Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Front Endocrinol (Lausanne) ; 12: 656831, 2021.
Article in English | MEDLINE | ID: mdl-33953699

ABSTRACT

Background: The metabolic syndrome (MetS) is correlated with disorders of the reproductive system, such as the polycystic ovary syndrome (PCOS). While consumption of a diet rich in carbohydrates is linked to the development of MetS, it is still unclear if this diet leads to ovarian dysfunction and PCOS. Objectives: We investigated the influence of a high-sucrose diet (HSD) on the ovarian milieu of Wistar rats and studied the correlation between high consumption of sugary drinks and the prevalence of PCOS in women. Methods: Wistar rats were given a standard laboratory diet (CTR, 10% sucrose, n = 8) or HSD (HSD, 25% sucrose, n = 8) from postnatal day 21 to 120. Animals were evaluated weekly to calculate food intake, feed efficiency and weight gain. Both onset of puberty and estrous cycle were monitored. Metabolic serum biochemistry, organ morphometry and ovarian histology were performed upon euthanasia. In parallel, a fixed-effects multiple linear regression analysis was performed using data from Brazilian states (459 state-year observations) to test the correlation between the consumption of sugar-sweetened beverages (surrogate for HSD intake) and the prevalence of PCOS (surrogate for ovarian dysfunction). Results: HSD animals showed increased adipose tissue accumulation, hyperglycaemia and insulin resistance when compared to CTR. Interestingly HSD rats also entered puberty earlier than CTR. Moreover, ovaries from HSD animals had an increased number of atretic antral follicles and cystic follicles, which were correlated with the hypertrophy of periovarian adipocytes. Finally, there was a positive correlation between the intake of sugary drinks and prevalence of PCOS in women of reproductive age. Conclusions: HSD ingestion leads to ovarian dysfunction in rats and could be correlated with PCOS in women, suggesting these alterations could lead to public health issues. Therefore, we reinforce the deleterious impact of HSD to the ovarian system and suggest that the reduction of added sugars intake could be beneficial to ovarian health.


Subject(s)
Dietary Sucrose/toxicity , Estrous Cycle , Metabolic Syndrome/pathology , Ovary/pathology , Polycystic Ovary Syndrome/pathology , Sexual Maturation , Animals , Body Weight , Female , Metabolic Syndrome/chemically induced , Metabolic Syndrome/metabolism , Ovary/drug effects , Polycystic Ovary Syndrome/chemically induced , Polycystic Ovary Syndrome/metabolism , Rats , Rats, Wistar
2.
Front Immunol ; 12: 669672, 2021.
Article in English | MEDLINE | ID: mdl-33995404

ABSTRACT

Background and Aims: Many nutritional and epidemiological studies have shown that high consumption of trans fatty acids can cause several adverse effects on human health, including cardiovascular disease, diabetes, and cancer. In the present study, we investigated the effect of trans fatty acids on innate immunity in the gut by observing mice fed with a diet high in trans fatty acids, which have been reported to cause dysbiosis. Methods: We used C57BL6/J mice and fed them with normal diet (ND) or high-fat, high-sucrose diet (HFHSD) or high-trans fatty acid, high-sucrose diet (HTHSD) for 12 weeks. 16S rRNA gene sequencing was performed on the mice stool samples, in addition to flow cytometry, real-time PCR, and lipidomics analysis of the mice serum and liver samples. RAW264.7 cells were used for the in vitro studies. Results: Mice fed with HTHSD displayed significantly higher blood glucose levels and advanced fatty liver and intestinal inflammation, as compared to mice fed with HFHSD. Furthermore, compared to mice fed with HFHSD, mice fed with HTHSD displayed a significant elevation in the expression of CD36 in the small intestine, along with a reduction in the expression of IL-22. Furthermore, there was a significant increase in the populations of ILC1s and T-bet-positive ILC3s in the lamina propria in mice fed with HTHSD. Finally, the relative abundance of the family Desulfovibrionaceae, which belongs to the phylum Proteobacteria, was significantly higher in mice fed with HFHSD or HTHSD, than in mice fed with ND; between the HFHSD and HTHSD groups, the abundance was slightly higher in the HTHSD group. Conclusions: This study revealed that compared to saturated fatty acid intake, trans fatty acid intake significantly exacerbated metabolic diseases such as diabetes and fatty liver.


Subject(s)
Blood Glucose/drug effects , Diet, High-Fat/adverse effects , Enteritis/chemically induced , Glucose Intolerance/chemically induced , Immunity, Innate/drug effects , Intestine, Small/drug effects , Lymphocytes/drug effects , Trans Fatty Acids/toxicity , Animals , Blood Glucose/metabolism , CD36 Antigens/metabolism , Dietary Sucrose/toxicity , Dysbiosis , Enteritis/immunology , Enteritis/metabolism , Enteritis/microbiology , Gastrointestinal Microbiome/drug effects , Glucose Intolerance/blood , Glucose Intolerance/immunology , Interleukins/metabolism , Intestine, Small/immunology , Intestine, Small/metabolism , Intestine, Small/microbiology , Liver/drug effects , Liver/immunology , Liver/metabolism , Lymphocytes/immunology , Lymphocytes/metabolism , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/chemically induced , Non-alcoholic Fatty Liver Disease/immunology , Non-alcoholic Fatty Liver Disease/metabolism , RAW 264.7 Cells , Interleukin-22
3.
Life Sci ; 269: 119048, 2021 Mar 15.
Article in English | MEDLINE | ID: mdl-33453246

ABSTRACT

AIMS: The present study investigated the potential effects of pterostilbene (PT) on glycemic and lipid profiles, fat storage, cardiovascular indices, and hepatic parameters of rats fed with sucrose solution. MAIN METHODS: 24 male Wistar rats received either drinking water or a 40% sucrose solution over a period of 140 days. After this period, animals were randomly allocated into four groups (n = 6): Control (C), C + Pterostilbene (PT), Sucrose (S), and S + PT. Pterostilbene (40 mg/kg) was given orally for 45 consecutive days. KEY FINDINGS: Pterostilbene did not influence morphometric and nutritional parameters. The insulin sensitivity index TyG was elevated in the C + PT group (p < 0.01) and reduced in S + PT group (p < 0.05). Basal glucose levels were lower in the S + PT group (p < 0.05), and the glycemic response was improved with PT treatment in glucose provocative tests. Conversely, rats from the C + PT group showed impaired glucose disposal during those tests. Lipid profile was partially improved by PT treatment. Hepatic oxidative stress in the S group was improved after PT treatment. In the C group, PT reduced SOD activity, glutathione levels, and increased catalase activity. Collagen content was reduced by PT treatment. SIGNIFICANCE: PT effects depends on the type of diet the animals were submitted. In rats fed with sucrose-solution, PT confirmed its positive effects, improving glucose and lipid profile, and acting as a potent antioxidant. The effects of PT on rats that consumed a normal diet were very discrete or even undesirable. We suggest caution with indiscriminate consume of natural compounds by healthy subjects.


Subject(s)
Antioxidants/pharmacology , Dietary Sucrose/toxicity , Hyperglycemia/drug therapy , Hyperlipidemias/drug therapy , Liver/drug effects , Oxidative Stress/drug effects , Stilbenes/pharmacology , Animals , Blood Glucose/metabolism , Hyperglycemia/chemically induced , Hyperglycemia/metabolism , Hyperglycemia/pathology , Hyperlipidemias/chemically induced , Hyperlipidemias/metabolism , Hyperlipidemias/pathology , Liver/metabolism , Liver/pathology , Male , Oxidation-Reduction , Rats , Rats, Wistar
4.
J Cereb Blood Flow Metab ; 41(7): 1734-1743, 2021 07.
Article in English | MEDLINE | ID: mdl-32757742

ABSTRACT

The hypothalamus is the central regulator of energy homeostasis. Hypothalamic neuronal circuits are disrupted upon overfeeding, and play a role in the development of metabolic disorders. While mouse models have been extensively employed for understanding the mechanisms of hypothalamic dysfunction, functional magnetic resonance imaging (fMRI) on hypothalamic nuclei has been challenging. We implemented a robust glucose-induced fMRI paradigm that allows to repeatedly investigate hypothalamic responses to glucose. This approach was used to test the hypothesis that hypothalamic nuclei functioning is impaired in mice exposed to a high-fat and high-sucrose diet (HFHSD) for seven days. The blood oxygen level-dependent (BOLD) fMRI signal was measured from brains of mice under light isoflurane anaesthesia, during which a 2.6 g/kg glucose load was administered. The mouse hypothalamus responded to glucose but not saline administration with a biphasic BOLD fMRI signal reduction. Relative to controls, HFHSD-fed mice showed attenuated or blunted responses in arcuate nucleus, lateral hypothalamus, ventromedial nucleus and dorsomedial nucleus, but not in paraventricular nucleus. In sum, we have developed an fMRI paradigm that is able to determine dysfunction of glucose-sensing neuronal circuits within the mouse hypothalamus in a non-invasive manner.


Subject(s)
Diet, High-Fat , Dietary Sucrose/toxicity , Glucose/administration & dosage , Hypothalamus/pathology , Magnetic Resonance Imaging/methods , Obesity/physiopathology , Animals , Hypothalamus/drug effects , Hypothalamus/metabolism , Male , Mice , Mice, Inbred C57BL
5.
Int J Mol Sci ; 22(1)2020 Dec 30.
Article in English | MEDLINE | ID: mdl-33396939

ABSTRACT

Liver fibrosis is a consequence of chronic liver injury associated with chronic viral infection, alcohol abuse, and nonalcoholic fatty liver. The evidence from clinical and animal studies indicates that transforming growth factor-ß (TGF-ß) signaling is associated with the development of liver fibrosis. Krüppel-like factor 10 (KLF10) is a transcription factor that plays a significant role in TGF-ß-mediated cell growth, apoptosis, and differentiation. In recent studies, it has been reported to be associated with glucose homeostasis and insulin resistance. In the present study, we investigated the role of KLF10 in the progression of liver disease upon a high-sucrose diet (HSD) in mice. Wild type (WT) and Klf10 knockout (KO) mice were fed either a control chow diet or HSD (50% sucrose) for eight weeks. Klf10 KO mice exhibited significant hepatic steatosis, inflammation, and liver injury upon HSD feeding, whereas the WT mice exhibited mild hepatic steatosis with no apparent liver injury. The livers of HSD-fed Klf10 KO mice demonstrated significantly increased endoplasmic reticulum stress, oxidative stress, and proinflammatory cytokines. Klf10 deletion led to the development of sucrose-induced hepatocyte cell death both in vivo and in vitro. Moreover, it significantly increased fibrogenic gene expression and collagen accumulation in the liver. Increased liver fibrosis was accompanied by increased phosphorylation and nuclear localization of Smad3. Here, we demonstrate that HSD-fed mice develop a severe liver injury in the absence of KLF10 due to the hyperactivation of the endoplasmic reticulum stress response and CCAAT/enhance-binding protein homologous protein (CHOP)-mediated apoptosis of hepatocytes. The current study suggests that KLF10 plays a protective role against the progression of hepatic steatosis into liver fibrosis in a lipogenic state.


Subject(s)
Dietary Sucrose/toxicity , Early Growth Response Transcription Factors/physiology , Endoplasmic Reticulum Stress , Gene Deletion , Inflammation/complications , Kruppel-Like Transcription Factors/physiology , Liver Cirrhosis/etiology , Animals , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidative Stress
6.
Am J Physiol Regul Integr Comp Physiol ; 316(5): R472-R485, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30758976

ABSTRACT

The main objective of these studies was to characterize metabolic, body composition, and cardiovascular responses to a free-choice high-fat, high-sucrose diet in female cycling and pregnant rats. In the nonpregnant state, female Sprague-Dawley rats offered a 3-wk free-choice high-fat, high-sucrose diet had greater energy intake, adiposity, serum leptin, and triglyceride concentrations compared with rats fed with standard chow and developed glucose intolerance. In addition, choice-diet-fed rats had larger cardiac ventricular weights, smaller kidney and pancreas weights, and higher blood pressure than chow-fed rats, but they did not exhibit resistance artery endothelial dysfunction. When the free-choice diet continued throughout pregnancy, rats remained hyperphagic, hyperleptinemic, and obese. Choice pregnant rats exhibited uterine artery endothelial dysfunction and had smaller fetuses compared with chow pregnant rats. Pregnancy normalized mean arterial blood pressure and pancreas weights in choice rats. These studies are the first to provide a comprehensive evaluation of free-choice high-fat, high-sucrose diet on metabolic and cardiovascular functions in female rats, extending the previous studies in males to female cycling and pregnant rodents. Free-choice diet may provide a new model of preconceptual maternal obesity to study the role of increased energy intake, individual food components, and preexisting maternal obesity on maternal and offspring physiological responses during pregnancy and after birth.


Subject(s)
Cardiovascular Diseases/etiology , Diet, High-Fat/adverse effects , Dietary Sucrose/toxicity , Energy Metabolism , Estrous Cycle , Fetal Growth Retardation/etiology , Hyperphagia/etiology , Obesity/etiology , Adiposity , Animal Nutritional Physiological Phenomena , Animals , Behavior, Animal , Biomarkers/blood , Cardiovascular Diseases/blood , Cardiovascular Diseases/physiopathology , Choice Behavior , Dietary Sucrose/metabolism , Feeding Behavior , Female , Fetal Growth Retardation/blood , Fetal Growth Retardation/physiopathology , Hemodynamics , Hyperphagia/blood , Hyperphagia/physiopathology , Hyperphagia/psychology , Maternal Nutritional Physiological Phenomena , Nutritional Status , Obesity/blood , Obesity/physiopathology , Pregnancy , Rats, Sprague-Dawley , Weight Gain
7.
J Pineal Res ; 66(2): e12545, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30586198

ABSTRACT

While physiological levels of glucocorticoids are required to ensure proper functions of the body, consistently high levels may engender several deleterious consequences. We have previously shown an increase in the activity of the hypothalamic-pituitary-adrenal (HPA) axis in rats fed sucrose-rich diets (SRD). The main goal of this study was to analyze the processes involved in the modulation of the pituitary production of ACTH by SRD, and to test melatonin as a possible therapeutic agent for the prevention of the HPA axis dysfunction. Male Wistar rats were fed standard chow and either SRD (30% sucrose in the drinking water) or plain water for three weeks. Melatonin was administered as subcutaneous pellets. Results showed that SRD treatment induced an increase in systemic ACTH and corticosterone levels and a decrease in melatonin levels. In the pituitary gland, we also detected an increase in the expression levels of proopiomelanocortin (POMC) that was accompanied by increased levels of: lipoperoxides, nitro-tyrosine modified proteins, catalase, heme oxygenase-1, interleukin-1ß mRNA, and by an increase in the tissue number of inflammatory cells (F4/80 and Iba-1 positive cells). Melatonin treatment prevented all these systemic and pituitary changes as well as the increase in POMC expression induced by incubation of AtT-20 corticotrophs with conditioned media obtained from stimulated macrophages. In conclusion, stimulation of POMC/ACTH production in rats fed a SRD could involve the generation of oxidative stress and inflammation in the pituitary gland. Melatonin treatment prevented these effects and normalized the activity of the HPA axis.


Subject(s)
Adrenocorticotropic Hormone/biosynthesis , Dietary Sucrose/toxicity , Melatonin/pharmacology , Pituitary Gland/drug effects , Animals , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/metabolism , Male , Pituitary Gland/metabolism , Pituitary-Adrenal System/drug effects , Pituitary-Adrenal System/metabolism , Rats , Rats, Wistar
8.
J Environ Pathol Toxicol Oncol ; 37(1): 63-80, 2018.
Article in English | MEDLINE | ID: mdl-29773001

ABSTRACT

In this study, we aimed to develop an experimental animal model for type 2 diabetes mellitus (T2DM) using a combination of monosodium glutamate (MSG) and high sucrose diet (HSD). Young male Wistar rats (20-30 g) were injected with MSG (2 or 4 mg/g, i.p. for 4 days). These rats were also fed an HSD, while the control group was fed a starch diet (SFD) for 150 days. Parameters assessed periodically were body weight, feed intake, blood glucose level, and oral glucose tolerance test (OGTT), lipid profile, liver and kidney function tests, skeletal muscle glucose uptake, cognitive function tests, and microvascular changes using isolated rat aorta. Histological changes in pancreas, liver, and kidney tissue were assessed using hematoxylin and eosin staining, whereas brain tissue was assessed using cresyl violet stain. Feeding MSG in combination with HSD in rats significantly increased body weight, and produced hyperglycemia, dyslipidemia, and hyperinsulinemia. Animals developed frank diabetic complications, which included insulin resistance in skeletal muscle, hypertension, vascular dysfunction, nephropathy, and dementia. Histological studies revealed neuronal loss with necrotic bodies in the brain, reduction in glomerular count in kidney, and severe hypertrophy and hyperplasia in the islets of Langerhans. These results indicate the successful induction of type-2 diabetes along with several diabetic complications by combining MSG with HSD.


Subject(s)
Diabetes Mellitus, Type 2/chemically induced , Dietary Sucrose/toxicity , Disease Models, Animal , Memory Disorders/chemically induced , Sodium Glutamate/toxicity , Vascular Diseases/chemically induced , Animals , Dose-Response Relationship, Drug , Flavoring Agents/toxicity , Male , Random Allocation , Rats , Rats, Wistar
9.
Am J Physiol Regul Integr Comp Physiol ; 314(4): R598-R610, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29351425

ABSTRACT

Male rats offered 30% sucrose solution in addition to chow develop leptin resistance without an increase in energy intake or body fat. This study tested whether the leptin resistance was dependent on the physical form of the sucrose. Sprague-Dawley rats were offered a sucrose-free (NS) diet, a 66.6% of energy as sucrose (HS) diet, or the NS diet + 30% sucrose solution (LS). Sucrose intake of LS rats equaled that of HS rats, but total carbohydrate intake exceeded that of HS rats. After 33 days, male and female LS rats were resistant to the inhibitory effect of peripherally administered leptin on food intake. LS rats drank small, frequent meals of sucrose during light and dark periods, whereas HS rats consumed more meals during the dark than the light period and remained responsive to leptin. Diet did not affect daily energy intake or insulin sensitivity. There was a small increase in body fat in the female rats. Leptin sensitivity was restored within 5 days of withdrawal from sucrose in male LS rats. This rapid reversal suggested that leptin resistance was associated with the metabolic impact of drinking sucrose. An experiment was carried out to test whether activity of the hexosamine biosynthetic pathway and glycation of leptin signaling proteins were increased in LS rats, but the results were equivocal. A final experiment determined that female LS rats were leptin-resistant within 18 days of access to sucrose solution and that the small, but significant, increase in body fat was associated with increased adipocyte glucose utilization and insulin responsiveness, which may have been secondary to adipocyte leptin resistance.


Subject(s)
Adipose Tissue, White/metabolism , Dietary Sucrose/toxicity , Leptin/blood , Adipocytes/metabolism , Adiposity , Administration, Oral , Animal Feed , Animal Nutritional Physiological Phenomena , Animals , Biomarkers/blood , Blood Glucose/metabolism , Dietary Sucrose/administration & dosage , Dietary Sucrose/blood , Energy Intake , Fatty Acids/blood , Female , Insulin/blood , Insulin Resistance , Male , Nutritional Status , Rats, Sprague-Dawley , Sex Factors , Time Factors
10.
Am J Physiol Heart Circ Physiol ; 312(4): H742-H751, 2017 Apr 01.
Article in English | MEDLINE | ID: mdl-28087518

ABSTRACT

Thirty percent of the world population is diagnosed with metabolic syndrome. High-fat/high-sucrose (HF/HS) diet (Western diet) correlates with metabolic syndrome prevalence. We characterized effects of the HF/HS diet on vascular (arterial stiffness, vasoreactivity, and coronary collateral development) and cardiac (echocardiography) function, oxidative stress, and inflammation in a rat model of metabolic syndrome (JCR rats). Furthermore, we determined whether male versus female animals were affected differentially by the Western diet. Cardiovascular function in JCR male rats was impaired versus normal Sprague-Dawley (SD) rats. HF/HS diet compromised cardiovascular (dys)function in JCR but not SD male rats. In contrast, cardiovascular function was minimally impaired in JCR female rats on normal chow. However, cardiovascular function in JCR female rats on the HF/HS diet deteriorated to levels comparable to JCR male rats on the HF/HS diet. Similarly, oxidative stress was markedly increased in male but not female JCR rats on normal chow but was equally exacerbated by the HF/HS diet in male and female JCR rats. These results indicate that the Western diet enhances oxidative stress and cardiovascular dysfunction in metabolic syndrome and eliminates the protective effect of female sex on cardiovascular function, implying that both males and females with metabolic syndrome are at equal risk for cardiovascular disease.NEW & NOTEWORTHY Western diet abolished protective effect of sex against cardiovascular disease (CVD) development in premenopausal animals with metabolic syndrome. Western diet accelerates progression of CVD in male and female animals with preexisting metabolic syndrome but not normal animals. Exacerbation of baseline oxidative stress correlates with accelerated progression of CVD in metabolic syndrome animals on Western diet.


Subject(s)
Diet, High-Fat/adverse effects , Dietary Sucrose/toxicity , Heart/physiopathology , Metabolic Syndrome/physiopathology , Animals , Cardiovascular Physiological Phenomena , Collateral Circulation , Coronary Circulation/drug effects , Echocardiography , Female , Heart/diagnostic imaging , Heart/drug effects , Inflammation/pathology , Male , Metabolic Syndrome/genetics , Oxidative Stress , Rats , Rats, Inbred Strains , Rats, Sprague-Dawley , Sex Characteristics , Vascular Stiffness/drug effects
11.
Pharm Biol ; 54(12): 2857-2863, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27241764

ABSTRACT

CONTEXT: Curcumin, an active principal of Curcuma longa Linn. (Zingiberaceae), has potent antioxidant and anti-inflammatory properties. OBJECTIVES: This study investigated the effects of curcumin on hyperlipidemia and hepatic steatosis in high-fructose-fed Wistar rats. MATERIALS AND METHODS: Forty male Wistar rats were divided into four groups with 10 rats in each. Two groups were fed with standard rodent diet and the other two with 60% high-fructose diet for 10 weeks. Curcumin (200 mg/kg body weight) was administered along with the diets simultaneously to each of the aforementioned diet groups. After 10 weeks of experiment, blood samples were collected from tail vein. Liver, adipose and epididymal tissues were collected after sacrifice of the animals and stored for further analyses. RESULTS: Administration of curcumin reduced body weight (280.6 ± 7.4 g), liver weight (2.5 ± 0.2 g/100 g BW), adipose weight (1.4 ± 0.3 g/100 g BW), plasma levels of TAG (86.1 ± 13.5 mg/dL), VLDL-C (17.2 ± 2.7 mg/dL), lipid ratios and increased HDL-C (28.4 ± 4.5 mg/dL) in fructose-fed rats. Curcumin supplementation significantly lowered TAG content and decreased the protein expression of LXR-α (43%) and SREBP1c (59%) in the liver. Furthermore, curcumin suppressed the expression of lipogenic enzymes, ACLY (95%), ACC (50%) and FAS (77%) in rats fed with high-fructose diet. No significant change was found in the expression of PPAR-α. DISCUSSION AND CONCLUSION: Curcumin prevented the high-fructose induced hyperlipidemia and hepatic steatosis.


Subject(s)
Curcumin/therapeutic use , Fatty Liver/drug therapy , Fructose/toxicity , Hyperlipidemias/prevention & control , Animals , Body Weight/drug effects , Body Weight/physiology , Dietary Sucrose/administration & dosage , Dietary Sucrose/toxicity , Eating/drug effects , Eating/physiology , Fatty Liver/chemically induced , Fatty Liver/pathology , Fructose/administration & dosage , Hyperlipidemias/chemically induced , Hyperlipidemias/pathology , Male , Rats , Rats, Wistar
12.
PLoS One ; 11(1): e0145730, 2016.
Article in English | MEDLINE | ID: mdl-26808523

ABSTRACT

Insulin resistance (IR) and type 2 diabetes mellitus (T2DM) have been found to be associated with postprandial hypertriglyceridemia (PPHTg). However, whether PPHTg can cause IR and diabetes is not clear. We therefore investigated the role of PPHTg in development of T2DM in rat model of T2DM. 96 male Wistar rats were randomized into four groups (24 rats each). Control Group A, high sucrose diet (HSD) Group B, HSD+Pioglitazone (10 mg/kg/day) Group C and HSD+Atorvastatin (20 mg/kg/day) Group D. Fat and glucose tolerance tests were done at regular intervals in all groups besides insulin and body weight measurement. At 26 weeks, low dose streptozotocin (15 mg/kg, i.p.) was given to half of the rats. All rats were followed up till 48 weeks. PPHTg developed as early as week 2 in Group B and stabilized by week 14. Group B displayed highest PPHTg compared to other groups. Atorvastatin treatment (Group D) abolished PPHTg which became comparable to controls, pioglitazone treatment partially blunted PPHTg resulting in intermediate PPHTg. Group B with highest PPHTg showed highest subsequent IR, glucose intolerance (GI) and highest incidence of prediabetes at week 26 and diabetes at week 34 and 46 compared to other groups. Group D rats displayed lower IR, GI, low incidence of prediabetes and diabetes at these time points compared to Groups B and C. ROC analysis showed that triglyceride area under the curve of each time point significantly predicts the risk of diabetes. Present study provides the evidence that PPHTg predicts the development of IR, GI and T2DM in rat model of diet induced T2DM.


Subject(s)
Diabetes Mellitus, Type 2/etiology , Dietary Fats/toxicity , Dietary Sucrose/toxicity , Glucose Intolerance/etiology , Hypertriglyceridemia/complications , Insulin Resistance , Postprandial Period , Prediabetic State/physiopathology , Animals , Atorvastatin/therapeutic use , Blood Glucose/analysis , Body Weight , Causality , Diabetes Mellitus, Type 2/physiopathology , Disease Progression , Glucose Intolerance/physiopathology , Glucose Tolerance Test , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Hypertriglyceridemia/drug therapy , Hypertriglyceridemia/physiopathology , Hypoglycemic Agents/therapeutic use , Intra-Abdominal Fat/pathology , Lipids/blood , Liver/chemistry , Liver/pathology , Male , Pancreas/pathology , Pioglitazone , Prediabetic State/drug therapy , Random Allocation , Rats , Rats, Wistar , Thiazolidinediones/therapeutic use
13.
Cancer Res ; 76(1): 24-9, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26729790

ABSTRACT

Epidemiologic studies have shown that dietary sugar intake has a significant impact on the development of breast cancer. One proposed mechanism for how sugar impacts cancer development involves inflammation. In the current study, we investigated the impact of dietary sugar on mammary gland tumor development in multiple mouse models, along with mechanisms that may be involved. We found that sucrose intake in mice comparable with levels of Western diets led to increased tumor growth and metastasis, when compared with a nonsugar starch diet. This effect was ascribed in part to increased expression of 12-lipoxygenase (12-LOX) and its arachidonate metabolite 12-hydroxy-5Z,8Z,10E,14Z-eicosatetraenoic acid (12-HETE). We determined that fructose derived from the sucrose was responsible for facilitating lung metastasis and 12-HETE production in breast tumors. Overall, our data suggested that dietary sugar induces 12-LOX signaling to increase risks of breast cancer development and metastasis.


Subject(s)
Arachidonate 12-Lipoxygenase/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carcinogenesis/metabolism , Dietary Sucrose/toxicity , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/pathology , Animals , Breast Neoplasms/enzymology , Cell Line, Tumor , Diet , Female , Heterografts , Humans , Mammary Glands, Animal/enzymology , Mammary Neoplasms, Experimental/enzymology , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Random Allocation , Risk Factors , Signal Transduction
14.
Neuroscience ; 315: 196-205, 2016 Feb 19.
Article in English | MEDLINE | ID: mdl-26704636

ABSTRACT

High sugar consumption is a risk factor for metabolic disturbances leading to memory impairment. Thus, rats subject to high sucrose intake (HSu) develop a metabolic syndrome and display memory deficits. We now investigated if these HSu-induced memory deficits were associated with metabolic and electrophysiological alterations in the hippocampus. Male Wistar rats were submitted for 9 weeks to a sucrose-rich diet (35% sucrose solution) and subsequently to a battery of behavioral tests; after sacrifice, their hippocampi were collected for ex vivo high-resolution magic angle spinning (HRMAS) metabolic characterization and electrophysiological extracellular recordings in slices. HSu rats displayed a decreased memory performance (object displacement and novel object recognition tasks) and helpless behavior (forced swimming test), without altered locomotion (open field). HRMAS analysis indicated a similar hippocampal metabolic profile of HSu and control rats. HSu rats also displayed no change of synaptic transmission and plasticity (long-term potentiation) in hippocampal Schaffer fibers-CA1 pyramid synapses, but had decreased amplitude of long-term depression in the temporoammonic (TA) pathway. Furthermore, HSu rats had an increased density of inhibitory adenosine A1 receptors (A1R), that translated into a greater potency of A1R in Schaffer fiber synapses, but not in the TA pathway, whereas the endogenous activation of A1R in HSu rats was preserved in the TA pathway but abolished in Schaffer fiber synapses. These results suggest that HSu triggers a hippocampal-dependent memory impairment that is not associated with altered hippocampal metabolism but is probably related to modified synaptic plasticity in hippocampal TA synapses.


Subject(s)
Diet/adverse effects , Dietary Sucrose/toxicity , Hippocampus/physiopathology , Memory Disorders/etiology , Memory Disorders/physiopathology , Animals , Disease Models, Animal , Emotions/physiology , Helplessness, Learned , Locomotion/physiology , Long-Term Potentiation/physiology , Long-Term Synaptic Depression/physiology , Male , Motor Activity/physiology , Rats, Wistar , Receptor, Adenosine A1/metabolism , Recognition, Psychology/physiology , Synapses/physiology , Synaptic Transmission/physiology
15.
PLoS One ; 10(6): e0131269, 2015.
Article in English | MEDLINE | ID: mdl-26102086

ABSTRACT

Elongation factor 1A-1 (eEF1A-1) has non-canonical functions in regulation of the actin cytoskeleton and apoptosis. It was previously identified through a promoter-trap screen as a mediator of fatty acid-induced cell death (lipotoxicity), and was found to participate in this process downstream of ER stress. Since ER stress is implicated in the pathogenesis of nonalcoholic fatty liver disease (NAFLD), we investigated the mechanism of action of eEF1A-1 in hepatocyte lipotoxicity. HepG2 cells were exposed to excess fatty acids, followed by assessments of ER stress, subcellular localization of eEF1A-1, and cell death. A specific inhibitor of eEF1A-1 elongation activity, didemnin B, was used to determine whether its function in protein synthesis is involved in lipotoxicity. Within 6 h, eEF1A-1 protein was modestly induced by high palmitate, and partially re-localized from its predominant location at the ER to polymerized actin at the cell periphery. This early induction and subcellular redistribution of eEF1A-1 coincided with the onset of ER stress, and was later followed by cell death. Didemnin B did not prevent the initiation of ER stress by high palmitate, as indicated by eIF2α phosphorylation. However, consistent with sustained inhibition of eEF1A-1-dependent elongation activity, didemnin B prevented the recovery of protein synthesis and increase in GRP78 protein that are normally associated with later phases of the response to ongoing ER stress. This resulted in decreased palmitate-induced cell death. Our data implicate eEF1A-1, and its function in protein synthesis, in hepatocyte lipotoxicity.


Subject(s)
Endoplasmic Reticulum Stress/drug effects , Hepatocytes/drug effects , Palmitates/toxicity , Peptide Chain Elongation, Translational , Peptide Elongation Factor 1/physiology , Animals , Apoptosis/drug effects , Depsipeptides/pharmacology , Dietary Fats/toxicity , Dietary Sucrose/toxicity , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Chaperone BiP , Gene Expression Regulation/drug effects , Heat-Shock Proteins/biosynthesis , Heat-Shock Proteins/genetics , Hep G2 Cells , Hepatocytes/metabolism , Humans , Leptin/deficiency , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Models, Animal , Non-alcoholic Fatty Liver Disease/chemically induced , Non-alcoholic Fatty Liver Disease/metabolism , Protein Transport
16.
PLoS Genet ; 11(2): e1004995, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25692475

ABSTRACT

Fatty acid (FA) metabolism is deregulated in several human diseases including metabolic syndrome, type 2 diabetes and cancers. Therefore, FA-metabolic enzymes are potential targets for drug therapy, although the consequence of these treatments must be precisely evaluated at the organismal and cellular levels. In healthy organism, synthesis of triacylglycerols (TAGs)-composed of three FA units esterified to a glycerol backbone-is increased in response to dietary sugar. Saturation in the storage and synthesis capacity of TAGs is associated with type 2 diabetes progression. Sugar toxicity likely depends on advanced-glycation-end-products (AGEs) that form through covalent bounding between amine groups and carbonyl groups of sugar or their derivatives α-oxoaldehydes. Methylglyoxal (MG) is a highly reactive α-oxoaldehyde that is derived from glycolysis through a non-enzymatic reaction. Glyoxalase 1 (Glo1) works to neutralize MG, reducing its deleterious effects. Here, we have used the power of Drosophila genetics to generate Fatty acid synthase (FASN) mutants, allowing us to investigate the consequence of this deficiency upon sugar-supplemented diets. We found that FASN mutants are lethal but can be rescued by an appropriate lipid diet. Rescued animals do not exhibit insulin resistance, are dramatically sensitive to dietary sugar and accumulate AGEs. We show that FASN and Glo1 cooperate at systemic and cell-autonomous levels to protect against sugar toxicity. We observed that the size of FASN mutant cells decreases as dietary sucrose increases. Genetic interactions at the cell-autonomous level, where glycolytic enzymes or Glo1 were manipulated in FASN mutant cells, revealed that this sugar-dependent size reduction is a direct consequence of MG-derived-AGE accumulation. In summary, our findings indicate that FASN is dispensable for cell growth if extracellular lipids are available. In contrast, FA-synthesis appears to be required to limit a cell-autonomous accumulation of MG-derived-AGEs, supporting the notion that MG is the most deleterious α-oxoaldehyde at the intracellular level.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Fatty Acid Synthase, Type I/genetics , Lactoylglutathione Lyase/genetics , Metabolic Syndrome/genetics , Neoplasms/genetics , Animals , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Dietary Sucrose/administration & dosage , Dietary Sucrose/toxicity , Drosophila , Fatty Acid Synthase, Type I/metabolism , Glycation End Products, Advanced/genetics , Glycation End Products, Advanced/metabolism , Humans , Insulin Resistance/genetics , Lactoylglutathione Lyase/metabolism , Lipids/administration & dosage , Metabolic Syndrome/metabolism , Metabolic Syndrome/pathology , Mutation , Neoplasms/metabolism , Neoplasms/pathology , Pyruvaldehyde/metabolism , Triglycerides/biosynthesis
17.
Clin Exp Pharmacol Physiol ; 41(7): 482-8, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24739055

ABSTRACT

The aim of the present study was to investigate Toll-like receptor-4 (TLR4) signalling at different stages of non-alcoholic fatty liver disease (NAFLD) induced by a high-fat, high-fructose (HFHFr) diet in mice. Both TLR4 wild-type (WT) and mutant (TLR4(mut) ) mice were fed either standard chow (SC) or the HFHFr diet for different periods of time from 4 to 16 weeks. Pathological characteristics and function of the liver were assessed. Simple steatosis, steatohepatitis and hepatic fibrosis occurred sequentially in Week 4, 8 and 16 in WT mice fed with the HFHFr. Expression of TLR4, myeloid differentiation factor 88 (MyD88), interferon regulatory factor (IRF) 3 and IRF7 started to increase at Week 4, peaked at Week 8 and then declined to basal levels at Week 16. This pattern was consistent with changes in inflammation in the liver revealed by haematoxylin and eosin staining. However, lipid accumulation, inflammation and fibrosis in livers of TLR4(mut) mice fed the HFHFr diet were significantly alleviated. In addition, the expression of activin A in WT mice fed the HFHFr diet increased at Week 16. The data suggest that TLR4 signalling mediates non-alcoholic steatohepatitis before fibrosis and that activin A is subsequently involved in NAFLD.


Subject(s)
Dietary Fats/toxicity , Dietary Sucrose/toxicity , Fructose/toxicity , Non-alcoholic Fatty Liver Disease/chemically induced , Toll-Like Receptor 4/metabolism , Activins/genetics , Activins/metabolism , Animals , Dietary Fats/administration & dosage , Dietary Sucrose/administration & dosage , Fructose/administration & dosage , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/metabolism , Interferon Regulatory Factor-7/genetics , Interferon Regulatory Factor-7/metabolism , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Signal Transduction , Toll-Like Receptor 4/genetics , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
18.
Brain Behav Immun ; 37: 134-41, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24309633

ABSTRACT

High energy diets have been shown to impair cognition however, the rapidity of these effects, and the dietary component/s responsible are currently unclear. We conducted two experiments in rats to examine the effects of short-term exposure to a diet rich in sugar and fat or rich in sugar on object (perirhinal-dependent) and place (hippocampal-dependent) recognition memory, and the role of inflammatory mediators in these responses. In Experiment 1, rats fed a cafeteria style diet containing chow supplemented with lard, cakes, biscuits, and a 10% sucrose solution performed worse on the place, but not the object recognition task, than chow fed control rats when tested after 5, 11, and 20 days. In Experiment 2, rats fed the cafeteria style diet either with or without sucrose and rats fed chow supplemented with sucrose also performed worse on the place, but not the object recognition task when tested after 5, 11, and 20 days. Rats fed the cafeteria diets consumed five times more energy than control rats and exhibited increased plasma leptin, insulin and triglyceride concentrations; these were not affected in the sucrose only rats. Rats exposed to sucrose exhibited both increased hippocampal inflammation (TNF-α and IL-1ß mRNA) and oxidative stress, as indicated by an upregulation of NRF1 mRNA compared to control rats. In contrast, these markers were not significantly elevated in rats that received the cafeteria diet without added sucrose. Hippocampal BDNF and neuritin mRNA were similar across all groups. These results show that relatively short exposures to diets rich in both fat and sugar or rich in sugar, impair hippocampal-dependent place recognition memory prior to the emergence of weight differences, and suggest a role for oxidative stress and neuroinflammation in this impairment.


Subject(s)
Diet, High-Fat/adverse effects , Dietary Sucrose/toxicity , Recognition, Psychology/physiology , Space Perception/physiology , Animals , Body Weight/physiology , Energy Intake/physiology , Hippocampus/metabolism , Male , Oxidative Stress , Rats , Rats, Sprague-Dawley
19.
J Clin Invest ; 123(12): 5061-70, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24177424

ABSTRACT

Energy and glucose homeostasis are regulated by central serotonin 2C receptors. These receptors are attractive pharmacological targets for the treatment of obesity; however, the identity of the serotonin 2C receptor-expressing neurons that mediate the effects of serotonin and serotonin 2C receptor agonists on energy and glucose homeostasis are unknown. Here, we show that mice lacking serotonin 2C receptors (Htr2c) specifically in pro-opiomelanocortin (POMC) neurons had normal body weight but developed glucoregulatory defects including hyperinsulinemia, hyperglucagonemia, hyperglycemia, and insulin resistance. Moreover, these mice did not show anorectic responses to serotonergic agents that suppress appetite and developed hyperphagia and obesity when they were fed a high-fat/high-sugar diet. A requirement of serotonin 2C receptors in POMC neurons for the maintenance of normal energy and glucose homeostasis was further demonstrated when Htr2c loss was induced in POMC neurons in adult mice using a tamoxifen-inducible POMC-cre system. These data demonstrate that serotonin 2C receptor-expressing POMC neurons are required to control energy and glucose homeostasis and implicate POMC neurons as the target for the effect of serotonin 2C receptor agonists on weight-loss induction and improved glycemic control.


Subject(s)
Energy Metabolism/physiology , Glucose/metabolism , Homeostasis/physiology , Neurons/physiology , Pro-Opiomelanocortin/physiology , Receptor, Serotonin, 5-HT2C/physiology , Animals , Appetite Depressants/pharmacology , Body Weight , Dietary Fats/toxicity , Dietary Sucrose/toxicity , Drug Resistance , Feeding Behavior/drug effects , Female , Glucagon/blood , Glucagon/metabolism , Hyperglycemia/genetics , Hyperglycemia/metabolism , Hyperinsulinism/genetics , Hyperinsulinism/metabolism , Hyperphagia/etiology , Insulin/metabolism , Insulin Resistance , Insulin Secretion , Male , Mice , Mice, Knockout , Obesity/etiology , Pro-Opiomelanocortin/biosynthesis , Pro-Opiomelanocortin/genetics , Receptor, Serotonin, 5-HT2C/deficiency , Receptor, Serotonin, 5-HT2C/genetics , Recombinant Fusion Proteins/biosynthesis , Serotonin/physiology , Serotonin Receptor Agonists/pharmacology
20.
Blood Coagul Fibrinolysis ; 24(1): 106-8, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23249568

ABSTRACT

Links between shift work and increases in metabolic risk factors for cardiovascular diseases have been documented in detail, although the underlying causes remain obscure. Plasminogen activator inhibitor-1 (PAI-1) is a key regulator of fibrinolysis that is also associated with an increased risk of cardiovascular diseases. We examined the effect of experimental chronic circadian clock disruption on PAI-1 expression in mice. Mice were exposed to chronic phase shifts and fed with a high-fat/high-sucrose diet. Chronic phase shifts resulted in increased plasma PAI-1 level through inducing PAI-1 mRNA expression and decreasing tissue-type plasminogen activator (tPA) mRNA expression in the liver. Chronic circadian clock disruption might induce hypofibrinolysis and increase the risk of cardiovascular events by inducing the PAI-1 gene expression in obese individuals.


Subject(s)
Cardiovascular Diseases/etiology , Chronobiology Disorders/blood , Gene Expression Regulation , Serpin E2/biosynthesis , Animals , Chronic Disease , Chronobiology Disorders/complications , Chronobiology Disorders/genetics , Corticosterone/blood , Diet, High-Fat/adverse effects , Dietary Sucrose/toxicity , Fibrinolysis , Liver/metabolism , Male , Mice , Mice, Inbred ICR , Obesity/complications , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Risk Factors , Serpin E2/blood , Serpin E2/genetics , Thrombophilia/etiology , Thrombophilia/physiopathology , Tissue Plasminogen Activator/biosynthesis , Tissue Plasminogen Activator/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...