Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 259
Filter
1.
J Dev Orig Health Dis ; 13(5): 527-540, 2022 10.
Article in English | MEDLINE | ID: mdl-34725018

ABSTRACT

In utero diet may be directly related to the risk of fetal hyperinsulinaemia and offspring metabolic health. This review examines the relationship between maternal dietary exposures and sub-clinical fetal hyperinsulinaemia and neonatal adiposity. Articles were identified in MEDLINE, Web of Science, Cochrane Controlled Register of Controlled Trials, Cumulative Index to Nursing and Allied Health Literature, SCOPUS, and SPORTDiscus (September 2019-March 2021) using the preferred reporting items for systematic reviews and meta-analyses guidelines. PROSPERO registration ID CRD42020146453. Studies were selected by two independent reviewers. Randomised controlled trials (RCT) involving a dietary intervention with pregnant women (healthy pregnancy, gestational diabetes mellitus and obesity) and reporting fetal cord-blood insulin, c-peptide, glucose or adiposity estimates were included. One author extracted all information on main study characteristics and outcomes. Risk of bias was assessed using the Cochrane Collaboration's bias risk assessment tool. A total of 733 articles were identified. Fourteen articles from 11 RCTs (3614 participants) were included. Studies reviewed showed no specific effect of maternal diet on neonatal cord blood insulin, c-peptide or glucose levels. Infants born to mothers who followed a low glycaemic load (GL) had lower skin fold thickness compared to controls. Interventions that provided individualised nutrition counselling to women with obesity or previous infant born > 4 kg were also associated with lower adiposity. The studies reviewed suggest that lifestyle-based dietary interventions to improve glycaemia (low GL) have a protective effect against excess adiposity. Future studies should incorporate multi-modal interventions with dietary counselling to support lifestyle changes throughout gestation and include assessments of maternal insulin resistance at recruitment.


Subject(s)
Adiposity , Hyperinsulinism , Blood Glucose , C-Peptide , Diet , Female , Humans , Hyperinsulinism/etiology , Hyperinsulinism/prevention & control , Infant , Infant, Newborn , Insulin , Obesity , Pregnancy
2.
Am J Physiol Lung Cell Mol Physiol ; 321(6): L1105-L1118, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34668415

ABSTRACT

Increased insulin is associated with obesity-related airway hyperreactivity and asthma. We tested whether the use of metformin, an antidiabetic drug used to reduce insulin resistance, can reduce circulating insulin, thereby preventing airway hyperreactivity in rats with dietary obesity. Male and female rats were fed a high- or low-fat diet for 5 wk. Some male rats were simultaneously treated with metformin (100 mg/kg orally). In separate experiments, after 5 wk of a high-fat diet, some rats were switched to a low-fat diet, whereas others continued a high-fat diet for an additional 5 wk. Bronchoconstriction and bradycardia in response to bilateral electrical vagus nerve stimulation or to inhaled methacholine were measured in anesthetized and vagotomized rats. Body weight, body fat, caloric intake, fasting glucose, and insulin were measured. Vagally induced bronchoconstriction was potentiated only in male rats on a high-fat diet. Males gained more body weight, body fat, and had increased levels of fasting insulin compared with females. Metformin prevented development of vagally induced airway hyperreactivity in male rats on high-fat diet, in addition to inhibiting weight gain, fat gain, and increased insulin. In contrast, switching rats to a low-fat diet for 5 wk reduced body weight and body fat, but it did not reverse fasting glucose, fasting insulin, or potentiation of vagally induced airway hyperreactivity. These data suggest that medications that target insulin may be effective treatment for obesity-related asthma.


Subject(s)
Asthma/drug therapy , Bronchial Hyperreactivity/drug therapy , Bronchoconstriction , Diet, High-Fat/adverse effects , Hyperinsulinism/prevention & control , Metformin/pharmacology , Obesity/complications , Animals , Asthma/chemically induced , Asthma/metabolism , Asthma/pathology , Bronchial Hyperreactivity/chemically induced , Bronchial Hyperreactivity/metabolism , Bronchial Hyperreactivity/pathology , Bronchoconstrictor Agents/toxicity , Female , Glucose/metabolism , Hyperinsulinism/etiology , Hyperinsulinism/metabolism , Hyperinsulinism/pathology , Hypoglycemic Agents/pharmacology , Male , Methacholine Chloride/toxicity , Rats , Rats, Sprague-Dawley , Vagus Nerve/drug effects , Weight Gain
3.
Front Endocrinol (Lausanne) ; 12: 701590, 2021.
Article in English | MEDLINE | ID: mdl-34484117

ABSTRACT

The beneficial effects of metformin, especially its capacity to ameliorate insulin resistance (IR) in polycystic ovary syndrome (PCOS), explains why it is widely prescribed. However, its effect on the offspring of patients with PCOS remains uncertain. This study investigated the impact of metformin treatment on the first- and second-generation female offspring born to letrozole-induced PCOS-IR rats. Forty-five female Wistar rats were implanted with continuous-release letrozole pellets or placebo and treated with metformin or vehicle control. Rats exposed to letrozole showed PCOS-like reproductive, endocrine, and metabolic phenotypes in contrast to the controls. Metformin significantly decreased the risk of body weight gain and increased INSR expression in F1 female offspring in PCOS-IR rats, contributing to the improvement in obesity, hyperinsulinemia, and IR. Decreased FSHR expression and increased LHCGR expression were observed in F1 female rats of the PCOS-IR and PCOS-IR+Metformin groups, suggesting that FSHR and LHCGR dysfunction might promote the development of PCOS. Nevertheless, we found no significant differences in INSR, FSHR, and LHCGR expression or other PCOS phenotypes in F2 female offspring of PCOS-IR rats. These findings indicated widespread reproductive, endocrine, and metabolic changes in the PCOS-IR rat model, but the PCOS phenotypes could not be stably inherited by the next generations. Metformin might have contributed to the improvement in obesity, hyperinsulinemia, and IR in F1 female offspring. The results of this study could be used as a theoretical basis in support of using metformin in the treatment of PCOS-IR patients.


Subject(s)
Hyperinsulinism/prevention & control , Insulin Resistance , Letrozole/toxicity , Metabolic Syndrome/prevention & control , Metformin/pharmacology , Obesity/prevention & control , Polycystic Ovary Syndrome/drug therapy , Animals , Animals, Newborn , Antineoplastic Agents/toxicity , Female , Hypoglycemic Agents/pharmacology , Polycystic Ovary Syndrome/chemically induced , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/pathology , Rats , Rats, Wistar
4.
Horm Metab Res ; 53(9): 616-624, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34496412

ABSTRACT

Insulin resistance (IR) is a pivotal process in various metabolic diseases. The well-known treatment is lifestyle modification and medication therapy, which may result in poor compliance and side effects. Melatonin has been suggested to have a role in glucose metabolism, yet the results across studies have been inconsistent. Therefore, we performed a systematic review to evaluate the effects of melatonin supplementation on insulin levels and IR. We searched PubMed, Embase, Cochrane Central Register of Controlled Trials, and ClinicalTrials.gov, and identified randomized controlled trials (RCTs) published prior to August 2020. Articles were reviewed, selected and extracted by two reviewers independently. In total, 8 RCTs of 376 participants were included. Data were pooled using a random-effects model, with mean differences (MDs) and 95% confidence intervals (CIs). Our results showed that melatonin administration significantly reduced insulin levels and homeostasis model assessment of insulin resistance (HOMA-IR), and increased the quantitative insulin sensitivity check index (QUICKI). We conclude that melatonin ameliorated hyperinsulinemia, insulin resistance, and insulin sensitivity, and the results are an update of a previous meta-analysis. Although more investigations are required, we clearly provide evidence for the use of melatonin as an adjuvant treatment for metabolic disorders involving IR.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Dietary Supplements , Hyperinsulinism/prevention & control , Insulin Resistance , Insulin/blood , Melatonin/administration & dosage , Antioxidants/administration & dosage , Humans , Prognosis , Randomized Controlled Trials as Topic
5.
J Endocrinol ; 250(3): 105-116, 2021 07 12.
Article in English | MEDLINE | ID: mdl-34156345

ABSTRACT

The well-balanced secretion between insulin and growth hormone (GH) is essential in regulating substrate metabolism, energy metabolism, and body composition. High insulin and low GH levels are often observed in obesity, contributing to reduced energy expenditure and further fat accumulation. Although suppression of hyperinsulinemia is proposed as a treatment for obesity, changes in GH secretion and energy metabolism following this treatment are not thoroughly studied. This leaves unexplained observations, such as unchanged lean mass following insulin reduction. In this study, high-fat diet-induced obese (DIO) and normal chow-fed lean mice on a C57BL/6J background were treated for 7 weeks with diazoxide (1250 mg/kg in food), a KATP channel opener that suppressed insulin secretion. Diazoxide treatment for 10 days was sufficient to increase pulsatile GH secretion in DIO mice before any significant body weight change. The restored insulin-GH balance in DIO mice was followed by improvement in substrate and energy metabolism in a prolonged treatment period (4-6 weeks), including reduced fat mass, increased lipid oxidation and energy expenditure, as well as improved insulin sensitivity and metabolic flexibility. These metabolic benefits occurred along with the changes in the expression level of genes regulated by the insulin-GH balance. When applying diazoxide to normal chow-fed normoinsulinemic lean mice, none of the above metabolic effects was observed, suggesting that the metabolic changes following diazoxide treatment were mediated through the suppression of hyperinsulinemia. These results suggest that suppression of hyperinsulinemia by diazoxide restores GH secretion and improves substrate and energy metabolism in DIO mice.


Subject(s)
Diazoxide/therapeutic use , Growth Hormone/metabolism , Hyperinsulinism/prevention & control , Obesity/metabolism , Animals , Body Composition/drug effects , Diazoxide/pharmacology , Diet, High-Fat , Energy Metabolism/drug effects , Hyperinsulinism/complications , Hyperinsulinism/metabolism , Insulin/metabolism , Insulin Resistance/physiology , Insulin Secretion/drug effects , Lipid Metabolism/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/complications , Obesity/drug therapy
6.
Radiol Oncol ; 55(1): 7-17, 2021 Jan 12.
Article in English | MEDLINE | ID: mdl-33885236

ABSTRACT

BACKGROUND: Scientific evidence has shown that an increase in regular physical activity is associated with a decrease in the development of many types of cancer. Potential mechanisms that link physical activity to reduced cancer risk include a decrease in systemic inflammation, hyperinsulinemia, insulin-like growth factor (IGF-I), sex hormones, pro-inflammatory leptin and other obesity-related cytokines, and a significant increase in anti-inflammatory adiponectin levels. In addition, physical activity improves immune function and the composition and diversity of the gastrointestinal microbiota. Moderate physical activity is important for cancer protection, but the most significant changes in the inflammatory profile are conferred by physical activity performed at higher intensities. Thus, there is a need for further investigation into the type, intensity, and duration of physical activity for the prevention of some types of cancer and the development of effective recommendations. CONCLUSIONS: There is a strong evidence that physical activity of moderate to vigorous intensity protects against colon and breast cancer, and probably against cancer at all other sites.


Subject(s)
Exercise , Neoplasms/prevention & control , Biomarkers, Tumor/blood , Breast Neoplasms/immunology , Breast Neoplasms/prevention & control , Colonic Neoplasms/immunology , Colonic Neoplasms/prevention & control , Humans , Hyperinsulinism/immunology , Hyperinsulinism/prevention & control , Inflammation/immunology , Inflammation/prevention & control , Neoplasms/immunology , Risk
7.
Nat Commun ; 12(1): 565, 2021 01 25.
Article in English | MEDLINE | ID: mdl-33495474

ABSTRACT

Accumulating evidence indicates that obesity with its associated metabolic dysregulation, including hyperinsulinemia and aberrant circadian rhythms, increases the risk for a variety of cancers including postmenopausal breast cancer. Caloric restriction can ameliorate the harmful metabolic effects of obesity and inhibit cancer progression but is difficult to implement and maintain outside of the clinic. In this study, we aim to test a time-restricted feeding (TRF) approach on mouse models of obesity-driven postmenopausal breast cancer. We show that TRF abrogates the obesity-enhanced mammary tumor growth in two orthotopic models in the absence of calorie restriction or weight loss. TRF also reduces breast cancer metastasis to the lung. Furthermore, TRF delays tumor initiation in a transgenic model of mammary tumorigenesis prior to the onset of obesity. Notably, TRF increases whole-body insulin sensitivity, reduces hyperinsulinemia, restores diurnal gene expression rhythms in the tumor, and attenuates tumor growth and insulin signaling. Importantly, inhibition of insulin secretion with diazoxide mimics TRF whereas artificial elevation of insulin through insulin pumps implantation reverses the effect of TRF, suggesting that TRF acts through modulating hyperinsulinemia. Our data suggest that TRF is likely to be effective in breast cancer prevention and therapy.


Subject(s)
Breast Neoplasms/prevention & control , Disease Models, Animal , Fasting , Hyperinsulinism/prevention & control , Obesity/prevention & control , Postmenopause/physiology , Animals , Breast Neoplasms/blood , Breast Neoplasms/physiopathology , Caloric Restriction/methods , Cell Line, Tumor , Diet, High-Fat , Female , Humans , Hyperinsulinism/blood , Hyperinsulinism/physiopathology , Insulin Resistance/physiology , Mice, Inbred C57BL , Mice, Obese , Obesity/blood , Obesity/physiopathology , Ovariectomy , Postmenopause/blood
8.
Front Endocrinol (Lausanne) ; 11: 599165, 2020.
Article in English | MEDLINE | ID: mdl-33324349

ABSTRACT

ARHGAP21 is a RhoGAP protein implicated in the modulation of insulin secretion and energy metabolism. ARHGAP21 transient-inhibition increase glucose-stimulated insulin secretion (GSIS) in neonatal islets; however, ARHGAP21 heterozygote mice have a reduced insulin secretion. These discrepancies are not totally understood, and it might be related to functional maturation of beta cells and peripheral sensitivity. Here, we investigated the real ARHGAP21 role in the insulin secretion process using an adult mouse model of acute ARHGAP21 inhibition, induced by antisense. After ARHGAP21 knockdown induction by antisense injection in 60-day old male mice, we investigated glucose and insulin tolerance test, glucose-induced insulin secretion, glucose-induced intracellular calcium dynamics, and gene expression. Our results showed that ARHGAP21 acts negatively in the GSIS of adult islet. This effect seems to be due to the modulation of important points of insulin secretion process, such as the energy metabolism (PGC1α), Ca2+ signalization (SYTVII), granule-extrusion (SNAP25), and cell-cell interaction (CX36). Therefore, based on these finds, ARHGAP21 may be an important target in Diabetes Mellitus (DM) treatment.


Subject(s)
GTPase-Activating Proteins/physiology , Gene Expression Regulation/drug effects , Glucose/pharmacology , Hyperinsulinism/prevention & control , Insulin Secretion , Insulin-Secreting Cells/drug effects , Animals , Homeostasis , Hyperinsulinism/metabolism , Hyperinsulinism/pathology , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Sweetening Agents/pharmacology
9.
FASEB J ; 34(8): 10640-10656, 2020 08.
Article in English | MEDLINE | ID: mdl-32579292

ABSTRACT

Eicosapentaenoic acid (EPA) has garnered attention after the success of the REDUCE-IT trial, which contradicted previous conclusions on EPA for cardiovascular disease risk. Here we first investigated EPA's preventative role on hyperglycemia and hyperinsulinemia. EPA ethyl esters prevented obesity-induced glucose intolerance, hyperinsulinemia, and hyperglycemia in C57BL/6J mice. Supporting NHANES analyses showed that fasting glucose levels of obese adults were inversely related to EPA intake. We next investigated how EPA improved murine hyperinsulinemia and hyperglycemia. EPA overturned the obesity-driven decrement in the concentration of 18-hydroxyeicosapentaenoic acid (18-HEPE) in white adipose tissue and liver. Treatment of obese inbred mice with RvE1, the downstream immunoresolvant metabolite of 18-HEPE, but not 18-HEPE itself, reversed hyperinsulinemia and hyperglycemia through the G-protein coupled receptor ERV1/ChemR23. To translate the findings, we determined if the effects of RvE1 were dependent on host genetics. RvE1's effects on hyperinsulinemia and hyperglycemia were divergent in diversity outbred mice that model human genetic variation. Secondary SNP analyses further confirmed extensive genetic variation in human RvE1/EPA-metabolizing genes. Collectively, the data suggest EPA prevents hyperinsulinemia and hyperglycemia, in part, through RvE1's activation of ERV1/ChemR23 in a host genetic manner. The studies underscore the need for personalized administration of RvE1 based on genetic/metabolic enzyme profiles.


Subject(s)
Eicosapentaenoic Acid/analogs & derivatives , Eicosapentaenoic Acid/pharmacology , Hyperglycemia/genetics , Hyperglycemia/prevention & control , Hyperinsulinism/genetics , Hyperinsulinism/prevention & control , Adipose Tissue, White/drug effects , Animals , Glucose Intolerance/genetics , Liver/drug effects , Male , Mice , Mice, Inbred C57BL , Obesity/genetics , Polymorphism, Single Nucleotide/genetics , Receptors, Chemokine/genetics , Receptors, G-Protein-Coupled/genetics
10.
J Endocrinol ; 246(1): 79-92, 2020 07.
Article in English | MEDLINE | ID: mdl-32369774

ABSTRACT

Glucocorticoids mediate numerous essential processes in the human body via binding to the glucocorticoid receptor (GR). Excessive GR signaling can cause disease, and GR antagonists can be used to treat many symptoms of glucocorticoid-induced pathology. The purpose of this study was to characterize the tissue-specific properties of the selective GR antagonist CORT125281. We evaluated the antagonistic effects of CORT125281 upon acute and subchronic corticosterone exposure in mice. In the acute corticosterone setting, hypothalamus-pituitary-adrenal-axis activity was investigated by measurement of basal- and stress-induced corticosterone levels, adrenocorticotropic hormone levels and pituitary proopiomelanocortin expression. GR signaling was evaluated by RT-PCR analysis of GR-responsive transcripts in liver, muscle, brown adipose tissue (BAT), white adipose tissue (WAT) and hippocampus. Pretreatment with a high dose of CORT125281 antagonized GR activity in a tissue-dependent manner. We observed complete inhibition of GR-induced target gene expression in the liver, partial blockade in muscle and BAT and no antagonism in WAT and hippocampus. Tissue distribution only partially explained the lack of effective antagonism. CORT125281 treatment did not disinhibit the hypothalamus-pituitary-adrenal neuroendocrine axis. In the subchronic corticosterone setting, CORT125281 partially prevented corticosterone-induced hyperinsulinemia, but not hyperlipidemia and immune suppression. In conclusion, CORT125281 antagonizes GR transcriptional activity in a tissue-dependent manner and improves corticosterone-induced hyperinsulinemia. Tailored dosing of CORT125281 may allow tissue-specific inhibition of GR transcriptional activity.


Subject(s)
Pituitary-Adrenal System/drug effects , Pituitary-Adrenal System/metabolism , Receptors, Glucocorticoid/antagonists & inhibitors , Receptors, Glucocorticoid/metabolism , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Animals , Corticosterone/pharmacology , Hippocampus/drug effects , Hippocampus/metabolism , Hyperinsulinism/chemically induced , Hyperinsulinism/prevention & control , Hyperlipidemias/metabolism , Hyperlipidemias/prevention & control , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/metabolism , Liver/drug effects , Liver/metabolism , Mice , Mifepristone/pharmacology , Muscles/drug effects , Muscles/metabolism
11.
PLoS Biol ; 18(2): e3000603, 2020 02.
Article in English | MEDLINE | ID: mdl-32092075

ABSTRACT

Type 2 diabetes (T2D) is characterized by insulin resistance along with pancreatic ß cell failure. ß cell factors are traditionally thought to control glucose homeostasis by modulating insulin levels, not insulin sensitivity. Exosomes are emerging as new regulators of intercellular communication. However, the role of ß-cell-derived exosomes in metabolic homeostasis is poorly understood. Here, we report that microRNA-26a (miR-26a) in ß cells not only modulates insulin secretion and ß cell replication in an autocrine manner but also regulates peripheral insulin sensitivity in a paracrine manner through circulating exosomes. MiR-26a is reduced in serum exosomes of overweight humans and is inversely correlated with clinical features of T2D. Moreover, miR-26a is down-regulated in serum exosomes and islets of obese mice. Using miR-26a knockin and knockout mouse models, we showed that miR-26a in ß cells alleviates obesity-induced insulin resistance and hyperinsulinemia. Mechanistically, miR-26a in ß cells enhances peripheral insulin sensitivity via exosomes. Meanwhile, miR-26a prevents hyperinsulinemia through targeting several critical regulators of insulin secretion and ß cell proliferation. These findings provide a new paradigm for the far-reaching systemic functions of ß cells and offer opportunities for the treatment of T2D.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Insulin Resistance , Insulin-Secreting Cells/metabolism , MicroRNAs/metabolism , Animals , Cell Proliferation , Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/genetics , Exosomes/metabolism , Gene Expression , Gene Expression Regulation , Glucose/metabolism , Humans , Hyperinsulinism/prevention & control , Hyperplasia/prevention & control , Insulin/metabolism , Insulin-Secreting Cells/pathology , Male , Mice , Mice, Obese , Mice, Transgenic , MicroRNAs/blood , MicroRNAs/genetics , Paracrine Communication , Signal Transduction
12.
Am J Physiol Endocrinol Metab ; 317(5): E783-E793, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31454257

ABSTRACT

Perturbations in postnatal leptin signaling have been associated with altered susceptibility to diet-induced obesity (DIO) under high-fat-diet (HFD), albeit with contradicting evidence. Previous studies have shown that alpha murine urokinase-type plasminogen activator (αMUPA) mice have a higher and longer postnatal leptin surge compared with their wild types (WTs) as well as lower body weight and food intake under regular diet (RD). Here we explored αMUPA's propensity for DIO and the effect of attenuating postnatal leptin signaling with leptin antagonist (LA) on energy homeostasis under both RD and HFD. Four-day-old αMUPA pups were treated on alternate days until postnatal day 18 with either vehicle or LA (10 or 20 mg·day-1·kg-1) and weaned into RD or HFD. Compared with RD-fed αMUPA males, HFD-fed αMUPA males showed higher energy intake, even when corrected for body weight difference, and became hyperinsulinemic and obese. Additionally, HFD-fed αMUPA males gained body weight at a higher rate than their WTs mainly because of strain differences in energy expenditure. LA administration did not affect strain differences under RD but attenuated αMUPA's hyperinsulinemia and DIO under HFD, most likely by mediating energy expenditure. Together with our previous findings, these results suggest that αMUPA's leptin surge underlies its higher susceptibility to obesity under HFD, highlighting the role of leptin-related developmental processes in inducing obesity in a postweaning obesogenic environment, at least in αMUPA males. This study therefore supports the use of αMUPA mice for elucidating developmental mechanisms of obesity and the efficacy of early-life manipulations via leptin surge axis in attenuating DIO.


Subject(s)
Diet, High-Fat , Disease Susceptibility , Leptin/antagonists & inhibitors , Obesity/prevention & control , Receptors, Urokinase Plasminogen Activator/genetics , Animals , Animals, Newborn , Body Weight , Eating , Energy Metabolism/drug effects , Female , Hyperinsulinism/prevention & control , Mice , Obesity/etiology , Pregnancy , Signal Transduction/drug effects , Species Specificity , Weight Gain/drug effects
13.
Am J Physiol Endocrinol Metab ; 317(2): E362-E373, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31237447

ABSTRACT

Skeletal muscle insulin resistance manifests shortly after high-fat feeding, yet mechanisms are not known. Here we set out to determine whether excess skeletal muscle membrane cholesterol and cytoskeletal derangement known to compromise glucose transporter (GLUT)4 regulation occurs early after high-fat feeding. We fed 6-wk-old male C57BL/6NJ mice either a low-fat (LF, 10% kcal) or a high-fat (HF, 45% kcal) diet for 1 wk. This HF feeding challenge was associated with an increase, albeit slight, in body mass, glucose intolerance, and hyperinsulinemia. Liver analyses did not reveal signs of hepatic insulin resistance; however, skeletal muscle immunoblots of triad-enriched regions containing transverse tubule membrane showed a marked loss of stimulated GLUT4 recruitment. An increase in cholesterol was also found in these fractions from HF-fed mice. These derangements were associated with a marked loss of cortical filamentous actin (F-actin) that is essential for GLUT4 regulation and known to be compromised by increases in membrane cholesterol. Both the withdrawal of the HF diet and two subcutaneous injections of the cholesterol-lowering agent methyl-ß-cyclodextrin at 3 and 6 days during the 1-wk HF feeding intervention completely mitigated cholesterol accumulation, cortical F-actin loss, and GLUT4 dysregulation. Moreover, these beneficial membrane/cytoskeletal changes occurred concomitant with a full restoration of metabolic responses. These results identify skeletal muscle membrane cholesterol accumulation as an early, reversible, feature of insulin resistance and suggest cortical F-actin loss as an early derangement of skeletal muscle insulin resistance.


Subject(s)
Cell Membrane/metabolism , Cholesterol/metabolism , Diet, High-Fat/adverse effects , Glucose Intolerance/etiology , Insulin Resistance , Muscle, Skeletal/metabolism , Animals , Cell Membrane/drug effects , Cholesterol/pharmacology , Diet, Western/adverse effects , Dietary Fats/pharmacology , Glucose Intolerance/metabolism , Glucose Intolerance/prevention & control , Hyperinsulinism/etiology , Hyperinsulinism/metabolism , Hyperinsulinism/prevention & control , Male , Mice , Mice, Inbred C57BL , Muscle, Skeletal/drug effects , beta-Cyclodextrins/pharmacology , beta-Cyclodextrins/therapeutic use
14.
Nutr Res ; 65: 89-98, 2019 05.
Article in English | MEDLINE | ID: mdl-30952505

ABSTRACT

Including carbohydrate/fructose-rich foods (predominantly fruit) in the diets of overweight individuals can improve chronic disease risk factors. We hypothesized dried plums (DP) would improve nutrient consumption, total antioxidant capacity (TAC), lipid and adipokine profiles, and would decrease adiposity and inflammation. To test this, we studied the effects of 8-weeks of twice-daily snacking of macronutrient-matched 100kcal servings of DP or refined carbohydrate-rich snack (low-fat muffins: LFM) on daily energy and nutrient consumption, and chronic disease risk factors in overweight adults. Body weight/composition, waist circumference, blood pressure, plasma glucose, insulin, c-peptide, lipids, TAC, adipokines and inflammation were measured at baseline and throughout the study. Postprandial glucose and insulin were assessed following assigned test foods at baseline and 8-weeks. Repeated measures ANOVAs were undertaken to examine group and time differences. Post-hoc independent and paired samples t-tests were conducted where necessary. DP increased (P<.05) overall intake of dietary fiber and potassium, and TAC, from baseline to 8-weeks. Baseline postprandial glycemia tended (P=.09) to be lower with DP versus LFM, while both groups had a decreased response after 8-weeks. Postprandial insulinemia was lower (P<.05) for DP at both time-points. No differences in body weight/composition, blood pressure, or fasting glucose, insulin, triglycerides, total cholesterol, HDL-C, inflammation or adipokines were detected. Low-density lipoprotein cholesterol (LDL-C) increased (P<.05) throughout the trial following LFM. Overall, DP lessened postprandial insulinemia, improved nutrient consumption and plasma TAC, and maintained plasma LDL-C compared to a macronutrient-matched refined carbohydrate snack, which could decrease chronic disease risk.


Subject(s)
Antioxidants/metabolism , Blood Glucose/metabolism , Cholesterol, LDL/blood , Diet , Feeding Behavior , Obesity/prevention & control , Prunus domestica , Adipokines/blood , Adiposity , Adult , Analysis of Variance , Cholesterol/blood , Female , Humans , Hyperglycemia/blood , Hyperglycemia/prevention & control , Hyperinsulinism/blood , Hyperinsulinism/prevention & control , Inflammation/blood , Insulin/blood , Male , Middle Aged , Nutrients/administration & dosage , Obesity/blood , Overweight/blood , Overweight/diet therapy , Postprandial Period , Young Adult
15.
Nutr Metab Cardiovasc Dis ; 29(5): 504-512, 2019 05.
Article in English | MEDLINE | ID: mdl-30948306

ABSTRACT

BACKGROUND AND AIMS: Higher fiber intake is associated with increased insulin sensitivity (IS) and reduced glucose-induced insulin secretion (GIIS) during isocaloric-diets; however, its role in hypocaloric-diets is unclear. We examined whether increased fiber intake predicts the amelioration in IS and GIIS following a hypocaloric-diet. METHODS AND RESULTS: This is a post-hoc analysis of 55 adult subjects (BMI > 27 kg/m2) who completed a 6-month hypocaloric-diet (-500 kcal/day). Dietary intake was assessed using 3-day food records at baseline and post-intervention. We evaluated glucose-induced insulin and C-peptide secretions as AUC of plasma insulin and C-peptide during intravenous-glucose-tolerance tests (IVGTT) and IS via hyperinsulinemic-euglycemic clamps. Data analysis employed regression models and 2-way RM ANOVAs. Post-intervention % change in fiber intake was associated positively with ISclamp (r = 0.30) and negatively with % change in total (r = -0.37) and 2nd phase GIISIVGTT (r = -0.44) but not C-peptide secretion. It remained associated with lower 2nd phase GIISIVGTT after adjustment for sex and % changes in BMI and energy-intake, independently of other macronutrients. Subjects who increased fiber intake (to 28.7 ± 9.0 g/day) had a greater decrease in 2nd phase GIISIVGTT, not C-peptide secretion, independently of sex or changes in adiposity or energy-intake compared to subjects who decreased intake (to 20.0 ± 6.8 g/day). CONCLUSION: Higher fiber intake is an independent predictor of reduced 2nd phase glucose-induced hyperinsulinemia after a hypocaloric-diet. It was not associated with plasma C-peptide, suggesting a role in faster insulin clearance rather reduced insulin secretion. Promoting high-fiber intake may increase the effectiveness of hypocaloric-diets in preventing type 2 diabetes. REGISTRATION: ISRCTN14476404, BioMedCentral.com. CLINICAL TRIAL REGISTRATION: This trial was registered at BioMed Central as ISRCTN14476404, on July 28th, 2017.


Subject(s)
Blood Glucose/metabolism , Caloric Restriction , Dietary Fiber/administration & dosage , Hyperinsulinism/prevention & control , Insulin/blood , Obesity/diet therapy , Adiposity , Biomarkers/blood , Caloric Restriction/adverse effects , Female , Humans , Hyperinsulinism/blood , Hyperinsulinism/diagnosis , Hyperinsulinism/etiology , Male , Middle Aged , Obesity/diagnosis , Obesity/physiopathology , Time Factors , Treatment Outcome , Weight Loss
16.
Nutrients ; 11(4)2019 Apr 24.
Article in English | MEDLINE | ID: mdl-31022865

ABSTRACT

Hepatic steatosis, an early stage of non-alcoholic fatty liver disease, is commonly present in obesity and type 2 diabetes, and is associated with reduced hepatic omega-3 polyunsaturated fatty acid (n3-PUFA) status that impacts on the anti-inflammatory and insulin sensitizing functions of n3-PUFA. Our objective was to directly compare plant- and marine-based n3-PUFA (α-linoleic acid (ALA)), eicosapentaenoic acid (EPA), and docosahexaenoic acid (DHA)) for their effects on hepatic steatosis, markers of hepatic inflammation and fibrosis, and insulinemia in obese rats. Fa/fa Zucker rats were provided diets containing ALA, EPA, DHA, or linoleic acid (LA, n6-PUFA) for eight weeks and compared to baseline fa/fa rats and lean Zucker rats fed LA-rich diet for eight weeks. Both DHA and EPA groups had liver lipid similar to baseline, however, DHA was more effective than EPA for reducing hepatic fatty acid synthase (FAS), increasing the proportion of smaller lipid droplets, reversing early fibrotic damage, and reducing fasting hyperinsulinemia. EPA was more effective for reducing FoxO1. Dietary ALA did not attenuate hepatic steatosis, most inflammatory markers or FAS. In summary, amongst the n3-PUFA, DHA was the most effective for elevating hepatic DHA levels, and preventing progression of hepatic steatosis via reductions in FAS and a marker of fibrosis.


Subject(s)
Docosahexaenoic Acids/pharmacology , Eicosapentaenoic Acid/pharmacology , Fatty Liver/prevention & control , Hyperinsulinism/prevention & control , Animal Feed/analysis , Animals , Biomarkers , Docosahexaenoic Acids/administration & dosage , Eicosapentaenoic Acid/administration & dosage , Feeding Behavior , Liver/chemistry , Liver/metabolism , Male , Phospholipids/chemistry , Phospholipids/metabolism , Rats , Rats, Zucker , Triglycerides/chemistry , Triglycerides/metabolism
17.
Int J Mol Sci ; 20(3)2019 Feb 01.
Article in English | MEDLINE | ID: mdl-30717287

ABSTRACT

Atypical antipsychotics, such as olanzapine, are commonly prescribed to patients with schizophrenic symptoms and other psychiatric disorders. However, weight gain and metabolic disturbance cause adverse effects, impair patient compliance and limit clinical utility. Thus, a better understanding of treatment-acquired adverse effects and identification of targets for therapeutic intervention are believed to offer more clinical benefits for patients with schizophrenia. Beyond its nutritional effects, studies have indicated that supplementation of chromium brings about beneficial outcomes against numerous metabolic disorders. In this study, we investigated whether olanzapine-induced weight gain and metabolic disturbance involved chromium dynamic mobilization in a female Sprague-Dawley rat model, and whether a dietary supplement of chromium improved olanzapine-acquired adverse effects. Olanzapine medicated rats experienced weight gain and adiposity, as well as the development of hyperglycemia, hyperinsulinemia, insulin resistance, hyperlipidemia, and inflammation. The olanzapine-induced metabolic disturbance was accompanied by a decrease in hepatic Akt and AMP-activated Protein Kinase (AMPK) actions, as well as an increase in serum interleukin-6 (IL-6), along with tissue chromium depletion. A daily intake of chromium supplements increased tissue chromium levels and thermogenic uncoupling protein-1 (UCP-1) expression in white adipose tissues, as well as improved both post-olanzapine weight gain and metabolic disturbance. Our findings suggest that olanzapine medicated rats showed a disturbance of tissue chromium homeostasis by inducing tissue depletion and urinary excretion. This loss may be an alternative mechanism responsible for olanzapine-induced weight gain and metabolic disturbance.


Subject(s)
Adiposity/drug effects , Antipsychotic Agents/adverse effects , Chlorides/pharmacology , Chromium Compounds/pharmacology , Hyperglycemia/metabolism , Hyperinsulinism/metabolism , Hyperlipidemias/metabolism , Olanzapine/adverse effects , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Adiposity/genetics , Administration, Oral , Animals , Chlorides/metabolism , Chromium Compounds/metabolism , Female , Gene Expression Regulation , Hyperglycemia/chemically induced , Hyperglycemia/genetics , Hyperglycemia/prevention & control , Hyperinsulinism/chemically induced , Hyperinsulinism/genetics , Hyperinsulinism/prevention & control , Hyperlipidemias/chemically induced , Hyperlipidemias/genetics , Hyperlipidemias/prevention & control , Inflammation , Insulin Resistance/genetics , Interleukin-6/genetics , Interleukin-6/metabolism , Liver/drug effects , Liver/metabolism , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism , Weight Gain/drug effects
18.
Nutr Res ; 58: 17-25, 2018 10.
Article in English | MEDLINE | ID: mdl-30340811

ABSTRACT

Dietary supplementation with melinjo (Gnetum gnemon L.) seed extract (MSE) has been proposed as an anti-obesity strategy. However, it remains unclear how MSE modulates energy balance. We tested the hypothesis that dietary MSE reduces energy intake and/or increases physical activity and metabolic thermogenesis in brown and white adipose tissue (BAT and WAT) in mice. Twenty-four C57BL/6 J mice were provided with normal diet, high-fat diet (HFD), or HFD with 1% MSE added, for 17 weeks. Food intake, spontaneous locomotor activity, hepatic triglyceride (TG) content, and blood parameters were examined. Mitochondrial thermogenesis-associated molecule and inflammatory marker expression levels in BAT and WAT were examined by quantitative PCR and western blotting. Dietary MSE did not affect energy intake or spontaneous locomotor activity, but significantly suppressed HFD-induced fat accumulation, hyperglycemia, and hyperinsulinemia. Homeostasis model assessment of insulin resistance score and hepatic TG content were both lower in the MSE-supplemented HFD-fed group than in the HFD-fed group, indicating reduced insulin resistance and a less fatty liver. Dietary MSE upregulated thermogenic uncoupling protein 1 (UCP1) and mitochondrial marker cytochrome c oxidase subunit IV protein expression in BAT; this was closely associated with sirtuin 1 mRNA induction. mRNAs of adipose inflammatory markers, such as monocyte chemotactic 1 and interleukin-1, were induced by HFD but suppressed by MSE. Considering that UCP1 protein expression is the most physiologically relevant parameter to assess the thermogenic capacities of BAT, our results indicate that dietary MSE supplementation induces BAT thermogenesis and reduces obesity-associated adipose tissue inflammation, hepatic steatosis, and insulin resistance.


Subject(s)
Adipose Tissue, Brown/metabolism , Gnetum , Inflammation/metabolism , Insulin Resistance , Obesity/metabolism , Plant Extracts/pharmacology , Uncoupling Protein 1/metabolism , Adipose Tissue, White/metabolism , Animals , Anti-Obesity Agents/pharmacology , Anti-Obesity Agents/therapeutic use , Diet, High-Fat , Dietary Supplements , Electron Transport Complex IV/metabolism , Fatty Liver/etiology , Fatty Liver/metabolism , Fatty Liver/prevention & control , Hyperglycemia/metabolism , Hyperglycemia/prevention & control , Hyperinsulinism/metabolism , Hyperinsulinism/prevention & control , Inflammation/etiology , Inflammation/prevention & control , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/metabolism , Obesity/etiology , Obesity/prevention & control , Plant Extracts/therapeutic use , Seeds , Sirtuin 1/metabolism , Thermogenesis/drug effects , Triglycerides/metabolism
19.
J Physiol Biochem ; 74(3): 441-454, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29797227

ABSTRACT

Physical training (PT) has been considered as a treatment in metabolic syndrome (MS), since it induces thermogenic activity in brown (BAT) and white (WAT) adipose tissues. We evaluated the therapeutic effect of PT on activity of WAT and BAT in rats with MS induced by high-fat diet (30% lard) for 13 weeks and submitted, for the last 6 weeks, to swimming or kept sedentary (SED) rats. MS-SED rats compared to control diet (CT-SED) rats showed low physical fitness and high levels of glucose, insulin, homeostasis evaluation of insulin resistance (HOMA-IR), homeostasis evaluation of the functional capacity of ß-cells (HOMA-ß), and blood pressure. The gastrocnemius muscle decreased in peroxisome proliferator-activated receptor gamma coactivator 1-alpha and beta (PGC-1α, PGC-1ß), and uncoupled protein 2 and 3 (UCP2 and UCP3) expressions. Both WAT and BAT increased in the adipocyte area and decreased in blood vessels and fibroblast numbers. WAT increased in expression of pro-inflammatory adipokines and decreased in anti-inflammatory adipokine and adiponectin. WAT and gastrocnemius showed impairment in the insulin signaling pathway. In response to PT, MS rats showed increased physical fitness and restoration of certain biometric and biochemical parameters and blood pressure. PT also induced thermogenic modulations in skeletal muscle, WAT and BAT, and also improved the insulin signaling pathway. Collectively, PT was effective in treating MS by inducing improvement in physical fitness and interchangeable effects between skeletal muscle, WAT and BAT, suggesting a development of brown-like adipocyte cells.


Subject(s)
Adipose Tissue, Brown/pathology , Adipose Tissue, White/pathology , Adiposity , Insulin Resistance , Metabolic Syndrome/therapy , Physical Conditioning, Animal , Thermogenesis , Adipokines/genetics , Adipokines/metabolism , Adipose Tissue, Brown/blood supply , Adipose Tissue, Brown/immunology , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/blood supply , Adipose Tissue, White/immunology , Adipose Tissue, White/metabolism , Animals , Biomarkers/blood , Diet, High-Fat/adverse effects , Dietary Fats/adverse effects , Gene Expression Regulation , Hyperglycemia/etiology , Hyperglycemia/prevention & control , Hyperinsulinism/etiology , Hyperinsulinism/prevention & control , Hypertension/etiology , Hypertension/prevention & control , Male , Metabolic Syndrome/metabolism , Metabolic Syndrome/pathology , Metabolic Syndrome/physiopathology , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscle, Skeletal/metabolism , Random Allocation , Rats, Inbred F344 , Weaning
20.
Sci Rep ; 8(1): 5899, 2018 04 12.
Article in English | MEDLINE | ID: mdl-29651002

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is a particularly deadly disease. Chronic conditions, including obesity and type-2 diabetes are risk factors, thus making PDAC amenable to preventive strategies. We aimed to characterize the chemo-preventive effects of metformin, a widely used anti-diabetic drug, on PDAC development using the KrasG12D mouse model subjected to a diet high in fats and calories (HFCD). LSL-KrasG12D/+;p48-Cre (KC) mice were given control diet (CD), HFCD, or HFCD with 5 mg/ml metformin in drinking water for 3 or 9 months. After 3 months, metformin prevented HFCD-induced weight gain, hepatic steatosis, depletion of intact acini, formation of advanced PanIN lesions, and stimulation of ERK and mTORC1 in pancreas. In addition to reversing hepatic and pancreatic histopathology, metformin normalized HFCD-induced hyperinsulinemia and hyperleptinemia among the 9-month cohort. Importantly, the HFCD-increased PDAC incidence was completely abrogated by metformin (p < 0.01). The obesogenic diet also induced a marked increase in the expression of TAZ in pancreas, an effect abrogated by metformin. In conclusion, administration of metformin improved the metabolic profile and eliminated the promoting effects of diet-induced obesity on PDAC formation in KC mice. Given the established safety profile of metformin, our findings have a strong translational potential for novel chemo-preventive strategies for PDAC.


Subject(s)
Carcinogenesis/drug effects , Carcinoma, Pancreatic Ductal/prevention & control , Fatty Liver/prevention & control , Hyperinsulinism/prevention & control , Metformin/pharmacology , Obesity/prevention & control , Pancreatic Neoplasms/prevention & control , Acyltransferases , Administration, Oral , Animals , Carcinogenesis/genetics , Carcinogenesis/metabolism , Carcinogenesis/pathology , Carcinoma, Pancreatic Ductal/etiology , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Chemoprevention/methods , Diet, High-Fat/adverse effects , Disease Models, Animal , Drinking Water , Fatty Liver/etiology , Fatty Liver/genetics , Fatty Liver/pathology , Female , Gene Expression Regulation , Humans , Hyperinsulinism/etiology , Hyperinsulinism/genetics , Hyperinsulinism/pathology , Male , Mechanistic Target of Rapamycin Complex 1/genetics , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Obesity/etiology , Obesity/genetics , Obesity/pathology , Pancreatic Neoplasms/etiology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/deficiency , Proto-Oncogene Proteins p21(ras)/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Weight Gain/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...