Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 853
1.
Nutrients ; 14(2)2022 Jan 13.
Article En | MEDLINE | ID: mdl-35057507

Lupins have a unique nutrient profile among legumes and may have beneficial health effects when included in the diet. The aim of this systematic review was to investigate the effects of lupin on a range of health outcome measures. Databases included MEDLINE, Embase and CINAHL, and focused on controlled intervention studies on healthy adults and those with chronic disease such as type 2 diabetes, cardiovascular disease and overweight. The Preferred Reporting Items for Systematic Reviews and Meta-Analyses protocol was followed. Investigated intervention diets utilised whole lupin, lupin protein or lupin fibre, and outcomes were measured by markers of chronic disease, body weight and satiety. Quality assessment of results was performed using the Cochrane revised risk of bias tool. Overall, 21 studies with 998 participants were included: 12 using whole lupin, four used lupin protein and five lupin fibre. Beneficial changes were observed in 71% of studies that measured blood pressure, 83% measuring satiety and 64% measuring serum lipids. Unintended weight loss occurred in 25% of studies. Whole lupin demonstrated more consistent beneficial effects for satiety, glycaemic control and blood pressure than lupin protein or lupin fibre. Heterogeneity, low study numbers and a small participant base indicated further studies are required to strengthen current evidence particularly regarding the protein and dietary fibre components of lupin.


Cardiovascular Diseases/diet therapy , Diabetes Mellitus, Type 2/diet therapy , Diet/methods , Lupinus , Overweight/diet therapy , Body Weight/drug effects , Female , Humans , Male , Satiation/drug effects
2.
Physiol Rep ; 9(17): e15002, 2021 09.
Article En | MEDLINE | ID: mdl-34435472

Pancreatic polypeptide (PP) is known to affect food intake. In this exploratory study, we set out to investigate its supraphysiological effect on food tolerance, gastric accommodation, and emptying. In 12 healthy volunteers, 0, 3, or 10 pmol*kg-1 *min-1 PP was administered intravenously (PP0, PP3 or PP10). Thirty minutes thereafter, nutrient drink infusion (60 ml*min-1 ) through a nasogastric feeding tube was started until maximum satiation. Gastric accommodation was assessed by measuring the intragastric pressure (IGP; nasogastric manometry). In a separate test, the effect of PP0 or PP10 on gastric emptying was tested in 10 healthy volunteers and assessed using the 13 C breath test. Results are presented as mean ± SEM, and p < 0.05 was considered significant. For the IGP test, PP increased ingested nutrient volume: 886 ± 93, 1059 ± 124, and 1025 ± 125 ml for PP0, PP3, and PP10, respectively (p = 0.048). In all groups, Nadir IGP values were reached upon food intake (transformed values: 1.5 ± 0.2, 1.7 ± 0.3, and 1.6 ± 0.3 mmHg for PP0, PP3, and PP10, respectively; NS) to return to baseline thereafter. For the gastric emptying study, volunteers ingested a similar nutrient volume: 802 ± 119 and 1089 ± 128 ml (p = 0.016), and gastric half-emptying time was 281 ± 52 and 249 ± 37 min for PP0 and PP10, respectively (NS). No significant correlation between tolerated nutrient volume and IGP drop (R² < 0.01; p = 0.88 for PP0 vs. PP3 and R² =0.07; p = 0.40 for PP0 vs. PP10, respectively) or gastric half-emptying time (R² = 0.12; p = 0.32) was found. A supraphysiological PP dose enhances food tolerance; however, this effect is not mediated through gastric motility. CLINICAL TRIAL REGISTRY NUMBER: NCT03854708 is obtained from clinicaltrials.gov.


Fasting/blood , Gastric Emptying/physiology , Nutrients/administration & dosage , Pancreatic Polypeptide/administration & dosage , Pancreatic Polypeptide/blood , Protein Precursors/administration & dosage , Protein Precursors/blood , Satiation/physiology , Cross-Over Studies , Gastric Emptying/drug effects , Humans , Manometry/methods , Satiation/drug effects , Single-Blind Method
3.
Nutrients ; 13(8)2021 Jul 29.
Article En | MEDLINE | ID: mdl-34444773

Structural differences in dietary fatty acids modify their rate of oxidation and effect on satiety, endpoints that may influence the development of obesity. This study tests the hypothesis that meals containing fat sources with elevated unsaturated fats will result in greater postprandial energy expenditure, fat oxidation, and satiety than meals containing fats with greater saturation. In a randomized, 5-way crossover design, healthy men and women (n = 23; age: 25.7 ± 6.6 years; BMI: 27.7 ± 3.8 kg/m2) consumed liquid meals containing 30 g of fat from heavy cream (HC), olive oil (OO), sunflower oil (SFO), flaxseed oil (FSO), and fish oil (FO). Energy expenditure and diet-induced thermogenesis (DIT) were determined by metabolic rate over a 240 min postprandial period. Serum concentrations of ghrelin, glucose, insulin, and triacylglycerol (TAG) were assessed. DIT induced by SFO was 5% lower than HC and FO (p = 0.04). Energy expenditure and substrate oxidation did not differ between fat sources. Postprandial TAG concentrations were significantly affected by fat source (p = 0.0001). Varying fat sources by the degree of saturation and PUFA type modified DIT but not satiety responses in normal to obese adult men and women.


Dietary Fats/pharmacology , Fatty Acids, Unsaturated/pharmacology , Fatty Acids/pharmacology , Satiation/drug effects , Thermogenesis/drug effects , Adolescent , Adult , Cross-Over Studies , Energy Metabolism/drug effects , Fats/chemistry , Fats/metabolism , Fats/pharmacology , Fatty Acids/chemistry , Fatty Acids/metabolism , Fatty Acids, Monounsaturated/pharmacology , Fatty Acids, Unsaturated/chemistry , Female , Humans , Male , Meals , Middle Aged , Obesity/metabolism , Olive Oil/pharmacology , Oxidation-Reduction , Postprandial Period/drug effects , Satiety Response/drug effects , Young Adult
4.
Front Endocrinol (Lausanne) ; 12: 676869, 2021.
Article En | MEDLINE | ID: mdl-34168616

Diet-induced gastrointestinal distension is known to evoke satiation and suppress postprandial hyperglycemia; however, the underlying mechanisms remain poorly understood. This study explored how gastrointestinal distension regulates energy homeostasis by using inflating stomach formulation (ISF), the carbonated solution containing pectin that forms stable gel bubbles under acidic condition in the stomach. Here we show that, in mice, oral administration of ISF induced distension of stomach and proximal intestine temporarily, stimulated intestinal glucagon-like peptide-1 (GLP-1) secretion, and activated vagal afferents and brainstem. ISF suppressed food intake and improved glucose tolerance via enhancing insulin sensitivity. The anorexigenic effect was partially inhibited, and the beneficial glycemic effect was blunted by pharmacological GLP-1 receptor blockade and chemical denervation of capsaicin-sensitive sensory nerves. In HFD-fed obese mice showing arrhythmic feeding and obesity, subchronic ISF treatment at the light period (LP) onset for 10 days attenuated LP hyperphagia and visceral fat accumulation. These results demonstrate that gastrointestinal distension by ISF stimulates GLP-1 secretion and the vagal afferent signaling to the brain, thereby regulating feeding behavior and glucose tolerance. Furthermore, subchronic ISF treatment ameliorates HFD-induced visceral obesity. We propose the diet that induces gastrointestinal distension as a novel treatment of hyperphagic obesity and diabetes.


Carbonated Beverages , Eating/drug effects , Glucagon-Like Peptide 1/metabolism , Insulin/blood , Intestines/drug effects , Pectins/administration & dosage , Vagus Nerve/drug effects , Animals , Diet, High-Fat , Feeding Behavior/drug effects , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Mice , Satiation/drug effects
5.
Endocrinology ; 162(9)2021 09 01.
Article En | MEDLINE | ID: mdl-34089599

The obesity pandemic requires effective preventative and therapeutic intervention strategies. Successful and sustained obesity treatment is currently limited to bariatric surgery. Modulating the release of gut hormones is considered promising to mimic bariatric surgery with its beneficial effects on food intake, body weight, and blood glucose levels. The gut peptide secretin was the first molecule to be termed a hormone; nevertheless, only recently has it been established as a legitimate anorexigenic peptide. In contrast to gut hormones that crosstalk with the brain either directly or by afferent neuronal projections, secretin mediates meal-associated brown fat thermogenesis to induce meal termination, thereby qualifying this physiological mechanism as an attractive, peripheral target for the treatment of obesity. In this perspective, it is of pivotal interest to deepen our as yet superficial knowledge on the physiological roles of secretin as well as meal-associated thermogenesis in energy balance and body weight regulation. Of note, the emerging differences between meal-associated thermogenesis and cold-induced thermogenesis must be taken into account. In fact, there is no correlation between these 2 entities. In addition, the investigation of potential effects of secretin in hedonic-driven food intake, bariatric surgery and chronic treatment using suitable application strategies to overcome pharmacokinetic limitations will provide further insight into its potential to influence energy balance. The aim of this article is to review the facts on secretin's metabolic effects, address prevailing gaps in our knowledge, and provide an overview on the opportunities and challenges of the therapeutic potential of secretin in body weight control.


Obesity/prevention & control , Satiation/drug effects , Secretin/pharmacology , Animals , Appetite Regulation/drug effects , Appetite Regulation/physiology , Energy Intake/drug effects , Energy Intake/physiology , Energy Metabolism/drug effects , Energy Metabolism/physiology , Humans , Obesity/etiology , Satiation/physiology , Secretin/physiology , Secretin/therapeutic use , Thermogenesis/drug effects
6.
Nutrients ; 13(2)2021 Feb 14.
Article En | MEDLINE | ID: mdl-33672963

Acacia gum (AG) is a non-viscous soluble fiber that is easily incorporated into beverages and foods. To determine its physiological effects in healthy human subjects, we fed 0, 20, and 40 g of acacia gum in orange juice along with a bagel and cream cheese after a 12 h fast and compared satiety, glycemic response, gastrointestinal tolerance, and food intake among treatments. Subjects (n = 48) reported less hunger and greater fullness at 15 min (p = 0.019 and 0.003, respectively) and 240 min (p = 0.036 and 0.05, respectively) after breakfast with the 40 g fiber treatment. They also reported being more satisfied at 15 min (p = 0.011) and less hungry with the 40 g fiber treatment at 30 min (p = 0.012). Subjects reported more bloating, flatulence, and GI rumbling on the 40 g fiber treatment compared to control, although values for GI tolerance were all low with AG treatment. No significant differences were found in area under the curve (AUC) or change from baseline for blood glucose response, although actual blood glucose with 20 g fiber at 30 min was significantly less than control. Individuals varied greatly in their postprandial glucose response to all treatments. AG improves satiety response and may lower peak glucose response at certain timepoints, and it is well tolerated in healthy human subjects. AG can be added to beverages and foods in doses that can help meet fiber recommendations.


Blood Glucose/drug effects , Dietary Fiber/administration & dosage , Gum Arabic/administration & dosage , Postprandial Period/drug effects , Satiation/drug effects , Adult , Area Under Curve , Citrus sinensis , Cross-Over Studies , Double-Blind Method , Eating/drug effects , Female , Fruit and Vegetable Juices , Healthy Volunteers , Humans , Hunger/drug effects , Male
7.
Nutrients ; 13(3)2021 Feb 26.
Article En | MEDLINE | ID: mdl-33652862

Bariatric surgery is an effective intervention for management of obesity through treating dysregulated appetite and achieving long-term weight loss maintenance. Moreover, significant changes in glucose homeostasis are observed after bariatric surgery including, in some cases, type 2 diabetes remission from the early postoperative period and postprandial hypoglycaemia. Levels of a number of gut hormones are dramatically increased from the early period after Roux-en-Y gastric bypass and sleeve gastrectomy-the two most commonly performed bariatric procedures-and they have been suggested as important mediators of the observed changes in eating behaviour and glucose homeostasis postoperatively. In this review, we summarise the current evidence from human studies on the alterations of gut hormones after bariatric surgery and their impact on clinical outcomes postoperatively. Studies which assess the role of gut hormones after bariatric surgery on food intake, hunger, satiety and glucose homeostasis through octreotide use (a non-specific inhibitor of gut hormone secretion) as well as with exendin 9-39 (a specific glucagon-like peptide-1 receptor antagonist) are reviewed. The potential use of gut hormones as biomarkers of successful outcomes of bariatric surgery is also evaluated.


Bariatric Surgery , Blood Glucose/drug effects , Gastrointestinal Hormones/pharmacology , Obesity, Morbid/therapy , Weight Loss/drug effects , Adult , Biomarkers/blood , Combined Modality Therapy , Eating/drug effects , Female , Gastrectomy , Gastric Bypass , Gastrointestinal Hormones/blood , Humans , Hunger/drug effects , Male , Middle Aged , Obesity, Morbid/blood , Octreotide/blood , Octreotide/pharmacology , Peptide Fragments/blood , Peptide Fragments/pharmacology , Postoperative Period , Satiation/drug effects , Treatment Outcome
8.
Neurogastroenterol Motil ; 33(8): e14083, 2021 08.
Article En | MEDLINE | ID: mdl-33615630

BACKGROUND: Gastric motor function alterations have been implicated in the pathogenesis of functional dyspepsia with postprandial distress syndrome (PDS). Prucalopride, a 5-TH4 agonist, is known to stimulate gastrointestinal motility. We aimed to evaluate the effect of prucalopride on gastric sensorimotor function in healthy subjects (HV). METHODS: Barostat and intragastric pressure (IGP) measurements were performed in 17 HV (59% females, age 29.4 ± 2.7 y) after treatment with placebo or prucalopride (2 mg) (single-blind cross-over). Isobaric stepwise distensions and gastric sensations were assessed to determine gastric compliance and sensitivity. Gastric accommodation (GA) with the barostat was quantified before and after ingestion of 200 ml of a nutrient drink (ND). GA measured by IGP was quantified as the drop of IGP from baseline during the intragastric infusion of ND until maximal satiation (60 ml/min). KEY RESULTS: Prucalopride did not affect barostat assessed gastric compliance or sensitivity. No differences were observed in GA after prucalopride. During the barostat study, 10 min after the meal, 7 HVs reported significantly higher ratings for nausea after prucalopride (p < 0.001), and vomiting was induced in 4 of the HVs. A positive correlation was observed between the delta mean perception of nausea with the delta mean increase of intra-balloon volume before and after meal ingestion (r = 0.37, p = 0.03). During IGP measurements, no effect on nutrient tolerance was observed and increased cramp severity scores were observed which were associated with a significant increase of distal IGP (r = 0.78, p < 0.0001). CONCLUSIONS & INFERENCES: Prucalopride does not enhances gastric accommodation but it might increase sensitivity to gastric distention. Furthermore, the increase in sensitivity seems to be related to an increase in nausea with distension. Clinicaltrials.gov: NCT04429802.


Benzofurans/pharmacology , Gastrointestinal Motility/drug effects , Satiation/drug effects , Serotonin 5-HT4 Receptor Agonists/pharmacology , Adult , Benzofurans/adverse effects , Female , Healthy Volunteers , Humans , Male , Nausea/chemically induced , Single-Blind Method
9.
Ned Tijdschr Geneeskd ; 1652021 01 19.
Article Nl | MEDLINE | ID: mdl-33560612

Obesity is a complex endocrine disease, mainly caused by environmental, behavioral and biological factors. Maintaining weight loss is extremely difficult due to the neuro-endocrine dysregulations that stimulate the body to return to the previous, increased, weight. Identifying underlying weight-gaining factors is needed, including medication-related, psychological and endocrine factors, as well as monogenic obesity. The cornerstone of treatment is optimization of lifestyle and all other contributing factors. Achieving at least 5% weight loss already has important health benefits. If combined lifestyle intervention (CLI) alone is not successful, pharmacotherapy or bariatric surgery can be added for patients with increased weight-related health risks. Recently, novel pharmacotherapy became available, among which, liraglutide 3 mg and the combination therapy naltrexone/bupropion, which leads to an additional 5-6% mean weight loss compared to CLI alone. For rare forms of obesity there are specific drugs that target defects in the regulation of hunger and satiety. Promising new pharmacotherapy for obesity is under development.


Anti-Obesity Agents/therapeutic use , Obesity/therapy , Bariatric Surgery , Bupropion/therapeutic use , Combined Modality Therapy , Drug Combinations , Drug Therapy, Combination , Humans , Hunger/drug effects , Life Style , Liraglutide/therapeutic use , Naltrexone/therapeutic use , Satiation/drug effects , Treatment Outcome , Weight Loss/drug effects
10.
Nutrients ; 13(2)2021 Jan 29.
Article En | MEDLINE | ID: mdl-33572825

Rice germ (RG) may be a safe and effective dietary supplement for obesity in menopause, considering its high protein content and considerable amounts of essential amino acids, good fatty acids, and fiber. This pilot randomized, blinded, parallel-group, placebo-controlled pilot trial investigated the effectiveness of 4-weeks RG supplementation (25 g twice a day) on body composition, as primary outcome, measured by Dual Energy X-Ray Absorptiometry (DXA), and metabolic parameters, as secondary outcomes, like amino acid profiles and satiating capacity, in obese postmenopausal women following a tailored hypocaloric diet (25-30% less than daily energy requirements). Twenty-seven women were randomly assigned to the supplemented group (14) or placebo group (13). There was a significant interaction between time and group for body mass index (BMI) (p < 0.0001), waist (p = 0.002) and hip circumferences (p = 0.01), total protein (0.008), albumin (0.005), Homeostasis Model Assessment index score (p = 0.04), glycine (p = 0.002), glutamine (p = 0.004), and histidine (p = 0.007). Haber's means over time showed a clearly greater feeling of satiety for the supplemented compared to the placebo group. These findings indicate that RG supplementation in addition to a tailored diet counterbalanced the metabolic changes typical of menopause, with improvements in BMI, body composition, insulin resistance, amino acid profiles, and satiety.


Amino Acids/blood , Body Composition/drug effects , Dietary Supplements , Obesity/therapy , Oryza , Satiation/drug effects , Absorptiometry, Photon , Aged , Body Mass Index , Diet, Reducing/methods , Double-Blind Method , Female , Humans , Insulin Resistance , Middle Aged , Obesity/physiopathology , Pilot Projects , Postmenopause/metabolism
11.
Endocr Res ; 46(2): 57-65, 2021.
Article En | MEDLINE | ID: mdl-33426974

The vagus nerve and the celiaco-mesenteric ganglia (CMG) are required for reduction of meal size (MS) and prolongation of the intermeal interval (IMI) by intraperitoneal (ip) sulfated cholecystokinin-8 (CCK-8). However, recently we have shown that the gut regulates these responses. Therefore, reevaluating the role of the vagus and the CMG in the feeding responses evoked by CCK is necessary because the gut contains the highest concentration of enteric, vagal and splanchnic afferents and CCK-A receptors, which are required for reduction of food intake by this peptide, compared to other abdominal organs. To address this necessity, we injected sulfated CCK-8 (0, 0.1, 0.5, 1 and 3 nmol/kg) in the aorta, near the gastrointestinal sites of action of the peptide, in three groups of free-feeding rats (n = 10 rats per group), subdiaphragmatic vagotomy (VGX), celiaco-mesenteric ganglionectomy (CMGX) and sham-operated, and recorded seven feeding responses. In the sham group, CCK-8 reduced MS (normal chow), prolonged the intermeal interval (IMI, time between first and second meals), increased satiety ratio (SR, IMI/MS), shortened duration of first meal, reduced total (24 hrs) food intake and reduced number of meals relative to saline vehicle. Vagotomy attenuated all of the previous responses except IMI length and SR, and CMGX attenuated all of those responses. In conclusion, the feeding responses evoked by sulfated CCK-8 require, independently, the vagus nerve and the CMG.


Behavior, Animal/physiology , Cholecystokinin/pharmacology , Feeding Behavior/physiology , Ganglia, Sympathetic/physiology , Peptide Fragments/pharmacology , Satiation/physiology , Sympathectomy , Vagotomy , Vagus Nerve/physiology , Animals , Celiac Artery , Cholecystokinin/administration & dosage , Feeding Behavior/drug effects , Ganglia, Sympathetic/surgery , Male , Peptide Fragments/administration & dosage , Rats , Rats, Sprague-Dawley , Satiation/drug effects , Time Factors , Vagus Nerve/surgery
12.
Appl Physiol Nutr Metab ; 46(8): 887-896, 2021 Aug.
Article En | MEDLINE | ID: mdl-33493023

Research investigating hemp protein consumption on glycemic response is limited. The effects of hemp protein consumption on blood glucose (BG), insulin, and satiety compared with soybean protein and a carbohydrate control were examined. Two acute randomized repeated-measures crossover experiments were conducted. In both, participants consumed the following isocaloric treatments: 40 g of hemp protein (hemp40), 20 g of hemp protein (hemp20), 40 g of soybean protein (soy40), 20 g of soybean protein (soy20), and a carbohydrate control. In experiments 1 (n = 27) and 2 (n = 16), appetite and BG were measured before (0-60 min, pre-pizza) and after a pizza meal (80-200 min, post-pizza). In experiment 1, food intake was measured at 60 min by ad libitum meal; in experiment 2 a fixed meal was provided (based on body weight) and insulin was measured pre-pizza and post-pizza. In both experiments, BG response was affected by treatment (p < 0.01), time (p < 0.001) and time-by-treatment (p < 0.001) from 0-200 min. Protein treatments lowered 0-60-min BG overall mean and area under the curve compared with control (p < 0.05) dose-dependently. In experiment 2, hemp40 and soy40 lowered (p < 0.05) overall mean insulin concentrations compared with hemp20, soy20, and control pre-meal. Results suggest that hemp protein, like soybean, dose-dependently lowers postprandial BG and insulin concentrations compared with a carbohydrate control. Clinical trial registry: NCT02366598 (experiment 1) and NCT02458027 (experiment 2). Novelty: Hemp protein concentrate dose-dependently leads to lower postprandial BG response compared with a carbohydrate control. No differences were seen between hemp and soy protein.


Blood Glucose/drug effects , Cannabis/metabolism , Diet/methods , Dietary Proteins/administration & dosage , Insulin/blood , Satiation/drug effects , Adolescent , Adult , Cross-Over Studies , Dietary Carbohydrates/administration & dosage , Dietary Carbohydrates/blood , Dietary Proteins/blood , Female , Humans , Male , Middle Aged , Soybean Proteins/administration & dosage , Soybean Proteins/blood , Young Adult
13.
Br J Nutr ; 125(1): 92-100, 2021 01 14.
Article En | MEDLINE | ID: mdl-32660667

Stimulation of gastrointestinal taste receptors affects eating behaviour. Intraduodenal infusion of tastants leads to increased satiation and reduced food intake, whereas intraileal infusion of tastants does not affect eating behaviour. Currently, it is unknown whether oral- or intragastric administration of tastants induces a larger effect on eating behaviour. This study investigated the effects of oral- and/or intragastric administration of quinine on food intake, appetite sensations and heart rate variability (HRV). In a blinded randomised crossover trial, thirty-two healthy volunteers participated in four interventions with a 1-week washout: oral placebo and intragastric placebo (OPGP), oral quinine and intragastric placebo (OQGP), oral placebo and intragastric quinine (OPGQ) and oral quinine and intragastric quinine (OQGQ). On test days, 150 min after a standardised breakfast, subjects ingested a capsule containing quinine or placebo and were sham-fed a mixture of quinine or placebo orally. At 50 min after intervention, subjects received an ad libitum meal to measure food intake. Visual analogue scales for appetite sensations were collected, and HRV measurements were performed at regular intervals. Oral and/or intragastric delivery of the bitter tastant quinine did not affect food intake (OPGP: 3273·6 (sem 131·8) kJ, OQGP: 3072·7 (sem 132·2) kJ, OPGQ: 3289·0 (sem 132·6) kJ and OQGQ: 3204·1 (sem 133·1) kJ, P = 0·069). Desire to eat and hunger decreased after OQGP and OPGQ compared with OPGP (P < 0·001 and P < 0·05, respectively), whereas satiation, fullness and HRV did not differ between interventions. In conclusion, sole oral sham feeding with and sole intragastric delivery of quinine decreased desire to eat and hunger, without affecting food intake, satiation, fullness or HRV.


Appetite/drug effects , Aversive Agents/administration & dosage , Eating/drug effects , Quinine/administration & dosage , Sensation/drug effects , Administration, Oral , Adolescent , Adult , Aged , Breakfast , Cross-Over Studies , Duodenum , Feeding Behavior/drug effects , Female , Healthy Volunteers , Heart Rate/drug effects , Humans , Hunger/drug effects , Ileum , Infusions, Parenteral , Male , Middle Aged , Satiation/drug effects , Single-Blind Method , Young Adult
14.
Clin Nutr ; 40(4): 1834-1842, 2021 04.
Article En | MEDLINE | ID: mdl-33092901

BACKGROUND & AIMS: Several studies have evaluated the effect of intragastric injection of botulinum toxin A to treat obesity, achieving mixed results. Our objective is to determine the effect of intragastric botulinum toxin A on weight loss, satiety, biomarkers, and quality of life of obese patients prior bariatric surgery. METHODS: Design: single-centre, randomised, double-blind, placebo-controlled clinical trial in 52 obese patients on bariatric surgery waiting lists. Two-arm parallel: the treatment group was administered intragastric botulinum toxin A by endoscopy, whereas the control group was administered physiological saline solution. Weight loss was evaluated at weeks 2, 4, 8, 16, and 24, as well as changes in body composition, satiety (Visual analogue scale (VAS) and GCSI questionnaire), quality of life (GIQLI questionnaire), and biomarkers of satiety and appetite. RESULTS: Weight loss at weeks 2, 4, 8, 16, and 24 after the endoscopy, with respect to the basal visit, was 0.6 ± 2 kg, 0.4 ± 2.7 kg, 0.4 ± 3.1 kg, 0.2 ± 4.5 kg, and 0.6 ± 4.3 kg for the control group vs 1.9 ± 2.1 kg, 2 ± 2.6 kg, 2.8 ± 4.1 kg, 3.5 ± 5.3 kg, and 4.5 ± 7 kg for the treatment group, respectively, being differences between groups significant at all times (p = 0.016, 0.031, 0.014, 0.021, and 0.023, respectively). Treatment group patients obtained a significantly higher score for GIQLI questionnaire compared with baseline (104.4 ± 13.9 points vs 97.7 ± 15.6 points; p = 0.024), showing a significant improvement in the section of subjective physical capacity. No significant differences were found regarding perception of satiety, or biomarkers of satiety and appetite. CONCLUSIONS: Intragastric injection of botulinum toxin A is an effective and safe procedure to achieve a moderate weight loss and improve quality of life. Registered under clinicaltrialsregister.eu Identifier EudraCT number 2015-004391-29 https://www.clinicaltrialsregister.eu/ctr-search/trial/2015-004391-29/ES.


Bariatric Surgery , Botulinum Toxins, Type A/therapeutic use , Endoscopy, Digestive System/methods , Obesity/drug therapy , Waiting Lists , Adult , Biomarkers/blood , Botulinum Toxins, Type A/administration & dosage , Double-Blind Method , Female , Follow-Up Studies , Humans , Injections , Male , Middle Aged , Obesity/blood , Quality of Life , Satiation/drug effects , Treatment Outcome , Weight Loss/drug effects
15.
Neuropharmacology ; 182: 108397, 2021 01.
Article En | MEDLINE | ID: mdl-33188843

The present study aimed to evaluate the effects of pharmacological manipulation of α-adrenergic agonists in the dorsal raphe nucleus (DR) on food intake in satiated rats. Adult male Wistar rats with chronically implanted cannula in the DR were injected with adrenaline (AD) or noradrenaline (NA) (both at doses of 6, 20 and 60 nmol), or α-1 adrenergic agonist phenylephrine (PHE) or α-2 adrenergic agonist clonidine (CLO) (both at doses of 6 and 20 nmol). The injections were followed by the evaluation of ingestive behaviors. Food and water intake were evaluated for 60 min. Administration of AD and NA at 60 nmol and CLO at 20 nmol increased food intake and decreased latency to start consumption in satiated rats. The ingestive behavior was not significantly affected by PHE treatment in the DR. CLO treatment increased Fos expression in the arcuate nucleus (ARC) and paraventricular nucleus of the hypothalamus (PVN) in rats that were allowed to eat during the experimental recording (AF group). However, when food was not offered during the experiment (WAF group), PVN neurons were not activated, whereas, neuronal activity remained high in the ARC when compared to control group. Noteworthy, ARC POMC neurons expressed Fos in the AF group. However, double-labeled POMC/Fos cells were absent in the ARC of the WAF group, although an increase in Fos expression was observed in non-POMC cells after CLO injections in the WAF group. In conclusion, the data from the present study highlight that the pharmacological activation of DR α-adrenoceptors affects food intake in satiated rats. The feeding response evoked by CLO injections into DR was similar to that induced by NA or AD injections, suggesting that the hyperphagia after NA or AD treatment depends on α-2 adrenoceptors activation. Finally, we have demonstrated that CLO injections into DR impact neuronal activity in the ARC, possibly evoking a homeostatic response toward food intake.


Adrenergic alpha-2 Receptor Agonists/administration & dosage , Clonidine/administration & dosage , Dorsal Raphe Nucleus/drug effects , Eating/drug effects , Receptors, Adrenergic, alpha-2 , Satiation/drug effects , Animals , Dorsal Raphe Nucleus/metabolism , Dose-Response Relationship, Drug , Eating/physiology , Injections, Intraventricular , Male , Rats , Rats, Wistar , Receptors, Adrenergic, alpha-2/metabolism , Satiation/physiology
16.
J Clin Endocrinol Metab ; 106(3): 654-664, 2021 03 08.
Article En | MEDLINE | ID: mdl-33300990

CONTEXT: Fructose compared to glucose has adverse effects on metabolic function, but endocrine responses to oral sucrose vs glucose is not well understood. OBJECTIVE: We investigated how oral sucrose vs glucose affected appetite-regulating hormones, and how biological factors (body mass index [BMI], insulin sensitivity, sex) influence endocrine responses to these 2 types of sugar. DESIGN: Sixty-nine adults (29 men; 23.22 ±â€…3.74 years; BMI 27.03 ±â€…4.96 kg/m2) completed the study. On 2 occasions, participants consumed 300-mL drinks containing 75 g of glucose or sucrose. Blood was sampled at baseline, 10, 35, and 120 minutes post drink for plasma glucose, insulin, glucagon-like peptide (GLP-1)(7-36), peptide YY (PYY)total, and acyl-ghrelin measures. Hormone levels were compared between conditions using a linear mixed model. Interaction models were performed, and results were stratified to assess how biological factors influence endocrine responses. RESULTS: Sucrose vs glucose ingestion provoked a less robust rise in glucose (P < .001), insulin (P < .001), GLP-1 (P < .001), and PYY (P = .02), whereas acyl-ghrelin suppression was similar between the sugars. We found BMI status by sugar interactions for glucose (P = .01) and PYY (P = .03); obese individuals had smaller increases in glucose and PYY levels after consuming sucrose vs glucose. There were interactions between insulin sensitivity and sugar for glucose (P = .003) and insulin (P = .04), and a sex by sugar interaction for GLP-1 (P = .01); men demonstrated smaller increases in GLP-1 in response to oral sucrose vs glucose. CONCLUSION: Sucrose is less efficient at signaling postprandial satiation than glucose, and biological factors influence differential hormone responses to sucrose vs glucose consumption.


Appetite Regulation/drug effects , Glucose/pharmacology , Sucrose/pharmacology , Administration, Oral , Adolescent , Adult , Appetite/drug effects , Appetite/physiology , Blood Glucose/drug effects , Blood Glucose/metabolism , Body Mass Index , Down-Regulation/drug effects , Eating/physiology , Female , Ghrelin/analogs & derivatives , Ghrelin/blood , Glucagon-Like Peptide 1/blood , Glucose/administration & dosage , Glucose Tolerance Test , Humans , Insulin Resistance/physiology , Male , Obesity/blood , Peptide YY/blood , Satiation/drug effects , Satiation/physiology , Sex Characteristics , South Carolina , Sucrose/administration & dosage , Young Adult
17.
Nutrients ; 12(12)2020 Dec 18.
Article En | MEDLINE | ID: mdl-33353080

Disturbed eating behaviours have been widely reported in psychotic disorders since the early 19th century. There is also evidence that antipsychotic (AP) treatment may induce binge eating or other related compulsive eating behaviours. It is therefore possible that abnormal eating patterns may contribute to the significant weight gain and other metabolic disturbances observed in patients with psychosis. In this scoping review, we aimed to explore the underlying psychopathological and neurobiological mechanisms of disrupted eating behaviours in psychosis spectrum disorders and the role of APs in this relationship. A systematic search identified 35 studies that met our eligibility criteria and were included in our qualitative synthesis. Synthesizing evidence from self-report questionnaires and food surveys, we found that patients with psychosis exhibit increased appetite and craving for fatty food, as well as increased caloric intake and snacking, which may be associated with increased disinhibition. Limited evidence from neuroimaging studies suggested that AP-naïve first episode patients exhibit similar neural processing of food to healthy controls, while chronic AP exposure may lead to decreased activity in satiety areas and increased activity in areas associated with reward anticipation. Overall, this review supports the notion that AP use can lead to disturbed eating patterns in patients, which may contribute to AP-induced weight gain. However, intrinsic illness-related effects on eating behaviors remain less well elucidated, and many confounding factors as well as variability in study designs limits interpretation of existing literature in this field and precludes firm conclusions from being made.


Antipsychotic Agents/adverse effects , Feeding Behavior/psychology , Psychotic Disorders/psychology , Antipsychotic Agents/therapeutic use , Appetite/drug effects , Brain/diagnostic imaging , Brain/physiopathology , Bulimia/chemically induced , Case-Control Studies , Clozapine/adverse effects , Clozapine/therapeutic use , Craving/drug effects , Diet Surveys , Energy Intake , Food Preferences , Humans , Hunger/drug effects , Neuroimaging , Olanzapine/adverse effects , Olanzapine/therapeutic use , Psychotic Disorders/drug therapy , Psychotic Disorders/physiopathology , Reward , Satiation/drug effects , Self Report , Snacks/psychology , Weight Gain/drug effects
18.
Nutrients ; 12(11)2020 Nov 08.
Article En | MEDLINE | ID: mdl-33171589

Metabolic syndrome (MetS) is characterised by metabolic abnormalities that increase the risk of developing type 2 diabetes mellitus and cardiovascular disease. Altered levels of circulating ghrelin, several adipokines and inflammatory markers secreted from adipose tissue, such as leptin, adiponectin, tumor necrosis factor alpha, are observed in overweight and obese individuals. We assessed the effect of supplementation with low doses of a cod protein hydrolysate (CPH) on fasting and postprandial levels of acylated ghrelin, as well as fasting levels of adiponectin, leptin and inflammatory markers in subjects with MetS. A multicentre, double-blinded, randomized controlled trial with a parallel group design was conducted. Subjects received a daily supplement of CPH (4 g protein, n = 15) or placebo (0 g protein, n = 15). We observed no effect on fasting or postprandial levels of acylated ghrelin, fasting levels of adiponectin (p = 0.089) or leptin (p = 0.967) after supplementation with CPH, compared to placebo. Overall, our study showed that 8 weeks supplementation with a low dose of CPH in subjects with MetS had no effect on satiety hormones or most of the inflammatory markers, but the levels of high-sensitivity C-reactive protein were statistically significantly different in the CPH-group compared to placebo group. The robustness and clinical relevance of these findings should be explored in future studies with a larger sample size.


Biomarkers/metabolism , Dietary Supplements , Fish Proteins/pharmacology , Inflammation/pathology , Metabolic Syndrome/pathology , Protein Hydrolysates/pharmacology , Satiation/drug effects , Adiponectin/blood , Adult , Female , Ghrelin/blood , Humans , Leptin/blood , Male , Metabolic Syndrome/blood , Middle Aged
19.
Curr Opin Clin Nutr Metab Care ; 23(6): 445-450, 2020 11.
Article En | MEDLINE | ID: mdl-32925180

PURPOSE OF REVIEW: Dietary fibers decrease risk of cardiovascular disease and obesity, but the most important mechanisms for fiber's protective properties are debated. The purpose of the review is to summarize the recent human studies that examine mechanisms how dietary fiber decreases risk of obesity-related disorders. RECENT FINDINGS: Dietary fiber has effects throughout the digestive tract that decrease risk of obesity-related diseases. Soluble, viscous fibers slow absorption of and decrease serum cholesterol. Intake of dietary fiber enhances satiety and reduces food intake at future meals. The importance of gut fermentation and changes in the gut microbiota and metabolites are linked to decrease risk for obesity-related disorders. Dietary fibers alter the gut microbiota and produce metabolites such as short-chain fatty acids that may explain fiber's role in obesity prevention and treatment. Dietary fiber encompasses many plant compounds, so conclusions that dietary fiber reduces or treats obesity-related disorders must be considered by the fiber was fed in the study. SUMMARY: Dietary fiber prevents and treats obesity-related disorders. Mechanisms for this protection include decreased absorption of macronutrients and enhanced satiety. Changes in the gut microbiota and short-chain fatty acids are emerging mechanisms to explain why high fiber diets protect against obesity and have a role in obesity treatment.


Dietary Fiber/pharmacology , Gastrointestinal Absorption/drug effects , Nutritional Physiological Phenomena/drug effects , Obesity/prevention & control , Satiation/drug effects , Diet, Healthy/methods , Fatty Acids, Volatile/metabolism , Gastrointestinal Microbiome/drug effects , Humans , Protective Factors
20.
Nutrients ; 12(9)2020 Sep 04.
Article En | MEDLINE | ID: mdl-32899700

Protein is the most satiating macronutrient, increasing secretion of gastrointestinal hormones and diet induced thermogenesis. In phenylketonuria (PKU), natural protein is restricted with approximately 80% of intake supplied by a synthetic protein source, which may alter satiety response. Casein glycomacropeptide (CGMP-AA), a carbohydrate containing peptide and alternative protein substitute to amino acids (AA), may enhance satiety mediated by its bioactive properties. AIM: In a three-year longitudinal; prospective study, the effect of AA and two different amounts of CGMP-AA (CGMP-AA only (CGMP100) and a combination of CGMP-AA and AA (CGMP50) on satiety, weight and body mass index (BMI) were compared. METHODS: 48 children with PKU completed the study. Median ages of children were: CGMP100; (n = 13), 9.2 years; CGMP50; (n = 16), 7.3 years; and AA (n = 19), 11.1 years. Semi-quantitative dietary assessments and anthropometry (weight, height and BMI) were measured every three months. RESULTS: The macronutrient contribution to total energy intake from protein, carbohydrate and fat was similar across the groups. Adjusting for age and gender, no differences in energy intake, weight, BMI, incidence of overweight or obesity was apparent between the groups. CONCLUSION: In this three-year longitudinal study, there was no indication to support a relationship between CGMP and satiety, as evidenced by decreased energy intake, thereby preventing overweight or obesity. Satiety is a complex multi-system process that is not fully understood.


Amino Acids/administration & dosage , Caseins/administration & dosage , Diet, Protein-Restricted/methods , Energy Intake/drug effects , Glycopeptides/administration & dosage , Phenylketonurias/diet therapy , Satiation/drug effects , Adolescent , Anthropometry , Body Mass Index , Body Weight/drug effects , Child , Child, Preschool , Diet Surveys , Female , Humans , Longitudinal Studies , Male , Nutrients , Phenylalanine/blood , Phenylketonurias/physiopathology , Prospective Studies
...