1
|
Frias-Toral E, Chapela S, Gonzalez V, Martinuzzi A, Locatelli J, Llobera N, Manrique E, Sarno G, Mingo M, Marchese F, Cuomo R, Romaniello L, Perna M, Giordano A, Santella B, Schiavo L. Optimizing Nutritional Management Before and After Bariatric Surgery: A Comprehensive Guide for Sustained Weight Loss and Metabolic Health. Nutrients 2025; 17:688. [PMID: 40005017 DOI: 10.3390/nu17040688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 01/24/2025] [Accepted: 02/12/2025] [Indexed: 02/27/2025] Open
Abstract
Obesity is associated with multiple comorbidities that contribute to increased mortality among affected individuals. There are multiple treatments for this condition, including nutritional interventions, pharmacological therapies, and surgical procedures. Within these, bariatric surgery is an effective treatment option that requires a multidisciplinary approach, both before and after surgery. Nutritional management prior to surgery aims to achieve metabolic control and reduce comorbidities associated with the procedure. Postoperative nutritional management focuses on preventing complications, ensuring adequate nourishment, and providing necessary supplementation for optimal recovery and long-term success. This narrative review examines all these critical aspects of nutritional management in bariatric surgery, including preoperative nutrition, postoperative nutrition and physical activity recommendation, different nutritional aspects according to the type of bariatric surgery, and future directions for investigation.
Collapse
Affiliation(s)
- Evelyn Frias-Toral
- Escuela de Medicina, Universidad Espíritu Santo, Samborondón 0901952, Ecuador
| | - Sebastián Chapela
- Departamento de Bioquímica Humana, Facultad de Medicina, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires C1121ABG, Argentina
- Unidad de Soporte Nutricional, Hospital Británico de Buenos Aires, Ciudad Autónoma de Buenos Aires C1280AEB, Argentina
| | - Victoria Gonzalez
- Unidad de Soporte Metabólico y Nutricional, Sanatorio Allende, Córdoba X5000BFB, Argentina
- Facultad de Ciencias de la Salud, Universidad Católica de Córdoba, Córdoba X5000IYG, Argentina
| | - Andres Martinuzzi
- Unidad de Soporte Nutricional, Sanatorio Rio Negro, Rio Negro R8500BAD, Argentina
- Asuntos Profesionales y Educación, Fresenius Kabi Argentina, Ciudad de Buenos Aires C1428AAU, Argentina
| | - Julieta Locatelli
- Instituto Alexander Fleming, Ciudad Autónoma de Buenos Aires C1426ANZ, Argentina
| | - Natalia Llobera
- Unidad de Soporte Nutricional, Hospital Británico de Buenos Aires, Ciudad Autónoma de Buenos Aires C1280AEB, Argentina
| | - Ezequiel Manrique
- Unidad de Soporte Nutricional, Hospital Privado Universitario de Córdoba, Córdoba X5016KEH, Argentina
- Nutrihome S.A., Ciudad de Buenos Aires C1428AAI, Argentina
| | - Gerardo Sarno
- Scuola Medica Salernitana, "San Giovanni di Dio e Ruggi D'Aragona" University Hospital, 84131 Salerno, Italy
| | - Monica Mingo
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, 84081 Baronissi, Italy
| | - Federica Marchese
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, 84081 Baronissi, Italy
| | - Raffaele Cuomo
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, 84081 Baronissi, Italy
| | - Ludovica Romaniello
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, 84081 Baronissi, Italy
| | - Martina Perna
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, 84081 Baronissi, Italy
| | - Annalisa Giordano
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, 84081 Baronissi, Italy
| | - Biagio Santella
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, 84081 Baronissi, Italy
- National Biodiversity Future Center (NBFC), 90133 Palermo, Italy
| | - Luigi Schiavo
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, 84081 Baronissi, Italy
- National Biodiversity Future Center (NBFC), 90133 Palermo, Italy
| |
Collapse
|
2
|
Martin LE, Lim J. Selective increases in taste sensitivity to glucose as a function of hunger status. Appetite 2025; 207:107901. [PMID: 39933656 DOI: 10.1016/j.appet.2025.107901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 01/06/2025] [Accepted: 02/06/2025] [Indexed: 02/13/2025]
Abstract
Glucose is critical for normal metabolic function in humans. Accordingly, the ability to sense glucose and glucose-containing saccharides is crucial for maintenance of energy homeostasis. Here, we report the evidence that glucose is perceived relatively stronger compared to fructose or sucralose when subjects are hungry. In the initial experiment, we measured the relative sensitivities between glucose and fructose when subjects were fasted vs. fed. Overnight fasted subjects (n = 22) completed a series of 3-AFC tests comparing one target (glucose from a range of concentrations) and two constants (200 mM fructose) before and after consuming mild-tasting breakfast sandwiches until satiated (600-1500 calories). We found that the relative sensitivity to glucose as compared to fructose was significantly higher when individuals were hungry vs. satiated (p < 0.001). We replicated this finding, by comparing the same range of glucose concentrations to a constant sucralose concentration (0.04mM) (N=19, p < 0.001). Importantly, when we compared a fixed concentration of sucralose (0.4mM) to a range of fructose concentrations, we saw no difference in iso-intense concentration before and after eating (N=19, p > 0.05). These findings support the hypothesis that hunger selectively increases taste sensitivity of glucose compared to other sweeteners.
Collapse
Affiliation(s)
- Laura E Martin
- Department of Food Science and Technology, Oregon State University, Corvallis OR, USA
| | - Juyun Lim
- Department of Food Science and Technology, Oregon State University, Corvallis OR, USA; Monell Chemical Senses Center, Philadelphia PA, USA.
| |
Collapse
|
3
|
Dubin RL, Heymsfield SB, Ravussin E, Greenway FL. Glucagon-like peptide-1 receptor agonist-based agents and weight loss composition: Filling the gaps. Diabetes Obes Metab 2024; 26:5503-5518. [PMID: 39344838 DOI: 10.1111/dom.15913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 08/15/2024] [Accepted: 08/16/2024] [Indexed: 10/01/2024]
Abstract
Excess adiposity is at the root of type 2 diabetes (T2D). Glucagon-like peptide-1 receptor agonists (GLP-1RAs) have emerged as first-line treatments for T2D based on significant weight loss results. The composition of weight loss using most diets consists of <25% fat-free mass (FFM) loss, with the remainder from fat stores. Higher amounts of weight loss (achieved with metabolic bariatric surgery) result in greater reductions in FFM. Our aim was to assess the impact that GLP-1RA-based treatments have on FFM. We analysed studies that reported changes in FFM with the following agents: exenatide, liraglutide, semaglutide, and the dual incretin receptor agonist tirzepatide. We performed an analysis of various weight loss interventions to provide a reference for expected changes in FFM. We evaluated studies using dual-energy X-ray absorptiometry (DXA) for measuring FFM (a crude surrogate for skeletal muscle). In evaluating the composition of weight loss, the percentage lost as fat-free mass (%FFML) was equal to ΔFFM/total weight change. The %FFML using GLP-1RA-based agents was between 20% and 40%. In the 28 clinical trials evaluated, the proportion of FFM loss was highly variable, but the majority reported %FFML exceeding 25%. Our review was limited to small substudies and the use of DXA, which does not measure skeletal muscle mass directly. Since FFM contains a variable amount of muscle (approximately 55%), this indirect measure may explain the heterogeneity in the data. Assessing quantity and quality of skeletal muscle using advanced imaging (magnetic resonance imaging) with functional testing will help fill the gaps in our current understanding.
Collapse
Affiliation(s)
- Robert L Dubin
- Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| | | | - Eric Ravussin
- Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| | - Frank L Greenway
- Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| |
Collapse
|
4
|
Badulescu S, Tabassum A, Le GH, Wong S, Phan L, Gill H, Llach CD, McIntyre RS, Rosenblat J, Mansur R. Glucagon-like peptide 1 agonist and effects on reward behaviour: A systematic review. Physiol Behav 2024; 283:114622. [PMID: 38945189 DOI: 10.1016/j.physbeh.2024.114622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 06/17/2024] [Accepted: 06/25/2024] [Indexed: 07/02/2024]
Abstract
INTRODUCTION The roles of metabolic signals, including Glucagon-like peptide 1 (GLP-1), have been implicated in multiple domains outside metabolic regulation. There is a growing interest in repurposing Glucagon-like peptide 1 receptor agonists (GLP-1RAs) as therapeutics for motivation and reward-related behavioural disturbances. Herein, we aim to systematically review the extant evidence on the potential effects of GLP-1RAs on the reward system. METHODS The study followed PRISMA guidelines using databases such as OVID, PubMed, Scopus, and Google Scholar. The search focused on "Reward Behavior" and "Glucagon Like Peptide 1 Receptor Agonists" and was restricted to human studies. Quality assessment achieved by the NIH's Quality Assessment of Controlled Intervention Studies RESULTS: GLP-1RAs consistently reduced energy intake and influenced reward-related behaviour. These agents have been associated with decreased neurocortical activation in response to higher rewards and food cues, particularly high-calorie foods, and lowered caloric intake and hunger levels. DISCUSSION GLP-1RAs show promise in addressing reward dysfunction linked to food stimuli, obesity, and T2DM. They normalize insulin resistance, and might also modulate dopaminergic signalling and reduce anhedonia. Their effects on glycemic variability and cravings suggest potential applications in addiction disorders.
Collapse
Affiliation(s)
- Sebastian Badulescu
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada; Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada.
| | - Aniqa Tabassum
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada; Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
| | - Gia Han Le
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada; Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
| | - Sabrina Wong
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada; Department of Pharmacology, University of Toronto, Toronto, ON, Canada
| | - Lee Phan
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
| | - Hartej Gill
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada; Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
| | - Cristian-Daniel Llach
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada; Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
| | - Roger S McIntyre
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada; Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada; Department of Pharmacology, University of Toronto, Toronto, ON, Canada
| | - Joshua Rosenblat
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada; Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada; Department of Pharmacology, University of Toronto, Toronto, ON, Canada
| | - Rodrigo Mansur
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada; Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
| |
Collapse
|
5
|
Yoshida R, Ninomiya Y. Mechanisms and Functions of Sweet Reception in Oral and Extraoral Organs. Int J Mol Sci 2024; 25:7398. [PMID: 39000505 PMCID: PMC11242429 DOI: 10.3390/ijms25137398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 06/27/2024] [Accepted: 07/03/2024] [Indexed: 07/16/2024] Open
Abstract
The oral detection of sugars relies on two types of receptor systems. The first is the G-protein-coupled receptor TAS1R2/TAS1R3. When activated, this receptor triggers a downstream signaling cascade involving gustducin, phospholipase Cβ2 (PLCβ2), and transient receptor potential channel M5 (TRPM5). The second type of receptor is the glucose transporter. When glucose enters the cell via this transporter, it is metabolized to produce ATP. This ATP inhibits the opening of KATP channels, leading to cell depolarization. Beside these receptor systems, sweet-sensitive taste cells have mechanisms to regulate their sensitivity to sweet substances based on internal and external states of the body. Sweet taste receptors are not limited to the oral cavity; they are also present in extraoral organs such as the gastrointestinal tract, pancreas, and brain. These extraoral sweet receptors are involved in various functions, including glucose absorption, insulin release, sugar preference, and food intake, contributing to the maintenance of energy homeostasis. Additionally, sweet receptors may have unique roles in certain organs like the trachea and bone. This review summarizes past and recent studies on sweet receptor systems, exploring the molecular mechanisms and physiological functions of sweet (sugar) detection in both oral and extraoral organs.
Collapse
Affiliation(s)
- Ryusuke Yoshida
- Department of Oral Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan
- Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan
| | - Yuzo Ninomiya
- Department of Oral Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan
- Graduate School of Dental Science, Kyushu University, Fukuoka 812-8582, Japan
- Monell Chemical Senses Center, Philadelphia, PA 19104, USA
| |
Collapse
|
6
|
Huber H, Schieren A, Holst JJ, Simon MC. Dietary impact on fasting and stimulated GLP-1 secretion in different metabolic conditions - a narrative review. Am J Clin Nutr 2024; 119:599-627. [PMID: 38218319 PMCID: PMC10972717 DOI: 10.1016/j.ajcnut.2024.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 01/03/2024] [Accepted: 01/09/2024] [Indexed: 01/15/2024] Open
Abstract
Glucagon-like peptide 1 (GLP-1), a gastrointestinal peptide and central mediator of glucose metabolism, is secreted by L cells in the intestine in response to food intake. Postprandial secretion of GLP-1 is triggered by nutrient-sensing via transporters and G-protein-coupled receptors (GPCRs). GLP-1 secretion may be lower in adults with obesity/overweight (OW) or type 2 diabetes mellitus (T2DM) than in those with normal glucose tolerance (NGT), but these findings are inconsistent. Because of the actions of GLP-1 on stimulating insulin secretion and promoting weight loss, GLP-1 and its analogs are used in pharmacologic preparations for the treatment of T2DM. However, physiologically stimulated GLP-1 secretion through the diet might be a preventive or synergistic method for improving glucose metabolism in individuals who are OW, or have impaired glucose tolerance (IGT) or T2DM. This narrative review focuses on fasting and postprandial GLP-1 secretion in individuals with different metabolic conditions and degrees of glucose intolerance. Further, the influence of relevant diet-related factors (e.g., specific diets, meal composition, and size, phytochemical content, and gut microbiome) that could affect fasting and postprandial GLP-1 secretion are discussed. Some studies showed diminished glucose- or meal-stimulated GLP-1 response in participants with T2DM, IGT, or OW compared with those with NGT, whereas other studies have reported an elevated or unchanged GLP-1 response in T2DM or IGT. Meal composition, especially the relationship between macronutrients and interventions targeting the microbiome can impact postprandial GLP-1 secretion, although it is not clear which macronutrients are strong stimulants of GLP-1. Moreover, glucose tolerance, antidiabetic treatment, grade of overweight/obesity, and sex were important factors influencing GLP-1 secretion. The results presented in this review highlight the potential of nutritional and physiologic stimulation of GLP-1 secretion. Further research on fasting and postprandial GLP-1 concentrations and the resulting metabolic consequences under different metabolic conditions is needed.
Collapse
Affiliation(s)
- Hanna Huber
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Institute of Neuroscience and Physiology, Mölndal, Sweden; Department Nutrition and Microbiota, University of Bonn, Institute of Nutrition and Food Science, Bonn, Germany
| | - Alina Schieren
- Department Nutrition and Microbiota, University of Bonn, Institute of Nutrition and Food Science, Bonn, Germany
| | - Jens Juul Holst
- Department of Biomedical Sciences, University of Copenhagen, Faculty of Health and Medical Sciences, Copenhagen, Denmark; The Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Faculty of Health and Medical Sciences, Copenhagen, Denmark
| | - Marie-Christine Simon
- Department Nutrition and Microbiota, University of Bonn, Institute of Nutrition and Food Science, Bonn, Germany.
| |
Collapse
|
7
|
Hirayama A, Iwata S, Oike A, Kawabata Y, Nagasato Y, Takai S, Sanematsu K, Takahashi I, Shigemura N. Cellular mechanisms of taste disturbance induced by the non-steroidal anti-inflammatory drug, diclofenac, in mice. Front Cell Neurosci 2023; 17:1279059. [PMID: 38164437 PMCID: PMC10757961 DOI: 10.3389/fncel.2023.1279059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 11/21/2023] [Indexed: 01/03/2024] Open
Abstract
Drug-induced taste disorders are a serious problem in an aging society. This study investigated the mechanisms underlying taste disturbances induced by diclofenac, a non-steroidal anti-inflammatory drug that reduces pain and inflammation by inhibiting the synthesis of prostaglandins by cyclooxygenase enzymes (COX-1 and COX-2). RT-PCR analyses demonstrated the expression of genes encoding arachidonic acid pathway components such as COX-1, COX-2 and prostaglandin synthases in a subset of mouse taste bud cells. Double-staining immunohistochemistry revealed that COX-1 and cytosolic prostaglandin E synthase (cPGES) were co-expressed with taste receptor type-1 member-3 (T1R3), a sweet/umami receptor component, or gustducin, a bitter/sweet/umami-related G protein, in a subset of taste bud cells. Long-term administration of diclofenac reduced the expression of genes encoding COX-1, gustducin and cPGES in mouse taste buds and suppressed both the behavioral and taste nerve responses to sweet and umami taste stimuli but not to other tastants. Furthermore, diclofenac also suppressed the responses of both mouse and human sweet taste receptors (T1R2/T1R3, expressed in HEK293 cells) to sweet taste stimuli. These results suggest that diclofenac may suppress the activation of sweet and umami taste cells acutely via a direct action on T1R2/T1R3 and chronically via inhibition of the COX/prostaglandin synthase pathway inducing down-regulated expression of sweet/umami responsive components. This dual inhibition mechanism may underlie diclofenac-induced taste alterations in humans.
Collapse
Affiliation(s)
- Ayaka Hirayama
- Section of Oral Neuroscience, Graduate School of Dental Science, Kyushu University, Fukuoka, Japan
- Section of Orthodontics and Dentofacial Orthopedics, Division of Oral Health, Growth and Development, Kyushu University, Fukuoka, Japan
| | - Shusuke Iwata
- Section of Oral Neuroscience, Graduate School of Dental Science, Kyushu University, Fukuoka, Japan
| | - Asami Oike
- Section of Oral Neuroscience, Graduate School of Dental Science, Kyushu University, Fukuoka, Japan
| | - Yuko Kawabata
- Section of Oral Neuroscience, Graduate School of Dental Science, Kyushu University, Fukuoka, Japan
| | - Yuki Nagasato
- Section of Oral Neuroscience, Graduate School of Dental Science, Kyushu University, Fukuoka, Japan
| | - Shingo Takai
- Section of Oral Neuroscience, Graduate School of Dental Science, Kyushu University, Fukuoka, Japan
| | - Keisuke Sanematsu
- Section of Oral Neuroscience, Graduate School of Dental Science, Kyushu University, Fukuoka, Japan
- Research and Development Center for Five-Sense Devices, Kyushu University, Fukuoka, Japan
- Oral Health/Brain Health/Total Health Research Center, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Ichiro Takahashi
- Section of Orthodontics and Dentofacial Orthopedics, Division of Oral Health, Growth and Development, Kyushu University, Fukuoka, Japan
| | - Noriatsu Shigemura
- Section of Oral Neuroscience, Graduate School of Dental Science, Kyushu University, Fukuoka, Japan
- Research and Development Center for Five-Sense Devices, Kyushu University, Fukuoka, Japan
| |
Collapse
|
8
|
Pullicin AJ, Wils D, Lim J. Oral glucose sensing in cephalic phase insulin release. Appetite 2023; 191:107070. [PMID: 37788735 DOI: 10.1016/j.appet.2023.107070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 09/07/2023] [Accepted: 09/30/2023] [Indexed: 10/05/2023]
Abstract
Oral stimulation with foods or food components elicits cephalic phase insulin release (CPIR), which limits postprandial hyperglycemia. Despite its physiological importance, the specific gustatory mechanisms that elicit CPIR have not been clearly defined. While most studies point to glucose and glucose-containing saccharides (e.g., sucrose, maltodextrins) as being the most consistent elicitors, it is not apparent whether this is due to the detection of glucose per se, or to the perceived taste cues associated with these stimuli (e.g., sweetness, starchiness). This study investigated potential sensory mechanisms involved with eliciting CPIR in humans, focusing on the role of oral glucose detection and associated taste. Four stimulus conditions possessing different carbohydrate and taste profiles were designed: 1) glucose alone; 2) glucose mixed with lactisole, a sweet taste inhibitor; 3) maltodextrin, which is digested to starchy- and sweet-tasting products during oral processing; and 4) maltodextrin mixed with lactisole and acarbose, an oral digestion inhibitor. Healthy adults (N = 22) attended four sessions where blood samples were drawn before and after oral stimulation with one of the target stimuli. Plasma c-peptide, insulin, and glucose concentrations were then analyzed. Whereas glucose alone elicited CPIR (one-sample t-test, p < 0.05), it did not stimulate the response in the presence of lactisole. Likewise, maltodextrin alone stimulated CPIR (p < 0.05), but maltodextrin with lactisole and acarbose did not. Together, these findings indicate that glucose is an effective CPIR stimulus, but that an associated taste sensation also serves as an important cue for triggering this response in humans.
Collapse
Affiliation(s)
- Alexa J Pullicin
- Department of Food Science and Technology, Oregon State University, Corvallis, OR, USA
| | - Daniel Wils
- Nutrition and Health Department, Roquette Frères, Lestrem, France
| | - Juyun Lim
- Department of Food Science and Technology, Oregon State University, Corvallis, OR, USA.
| |
Collapse
|
9
|
Kawabata Y, Takai S, Sanematsu K, Yoshida R, Kawabata F, Shigemura N. The Antiarrhythmic Drug Flecainide Enhances Aversion to HCl in Mice. eNeuro 2023; 10:ENEURO.0048-23.2023. [PMID: 37696662 PMCID: PMC10515741 DOI: 10.1523/eneuro.0048-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 08/21/2023] [Accepted: 09/04/2023] [Indexed: 09/13/2023] Open
Abstract
Drug-induced taste disorders reduce quality of life, but little is known about the molecular mechanisms by which drugs induce taste disturbances. In this study, we investigated the short-term and long-term effects of the antiarrhythmic drug flecainide, which is known to cause taste dysfunction. Analyses of behavioral responses (licking tests) revealed that mice given a single intraperitoneal injection of flecainide exhibited a significant reduction in preference for a sour tastant (HCl) but not for other taste solutions (NaCl, quinine, sucrose, KCl and monopotassium glutamate) when compared with controls. Mice administered a single dose of flecainide also had significantly higher taste nerve responses to HCl but not to other taste solutions. Compared with controls, mice administered flecainide once-daily for 30 d showed a reduced preference for HCl without any changes in the behavioral responses to other taste solutions. The electrophysiological experiments using HEK293T cells transiently expressing otopetrin-1 (Otop1; the mouse sour taste receptor) showed that flecainide did not alter the responses to HCl. Taken together, our results suggest that flecainide specifically enhances the response to HCl in mice during short-term and long-term administration. Although further studies will be needed to elucidate the molecular mechanisms, these findings provide new insights into the pathophysiology of drug-induced taste disorders.
Collapse
Affiliation(s)
- Yuko Kawabata
- Section of Oral Neuroscience, Graduate School of Dental Science, Kyushu University, Fukuoka 812-8582, Japan
| | - Shingo Takai
- Section of Oral Neuroscience, Graduate School of Dental Science, Kyushu University, Fukuoka 812-8582, Japan
| | - Keisuke Sanematsu
- Section of Oral Neuroscience, Graduate School of Dental Science, Kyushu University, Fukuoka 812-8582, Japan
- Research and Development Center for Five-Sense Devices, Kyushu University, Fukuoka 819-0395, Japan
- Oral Health/Brain Health/Total Health Research Center, Graduate School of Dental Science, Kyushu University, Fukuoka 812-8582, Japan
| | - Ryusuke Yoshida
- Department of Oral Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan
| | - Fuminori Kawabata
- Physiology of Domestic Animals, Faculty of Agriculture and Life Science, Hirosaki University, Hirosaki 036-8561, Japan
| | - Noriatsu Shigemura
- Section of Oral Neuroscience, Graduate School of Dental Science, Kyushu University, Fukuoka 812-8582, Japan
- Research and Development Center for Five-Sense Devices, Kyushu University, Fukuoka 819-0395, Japan
| |
Collapse
|
10
|
Chuong V, Farokhnia M, Khom S, Pince CL, Elvig SK, Vlkolinsky R, Marchette RC, Koob GF, Roberto M, Vendruscolo LF, Leggio L. The glucagon-like peptide-1 (GLP-1) analogue semaglutide reduces alcohol drinking and modulates central GABA neurotransmission. JCI Insight 2023; 8:e170671. [PMID: 37192005 PMCID: PMC10371247 DOI: 10.1172/jci.insight.170671] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 05/10/2023] [Indexed: 05/17/2023] Open
Abstract
Growing evidence indicates that the glucagon-like peptide-1 (GLP-1) system is involved in the neurobiology of addictive behaviors, and GLP-1 analogues may be used for the treatment of alcohol use disorder (AUD). Here, we examined the effects of semaglutide, a long-acting GLP-1 analogue, on biobehavioral correlates of alcohol use in rodents. A drinking-in-the-dark procedure was used to test the effects of semaglutide on binge-like drinking in male and female mice. We also tested the effects of semaglutide on binge-like and dependence-induced alcohol drinking in male and female rats, as well as acute effects of semaglutide on spontaneous inhibitory postsynaptic currents (sIPSCs) from central amygdala (CeA) and infralimbic cortex (ILC) neurons. Semaglutide dose-dependently reduced binge-like alcohol drinking in mice; a similar effect was observed on the intake of other caloric/noncaloric solutions. Semaglutide also reduced binge-like and dependence-induced alcohol drinking in rats. Semaglutide increased sIPSC frequency in CeA and ILC neurons from alcohol-naive rats, suggesting enhanced GABA release, but had no overall effect on GABA transmission in alcohol-dependent rats. In conclusion, the GLP-1 analogue semaglutide decreased alcohol intake across different drinking models and species and modulated central GABA neurotransmission, providing support for clinical testing of semaglutide as a potentially novel pharmacotherapy for AUD.
Collapse
Affiliation(s)
- Vicky Chuong
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program (NIDA IRP) and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research (NIAAA DICBR), NIH, Baltimore and Bethesda, Maryland, USA
- Neurobiology of Addiction Section, NIDA IRP, NIH, Baltimore, Maryland, USA
| | - Mehdi Farokhnia
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program (NIDA IRP) and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research (NIAAA DICBR), NIH, Baltimore and Bethesda, Maryland, USA
| | - Sophia Khom
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
- Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
| | - Claire L. Pince
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program (NIDA IRP) and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research (NIAAA DICBR), NIH, Baltimore and Bethesda, Maryland, USA
- Neurobiology of Addiction Section, NIDA IRP, NIH, Baltimore, Maryland, USA
| | - Sophie K. Elvig
- Neurobiology of Addiction Section, NIDA IRP, NIH, Baltimore, Maryland, USA
| | - Roman Vlkolinsky
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | | | - George F. Koob
- Neurobiology of Addiction Section, NIDA IRP, NIH, Baltimore, Maryland, USA
| | - Marisa Roberto
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - Leandro F. Vendruscolo
- Stress and Addiction Neuroscience Unit, NIDA IRP and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore, Maryland, USA
| | - Lorenzo Leggio
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program (NIDA IRP) and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research (NIAAA DICBR), NIH, Baltimore and Bethesda, Maryland, USA
| |
Collapse
|
11
|
Cawthon CR, Blonde GD, Nisi AV, Bloomston HM, Krubitski B, le Roux CW, Spector AC. Chronic Semaglutide Treatment in Rats Leads to Daily Excessive Concentration-Dependent Sucrose Intake. J Endocr Soc 2023; 7:bvad074. [PMID: 37388574 PMCID: PMC10306276 DOI: 10.1210/jendso/bvad074] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Indexed: 07/01/2023] Open
Abstract
Context The glucagon-like peptide-1 receptor (GLP-1R) agonist semaglutide (SEMA) produces 15% weight loss when chronically administered to humans with obesity. Methods In 2 separate experiments, rats received daily injections of either vehicle (VEH) or SEMA starting at 7 µg/kg body weight (BW) and increasing over 10 days to the maintenance dose (70 µg/kg-BW), emulating clinical dose escalation strategies. Results During dose escalation and maintenance, SEMA rats reduced chow intake and bodyweight. Experiment 2 meal pattern analysis revealed that meal size, not number, mediated these SEMA-induced changes in chow intake. This suggests SEMA affects neural processes controlling meal termination and not meal initiation. Two-bottle preference tests (vs water) began after 10 to 16 days of maintenance dosing. Rats received either an ascending sucrose concentration series (0.03-1.0 M) and 1 fat solution (Experiment 1) or a 4% and 24% sucrose solution in a crossover design (Experiment 2). At lower sucrose concentrations, SEMA-treated rats in both experiments drank sometimes >2× the volume consumed by VEH controls; at higher sucrose concentrations (and 10% fat), intake was similar between treatment groups. Energy intake of SEMA rats became similar to VEH rats. This was unexpected because GLP-1R agonism is thought to decrease the reward and/or increase the satiating potency of palatable foods. Despite sucrose-driven increases in both groups, a significant bodyweight difference between SEMA- and VEH-treated rats remained. Conclusion The basis of the SEMA-induced overconsumption of sucrose at lower concentrations relative to VEH controls remains unclear, but the effects of chronic SEMA treatment on energy intake and BW appear to depend on the caloric sources available.
Collapse
Affiliation(s)
- Carolina R Cawthon
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL 32306, USA
| | - Ginger D Blonde
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL 32306, USA
| | - A Valentina Nisi
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL 32306, USA
| | - Haley M Bloomston
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL 32306, USA
| | - Belle Krubitski
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL 32306, USA
| | - Carel W le Roux
- Diabetes Complications Research Center, Conway Institute, School of Medicine, University College Dublin, Dublin, D04 C1P1, Ireland
| | - Alan C Spector
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL 32306, USA
| |
Collapse
|
12
|
Abstract
Taste is one of our five primary senses, and taste impairment has been shown to increase with aging. The ability to taste allows us to enjoy the food we eat and to avoid foods that are potentially spoiled or poisonous. Recent advances in our understanding of the molecular mechanisms of taste receptor cells located within taste buds help us decipher how taste works. The discoveries of "classic" endocrine hormones in taste receptor cells point toward taste buds being actual endocrine organs. A better understanding of how taste works may help in reversing taste impairment associated with aging.
Collapse
Affiliation(s)
- Chee W Chia
- Intramural Research Program, National Institute on Aging, National Institutes of Health, 3001 S. Hanover Street, 5th Floor, Room NM536, Baltimore, MD 21225, USA
| | - Shayna M Yeager
- Intramural Research Program, National Institute on Aging, National Institutes of Health, 3001 S. Hanover Street, 5th Floor, Room NM547, Baltimore, MD 21225, USA
| | - Josephine M Egan
- Intramural Research Program, National Institute on Aging, National Institutes of Health, 3001 S. Hanover Street, 5th Floor, Room NM527, Baltimore, MD 21225, USA.
| |
Collapse
|
13
|
Al-Ghurayr NK, Al-Mowalad AM, Omar UM, Ashi HM, Al-Shehri SS, AlShaikh AA, AlHarbi SM, Alsufiani HM. Salivary Hormones Leptin, Ghrelin, Glucagon, and Glucagon-Like Peptide 1 and Their Relation to Sweet Taste Perception in Diabetic Patients. J Diabetes Res 2023; 2023:7559078. [PMID: 37223639 PMCID: PMC10202606 DOI: 10.1155/2023/7559078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 05/01/2023] [Accepted: 05/03/2023] [Indexed: 05/25/2023] Open
Abstract
Diabetes mellitus (DM) is one of the most common diseases worldwide. DM may disrupt hormone regulation. Metabolic hormones, leptin, ghrelin, glucagon, and glucagon-like peptide 1, are produced by the salivary glands and taste cells. These salivary hormones are expressed at different levels in diabetic patients compared to control group and may cause differences in the perception of sweetness. This study is aimed at assessing the concentrations of salivary hormones leptin, ghrelin, glucagon, and GLP-1 and their correlations with sweet taste perception (including thresholds and preferences) in patients with DM. A total of 155 participants were divided into three groups: controlled DM, uncontrolled DM, and control groups. Saliva samples were collected to determine salivary hormone concentrations by ELISA kits. Varying sucrose concentrations (0.015, 0.03, 0.06, 0.12, 0.25, 0.5, and 1 mol/l) were used to assess sweetness thresholds and preferences. Results showed a significant increase in salivary leptin concentrations in the controlled DM and uncontrolled DM compared to the control group. In contrast, salivary ghrelin and GLP-1 concentrations were significantly lower in the uncontrolled DM group than in the control group. In general, HbA1c was positively correlated with salivary leptin concentrations and negatively correlated with salivary ghrelin concentrations. Additionally, in both the controlled and uncontrolled DM groups, salivary leptin was negatively correlated with the perception of sweetness. Salivary glucagon concentrations were negatively correlated with sweet taste preferences in both controlled and uncontrolled DM. In conclusion, the salivary hormones leptin, ghrelin, and GLP-1 are produced either higher or lower in patients with diabetes compared to the control group. In addition, salivary leptin and glucagon are inversely associated with sweet taste preference in diabetic patients.
Collapse
Affiliation(s)
- Nada K. Al-Ghurayr
- Biochemistry Department, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ashjan M. Al-Mowalad
- Biochemistry Department, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ulfat M. Omar
- Biochemistry Department, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Princess Dr. Najla Bint Saud Al-Saud Center for Excellence Research in Biotechnology, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Heba M. Ashi
- Department of Dental Public Health, Faculty of Dentistry, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Saad S. Al-Shehri
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Abdelrahman A. AlShaikh
- Department of Medicine, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Shada M. AlHarbi
- Albawadi 1 Primary Health Care Center, King Fahad Hospital, Ministry of Health, Jeddah, Saudi Arabia
| | - Hadeil M. Alsufiani
- Biochemistry Department, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Experimental Biochemistry Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
14
|
Chometton S, Tsan L, Hayes AMR, Kanoski SE, Schier LA. Early-life influences of low-calorie sweetener consumption on sugar taste. Physiol Behav 2023; 264:114133. [PMID: 36801464 PMCID: PMC11062773 DOI: 10.1016/j.physbeh.2023.114133] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 02/14/2023] [Accepted: 02/16/2023] [Indexed: 02/21/2023]
Abstract
Children and adolescents are the highest consumers of added sugars, particularly from sugar-sweetened beverages (SSB). Regular consumption of SSB early in life induces a variety of negative consequences on health that can last into adulthood. Low-calorie sweeteners (LCS) are increasingly used as an alternative to added sugars because they provide a sweet sensation without adding calories to the diet. However, the long-term effects of early-life consumption of LCS are not well understood. Considering LCS engage at least one of the same taste receptors as sugars and potentially modulate cellular mechanisms of glucose transport and metabolism, it is especially important to understand how early-life LCS consumption impacts intake of and regulatory responses to caloric sugars. In our recent study, we found that habitual intake of LCS during the juvenile-adolescence period significantly changed how rats responded to sugar later in life. Here, we review evidence that LCS and sugars are sensed via common and distinct gustatory pathways, and then discuss the implications this has for shaping sugar-associated appetitive, consummatory, and physiological responses. Ultimately, the review highlights the diverse gaps in knowledge that will be necessary to fill to understand the consequences of regular LCS consumption during important phases of development.
Collapse
Affiliation(s)
- Sandrine Chometton
- Department of Biological Sciences, Human and Evolutionary Biology Section, University of Southern California, Los Angeles, CA, USA
| | - Linda Tsan
- Department of Biological Sciences, Human and Evolutionary Biology Section, University of Southern California, Los Angeles, CA, USA
| | - Anna M R Hayes
- Department of Biological Sciences, Human and Evolutionary Biology Section, University of Southern California, Los Angeles, CA, USA
| | - Scott E Kanoski
- Department of Biological Sciences, Human and Evolutionary Biology Section, University of Southern California, Los Angeles, CA, USA
| | - Lindsey A Schier
- Department of Biological Sciences, Human and Evolutionary Biology Section, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
15
|
Oike A, Iwata S, Hirayama A, Ono Y, Nagasato Y, Kawabata Y, Takai S, Sanematsu K, Wada N, Shigemura N. Bisphosphonate affects the behavioral responses to HCl by disrupting farnesyl diphosphate synthase in mouse taste bud and tongue epithelial cells. Sci Rep 2022; 12:21246. [PMID: 36481783 PMCID: PMC9732047 DOI: 10.1038/s41598-022-25755-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022] Open
Abstract
Little is known about the molecular mechanisms underlying drug-induced taste disorders, which can cause malnutrition and reduce quality of life. One of taste disorders is known adverse effects of bisphosphonates, which are administered as anti-osteoporotic drugs. Therefore, the present study evaluated the effects of risedronate (a bisphosphonate) on taste bud cells. Expression analyses revealed that farnesyl diphosphate synthase (FDPS, a key enzyme in the mevalonate pathway) was present in a subset of mouse taste bud and tongue epithelial cells, especially type III sour-sensitive taste cells. Other mevalonate pathway-associated molecules were also detected in mouse taste buds. Behavioral analyses revealed that mice administered risedronate exhibited a significantly enhanced aversion to HCl but not for other basic taste solutions, whereas the taste nerve responses were not affected by risedronate. Additionally, the taste buds of mice administered risedronate exhibited significantly lower mRNA expression of desmoglein-2, an integral component of desmosomes. Taken together, these findings suggest that risedronate may interact directly with FDPS to inhibit the mevalonate pathway in taste bud and tongue epithelial cells, thereby affecting the expression of desmoglein-2 related with epithelial barrier function, which may lead to alterations in behavioral responses to HCl via somatosensory nerves.
Collapse
Affiliation(s)
- Asami Oike
- grid.177174.30000 0001 2242 4849Section of Oral Neuroscience, Graduate School of Dental Science, Kyushu University, Fukuoka, Japan ,grid.177174.30000 0001 2242 4849Section of Interdisciplinary Dentistry, Graduate School of Dental Science, Kyushu University, Fukuoka, Japan
| | - Shusuke Iwata
- grid.177174.30000 0001 2242 4849Section of Oral Neuroscience, Graduate School of Dental Science, Kyushu University, Fukuoka, Japan ,grid.177174.30000 0001 2242 4849Research and Development Center for Five-Sense Devices, Kyushu University, Fukuoka, Japan
| | - Ayaka Hirayama
- grid.177174.30000 0001 2242 4849Section of Oral Neuroscience, Graduate School of Dental Science, Kyushu University, Fukuoka, Japan
| | - Yurika Ono
- grid.177174.30000 0001 2242 4849Section of Oral Neuroscience, Graduate School of Dental Science, Kyushu University, Fukuoka, Japan
| | - Yuki Nagasato
- grid.177174.30000 0001 2242 4849Section of Oral Neuroscience, Graduate School of Dental Science, Kyushu University, Fukuoka, Japan
| | - Yuko Kawabata
- grid.177174.30000 0001 2242 4849Department of Cell Biology, Aging Science, and Pharmacology, Division of Oral Biological Sciences, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Shingo Takai
- grid.177174.30000 0001 2242 4849Section of Oral Neuroscience, Graduate School of Dental Science, Kyushu University, Fukuoka, Japan
| | - Keisuke Sanematsu
- grid.177174.30000 0001 2242 4849Section of Oral Neuroscience, Graduate School of Dental Science, Kyushu University, Fukuoka, Japan ,grid.177174.30000 0001 2242 4849Research and Development Center for Five-Sense Devices, Kyushu University, Fukuoka, Japan ,grid.177174.30000 0001 2242 4849Oral Health/Brain Health/Total Health Research Center, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Naohisa Wada
- grid.177174.30000 0001 2242 4849Section of Interdisciplinary Dentistry, Graduate School of Dental Science, Kyushu University, Fukuoka, Japan
| | - Noriatsu Shigemura
- grid.177174.30000 0001 2242 4849Section of Oral Neuroscience, Graduate School of Dental Science, Kyushu University, Fukuoka, Japan ,grid.177174.30000 0001 2242 4849Research and Development Center for Five-Sense Devices, Kyushu University, Fukuoka, Japan
| |
Collapse
|
16
|
Васюкова ОВ, Окороков ПЛ, Безлепкина ОБ. [Modern strategies for the treatment of childhood obesity]. PROBLEMY ENDOKRINOLOGII 2022; 68:131-136. [PMID: 36689718 PMCID: PMC9939972 DOI: 10.14341/probl13208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 12/07/2022] [Accepted: 12/07/2022] [Indexed: 01/25/2023]
Abstract
The prevalence of obesity and related metabolic disorders in children and adolescents in the Russian Federation is steadily increasing, which requires healthcare professionals to search for new methods of treatment and prevention. The treatment of childhood obesity should be based on a comprehensive approach, including diet therapy, increased physical activity, behavioral therapy and psychological support. To increase the effectiveness of the formation of new eating habits and proper eating behavior, as well as to increase the adherence of children and adolescents to treatment, drug therapy of obesity is used, aimed primarily at reducing appetite. Considering the efficacy and safety of glucagon-like peptide 1 analog (Liraglutide) in adolescents, as well as a small number of gastrointestinal side effects, this drug is promising in the complex treatment of childhood obesity. This review presents an analysis of the literature on non-medicated and drug-based methods of treatment of childhood obesity.
Collapse
|
17
|
Hurley MM, Smith KR, Harris C, Goodman EJ, Carnell S, Kamath V, Moran TH, Steele KE. Investigating relationships between post-prandial gut hormone responses and taste liking ratings prior to and following bariatric surgery: a pilot study. Int J Obes (Lond) 2022; 46:2114-2119. [PMID: 36045151 PMCID: PMC10805172 DOI: 10.1038/s41366-022-01214-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 08/08/2022] [Accepted: 08/10/2022] [Indexed: 11/08/2022]
Abstract
BACKGROUND Alterations in gut hormone secretion and reported changes in taste preferences have been suggested to contribute to the weight-reducing effects of bariatric surgery. However, a link between changes in gut hormone secretion and taste preferences following bariatric surgery has yet to be elucidated. METHODS Here we examined the potential relationships between gut hormone responses (GLP-1 and PYY3-36 peak, ghrelin trough) to a test meal of Ensure and liking ratings for taste mixtures varying in sugar and fat content before and following bariatric surgery (vertical sleeve gastrectomy (VSG): N = 4; Roux-en Y gastric bypass (RYGB): N = 8). RESULTS Significant increases in GLP-1 and PYY3-36 peak and a significant drop in ghrelin trough were observed following surgery. Pre- and postoperation, patients with higher postprandial GLP-1 or PYY3-36 peaks gave lower liking ratings for mixtures containing a combination of fat and sugar (half and half + 20% added sugar) whereas, for the combined surgery analyses, no relationships were found with solutions comprised of high fat (half and half + 0% sugar), predominantly high sugar (skim milk + 20% added sugar), or low fat and low sugar (skim milk + 0% added sugar). Within the RYGB patients, patients with the greatest increase in postprandial GLP-1 peak from preoperation to postoperation also demonstrated the greatest decrease in liking for half & half + 20% added sugar and skim milk + 20% added sugar, but not the unsweetened version of each solution. No pre- or postoperative relationship between ghrelin and liking ratings were observed. CONCLUSION Gut hormone responses following bariatric surgery may contribute to taste processing of sugar+fat mixtures and together influence weight loss.
Collapse
Affiliation(s)
- Matthew M Hurley
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kimberly R Smith
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Civonnia Harris
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ethan J Goodman
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Susan Carnell
- Division of Child and Adolescent Psychiatry, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Vidyulata Kamath
- Division of Medical Psychology, Department of Psychiatry & Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Timothy H Moran
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kimberley E Steele
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
18
|
Sweet Taste Signaling: The Core Pathways and Regulatory Mechanisms. Int J Mol Sci 2022; 23:ijms23158225. [PMID: 35897802 PMCID: PMC9329783 DOI: 10.3390/ijms23158225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/23/2022] [Accepted: 07/25/2022] [Indexed: 12/10/2022] Open
Abstract
Sweet taste, a proxy for sugar-derived calories, is an important driver of food intake, and animals have evolved robust molecular and cellular machinery for sweet taste signaling. The overconsumption of sugar-derived calories is a major driver of obesity and other metabolic diseases. A fine-grained appreciation of the dynamic regulation of sweet taste signaling mechanisms will be required for designing novel noncaloric sweeteners with better hedonic and metabolic profiles and improved consumer acceptance. Sweet taste receptor cells express at least two signaling pathways, one mediated by a heterodimeric G-protein coupled receptor encoded by taste 1 receptor members 2 and 3 (TAS1R2 + TAS1R3) genes and another by glucose transporters and the ATP-gated potassium (KATP) channel. Despite these important discoveries, we do not fully understand the mechanisms regulating sweet taste signaling. We will introduce the core components of the above sweet taste signaling pathways and the rationale for having multiple pathways for detecting sweet tastants. We will then highlight the roles of key regulators of the sweet taste signaling pathways, including downstream signal transduction pathway components expressed in sweet taste receptor cells and hormones and other signaling molecules such as leptin and endocannabinoids.
Collapse
|
19
|
Behavioral responses to sweet compounds via T1R2-independent pathways in chickens. Poult Sci 2022; 101:101928. [PMID: 35679679 PMCID: PMC9189227 DOI: 10.1016/j.psj.2022.101928] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 04/05/2022] [Accepted: 04/18/2022] [Indexed: 11/23/2022] Open
Abstract
Elucidating the taste sensing systems in chickens will enhance our understanding of poultry nutrition and improve the feeding strategies used in poultry farming. It is known that chickens lack the sweet taste receptor subunit, taste receptor type 1 member 2 (T1R2), in their genome. Thus, the present study investigated T1R2-independent sweet-sensing pathways in chickens. RT-PCR analysis revealed that glucose transporters known to play an important role in T1R2-independent sweet sensing in mammals—namely sodium-glucose cotransporter 1 (SGLT1) and ATP-gated K+ channel subunits—are expressed in the palate, the main taste organ in chickens. In behavioral tests, chickens slightly preferred glucose, galactose, sucrose, maltose, lactose, and stevioside, while high doses of sucrose and fructose were rejected. Chickens did not show any preference for noncaloric sweeteners or sugar alcohol, such as acesulfame K, aspartame, saccharin, sucralose, or sorbitol. The preference for galactose was inhibited by an inhibitor of SGLT1 in a dose-dependent manner. In addition, we found that glucagon-like peptide 1 (GLP-1) and mRNA of the GLP-1 receptor, which are involved specifically in sweet transmission in mice, are also present in the oral tissues of chickens. The present results imply that chickens can sense various sweet compounds via T1R2-independent pathways in oral tissues.
Collapse
|
20
|
Caretta A, Mucignat-Caretta C. Not Only COVID-19: Involvement of Multiple Chemosensory Systems in Human Diseases. Front Neural Circuits 2022; 16:862005. [PMID: 35547642 PMCID: PMC9081982 DOI: 10.3389/fncir.2022.862005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 03/21/2022] [Indexed: 11/13/2022] Open
Abstract
Chemosensory systems are deemed marginal in human pathology. In appraising their role, we aim at suggesting a paradigm shift based on the available clinical and experimental data that will be discussed. Taste and olfaction are polymodal sensory systems, providing inputs to many brain structures that regulate crucial visceral functions, including metabolism but also endocrine, cardiovascular, respiratory, and immune systems. Moreover, other visceral chemosensory systems monitor different essential chemical parameters of “milieu intérieur,” transmitting their data to the brain areas receiving taste and olfactory inputs; hence, they participate in regulating the same vital functions. These chemosensory cells share many molecular features with olfactory or taste receptor cells, thus they may be affected by the same pathological events. In most COVID-19 patients, taste and olfaction are disturbed. This may represent only a small portion of a broadly diffuse chemosensory incapacitation. Indeed, many COVID-19 peculiar symptoms may be explained by the impairment of visceral chemosensory systems, for example, silent hypoxia, diarrhea, and the “cytokine storm”. Dysregulation of chemosensory systems may underlie the much higher mortality rate of COVID-19 Acute Respiratory Distress Syndrome (ARDS) compared to ARDSs of different origins. In chronic non-infectious diseases like hypertension, diabetes, or cancer, the impairment of taste and/or olfaction has been consistently reported. This may signal diffuse chemosensory failure, possibly worsening the prognosis of these patients. Incapacitation of one or few chemosensory systems has negligible effects on survival under ordinary life conditions but, under stress, like metabolic imbalance or COVID-19 pneumonia, the impairment of multiple chemosensory systems may lead to dire consequences during the course of the disease.
Collapse
Affiliation(s)
- Antonio Caretta
- National Institute for Biostructures and Biosystems (NIBB), Rome, Italy
- Department of Food and Drug Science, University of Parma, Parma, Italy
| | - Carla Mucignat-Caretta
- National Institute for Biostructures and Biosystems (NIBB), Rome, Italy
- Department of Molecular Medicine, University of Padova, Padua, Italy
- *Correspondence: Carla Mucignat-Caretta,
| |
Collapse
|
21
|
Al-Alsheikh AS, Alabdulkader S, Johnson B, Goldstone AP, Miras AD. Effect of Obesity Surgery on Taste. Nutrients 2022; 14:866. [PMID: 35215515 PMCID: PMC8878262 DOI: 10.3390/nu14040866] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 02/07/2022] [Accepted: 02/15/2022] [Indexed: 12/03/2022] Open
Abstract
Obesity surgery is a highly efficacious treatment for obesity and its comorbidities. The underlying mechanisms of weight loss after obesity surgery are not yet fully understood. Changes to taste function could be a contributing factor. However, the pattern of change in different taste domains and among obesity surgery operations is not consistent in the literature. A systematic search was performed to identify all articles investigating gustation in human studies following bariatric procedures. A total of 3323 articles were identified after database searches, searching references and deduplication, and 17 articles were included. These articles provided evidence of changes in the sensory and reward domains of taste following obesity procedures. No study investigated the effect of obesity surgery on the physiological domain of taste. Taste detection sensitivity for sweetness increases shortly after Roux-en-Y gastric bypass. Additionally, patients have a reduced appetitive reward value to sweet stimuli. For the subgroup of patients who experience changes in their food preferences after Roux-en-Y gastric bypass or vertical sleeve gastrectomy, changes in taste function may be underlying mechanisms for changing food preferences which may lead to weight loss and its maintenance. However, data are heterogeneous; the potential effect dilutes over time and varies significantly between different procedures.
Collapse
Affiliation(s)
- Alhanouf S. Al-Alsheikh
- Department of Metabolism, Digestion and Reproduction, Imperial College London, Hammersmith Hospital, London W12 0NN, UK; (A.S.A.-A.); (S.A.); (B.J.); (A.D.M.)
- Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, Riyadh 11451, Saudi Arabia
| | - Shahd Alabdulkader
- Department of Metabolism, Digestion and Reproduction, Imperial College London, Hammersmith Hospital, London W12 0NN, UK; (A.S.A.-A.); (S.A.); (B.J.); (A.D.M.)
- Department of Health Sciences, College of Health and Rehabilitation Sciences, Princess Nourah Bint Abdulrahman University, Riyadh 84428, Saudi Arabia
| | - Brett Johnson
- Department of Metabolism, Digestion and Reproduction, Imperial College London, Hammersmith Hospital, London W12 0NN, UK; (A.S.A.-A.); (S.A.); (B.J.); (A.D.M.)
| | - Anthony P. Goldstone
- PsychoNeuroEndocrinology Research Group, Division of Psychiatry, Department of Brain Sciences, Imperial College London, Hammersmith Hospital, London W12 0NN, UK
| | - Alexander Dimitri Miras
- Department of Metabolism, Digestion and Reproduction, Imperial College London, Hammersmith Hospital, London W12 0NN, UK; (A.S.A.-A.); (S.A.); (B.J.); (A.D.M.)
| |
Collapse
|
22
|
Treesukosol Y, Moran TH. Administration of Exendin-4 but not CCK alters lick responses and trial initiation to sucrose and intralipid during brief-access tests. Chem Senses 2022; 47:bjac004. [PMID: 35427413 PMCID: PMC9012268 DOI: 10.1093/chemse/bjac004] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Administration of cholecystokinin (CCK) or the glucagon-like peptide 1 (GLP-1) receptor agonist Exendin-4 (Ex-4) reduces food intake. Findings in the literature suggest CCK reduces intake primarily as a satiety signal whereas GLP-1 may play a role in both satiety and reward-related feeding signals. Compounds that humans describe as âsweetâ and âfattyâ are palatable yet are signaled via separate transduction pathways. Here, unconditioned lick responses to sucrose and intralipid were measured in a brief-access lick procedure in food-restricted male rats in response to i.p. administration of Ex-4 (3 h before test), CCK (30 min before test), or a combination of both. The current experimental design measures lick responses to water and varying concentrations of both sucrose (0.03, 0.1, and 0.5 M) and intralipid (0.2%, 2%, and 20%) during 10-s trials across a 30-min single test session. This design minimized postingestive influences. Compared with saline-injected controls, CCK (1.0, 3.0, or 6.0 µg/kg) did not change lick responses to sucrose or intralipid. Number of trials initiated and lick responses to both sucrose and intralipid were reduced in rats injected with 3.0 µg/kg, but not 1.0 µg/kg Ex-4. The supplement of CCK did not alter lick responses or trials initiated compared with Ex-4 administration alone. These findings support a role for GLP-1 but not CCK in the oral responsiveness to palatable stimuli. Furthermore, Ex-4-induced reductions were observed for both sucrose and intralipid, compounds representing âsweetâ and âfat,â respectively.
Collapse
Affiliation(s)
- Yada Treesukosol
- Department of Psychology, California State University Long Beach, Long Beach, CA 90840, United States
| | - Timothy H Moran
- Department of Psychiatry and Behavioral Sciences, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
23
|
Jensterle M, DeVries JH, Battelino T, Battelino S, Yildiz B, Janez A. Glucagon-like peptide-1, a matter of taste? Rev Endocr Metab Disord 2021; 22:763-775. [PMID: 33123893 DOI: 10.1007/s11154-020-09609-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/26/2020] [Indexed: 01/22/2023]
Abstract
Understanding of gustatory coding helps to predict, and perhaps even modulate the ingestive decision circuitry, especially when eating behaviour becomes dysfunctional. Preclinical research demonstrated that glucagon like peptide 1 (GLP-1) is locally synthesized in taste bud cells in the tongue and that GLP-1 receptor exists on the gustatory nerves in close proximity to GLP-1 containing taste bud cells. In humans, the tongue has not yet been addressed as clinically relevant target for GLP-1 based therapies. The primary aim of the current review was to elaborate on the role of GLP- 1 in mammalian gustatory system, in particular in the perception of sweet. Secondly, we aimed to explore what modulates gustatory coding and whether the GLP-1 based therapies might be involved in regulation of taste perception. We performed a series of PubMed, Medline and Embase databases systemic searches. The Population-Intervention-Comparison-Outcome (PICO) framework was used to identify interventional studies. Based on the available data, GLP-1 is specifically involved in the perception of sweet. Aging, diabetes and obesity are characterized by diminished taste and sweet perception. Calorie restriction and bariatric surgery are associated with a diminished appreciation of sweet food. GLP-1 receptor agonists (RAs) modulate food preference, yet its modulatory potential in gustatory coding is currently unknown. Future studies should explore whether GLP-1 RAs modulate taste perception to the extent that changes of food preference and consumption ensue.
Collapse
Affiliation(s)
- Mojca Jensterle
- Division of Internal Medicine, Department of Endocrinology, Diabetes and Metabolic Diseases, University Medical Centre Ljubljana, Zaloška cesta, 7, 1000, Ljubljana, Slovenia
- Department of Internal Medicine, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia, Zaloška cesta 7, 1000, Ljubljana, Slovenia
| | - J Hans DeVries
- Department of Endocrinology and Metabolism, Amsterdam University Medical Center, University of Amsterdam, Meibergdreef 9, 1105, AZ, Amsterdam, The Netherlands
| | - Tadej Battelino
- Department of Endocrinology, Diabetes and Metabolism, University Children's Hospital, University Medical Centre Ljubljana, Bohoričeva 20, SI-1000, Ljubljana, Slovenia
- Department of Pediatrics, Faculty of Medicine, University of Ljubljana, Bohoričeva 20, SI-1000, Ljubljana, Slovenia
| | - Saba Battelino
- Department of Otorhinolaryngology and Cervicofacial Surgery, University Medical Centre Ljubljana, Zaloska cesta 2, 1000, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Bulent Yildiz
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Hacettepe University School of Medicine, Hacettepe, 06100, Ankara, Turkey
| | - Andrej Janez
- Division of Internal Medicine, Department of Endocrinology, Diabetes and Metabolic Diseases, University Medical Centre Ljubljana, Zaloška cesta, 7, 1000, Ljubljana, Slovenia.
- Department of Internal Medicine, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia, Zaloška cesta 7, 1000, Ljubljana, Slovenia.
| |
Collapse
|
24
|
Ben‐Chetrit E, Ben‐Ya'acov A, Quitina A, Atia O, Regev E, Shteyer E, Nashef R. Anosmia and dysgeusia amongst COVID-19 patients are associated with low levels of serum glucagon-like peptide 1. Int J Clin Pract 2021; 75:e14996. [PMID: 34714940 PMCID: PMC8646360 DOI: 10.1111/ijcp.14996] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 10/27/2021] [Indexed: 12/25/2022] Open
Abstract
PURPOSE Anosmia and dysgeusia (AD) are common amongst COVID-19 patients. These symptoms are not frequently associated with rhinorrhea or nasal congestion and the underlying mechanism is unclear. Previous reports suggested that glucagon-like peptide-1 (GLP-1) signalling plays a role in the modulation of olfaction and ageusia. We aimed to assess the correlation between GLP-1 and COVID-19-associated AD. METHODS Blood samples obtained from COVID-19 patients with and without AD were tested for serum GLP-1 levels using enzyme-linked immunosorbent assay (ELISA). A second control group comprised of COVID-19-negative volunteers. RESULTS Forty-nine subjects were included in the study. Nineteen were positive for COVID-19. Of the 19 patients, 10 had AD and 9 declined such complaints. Age and basic metabolic rate were similar amongst all study groups. Serum GLP-1 levels were significantly lower amongst patients with AD compared with patients without AD and COVID-19-negative individuals (1820 pg/mL vs 3536 pg/mL vs 3014 pg/mL, respectively, P < .02). CONCLUSION COVID-19 patients who reported AD had lower serum levels of GLP-1 compared with those lacking AD symptoms and COVID-19-negative individuals. These results suggest that GLP-1 may be involved in the pathogenesis of AD. However, further larger scale studies should corroborate our findings.
Collapse
Affiliation(s)
- Eli Ben‐Chetrit
- Infectious Diseases UnitShaare Zedek Medical CenterHebrew University School of MedicineJerusalemIsrael
| | - Ami Ben‐Ya'acov
- The Juliet Keidan Institute of Pediatric Gastroenterology Hepatology and NutritionShaare Zedek Medical CenterThe Hebrew UniversityJerusalemIsrael
| | - Ahmad Quitina
- The Juliet Keidan Institute of Pediatric Gastroenterology Hepatology and NutritionShaare Zedek Medical CenterThe Hebrew UniversityJerusalemIsrael
| | - Ohad Atia
- The Juliet Keidan Institute of Pediatric Gastroenterology Hepatology and NutritionShaare Zedek Medical CenterThe Hebrew UniversityJerusalemIsrael
| | - Eran Regev
- Oral and Maxillofacial UnitShaare Zedek Medical CenterHebrew University School of MedicineJerusalemIsrael
| | - Eyal Shteyer
- The Juliet Keidan Institute of Pediatric Gastroenterology Hepatology and NutritionShaare Zedek Medical CenterThe Hebrew UniversityJerusalemIsrael
| | - Rizan Nashef
- Oral and Maxillofacial UnitShaare Zedek Medical CenterHebrew University School of MedicineJerusalemIsrael
| |
Collapse
|
25
|
Nielsen MS, Ritz C, Chenchar A, Bredie WLP, Gillum MP, Sjödin A. Does FGF21 Mediate the Potential Decrease in Sweet Food Intake and Preference Following Bariatric Surgery? Nutrients 2021; 13:nu13113840. [PMID: 34836096 PMCID: PMC8624965 DOI: 10.3390/nu13113840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 10/22/2021] [Accepted: 10/25/2021] [Indexed: 11/17/2022] Open
Abstract
The liver-derived hormone fibroblast growth factor 21 (FGF21) has recently been linked to preference for sweet-tasting food. We hypothesized, that surgery-induced changes in FGF21 could mediate the reduction in sweet food intake and preference following bariatric surgery. Forty participants (35 females) with severe obesity (BMI ≥ 35 kg/m2) scheduled for roux-en-y gastric bypass (n = 30) or sleeve gastrectomy (n = 10) were included. Pre- and postprandial responses of intact plasma FGF21 as well as intake of sweet-tasting food assessed at a buffet meal test, the hedonic evaluation of sweet taste assessed using an apple juice with added sucrose and visual analog scales, and sweet taste sensitivity were assessed before and 6 months after bariatric surgery. In a cross-sectional analysis pre-surgery, pre- and postprandial intact FGF21 levels were negatively associated with the hedonic evaluation of a high-sucrose juice sample (p = 0.03 and p = 0.02). However, no changes in pre- (p = 0.24) or postprandial intact FGF21 levels were found 6 months after surgery (p = 0.11), and individual pre- to postoperative changes in pre- and postprandial intact FGF21 levels were not found to be associated with changes in intake of sweet foods, the hedonic evaluation of sweet taste or sweet taste sensitivity (all p ≥ 0.10). In conclusion, we were not able to show an effect of bariatric surgery on circulating FGF21, and individual postoperative changes in FGF21 were not found to mediate an effect of surgery on sweet food intake and preference.
Collapse
Affiliation(s)
- Mette S. Nielsen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark; (M.S.N.); (A.C.); (M.P.G.)
| | - Christian Ritz
- National Institute of Public Health, University of Southern Denmark, 1455 Copenhagen, Denmark;
| | - Anne Chenchar
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark; (M.S.N.); (A.C.); (M.P.G.)
- School of Pharmacy, College of Health Science, University of Wyoming, Laramie, WY 82071, USA
| | - Wender L. P. Bredie
- Department of Food Science, Faculty of Science, University of Copenhagen, 1958 Frederiksberg, Denmark;
| | - Matthew P. Gillum
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark; (M.S.N.); (A.C.); (M.P.G.)
| | - Anders Sjödin
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, 1958 Frederiksberg, Denmark
- Correspondence:
| |
Collapse
|
26
|
Abstract
Gut microbiota has emerged as a major metabolically active organ with critical functions in both health and disease. The trillions of microorganisms hosted by the gastrointestinal tract are involved in numerous physiological and metabolic processes including modulation of appetite and regulation of energy in the host spanning from periphery to the brain. Indeed, bacteria and their metabolic byproducts are working in concert with the host chemosensory signaling pathways to affect both short- and long-term ingestive behavior. Sensing of nutrients and taste by specialized G protein-coupled receptor cells is important in transmitting food-related signals, optimizing nutrition as well as in prevention and treatment of several diseases, notably obesity, diabetes and associated metabolic disorders. Further, bacteria metabolites interact with specialized receptors cells expressed by gut epithelium leading to taste and appetite response changes to nutrients. This review describes recent advances on the role of gut bacteria in taste perception and functions. It further discusses how intestinal dysbiosis characteristic of several pathological conditions may alter and modulate taste preference and food consumption via changes in taste receptor expression.
Collapse
|
27
|
Jensterle M, Ferjan S, Battelino T, Kovač J, Battelino S, Šuput D, Vovk A, Janež A. Does intervention with GLP-1 receptor agonist semaglutide modulate perception of sweet taste in women with obesity: study protocol of a randomized, single-blinded, placebo-controlled clinical trial. Trials 2021; 22:464. [PMID: 34281590 PMCID: PMC8287101 DOI: 10.1186/s13063-021-05442-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 07/09/2021] [Indexed: 11/30/2022] Open
Abstract
Background Preclinical studies demonstrated that glucagon-like peptide 1 (GLP-1) is locally synthesized in taste bud cells and that GLP-1 receptor exists on the gustatory nerves in close proximity to GLP-1-containing taste bud cells. This local paracrine GLP-1 signalling seems to be specifically involved in the perception of sweets. However, the role of GLP-1 in taste perception remains largely unaddressed in clinical studies. Whether any weight-reducing effects of GLP-1 receptor agonists are mediated through the modulation of taste perception is currently unknown. Methods and analysis This is an investigator-initiated, randomized single-blind, placebo-controlled clinical trial. We will enrol 30 women with obesity and polycystic ovary syndrome (PCOS). Participants will be randomized in a 1:1 ratio to either semaglutide 1.0 mg or placebo for 16 weeks. The primary endpoints are alteration of transcriptomic profile of tongue tissue as changes in expression level from baseline to follow-up after 16 weeks of treatment, measured by RNA sequencing, and change in taste sensitivity as detected by chemical gustometry. Secondary endpoints include change in neural response to visual food cues and to sweet-tasting substances as assessed by functional MRI, change in body weight, change in fat mass and change in eating behaviour and food intake. Discussion This is the first study to investigate the role of semaglutide on taste perception, along with a neural response to visual food cues in reward processing regions. The study may identify the tongue and the taste perception as a novel target for GLP-1 receptor agonists. Ethics and disseminations The study has been approved by the Slovene National Medical Ethics Committee and will be conducted in accordance with the Declaration of Helsinki and Good Clinical Practice guidelines. Results will be submitted for publication in an international peer-reviewed scientific journal. Trial registration ClinicalTrials.govNCT04263415. Retrospectively registered on 10 February 2020 Supplementary Information The online version contains supplementary material available at 10.1186/s13063-021-05442-y.
Collapse
Affiliation(s)
- Mojca Jensterle
- Department of Endocrinology, Diabetes and Metabolic Diseases, Division of Internal Medicine, University Medical Centre Ljubljana, Zaloška cesta 7, SI-1000, Ljubljana, Slovenia.,Faculty of Medicine, University of Ljubljana, Vrazov trg 2, SI-1000, Ljubljana, Slovenia
| | - Simona Ferjan
- Department of Endocrinology, Diabetes and Metabolic Diseases, Division of Internal Medicine, University Medical Centre Ljubljana, Zaloška cesta 7, SI-1000, Ljubljana, Slovenia.,Faculty of Medicine, University of Ljubljana, Vrazov trg 2, SI-1000, Ljubljana, Slovenia
| | - Tadej Battelino
- Faculty of Medicine, University of Ljubljana, Vrazov trg 2, SI-1000, Ljubljana, Slovenia.,Department of Endocrinology, Diabetes and Metabolism, University Children's Hospital, University Medical Centre Ljubljana, Bohoričeva 20, SI-1000, Ljubljana, Slovenia
| | - Jernej Kovač
- Faculty of Medicine, University of Ljubljana, Vrazov trg 2, SI-1000, Ljubljana, Slovenia.,Department of Endocrinology, Diabetes and Metabolism, University Children's Hospital, University Medical Centre Ljubljana, Bohoričeva 20, SI-1000, Ljubljana, Slovenia
| | - Saba Battelino
- Faculty of Medicine, University of Ljubljana, Vrazov trg 2, SI-1000, Ljubljana, Slovenia.,Department of Otorhinolaryngology and Cervicofacial Surgery, University Medical Centre Ljubljana, Zaloška cesta 2, SI-1000, Ljubljana, Slovenia
| | - Dušan Šuput
- Faculty of Medicine, University of Ljubljana, Vrazov trg 2, SI-1000, Ljubljana, Slovenia
| | - Andrej Vovk
- Faculty of Medicine, University of Ljubljana, Vrazov trg 2, SI-1000, Ljubljana, Slovenia
| | - Andrej Janež
- Department of Endocrinology, Diabetes and Metabolic Diseases, Division of Internal Medicine, University Medical Centre Ljubljana, Zaloška cesta 7, SI-1000, Ljubljana, Slovenia. .,Faculty of Medicine, University of Ljubljana, Vrazov trg 2, SI-1000, Ljubljana, Slovenia.
| |
Collapse
|
28
|
von Molitor E, Riedel K, Krohn M, Hafner M, Rudolf R, Cesetti T. Sweet Taste Is Complex: Signaling Cascades and Circuits Involved in Sweet Sensation. Front Hum Neurosci 2021; 15:667709. [PMID: 34239428 PMCID: PMC8258107 DOI: 10.3389/fnhum.2021.667709] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 05/20/2021] [Indexed: 12/13/2022] Open
Abstract
Sweetness is the preferred taste of humans and many animals, likely because sugars are a primary source of energy. In many mammals, sweet compounds are sensed in the tongue by the gustatory organ, the taste buds. Here, a group of taste bud cells expresses a canonical sweet taste receptor, whose activation induces Ca2+ rise, cell depolarization and ATP release to communicate with afferent gustatory nerves. The discovery of the sweet taste receptor, 20 years ago, was a milestone in the understanding of sweet signal transduction and is described here from a historical perspective. Our review briefly summarizes the major findings of the canonical sweet taste pathway, and then focuses on molecular details, about the related downstream signaling, that are still elusive or have been neglected. In this context, we discuss evidence supporting the existence of an alternative pathway, independent of the sweet taste receptor, to sense sugars and its proposed role in glucose homeostasis. Further, given that sweet taste receptor expression has been reported in many other organs, the physiological role of these extraoral receptors is addressed. Finally, and along these lines, we expand on the multiple direct and indirect effects of sugars on the brain. In summary, the review tries to stimulate a comprehensive understanding of how sweet compounds signal to the brain upon taste bud cells activation, and how this gustatory process is integrated with gastro-intestinal sugar sensing to create a hedonic and metabolic representation of sugars, which finally drives our behavior. Understanding of this is indeed a crucial step in developing new strategies to prevent obesity and associated diseases.
Collapse
Affiliation(s)
- Elena von Molitor
- Institute of Molecular and Cell Biology, Hochschule Mannheim, Mannheim, Germany
| | | | | | - Mathias Hafner
- Institute of Molecular and Cell Biology, Hochschule Mannheim, Mannheim, Germany
| | - Rüdiger Rudolf
- Institute of Molecular and Cell Biology, Hochschule Mannheim, Mannheim, Germany.,Interdisciplinary Center for Neurosciences, Heidelberg University, Heidelberg, Germany
| | - Tiziana Cesetti
- Institute of Molecular and Cell Biology, Hochschule Mannheim, Mannheim, Germany
| |
Collapse
|
29
|
Decarie-Spain L, Kanoski SE. Ghrelin and Glucagon-Like Peptide-1: A Gut-Brain Axis Battle for Food Reward. Nutrients 2021; 13:977. [PMID: 33803053 PMCID: PMC8002922 DOI: 10.3390/nu13030977] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/14/2021] [Accepted: 03/14/2021] [Indexed: 12/17/2022] Open
Abstract
Eating behaviors are influenced by the reinforcing properties of foods that can favor decisions driven by reward incentives over metabolic needs. These food reward-motivated behaviors are modulated by gut-derived peptides such as ghrelin and glucagon-like peptide-1 (GLP-1) that are well-established to promote or reduce energy intake, respectively. In this review we highlight the antagonizing actions of ghrelin and GLP-1 on various behavioral constructs related to food reward/reinforcement, including reactivity to food cues, conditioned meal anticipation, effort-based food-motivated behaviors, and flavor-nutrient preference and aversion learning. We integrate physiological and behavioral neuroscience studies conducted in both rodents and human to illustrate translational findings of interest for the treatment of obesity or metabolic impairments. Collectively, the literature discussed herein highlights a model where ghrelin and GLP-1 regulate food reward-motivated behaviors via both competing and independent neurobiological and behavioral mechanisms.
Collapse
Affiliation(s)
- Lea Decarie-Spain
- Human & Evolutionary Biology Section, Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089, USA;
| | - Scott E. Kanoski
- Human & Evolutionary Biology Section, Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089, USA;
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA 90089, USA
| |
Collapse
|
30
|
Jensterle M, Rizzo M, Janez A. Glucagon-Like Peptide 1 and Taste Perception: From Molecular Mechanisms to Potential Clinical Implications. Int J Mol Sci 2021; 22:ijms22020902. [PMID: 33477478 PMCID: PMC7830704 DOI: 10.3390/ijms22020902] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 01/03/2021] [Accepted: 01/15/2021] [Indexed: 12/14/2022] Open
Abstract
Preclinical studies provided some important insights into the action of glucagon-like peptide 1 (GLP-1) in taste perception. This review examines the literature to uncover some molecular mechanisms and connections between GLP-1 and the gustatory coding. Local GLP-1 production in the taste bud cells, the expression of GLP-1 receptor on the adjacent nerves, a functional continuum in the perception of sweet chemicals from the gut to the tongue and an identification of GLP-1 induced signaling pathways in peripheral and central gustatory coding all strongly suggest that GLP-1 is involved in the taste perception, especially sweet. However, the impact of GLP-1 based therapies on gustatory coding in humans remains largely unaddressed. Based on the molecular background we encourage further exploration of the tongue as a new treatment target for GLP-1 receptor agonists in clinical studies. Given that pharmacological manipulation of gustatory coding may represent a new potential strategy against obesity and diabetes, the topic is of utmost clinical relevance.
Collapse
Affiliation(s)
- Mojca Jensterle
- Diabetes and Metabolic Diseases, Division of Internal Medicine, Department of Endocrinology, University Medical Centre Ljubljana, Zaloška Cesta 7, 1000 Ljubljana, Slovenia;
- Department of Internal Medicine, Faculty of Medicine, University of Ljubljana, Zaloška Cesta 7, 1000 Ljubljana, Slovenia
| | - Manfredi Rizzo
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of South Carolina, Columbia, SC 29208, USA;
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, 90133 Palermo, Italy
| | - Andrej Janez
- Diabetes and Metabolic Diseases, Division of Internal Medicine, Department of Endocrinology, University Medical Centre Ljubljana, Zaloška Cesta 7, 1000 Ljubljana, Slovenia;
- Department of Internal Medicine, Faculty of Medicine, University of Ljubljana, Zaloška Cesta 7, 1000 Ljubljana, Slovenia
- Correspondence: ; Tel.: +386-1-522-3114; Fax: +386-1-522-9359
| |
Collapse
|
31
|
Have a heart: failure to increase GLP-1 caused by heart failure increases the risk of diabetes. Clin Sci (Lond) 2020; 134:3119-3121. [PMID: 33269792 DOI: 10.1042/cs20201029] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 11/11/2020] [Accepted: 11/12/2020] [Indexed: 11/17/2022]
Abstract
Incretins represent a group of gut-derived peptide hormones that, at physiological concentrations, potentiate the release of insulin. Work leading to the discovery of incretins began as early as the late 1800s where scientists, including Claude Bernard who is widely considered the father of modern physiology (Rehfeld, J.F. The Origin and Understanding of the Incretin Concept. Front. Endocrinol. (Lausanne) (2018) 9, 387; Robin, E.D. Claude Bernard. Pioneer of regulatory biology. JAMA (1979) 242, 1283-1284), attempted to understand the pancreas as an important organ in the development of diabetes mellitus and blood glucose control. After the seminal work of Paulescu and Banting and Best in the early 1920s that led to the discovery of insulin (Murray I. Paulesco and the isolation of insulin. J. Hist. Med. Allied Sci. (1971) 26, 150-157; Raju T.N. The Nobel Chronicles. 1923: Frederick G. Banting (1891-1941), John J.R. Macleod (1876-1935). Lancet (1998) 352, 1482), attention was turned toward understanding gastrointestinal factors that might regulate insulin secretion. A series of experiments by Jean La Barre showed that a specific fraction of intestinal extract caused a reduction in blood glucose. La Barre posited that the fraction's glucose lowering actions occurred by increasing insulin release, after which he coined the term 'incretin'. In the 1970s, the first incretin was purified, glucose insulinotropic polypeptide (GIP) (Gupta K. and Raja A. Physiology, Gastric Inhibitory Peptide StatPearls Treasure Island (FL); 2020), followed by the discovery of a second incretin in the 1980s, glucagon-like peptide-1 (GLP-1). Interest and understanding of the incretins, has grown since that time.
Collapse
|
32
|
Yasumatsu K, Ohkuri T, Yoshida R, Iwata S, Margolskee RF, Ninomiya Y. Sodium-glucose cotransporter 1 as a sugar taste sensor in mouse tongue. Acta Physiol (Oxf) 2020; 230:e13529. [PMID: 32599649 DOI: 10.1111/apha.13529] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 06/20/2020] [Accepted: 06/22/2020] [Indexed: 12/20/2022]
Abstract
AIM We investigated potential neuron types that code sugar information and how sodium-glucose cotransporters (SGLTs) and T1Rs are involved. METHODS Whole-nerve recordings in the chorda tympani (CT) and the glossopharyngeal (GL) nerves and single-fibre recordings in the CT were performed in T1R3-KO and wild-type (WT) mice. Behavioural response measurements were conducted in T1R3-KO mice using phlorizin (Phl), a competitive inhibitor of SGLTs. RESULTS Results indicated that significant enhancement occurred in responses to sucrose and glucose (Glc) by adding 10 mmol/L NaCl but not in responses to KCl, monopotassium glutamate, citric acid, quinine sulphate, SC45647(SC) or polycose in both CT and GL nerves. These enhancements were abolished by lingual application of Phl. In single-fibre recording, fibres showing maximal response to sucrose could be classified according to responses to SC and Glc with or without 10 mmol/L NaCl in the CT of WT mice, namely, Phl-insensitive type, Phl-sensitive Glc-type and Mixed (Glc and SC responding)-type fibres. In T1R3-KO mice, Phl-insensitive-type fibres disappeared. Results from behavioural experiments showed that the number of licks and amount of intake for Glc with or without 10 mmol/L NaCl were significantly suppressed by Phl. CONCLUSION We found evidence for the contribution of SGLTs in sugar sensing in taste cells of mouse tongue. Moreover, we found T1R-dependent (Phl-insensitive) type, Glc-type and Mixed (SGLTs and T1Rs)-type fibres. SGLT1 may be involved in the latter two types and may play important roles in the glucose-specific cephalic phase of digestion and palatable food intake.
Collapse
Affiliation(s)
- Keiko Yasumatsu
- Tokyo Dental Junior College Chiyoda‐ku Tokyo Japan
- Division of Sensory Physiology and Medical Application Sensing, Research and Development Centre for Five‐Sense Devices Kyushu University Fukuoka Japan
| | - Tadahiro Ohkuri
- Section of Oral Neuroscience Graduate School of Dental Sciences Kyushu University Fukuoka Japan
| | - Ryusuke Yoshida
- Section of Oral Neuroscience Graduate School of Dental Sciences Kyushu University Fukuoka Japan
- Department of Oral Physiology Graduate School of Medicine, Dentistry and Pharmaceutical Sciences Okayama University Okayama Japan
| | - Shusuke Iwata
- Division of Sensory Physiology and Medical Application Sensing, Research and Development Centre for Five‐Sense Devices Kyushu University Fukuoka Japan
- Section of Oral Neuroscience Graduate School of Dental Sciences Kyushu University Fukuoka Japan
| | | | - Yuzo Ninomiya
- Division of Sensory Physiology and Medical Application Sensing, Research and Development Centre for Five‐Sense Devices Kyushu University Fukuoka Japan
- Monell Chemical Senses Centre Philadelphia PA USA
| |
Collapse
|
33
|
Nunez-Salces M, Li H, Feinle-Bisset C, Young RL, Page AJ. Nutrient-sensing components of the mouse stomach and the gastric ghrelin cell. Neurogastroenterol Motil 2020; 32:e13944. [PMID: 32666613 DOI: 10.1111/nmo.13944] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 04/22/2020] [Accepted: 06/22/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND The ability of the gut to detect nutrients is critical to the regulation of gut hormone secretion, food intake, and postprandial blood glucose control. Ingested nutrients are detected by specific gut chemosensors. However, knowledge of these chemosensors has primarily been derived from the intestine, while available information on gastric chemosensors is limited. This study aimed to investigate the nutrient-sensing repertoire of the mouse stomach with particular emphasis on ghrelin cells. METHODS Quantitative RT-PCR was used to determine mRNA levels of nutrient chemosensors (protein: G protein-coupled receptor 93 [GPR93], calcium-sensing receptor [CaSR], metabotropic glutamate receptor type 4 [mGluR4]; fatty acids: CD36, FFAR2&4; sweet/umami taste: T1R3), taste transduction components (TRPM5, GNAT2&3), and ghrelin and ghrelin-processing enzymes (PC1/3, ghrelin O-acyltransferase [GOAT]) in the gastric corpus and antrum of adult male C57BL/6 mice. Immunohistochemistry was performed to assess protein expression of chemosensors (GPR93, T1R3, CD36, and FFAR4) and their co-localization with ghrelin. KEY RESULTS Most nutrient chemosensors had higher mRNA levels in the antrum compared to the corpus, except for CD36, GNAT2, ghrelin, and GOAT. Similar regional distribution was observed at the protein level. At least 60% of ghrelin-positive cells expressed T1R3 and FFAR4, and over 80% expressed GPR93 and CD36. CONCLUSIONS AND INFERENCES The cellular mechanisms for the detection of nutrients are expressed in a region-specific manner in the mouse stomach and gastric ghrelin cells. These gastric nutrient chemosensors may play a role modulating gastrointestinal responses, such as the inhibition of ghrelin secretion following food intake.
Collapse
Affiliation(s)
- Maria Nunez-Salces
- Vagal Afferent Research Group, Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia.,Centre of Research Excellence in Translating Nutritional Science to Good Health, Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia.,Nutrition, Diabetes & Gut Health, Lifelong Health Theme, South Australian Health & Medical Research Institute (SAHMRI), Adelaide, SA, Australia
| | - Hui Li
- Vagal Afferent Research Group, Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia.,Centre of Research Excellence in Translating Nutritional Science to Good Health, Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia.,Nutrition, Diabetes & Gut Health, Lifelong Health Theme, South Australian Health & Medical Research Institute (SAHMRI), Adelaide, SA, Australia
| | - Christine Feinle-Bisset
- Centre of Research Excellence in Translating Nutritional Science to Good Health, Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
| | - Richard L Young
- Centre of Research Excellence in Translating Nutritional Science to Good Health, Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia.,Nutrition, Diabetes & Gut Health, Lifelong Health Theme, South Australian Health & Medical Research Institute (SAHMRI), Adelaide, SA, Australia.,Intestinal Nutrient Sensing Group, Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
| | - Amanda J Page
- Vagal Afferent Research Group, Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia.,Centre of Research Excellence in Translating Nutritional Science to Good Health, Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia.,Nutrition, Diabetes & Gut Health, Lifelong Health Theme, South Australian Health & Medical Research Institute (SAHMRI), Adelaide, SA, Australia
| |
Collapse
|
34
|
An alternative pathway for sweet sensation: possible mechanisms and physiological relevance. Pflugers Arch 2020; 472:1667-1691. [PMID: 33030576 DOI: 10.1007/s00424-020-02467-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 09/14/2020] [Accepted: 09/23/2020] [Indexed: 12/12/2022]
Abstract
Sweet substances are detected by taste-bud cells upon binding to the sweet-taste receptor, a T1R2/T1R3 heterodimeric G protein-coupled receptor. In addition, experiments with mouse models lacking the sweet-taste receptor or its downstream signaling components led to the proposal of a parallel "alternative pathway" that may serve as metabolic sensor and energy regulator. Indeed, these mice showed residual nerve responses and behavioral attraction to sugars and oligosaccharides but not to artificial sweeteners. In analogy to pancreatic β cells, such alternative mechanism, to sense glucose in sweet-sensitive taste cells, might involve glucose transporters and KATP channels. Their activation may induce depolarization-dependent Ca2+ signals and release of GLP-1, which binds to its receptors on intragemmal nerve fibers. Via unknown neuronal and/or endocrine mechanisms, this pathway may contribute to both, behavioral attraction and/or induction of cephalic-phase insulin release upon oral sweet stimulation. Here, we critically review the evidence for a parallel sweet-sensitive pathway, involved signaling mechanisms, neural processing, interactions with endocrine hormonal mechanisms, and its sensitivity to different stimuli. Finally, we propose its physiological role in detecting the energy content of food and preparing for digestion.
Collapse
|
35
|
Crosson SM, Marques A, Dib P, Dotson CD, Munger SD, Zolotukhin S. Taste Receptor Cells in Mice Express Receptors for the Hormone Adiponectin. Chem Senses 2020; 44:409-422. [PMID: 31125082 DOI: 10.1093/chemse/bjz030] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The metabolic hormone adiponectin is secreted into the circulation by adipocytes and mediates key biological functions, including insulin sensitivity, adipocyte development, and fatty acid oxidation. Adiponectin is also abundant in saliva, where its functions are poorly understood. Here we report that murine taste receptor cells (TRCs) express specific adiponectin receptors and may be a target for salivary adiponectin. This is supported by the presence of all three known adiponectin receptors in transcriptomic data obtained by RNA-seq analysis of purified circumvallate (CV) taste buds. As well, immunohistochemical analysis of murine CV papillae showed that two adiponectin receptors, ADIPOR1 and T-cadherin, are localized to subsets of TRCs. Immunofluorescence for T-cadherin was primarily co-localized with the Type 2 TRC marker phospholipase C β2, suggesting that adiponectin signaling could impact sweet, bitter, or umami taste signaling. However, adiponectin null mice showed no differences in behavioral lick responsiveness compared with wild-type controls in brief-access lick testing. AAV-mediated overexpression of adiponectin in the salivary glands of adiponectin null mice did result in a small but significant increase in behavioral lick responsiveness to the fat emulsion Intralipid. Together, these results suggest that salivary adiponectin can affect TRC function, although its impact on taste responsiveness and peripheral taste coding remains unclear.
Collapse
Affiliation(s)
- Sean M Crosson
- Department of Pediatrics, Division of Cellular and Molecular Therapy, University of Florida, Gainesville, FL, USA.,Center for Smell and Taste, University of Florida, Gainesville, FL, USA.,Graduate Program in Biomedical Sciences, University of Florida, Gainesville, FL, USA
| | - Andrew Marques
- Department of Pediatrics, Division of Cellular and Molecular Therapy, University of Florida, Gainesville, FL, USA
| | - Peter Dib
- Graduate Program in Biomedical Sciences, University of Florida, Gainesville, FL, USA.,Department of Anatomy and Cell Biology, University of Florida, Gainesville, FL, USA
| | - Cedrick D Dotson
- Center for Smell and Taste, University of Florida, Gainesville, FL, USA.,Department of Neuroscience, University of Florida, Gainesville, FL, USA
| | - Steven D Munger
- Center for Smell and Taste, University of Florida, Gainesville, FL, USA.,Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, USA.,Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism; University of Florida, Gainesville, FL, USA
| | - Sergei Zolotukhin
- Department of Pediatrics, Division of Cellular and Molecular Therapy, University of Florida, Gainesville, FL, USA.,Center for Smell and Taste, University of Florida, Gainesville, FL, USA
| |
Collapse
|
36
|
Abstract
Bariatric surgery (BS) is today the most effective therapy for inducing long-term weight loss and for reducing comorbidity burden and mortality in patients with severe obesity. On the other hand, BS may be associated to new clinical problems, complications and side effects, in particular in the nutritional domain. Therefore, the nutritional management of the bariatric patients requires specific nutritional skills. In this paper, a brief overview of the nutritional management of the bariatric patients will be provided from pre-operative to post-operative phase. Patients with severe obesity often display micronutrient deficiencies when compared to normal weight controls. Therefore, nutritional status should be checked in every patient and correction of deficiencies attempted before surgery. At present, evidences from randomized and retrospective studies do not support the hypothesis that pre-operative weight loss could improve weight loss after BS surgery, and the insurance-mandated policy of a preoperative weight loss as a pre-requisite for admission to surgery is not supported by medical evidence. On the contrary, some studies suggest that a modest weight loss of 5-10% in the immediate preoperative period could facilitate surgery and reduce the risk of complications. Very low calories diet (VLCD) and very low calories ketogenic diets (VLCKD) are the most frequently used methods for the induction of a pre-operative weight loss today. After surgery, nutritional counselling is recommended in order to facilitate the adaptation of the eating habits to the new gastro-intestinal physiology. Nutritional deficits may arise according to the type of bariatric procedure and they should be prevented, diagnosed and eventually treated. Finally, specific nutritional problems, like dumping syndrome and reactive hypoglycaemia, can occur and should be managed largely by nutritional manipulation. In conclusion, the nutritional management of the bariatric patients requires specific nutritional skills and the intervention of experienced nutritionists and dieticians.
Collapse
Affiliation(s)
- Silvia Bettini
- Luca Busetto Center for the Study and the Integrated Management of Obesity, Padova University Hospital, Padova, Italy
| | - Anna Belligoli
- Luca Busetto Center for the Study and the Integrated Management of Obesity, Padova University Hospital, Padova, Italy
| | - Roberto Fabris
- Luca Busetto Center for the Study and the Integrated Management of Obesity, Padova University Hospital, Padova, Italy
| | - Luca Busetto
- Luca Busetto Center for the Study and the Integrated Management of Obesity, Padova University Hospital, Padova, Italy.
- Clinica Medica 3, Azienda Ospedaliera di Padova, Via Giustiniani 2, Padova, 35128, Italy.
| |
Collapse
|
37
|
Rohde K, Schamarek I, Blüher M. Consequences of Obesity on the Sense of Taste: Taste Buds as Treatment Targets? Diabetes Metab J 2020; 44:509-528. [PMID: 32431111 PMCID: PMC7453985 DOI: 10.4093/dmj.2020.0058] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 03/25/2020] [Indexed: 12/19/2022] Open
Abstract
Premature obesity-related mortality is caused by cardiovascular and pulmonary diseases, type 2 diabetes mellitus, physical disabilities, osteoarthritis, and certain types of cancer. Obesity is caused by a positive energy balance due to hyper-caloric nutrition, low physical activity, and energy expenditure. Overeating is partially driven by impaired homeostatic feedback of the peripheral energy status in obesity. However, food with its different qualities is a key driver for the reward driven hedonic feeding with tremendous consequences on calorie consumption. In addition to visual and olfactory cues, taste buds of the oral cavity process the earliest signals which affect the regulation of food intake, appetite and satiety. Therefore, taste buds may play a crucial role how food related signals are transmitted to the brain, particularly in priming the body for digestion during the cephalic phase. Indeed, obesity development is associated with a significant reduction in taste buds. Impaired taste bud sensitivity may play a causal role in the pathophysiology of obesity in children and adolescents. In addition, genetic variation in taste receptors has been linked to body weight regulation. This review discusses the importance of taste buds as contributing factors in the development of obesity and how obesity may affect the sense of taste, alterations in food preferences and eating behavior.
Collapse
Affiliation(s)
- Kerstin Rohde
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Center Munich at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany.
| | - Imke Schamarek
- Medical Department III (Endocrinology, Nephrology and Rheumatology), University of Leipzig, Leipzig, Germany
| | - Matthias Blüher
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Center Munich at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany.
- Medical Department III (Endocrinology, Nephrology and Rheumatology), University of Leipzig, Leipzig, Germany
| |
Collapse
|
38
|
Hyde KM, Blonde GD, Bueter M, le Roux CW, Spector AC. Gastric bypass in female rats lowers concentrated sugar solution intake and preference without affecting brief-access licking after long-term sugar exposure. Am J Physiol Regul Integr Comp Physiol 2020; 318:R870-R885. [PMID: 32083966 DOI: 10.1152/ajpregu.00240.2019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
In rodents, Roux-en-Y gastric bypass (RYGB) decreases intake of, and preference for, foods or fluids that are high in sugar. Whether these surgically induced changes are due to decreases in the palatability of sugar stimuli is controversial. We used RYGB and sham-operated (SHAM) female rats to test the influence of prolonged ingestive experience with sugar solutions on the motivational potency of these stimuli to drive licking in brief-access (BA) tests. In experiment 1, RYGB attenuated intake of, and caloric preference for, 0.3 M sucrose during five consecutive, 46-h two-bottle tests (TBTs; sucrose). A second series of TBTs (5 consecutive, 46-h tests) with 1.0 M sucrose revealed similar results, except fluid preference for 1.0 M sucrose also significantly decreased. Before, between, and after the two series of TBTs, two sessions of BA tests (30 min; 10-s trials) with an array of sucrose concentrations (0 and 0.01-1.0 M) were conducted. Concentration-dependent licking and overall trial initiation did not differ between surgical groups in any test. In a similar experimental design in a second cohort of female rats, 0.6 and 2.0 M glucose (isocaloric with sucrose concentrations in experiment 1) were used in the TBTs; 0 and 0.06-2.0 M glucose were used in the BA tests. Outcomes were similar to those for experiment 1, except RYGB rats initiated fewer trials during the BA tests. Although RYGB profoundly affected intake of, and caloric preference for, sugar solutions and, with high concentrations, fluid preference, RYGB never influenced the motivational potency of sucrose or glucose to drive concentration-dependent licking in BA tests.
Collapse
Affiliation(s)
- Kellie M Hyde
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, Florida
| | - Ginger D Blonde
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, Florida
| | - Marco Bueter
- Division of Visceral and Transplantation Surgery, Department of Surgery, University of Zürich, Zürich, Switzerland
| | - Carel W le Roux
- Diabetes Complications Research Centre, Conway Institute, School of Medicine, University College Dublin, Dublin, Ireland
| | - Alan C Spector
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, Florida
| |
Collapse
|
39
|
Sensing Senses: Optical Biosensors to Study Gustation. SENSORS 2020; 20:s20071811. [PMID: 32218129 PMCID: PMC7180777 DOI: 10.3390/s20071811] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 03/19/2020] [Accepted: 03/21/2020] [Indexed: 12/11/2022]
Abstract
The five basic taste modalities, sweet, bitter, umami, salty and sour induce changes of Ca2+ levels, pH and/or membrane potential in taste cells of the tongue and/or in neurons that convey and decode gustatory signals to the brain. Optical biosensors, which can be either synthetic dyes or genetically encoded proteins whose fluorescence spectra depend on levels of Ca2+, pH or membrane potential, have been used in primary cells/tissues or in recombinant systems to study taste-related intra- and intercellular signaling mechanisms or to discover new ligands. Taste-evoked responses were measured by microscopy achieving high spatial and temporal resolution, while plate readers were employed for higher throughput screening. Here, these approaches making use of fluorescent optical biosensors to investigate specific taste-related questions or to screen new agonists/antagonists for the different taste modalities were reviewed systematically. Furthermore, in the context of recent developments in genetically encoded sensors, 3D cultures and imaging technologies, we propose new feasible approaches for studying taste physiology and for compound screening.
Collapse
|
40
|
Hankir MK, Al-Bas S, Rullmann M, Chakaroun R, Seyfried F, Pleger B. Homeostatic, reward and executive brain functions after gastric bypass surgery. Appetite 2020; 146:104419. [DOI: 10.1016/j.appet.2019.104419] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 07/01/2019] [Accepted: 08/23/2019] [Indexed: 12/15/2022]
|
41
|
Abstract
In the last few years, single-cell profiling of taste cells and ganglion cells has advanced our understanding of transduction, encoding, and transmission of information from taste buds as relayed to the central nervous system. This review focuses on new knowledge from these molecular approaches and attempts to place this in the context of previous questions and findings in the field. The individual taste cells within a taste bud are molecularly specialized for detection of one of the primary taste qualities: salt, sour, sweet, umami, and bitter. Transduction and transmitter release mechanisms differ substantially for taste cells transducing sour (Type III cells) compared with those transducing the qualities of sweet, umami, or bitter (Type II cells), although ultimately all transmission of taste relies on activation of purinergic P2X receptors on the afferent nerves. The ganglion cells providing innervation to the taste buds also appear divisible into functional and molecular subtypes, and each ganglion cell is primarily but not exclusively responsive to one taste quality.
Collapse
Affiliation(s)
- Sue C. Kinnamon
- Rocky Mountain Taste & Smell Center, Department of Otolaryngology and Department of Cell & Developmental Biology, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Thomas E. Finger
- Rocky Mountain Taste & Smell Center, Department of Otolaryngology and Department of Cell & Developmental Biology, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| |
Collapse
|
42
|
Abstract
The regulation of glycemia is under a tight neuronal detection of glucose levels performed by the gut-brain axis and an efficient efferent neuronal message sent to the peripheral organs, as the pancreas to induce insulin and inhibit glucagon secretions. The neuronal detection of glucose levels is performed by the autonomic nervous system including the enteric nervous system and the vagus nerve innervating the gastro-intestinal tractus, from the mouth to the anus. A dysregulation of this detection leads to the one of the most important current health issue around the world i.e. diabetes mellitus. Furthemore, the consequences of diabetes mellitus on neuronal homeostasis and activities participate to the aggravation of the disease establishing a viscious circle. Prokaryotic cells as bacteria, reside in our gut. The strong relationship between prokaryotic cells and our eukaryotic cells has been established long ago, and prokaryotic and eukaryotic cells in our body have evolved synbiotically. For the last decades, studies demonstrated the critical role of the gut microbiota on the metabolic control and how its shift can induce diseases such as diabetes. Despite an important increase of knowledge, few is known about 1) how the gut microbiota influences the neuronal detection of glucose and 2) how the diabetes mellitus-induced gut microbiota shift observed participates to the alterations of autonomic nervous system and the gut-brain axis activity.
Collapse
Affiliation(s)
- Estelle Grasset
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, University of Gothenburg, 41345, Gothenburg, Sweden.
| | - Remy Burcelin
- Institut National de la Santé et de la Recherche Médicale (INSERM), Toulouse, France
- Unité Mixte de Recherche (UMR) 1048, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Team 2 : 'Intestinal Risk Factors, Diabetes, Université Paul Sabatier (UPS), Dyslipidemia', F-31432, Toulouse, Cedex 4, France
| |
Collapse
|
43
|
Brindisi MC, Brondel L, Meillon S, Barthet S, Grall S, Fenech C, Liénard F, Schlich P, Astruc K, Mouillot T, Jacquin-Piques A, Leloup C, Vergès B, Pénicaud L. Proof of concept: Effect of GLP-1 agonist on food hedonic responses and taste sensitivity in poor controlled type 2 diabetic patients. Diabetes Metab Syndr 2019; 13:2489-2494. [PMID: 31405666 DOI: 10.1016/j.dsx.2019.06.021] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 06/27/2019] [Indexed: 12/14/2022]
Abstract
AIMS GLP-1 analogues decrease food intake and have great promise for the fight against obesity. Little is known about their effects on food hedonic sensations and taste perception in poor controlled patients with type 2 diabetes (T2D). MATERIALS AND METHODS Eighteen T2D patients with BMI ≥25 kg/m2 and poor controlled glycemia were studied before and after 3 months of treatment with Liraglutide. Detection thresholds for salty, sweet and bitter tastes, optimal preferences, olfactory liking, wanting and recalled liking for several food items were assessed. Subjects also answered questionnaires to measure their attitudes to food. RESULTS T2D patients had a significant decrease in bodyweight and HbA1c after treatment with Liraglutide. Liraglutide improved gustative detection threshold of sweet flavors, and decreased wanting for sweet foods and recalled liking for fatty foods. It also led to a decrease in feelings of hunger. CONCLUSIONS Liraglutide increases sensitivity to sweet tastes and decreases pleasure responses for fatty foods in poor controlled T2D patients, and is of particular interest in the understanding of the mechanisms of weight loss. CLINICAL TRIAL NCT02674893.
Collapse
Affiliation(s)
- Marie-Claude Brindisi
- CNRS UMR 6265 Centre des Sciences du Goût et de l'Alimentation, F-21000, Dijon, France; INRA UMR 1324 Centre des Sciences du Goût et de l'Alimentation, F-21000, Dijon, France; Diabetes and Endocrinology Unit, Dijon University Hospital, F-21000, France.
| | - Laurent Brondel
- CNRS UMR 6265 Centre des Sciences du Goût et de l'Alimentation, F-21000, Dijon, France; INRA UMR 1324 Centre des Sciences du Goût et de l'Alimentation, F-21000, Dijon, France
| | - Sophie Meillon
- CNRS UMR 6265 Centre des Sciences du Goût et de l'Alimentation, F-21000, Dijon, France; INRA UMR 1324 Centre des Sciences du Goût et de l'Alimentation, F-21000, Dijon, France
| | - Sophie Barthet
- CNRS UMR 6265 Centre des Sciences du Goût et de l'Alimentation, F-21000, Dijon, France; INRA UMR 1324 Centre des Sciences du Goût et de l'Alimentation, F-21000, Dijon, France
| | - Sylvie Grall
- CNRS UMR 6265 Centre des Sciences du Goût et de l'Alimentation, F-21000, Dijon, France; INRA UMR 1324 Centre des Sciences du Goût et de l'Alimentation, F-21000, Dijon, France
| | - Claire Fenech
- CNRS UMR 6265 Centre des Sciences du Goût et de l'Alimentation, F-21000, Dijon, France; INRA UMR 1324 Centre des Sciences du Goût et de l'Alimentation, F-21000, Dijon, France
| | - Fabienne Liénard
- CNRS UMR 6265 Centre des Sciences du Goût et de l'Alimentation, F-21000, Dijon, France; INRA UMR 1324 Centre des Sciences du Goût et de l'Alimentation, F-21000, Dijon, France
| | - Pascal Schlich
- CNRS UMR 6265 Centre des Sciences du Goût et de l'Alimentation, F-21000, Dijon, France; INRA UMR 1324 Centre des Sciences du Goût et de l'Alimentation, F-21000, Dijon, France
| | | | - Thomas Mouillot
- CNRS UMR 6265 Centre des Sciences du Goût et de l'Alimentation, F-21000, Dijon, France; Gastroenterology Department, Dijon University Hospital, F-21000, France
| | - Agnès Jacquin-Piques
- CNRS UMR 6265 Centre des Sciences du Goût et de l'Alimentation, F-21000, Dijon, France; Neurology Department, Dijon University Hospital, F-21000, France
| | - Corinne Leloup
- CNRS UMR 6265 Centre des Sciences du Goût et de l'Alimentation, F-21000, Dijon, France; INRA UMR 1324 Centre des Sciences du Goût et de l'Alimentation, F-21000, Dijon, France
| | - Bruno Vergès
- Diabetes and Endocrinology Unit, Dijon University Hospital, F-21000, France
| | - Luc Pénicaud
- CNRS UMR 6265 Centre des Sciences du Goût et de l'Alimentation, F-21000, Dijon, France; INRA UMR 1324 Centre des Sciences du Goût et de l'Alimentation, F-21000, Dijon, France; Stromalab ERL 5311 CNRS, F-31432, Toulouse, France
| |
Collapse
|
44
|
A Preventive Prebiotic Supplementation Improves the Sweet Taste Perception in Diet-Induced Obese Mice. Nutrients 2019; 11:nu11030549. [PMID: 30841548 PMCID: PMC6471995 DOI: 10.3390/nu11030549] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 02/27/2019] [Accepted: 02/27/2019] [Indexed: 12/23/2022] Open
Abstract
Orosensory perception of sweet stimulus is blunted in diet-induced obese (DIO) rodents. Although this alteration might contribute to unhealthy food choices, its origin remains to be understood. Cumulative evidence indicates that prebiotic manipulations of the gut microbiota are associated with changes in food intake by modulating hedonic and motivational drive for food reward. In the present study, we explore whether a prebiotic supplementation can also restore the taste sensation in DIO mice. The preference and licking behavior in response to various sucrose concentrations were determined using respectively two-bottle choice tests and gustometer analysis in lean and obese mice supplemented or not with 10% inulin-type fructans prebiotic (P) in a preventive manner. In DIO mice, P addition reduced the fat mass gain and energy intake, limited the gut dysbiosis and partially improved the sweet taste perception (rise both of sucrose preference and number of licks/10 s vs. non-supplemented DIO mice). No clear effect on orosensory perception of sucrose was found in the supplemented control mice. Therefore, a preventive P supplementation can partially correct the loss of sweet taste sensitivity found in DIO mice, with the efficiency of treatment being dependent from the nutritional status of mice (high fat diet vs. regular chow).
Collapse
|
45
|
Identification and comparison of oligopeptides during withering process of White tea by ultra-high pressure liquid chromatography coupled with quadrupole-orbitrap ultra-high resolution mass spectrometry. Food Res Int 2019; 121:825-834. [PMID: 31108814 DOI: 10.1016/j.foodres.2019.01.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 12/28/2018] [Accepted: 01/03/2019] [Indexed: 11/21/2022]
Abstract
Peptides could have specific tastes or bioactivities depending on the length and sequence of amino acids. Till date it remains unknown what peptides are formed during the white tea manufacturing process and whether they contribute to the flavor or bio-activities of white tea. As a first step to address these questions, we applied ultra-high pressure liquid chromatography coupled with quadrupole-orbitrap ultra-high resolution mass spectrometry (UPLC-Quadrupole-Orbitrap-UHRMS) to monitor peptides dynamic changes during the withering process. A total of 196 abundant peptides were identified. Most of them were oligopeptides within a molecular weight of 1000 Da. Four of them were randomly selected, synthesized peptides were applied for further confirmation and quantification. Sequence analysis suggested that some of them were potential taste contributors. Proteinase cleave site analysis identified two separate periods of active proteins degradation at 0-12 h and 30-42 h of the withering processes. Further analysis of cleavage sites also suggested that protein degradation during withering steps were random rather than a stepwise reaction.
Collapse
|
46
|
Sakai C, Abe S, Kouzuki M, Shimohiro H, Ota Y, Sakinada H, Takeuchi T, Okura T, Kasagi T, Hanaki K. A Randomized Placebo-controlled Trial of an Oral Preparation of High Molecular Weight Fucoidan in Patients with Type 2 Diabetes with Evaluation of Taste Sensitivity. Yonago Acta Med 2019. [DOI: 10.33160/yam.2019.03.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Chieko Sakai
- *Department of Adult and Elderly Nursing, School of Health Sciences, Tottori University Faculty of Medicine, Yonago 683-8503, Japan
| | - Sunao Abe
- †Marine Products Kimuraya Co., Ltd., Sakaiminato 684-0072, Japan
| | - Minoru Kouzuki
- ‡Department of Biological Regulation, School of Health Sciences, Tottori University Faculty of Medicine, Yonago 683-8503, Japan
| | - Hisashi Shimohiro
- §Department of Pathobiological Science and Technology, School of Health Sciences, Tottori University Faculty of Medicine, Yonago 683-8503, Japan
| | - Yoshie Ota
- Clinical Laboratory Department, Hakuai Hospital, Yonago 683-0853, Japan
| | - Hironori Sakinada
- Clinical Laboratory Department, Hakuai Hospital, Yonago 683-0853, Japan
| | - Tatsuo Takeuchi
- ¶Department of Endocrinology and Metabolism, Hakuai Hospital, Yonago 683-0853, Japan
| | - Tsuyoshi Okura
- **Division of Molecular Medicine and Therapeutics, Tottori University Faculty of Medicine, Yonago 683-8503, Japan
| | - Takeshi Kasagi
- †Marine Products Kimuraya Co., Ltd., Sakaiminato 684-0072, Japan
| | - Keiichi Hanaki
- ††Department of Women’s and Children’s Family Nursing, School of Health Sciences, Tottori University Faculty of Medicine, Yonago 683-8503, Japan
| |
Collapse
|
47
|
Al-Najim W, Docherty NG, le Roux CW. Food Intake and Eating Behavior After Bariatric Surgery. Physiol Rev 2018; 98:1113-1141. [PMID: 29717927 DOI: 10.1152/physrev.00021.2017] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Obesity is an escalating global chronic disease. Bariatric surgery is a very efficacious treatment for obesity and its comorbidities. Alterations to gastrointestinal anatomy during bariatric surgery result in neurological and physiological changes affecting hypothalamic signaling, gut hormones, bile acids, and gut microbiota, which coalesce to exert a profound influence on eating behavior. A thorough understanding of the mechanisms underlying eating behavior is essential in the management of patients after bariatric surgery. Studies investigating candidate mechanisms have expanded dramatically in the last decade. Herein we review the proposed mechanisms governing changes in eating behavior, food intake, and body weight after bariatric surgery. Additive or synergistic effects of both conditioned and unconditioned factors likely account for the complete picture of changes in eating behavior. Considered application of strategies designed to support the underlying principles governing changes in eating behavior holds promise as a means of optimizing responses to surgery and long-term outcomes.
Collapse
Affiliation(s)
- Werd Al-Najim
- Diabetes Complications Research Centre, Conway Institute, School of Medicine and Medical Sciences, University College Dublin , Dublin , Ireland ; Department of Gastrosurgical Research and Education, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg , Gothenburg , Sweden ; and Investigative Science, Imperial College London , London , United Kingdom
| | - Neil G Docherty
- Diabetes Complications Research Centre, Conway Institute, School of Medicine and Medical Sciences, University College Dublin , Dublin , Ireland ; Department of Gastrosurgical Research and Education, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg , Gothenburg , Sweden ; and Investigative Science, Imperial College London , London , United Kingdom
| | - Carel W le Roux
- Diabetes Complications Research Centre, Conway Institute, School of Medicine and Medical Sciences, University College Dublin , Dublin , Ireland ; Department of Gastrosurgical Research and Education, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg , Gothenburg , Sweden ; and Investigative Science, Imperial College London , London , United Kingdom
| |
Collapse
|
48
|
Orosensory Detection of Dietary Fatty Acids Is Altered in CB₁R -/- Mice. Nutrients 2018; 10:nu10101347. [PMID: 30241419 PMCID: PMC6213063 DOI: 10.3390/nu10101347] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Revised: 09/14/2018] [Accepted: 09/18/2018] [Indexed: 12/25/2022] Open
Abstract
Obesity is one of the major public health issues, and its prevalence is steadily increasing all the world over. The endocannabinoid system (ECS) has been shown to be involved in the intake of palatable food via activation of cannabinoid 1 receptor (CB1R). However, the involvement of lingual CB1R in the orosensory perception of dietary fatty acids has never been investigated. In the present study, behavioral tests on CB1R−/− and wild type (WT) mice showed that the invalidation of Cb1r gene was associated with low preference for solutions containing rapeseed oil or a long-chain fatty acid (LCFA), such as linoleic acid (LA). Administration of rimonabant, a CB1R inverse agonist, in mice also brought about a low preference for dietary fat. No difference in CD36 and GPR120 protein expressions were observed in taste bud cells (TBC) from WT and CB1R−/− mice. However, LCFA induced a higher increase in [Ca2+]i in TBC from WT mice than that in TBC from CB1R−/− mice. TBC from CB1R−/− mice also exhibited decreased Proglucagon and Glp-1r mRNA and a low GLP-1 basal level. We report that CB1R is involved in fat taste perception via calcium signaling and GLP-1 secretion.
Collapse
|
49
|
Taste of glucose elicits cephalic-phase insulin release in mice. Physiol Behav 2018; 192:200-205. [DOI: 10.1016/j.physbeh.2018.04.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 03/31/2018] [Accepted: 04/01/2018] [Indexed: 01/08/2023]
|
50
|
O'Brien P, Hewett R, Corpe C. Sugar sensor genes in the murine gastrointestinal tract display a cephalocaudal axis of expression and a diurnal rhythm. Physiol Genomics 2018; 50:448-458. [PMID: 29625018 DOI: 10.1152/physiolgenomics.00139.2017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Distributed along the length of the gastrointestinal (GI) tract are nutrient sensing cells that release numerous signaling peptides influencing GI function, nutrient homeostasis and energy balance. Recent studies have shown a diurnal rhythm in GI nutrient sensing, but the mechanisms responsible for rhythmicity are poorly understood. In this report we studied murine GI sugar sensor gene and protein expression levels in the morning (7 AM) and evening (7 PM). Sweet taste receptor ( tas1r2/tas1r3/gnat3/gnat1) sugar transporter ( slc5a1, slc2a2, slc2a5) and putative sugar sensor ( slc5a4a and slc5a4b) gene expression levels were highest in tongue and proximal and distal small intestine, respectively. Clock gene ( cry2/arntl) activity was detected in all regions studied. Slc5a4a and slc5a4b gene expression showed clear diurnal rhythmicity in the small intestine and stomach, respectively, although no rhythmicity was detected in SGLT3 protein expression. Tas1r2, tas1r3, gnat1, and gcg displayed a limited rhythm in gene expression in proximal small intestine. Microarray analysis revealed a diurnal rhythm in gut peptide gene expression in tongue (7 AM vs. 7 PM) and in silico promoter analysis indicated intestinal sugar sensors and transporters possessed the canonical E box elements necessary for clock gene control over gene transcription. In this report we present evidence of a diurnal rhythm in genes that are responsible for intestinal nutrient sensing that is most likely controlled by clock gene activity. Disturbances in clock gene/nutrient sensing interactions may be important in the development of diet-related diseases, such as obesity and diabetes.
Collapse
Affiliation(s)
- Patrick O'Brien
- Department of Nutritional Sciences, School of Medicine, King's College London , London , United Kingdom
| | - Rhys Hewett
- Department of Nutritional Sciences, School of Medicine, King's College London , London , United Kingdom
| | - Christopher Corpe
- Department of Nutritional Sciences, School of Medicine, King's College London , London , United Kingdom
| |
Collapse
|