry , Insulin/blood , Liver X Receptors , Male , Microscopy, Confocal , Rats , Rats, Sprague-Dawley , Tissue Distribution , Triglycerides/blood
SELECTION OF CITATIONS
SEARCH DETAIL