1
|
Cordero P, Ramírez-Toloza G, Dufflocq P, Herrera-Alcaíno S, Guzmán-Pino SA. Reduced Dietary Protein and Essential Amino Acids Impair Growth Performance and Increase Lysine Sensitivity in Broiler Chickens. Animals (Basel) 2025; 15:1027. [PMID: 40218420 PMCID: PMC11988050 DOI: 10.3390/ani15071027] [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: 03/09/2025] [Revised: 03/30/2025] [Accepted: 03/31/2025] [Indexed: 04/14/2025] Open
Abstract
Nutrition plays a fundamental role in poultry production. Developing high-quality diets that ensure positive feed perception by birds is a key strategy for enhancing production performance within the industry. This study evaluated the effect of crude protein (CP) and four essential amino acid (AA) reductions on broiler chickens' growth performance and taste sensitivity for Lysine, Methionine, Threonine, and Tryptophan. Sixty-four one-day-old male broiler chickens (Ross 308) were exposed to four dietary treatments consisting of a balanced (control) diet with a total inclusion of CP and Lysine, Methionine, Threonine, and Tryptophan (T1); diets with a 30 g/kg reduction in CP maintaining 100% exogenous incorporation of the four aforementioned AAs (T2); diets with a 30 g/kg reduction in CP and 50% exogenous incorporation of the same four AAs (T3); and diets with a 30 g/kg reduction in CP with no exogenous incorporation of the four previously mentioned AAs (T4). After 7 days of acclimatization to the diets, the birds were offered Lysine, Methionine, Threonine, and Tryptophan at eight concentrations (0.1, 0.5, 1.0, 1.5, 2.0, 2.5, 3.0, and 3.5%) each for 4 h/day in a double-choice test for 32 days to determine the preferences and sensory-motivated intake (SMI) of the birds. During the 39-day trial, the birds in the T4 group showed a lower (p < 0.050) average daily feed intake (ADFI), average daily gain (ADG), feed conversion ratio (FCR), and body weight (BW) than the birds of the rest of the treatments. No significant effects of the different dietary treatments, CP content, or AA levels were observed on the preferences or SMI of the birds (p > 0.050). However, differences were found in the preferences and SMI between birds from the different treatments when evaluating the treatment × AA × concentration interaction (p < 0.050), reflecting that dietary treatments generate changes in the consumption behavior of birds in response to the delivery of AAs at different concentrations. The preference and SMI thresholds for Lysine decreased in the birds in the T4 group, while the thresholds for Methionine and Tryptophan increased compared to the birds in the control group (p < 0.050). We concluded that the reduction in CP and AAs in the diet of broiler chickens affects their productive parameters and increases their taste sensitivity, which was reflected in lower weight gain and feed conversion efficiency, along with increased preferences and higher SMI values in the birds exposed to diets with more significant nutritional deficits.
Collapse
Affiliation(s)
- Paloma Cordero
- Programa de Doctorado en Ciencias Silvoagropecuarias y Veterinarias, Campus Sur, Universidad de Chile, Santiago 8820808, Chile; (P.C.); (S.H.-A.)
- Departamento de Fomento de la Producción Animal, Facultad de Ciencias Veterinarias y Pecuarias, Universidad de Chile, Santiago 8820808, Chile
| | - Galia Ramírez-Toloza
- Departamento de Medicina Preventiva Animal, Facultad de Ciencias Veterinarias y Pecuarias, Universidad de Chile, Santiago 8820808, Chile;
| | - Pablo Dufflocq
- Food Quality Research Center, Universidad de Chile, Santiago 882080, Chile;
| | - Sofía Herrera-Alcaíno
- Programa de Doctorado en Ciencias Silvoagropecuarias y Veterinarias, Campus Sur, Universidad de Chile, Santiago 8820808, Chile; (P.C.); (S.H.-A.)
- Departamento de Fomento de la Producción Animal, Facultad de Ciencias Veterinarias y Pecuarias, Universidad de Chile, Santiago 8820808, Chile
| | - Sergio A. Guzmán-Pino
- Departamento de Fomento de la Producción Animal, Facultad de Ciencias Veterinarias y Pecuarias, Universidad de Chile, Santiago 8820808, Chile
| |
Collapse
|
2
|
Muthuswamy K, Vasanthakumar K, Panneerselvan P, Thangamani L, Krishnan V, Piramanayagam S, Subramaniam S. FAHFA promotes intracellular calcium signaling via activating the fat taste receptor, CD36 and Src protein kinases in mice taste bud cells. Biochim Biophys Acta Gen Subj 2024; 1868:130722. [PMID: 39426759 DOI: 10.1016/j.bbagen.2024.130722] [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: 04/23/2024] [Revised: 08/30/2024] [Accepted: 10/08/2024] [Indexed: 10/21/2024]
Abstract
Two lipid sensors, CD36 and GPR120, are crucial for the orosensory detection of fat taste and for mediating fat preference. However, the mechanism by which endogenous lipid (FAHFA) binds to CD36 to initiate intracellular signaling remains unexplained. Hence, the primary objective of this study is to investigate the binding mechanism of FAHFA to CD36 and its role in isolated mouse taste bud cells (mTBCs). The Schrodinger platform was used to assess the molecular dynamics of protein and ligand interactions, and an in vitro experiment was used to validate the findings. Based on the docking score of the ligand, the molecular mechanistic activities of the targeted complexes, CD36-5-POHSA (-8.2 kcal/mol), were investigated using the dynamic simulation. In comparison to linoleic acid (LA), POHSA rapidly increased [Ca2+]i via acting on CD36, and 5-POHSA treatment in mTBCs activated src-kinase at 20 μM. CD36 siRNA transfection in TBCs downregulate the CD36 protein expression as well as [Ca2+]i flux. This study suggests that 5-POHSA may help combat taste abnormalities and the adverse effects of obesity by binding to the lingual CD36 receptor and activating the tongue-brain axis.
Collapse
Affiliation(s)
- Karthi Muthuswamy
- Molecular Physiology Laboratory, Department of Biochemistry, Bharathiar University, Coimbatore, Tamilnadu 641046, India; Men's Health Research Unit, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Keerthana Vasanthakumar
- Molecular Physiology Laboratory, Department of Biochemistry, Bharathiar University, Coimbatore, Tamilnadu 641046, India
| | - Prabha Panneerselvan
- Molecular Physiology Laboratory, Department of Biochemistry, Bharathiar University, Coimbatore, Tamilnadu 641046, India
| | - Lokesh Thangamani
- Computational Biology Lab, Department of Bioinformatics, Bharathiar University, Coimbatore, Tamilnadu 641046, India
| | - Vasanth Krishnan
- Molecular Biology Laboratory, Department of Botany, Bharathiar University, Coimbatore, Tamilnadu 641046, India
| | - Shanmughavel Piramanayagam
- Computational Biology Lab, Department of Bioinformatics, Bharathiar University, Coimbatore, Tamilnadu 641046, India
| | - Selvakumar Subramaniam
- Molecular Physiology Laboratory, Department of Biochemistry, Bharathiar University, Coimbatore, Tamilnadu 641046, India.
| |
Collapse
|
3
|
Primeaux SD, Dubin R, Greenway FL. Orlistat mouth rinse: Using the tongue to deliver antiobesity medication in a double-blind randomized crossover pilot trial. Diabetes Obes Metab 2023; 25:2236-2242. [PMID: 37132340 PMCID: PMC10330229 DOI: 10.1111/dom.15101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 04/11/2023] [Accepted: 04/15/2023] [Indexed: 05/04/2023]
Abstract
AIM To investigate the effects of an orlistat mouth rinse on the intake of a high-fat meal. METHODS A double-blind, balanced order, crossover study was conducted in participants (n = 10, body mass index 25-30 kg/m2 ) assigned to receive placebo or orlistat (24 mg/mL) prior to a high-fat meal. Participants were divided into low- or high-fat consumers based on calories consumed from fat following placebo administration. RESULTS The orlistat mouth rinse decreased total and fat calories consumed during the high-fat meal in high-fat consumers, and did not alter calories consumed in low-fat consumers (P < 0.05). CONCLUSIONS Orlistat decreases long-chain fatty acid (LCFA) absorption by inhibiting lipases that breakdown triglycerides. Orlistat mouth rinse decreased fat intake in high-fat consumers, suggesting that orlistat inhibited the detection of LCFAs from the high-fat test meal. Lingual delivery of orlistat is predicted to eliminate the risk of oil incontinence and promote weight loss in individuals who prefer fat.
Collapse
Affiliation(s)
- Stefany D Primeaux
- Department of Physiology, LSU Health-NO, New Orleans, Louisiana, USA
- Joint Diabetes, Endocrinology & Metabolism Program, Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| | - Robert Dubin
- Florida State University College of Medicine, Venice, Florida, USA
| | - Frank L Greenway
- Clinical Trials Department, Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| |
Collapse
|
4
|
Hichami A, Saidi H, Khan AS, Degbeni P, Khan NA. In Vitro Functional Characterization of Type-I Taste Bud Cells as Monocytes/Macrophages-like Which Secrete Proinflammatory Cytokines. Int J Mol Sci 2023; 24:10325. [PMID: 37373472 DOI: 10.3390/ijms241210325] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 06/14/2023] [Accepted: 06/17/2023] [Indexed: 06/29/2023] Open
Abstract
The sense of taste determines the choice of nutrients and food intake and, consequently, influences feeding behaviors. The taste papillae are primarily composed of three types of taste bud cells (TBC), i.e., type I, type II, and type III. The type I TBC, expressing GLAST (glutamate--aspartate transporter), have been termed as glial-like cells. We hypothesized that these cells could play a role in taste bud immunity as glial cells do in the brain. We purified type I TBC, expressing F4/80, a specific marker of macrophages, from mouse fungiform taste papillae. The purified cells also express CD11b, CD11c, and CD64, generally expressed by glial cells and macrophages. We further assessed whether mouse type I TBC can be polarized toward M1 or M2 macrophages in inflammatory states like lipopolysaccharide (LPS)-triggered inflammation or obesity, known to be associated with low-grade inflammation. Indeed, LPS-treatment and obesity state increased TNFα, IL-1β, and IL-6 expression, both at mRNA and protein levels, in type I TBC. Conversely, purified type I TBC treated with IL-4 showed a significant increase in arginase 1 and IL-4. These findings provide evidence that type I gustatory cells share many features with macrophages and may be involved in oral inflammation.
Collapse
Affiliation(s)
- Aziz Hichami
- Physiologie de la Nutrition & Toxicologie, UMR INSERM U1231 Lipide, Nutrition & Cancer, Université de Bourgogne, 21000 Dijon, France
| | - Hamza Saidi
- Physiologie de la Nutrition & Toxicologie, UMR INSERM U1231 Lipide, Nutrition & Cancer, Université de Bourgogne, 21000 Dijon, France
- Bioenergetics and Intermediary Metabolism Team, Laboratory of Biology and Organisms Physiology, University of Sciences and Technology Houari Boumediene, Algiers 16111, Algeria
| | - Amira Sayed Khan
- Physiologie de la Nutrition & Toxicologie, UMR INSERM U1231 Lipide, Nutrition & Cancer, Université de Bourgogne, 21000 Dijon, France
| | - Pernelle Degbeni
- Physiologie de la Nutrition & Toxicologie, UMR INSERM U1231 Lipide, Nutrition & Cancer, Université de Bourgogne, 21000 Dijon, France
| | - Naim Akhtar Khan
- Physiologie de la Nutrition & Toxicologie, UMR INSERM U1231 Lipide, Nutrition & Cancer, Université de Bourgogne, 21000 Dijon, France
| |
Collapse
|
5
|
Lin F, Liu Y, Rudeski-Rohr T, Dahir N, Calder A, Gilbertson TA. Adiponectin Enhances Fatty Acid Signaling in Human Taste Cells by Increasing Surface Expression of CD36. Int J Mol Sci 2023; 24:ijms24065801. [PMID: 36982874 PMCID: PMC10059208 DOI: 10.3390/ijms24065801] [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: 02/24/2023] [Revised: 03/10/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023] Open
Abstract
Adiponectin, a key metabolic hormone, is secreted into the circulation by fat cells where it enhances insulin sensitivity and stimulates glucose and fatty acid metabolism. Adiponectin receptors are highly expressed in the taste system; however, their effects and mechanisms of action in the modulation of gustatory function remain unclear. We utilized an immortalized human fungiform taste cell line (HuFF) to investigate the effect of AdipoRon, an adiponectin receptor agonist, on fatty acid-induced calcium responses. We showed that the fat taste receptors (CD36 and GPR120) and taste signaling molecules (Gα-gust, PLCβ2, and TRPM5) were expressed in HuFF cells. Calcium imaging studies showed that linoleic acid induced a dose-dependent calcium response in HuFF cells, and it was significantly reduced by the antagonists of CD36, GPR120, PLCβ2, and TRPM5. AdipoRon administration enhanced HuFF cell responses to fatty acids but not to a mixture of sweet, bitter, and umami tastants. This enhancement was inhibited by an irreversible CD36 antagonist and by an AMPK inhibitor but was not affected by a GPR120 antagonist. AdipoRon increased the phosphorylation of AMPK and the translocation of CD36 to the cell surface, which was eliminated by blocking AMPK. These results indicate that AdipoRon acts to increase cell surface CD36 in HuFF cells to selectively enhance their responses to fatty acids. This, in turn, is consistent with the ability of adiponectin receptor activity to alter taste cues associated with dietary fat intake.
Collapse
Affiliation(s)
- Fangjun Lin
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Yan Liu
- Department of Internal Medicine, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Trina Rudeski-Rohr
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Naima Dahir
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Ashley Calder
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Timothy A Gilbertson
- Department of Internal Medicine, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| |
Collapse
|
6
|
Variability of oral/taste sensitivity to fat: An investigation of attribution from detection threshold methods with repeated measurements. Food Res Int 2023; 165:112432. [PMID: 36869468 DOI: 10.1016/j.foodres.2022.112432] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 12/27/2022] [Accepted: 12/29/2022] [Indexed: 01/09/2023]
Abstract
Accumulating psychophysical evidence suggests substantial individual variability in oral/taste sensitivity to non-esterified, long-chain fatty acids (NEFA), which is commonly referred to as fat taste or oleogustus. Recent studies have sought to determine its associations with human factors such as body mass index (BMI) and food preferences, as it has been claimed that excessive fat consumption is related to several health conditions, including obesity. Yet, the findings are controversial. On the other hand, it has been noted that considerable variability also occurs based on the methodology used to measure the fatty acid taste. Specifically, learning effects have been observed over repeated measurements of the detection threshold of NEFA, yet there has been no methodology available to take into account these learning effects. Accordingly, in the present study, a novel methodology using a descending-block dual reminder A-Not A (DR A-Not A) method with a warm-up has been proposed to measure the NEFA detection threshold based on the signal detection theory and considering NEFA taste learning effects over repeated sessions. Homogeneous subjects (young adult Korean females within the normal BMI range, non-vegetarians) were randomized to either the novel descending-block DR A-Not A method or ascending triangle method that is commonly used for fat perception studies. Pure oleic acid emulsions were used as fat taste stimuli to be discriminated from pure mineral water. Each subject completed 14 repeated visits. For the ascending triangle method, 14 thresholds were determined using a stopping rule, while for the novel method, 7 thresholds were determined each per two consecutive days, using a criterion of a lower limit of 50% confidence interval of d' = 0.5, considering the practical aspects of taste studies in food sensory science. Based on the group median results of the last two visits, the variability of the detection thresholds was reduced using the novel descending-block DR A-Not A method due to better learning effects over repeated sessions. This shows the potential of the descending-block DR A-Not A threshold method for further studies on oral/taste sensitivity to fat.
Collapse
|
7
|
Aukan MI, Coutinho S, Pedersen SA, Simpson MR, Martins C. Differences in gastrointestinal hormones and appetite ratings between individuals with and without obesity-A systematic review and meta-analysis. Obes Rev 2023; 24:e13531. [PMID: 36416279 PMCID: PMC10078575 DOI: 10.1111/obr.13531] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 08/26/2022] [Accepted: 10/27/2022] [Indexed: 11/24/2022]
Abstract
Determining if gastrointestinal (GI) hormone response to food intake differs between individuals with, and without, obesity may improve our understanding of obesity pathophysiology. A systematic review and meta-analysis of studies assessing the concentrations of GI hormones, as well as appetite ratings, following a test meal, in individuals with and without obesity was undertaken. Systematic searches were conducted in the databases MEDLINE, Embase, Cochrane Library, PsycINFO, Web of Science, and ClinicalTrials.gov. A total of 7514 unique articles were retrieved, 115 included in the systematic review, and 70 in the meta-analysis. The meta-analysis compared estimated standardized mean difference in GI hormones' concentration, as well as appetite ratings, between individuals with and without obesity. Basal and postprandial total ghrelin concentrations were lower in individuals with obesity compared with controls, and this was reflected by lower postprandial hunger ratings in the former. Individuals with obesity had a lower postprandial concentration of total peptide YY compared with controls, but no significant differences were found for glucagon-like peptide 1, cholecystokinin, or other appetite ratings. A large methodological and statistical heterogeneity among studies was found. More comprehensive studies are needed to understand if the differences observed are a cause or a consequence of obesity.
Collapse
Affiliation(s)
- Marthe Isaksen Aukan
- Obesity Research Group, Department of Clinical and Molecular Medicine, Faculty of Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.,Centre of Obesity and Innovation (ObeCe), Clinic of Surgery, St. Olav University Hospital, Trondheim, Norway
| | - Silvia Coutinho
- Obesity Research Group, Department of Clinical and Molecular Medicine, Faculty of Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.,Department of Public Health Nutrition at the Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo (UiO), Oslo, Norway
| | - Sindre Andre Pedersen
- Library Section for Research Support, Data and Analysis, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Melanie Rae Simpson
- Department of Public Health and Nursing, Norwegian University of Science and Technology, Trondheim, Norway.,Clinical Research Unit Central Norway, St. Olavs Hospital, Trondheim, Norway
| | - Catia Martins
- Obesity Research Group, Department of Clinical and Molecular Medicine, Faculty of Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.,Centre of Obesity and Innovation (ObeCe), Clinic of Surgery, St. Olav University Hospital, Trondheim, Norway.,Department of Nutrition Sciences, the University of Alabama at Birmingham (UAB), Birmingham, Alabama, USA
| |
Collapse
|
8
|
The effect of gastrointestinal bitter sensing on appetite regulation and energy intake: A systematic review. Appetite 2023; 180:106336. [PMID: 36216215 DOI: 10.1016/j.appet.2022.106336] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 09/28/2022] [Accepted: 09/29/2022] [Indexed: 11/05/2022]
Abstract
Taste receptors are located on the epithelial surface throughout the alimentary canal to identify nutrients and potential toxins. In the oral cavity, the role of taste is to encourage or discourage ingestion, while in the gastrointestinal (GI) tract, the taste receptors help the body prepare for an appropriate response to the ingested foods. The GI sensing of bitter compounds may alter the secretion of appetite-related hormones thereby reducing food intake, which may have potential use for managing health outcomes. This systematic literature review investigated the acute effects of administering different bitter tasting compounds on circulating levels of selected GI hormones, subjective appetite, and energy intake in humans. A literature search was conducted using Medline, CINAHL and Web of Science databases. Of 290 articles identified, 16 met the inclusion criteria. Twelve studies assessed food intake; four of these found bitter administration decreased food intake and eight did not. Fourteen studies assessed subjective appetite; seven found bitter administration affected at least one measure of appetite and seven detected no significant changes. Nine studies included measures of GI hormones; no significant effects were found for changes in GLP-1, CCK or PYY. Four studies measured motilin and ghrelin and found mostly consistent changes in either food intake or subjective appetite. Overall, the data on food intake and subjective appetite were inconsistent, with only motilin and ghrelin responsive to post-oral bitter administration. There is limited consistent conclusive evidence that bitter compounds influence food intake, appetite or hormones with the reasons for this discussed within. SYSTEMATIC REVIEW REGISTRATION: CRD42021226102.
Collapse
|
9
|
Sobek G, Dąbrowski M. The importance of the taste preferences and sensitivity of mothers and their children in the aspect of excessive body weight of children. Front Endocrinol (Lausanne) 2022; 13:1031884. [PMID: 36465664 PMCID: PMC9708877 DOI: 10.3389/fendo.2022.1031884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 10/31/2022] [Indexed: 11/17/2022] Open
Abstract
INTRODUCTION Food selection among adults and mostly children depends mainly on the taste of a dish. Poor taste sensitivity as well as strong preferences for sweet and fat taste may be the factors predisposing children to become overweight and/or develop obesity. Family environment, including mothers' eating habits and preferences, may affect children's taste perception and preferences. The aim of the study was to assess taste perception and preferences in children and their mothers in relation to their weight status. METHODS Sensory tests were carried out using puddings with different sugar and fat content. In all study participants anthropometric measurements (weight and height with BMI calculation) were performed. RESULTS The study results did not reveal any differences in the taste sensitivity of overweight/obese and normative body weight children. Similarity was found in the perception of different levels of sweet/fat flavors among children and parents. Overweight/obese children were two times more likely to choose a very fat and very sweet taste compared to normal weight children. The results showed that children prefer a sweet taste more often than their mothers. Mothers' fat taste preferences were important - the fatter the taste they selected, the greater the percentage of children with obesity. DISCUSSION Mothers' taste sensitivity may affect children's perception of the quality (intensity) of flavors. Normal-weight children chose a low fat and low sweet taste more frequently than those with excess of body weight. The role of parents in shaping taste preferences is of utmost importance and should be based on limiting the consumption of products rich in sugar and/or fat.
Collapse
Affiliation(s)
- Grzegorz Sobek
- Institute of Health Sciences, College of Medical Sciences, University of Rzeszow, Rzeszów, Poland
| | - Mariusz Dąbrowski
- Institute of Medical Sciences, College of Medical Sciences, University of Rzeszow, Rzeszów, Poland
| |
Collapse
|
10
|
Geary N, Asarian L, Graf G, Gobbi S, Tobler PN, Rehfeld JF, Leeners B. Increased Meal Size but Reduced Meal-Stimulated Plasma Cholecystokinin Concentrations in Women With Obesity. Endocrinology 2022; 164:6845692. [PMID: 36423205 DOI: 10.1210/endocr/bqac192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Indexed: 11/26/2022]
Abstract
To better understand the physiological basis of obesity in women, we investigated whether obesity or menstrual cycle phase affects laboratory test-meal size or meal-stimulated plasma cholecystokinin (CCK) concentration. Women with healthy weight (body mass index [BMI] of 18.5-24.9 kg/m2, N = 16) or obesity (BMI 30-39.9 kg/m2, N = 20) were tested once in the late-follicular or peri-ovulatory phase (LF/PO) and once in the mid-luteal phase (ML). Meals of ham sandwiches were offered and blood was sampled. Menstrual cycle phases were verified with participants' reports of menses and measurements of progesterone and luteinizing hormone (LH) concentrations. Women with obesity ate significantly larger meals than women with healthy weight, (mean, 711 [95% CI, 402-1013] kJ, P = 0.001, during the LF/PO and 426 [105-734] kJ, P = 0.027, larger during the ML). Women with healthy weight ate smaller meals during LF/PO than ML (decrease, 510 [192-821 kJ], P = 0.008), but women with obesity did not (decrease, 226 [-87-542] kJ, P = 0.15). CCK concentrations 18 to 30 minutes after meal onset were lower in women with obesity than in women with healthy weight during LF/PO (3.6 [3.1-4.1] vs 6.1 [4.5-7.7] pmol/L; P = 0.004), but not during ML, with a significant interaction effect (1.8 [1.2-2.4] pmol/L, P = 0.048). Women with obesity consumed larger meals than women with healthy weight but displayed reduced meal-stimulated plasma CCK concentrations. These data are consistent with the hypothesis that a defect in CCK secretion compromises satiation in obese women and contributes to the development or maintenance of obesity.
Collapse
Affiliation(s)
- Nori Geary
- Department of Psychiatry, Weill Cornell Medical College, New York, NY 10021, USA†
| | - Lori Asarian
- Institute of Veterinary Physiology, University of Zurich, 8057 Zurich, Switzerland
| | - Gwendolyn Graf
- Department of Reproductive Endocrinology, University Hospital of Zurich, 8091 Zurich, Switzerland
| | - Susanna Gobbi
- Zurich Center for Neuroeconomics, University of Zurich, 8006 Zurich, Switzerland
| | - Philippe N Tobler
- Zurich Center for Neuroeconomics, University of Zurich, 8006 Zurich, Switzerland
| | - Jens F Rehfeld
- Department of Clinical Biochemistry, Rigshospitalet, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Brigitte Leeners
- Department of Reproductive Endocrinology, University Hospital of Zurich, 8091 Zurich, Switzerland
| |
Collapse
|
11
|
Garcia‐Burgos D, Andres F, Trier S, Vögele C, Munsch S. Discrimination of sweet‐fat ingredients in people with weight‐ and eating‐related problems using a signal detection theory. J SENS STUD 2022. [DOI: 10.1111/joss.12783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
| | - Fabienne Andres
- Department of Psychology University of Fribourg Fribourg Switzerland
| | | | - Claus Vögele
- Institute for Health and Behaviour, Department of Behavioural and Cognitive Sciences University of Luxembourg Luxembourg Luxembourg
| | - Simone Munsch
- Department of Psychology University of Fribourg Fribourg Switzerland
| |
Collapse
|
12
|
Rezaie P, Bitarafan V, Rose BD, Lange K, Rehfeld JF, Horowitz M, Feinle-Bisset C. Quinine Effects on Gut and Pancreatic Hormones and Antropyloroduodenal Pressures in Humans-Role of Delivery Site and Sex. J Clin Endocrinol Metab 2022; 107:e2870-e2881. [PMID: 35325161 PMCID: PMC9250303 DOI: 10.1210/clinem/dgac182] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Indexed: 02/07/2023]
Abstract
CONTEXT The bitter substance quinine modulates the release of a number of gut and gluco-regulatory hormones and upper gut motility. As the density of bitter receptors may be higher in the duodenum than the stomach, direct delivery to the duodenum may be more potent in stimulating these functions. The gastrointestinal responses to bitter compounds may also be modified by sex. BACKGROUND We have characterized the effects of intragastric (IG) versus intraduodenal (ID) administration of quinine hydrochloride (QHCl) on gut and pancreatic hormones and antropyloroduodenal pressures in healthy men and women. METHODS 14 men (26 ± 2 years, BMI: 22.2 ± 0.5 kg/m2) and 14 women (28 ± 2 years, BMI: 22.5 ± 0.5 kg/m2) received 600 mg QHCl on 2 separate occasions, IG or ID as a 10-mL bolus, in randomized, double-blind fashion. Plasma ghrelin, cholecystokinin, peptide YY, glucagon-like peptide-1 (GLP-1), insulin, glucagon, and glucose concentrations and antropyloroduodenal pressures were measured at baseline and for 120 minutes following QHCl. RESULTS Suppression of ghrelin (P = 0.006), stimulation of cholecystokinin (P = 0.030), peptide YY (P = 0.017), GLP-1 (P = 0.034), insulin (P = 0.024), glucagon (P = 0.030), and pyloric pressures (P = 0.050), and lowering of glucose (P = 0.001) were greater after ID-QHCl than IG-QHCl. Insulin stimulation (P = 0.021) and glucose reduction (P = 0.001) were greater in females than males, while no sex-associated effects were found for cholecystokinin, peptide YY, GLP-1, glucagon, or pyloric pressures. CONCLUSION ID quinine has greater effects on plasma gut and pancreatic hormones and pyloric pressures than IG quinine in healthy subjects, consistent with the concept that stimulation of small intestinal bitter receptors is critical to these responses. Both insulin stimulation and glucose lowering were sex-dependent.
Collapse
Affiliation(s)
- Peyman Rezaie
- Adelaide Medical School and Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide SA 5005, Australia
| | - Vida Bitarafan
- Adelaide Medical School and Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide SA 5005, Australia
| | - Braden D Rose
- Adelaide Medical School and Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide SA 5005, Australia
| | - Kylie Lange
- Adelaide Medical School and Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide SA 5005, Australia
| | - Jens F Rehfeld
- Department of Clinical Biochemistry, Rigshospitalet, 2100 Copenhagen, Denmark
| | - Michael Horowitz
- Adelaide Medical School and Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide SA 5005, Australia
- Endocrine and Metabolic Unit, Royal Adelaide Hospital, Adelaide SA 5005, Australia
| | - Christine Feinle-Bisset
- Correspondence: Prof Christine Feinle-Bisset, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Corner of North Tce and George St, Adelaide SA 5005, Australia.
| |
Collapse
|
13
|
Yazdanpanah Z, Mozaffari-Khosravi H, Mirzaei M, Sheikhha MH, Salehi-Abargouei A. A systematic review and meta-analysis on the association between CD36 rs1761667 polymorphism and cardiometabolic risk factors in adults. Sci Rep 2022; 12:5916. [PMID: 35396566 PMCID: PMC8993862 DOI: 10.1038/s41598-022-09908-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 03/21/2022] [Indexed: 12/04/2022] Open
Abstract
The cluster of differentiation 36 (CD36) is one of the main receptors implicated in the pathogenesis of the cardiovascular disease. This study aimed to assess the association between CD36 rs1761667 polymorphism and cardiometabolic risk factors including body mass index (BMI), waist circumference (WC), total cholesterol (TC), triglyceride, HDL-C, LDL-C, blood pressure and fasting blood glucose (FBG). PubMed, EMBASE, Scopus, web of science, and Google Scholar were searched up to December 2021. Subgroup and meta-regression analyses were conducted to explore sources of heterogeneity. Eighteen eligible studies (6317 participants) were included in the study. In the overall analysis, a significant association was found between rs1761667 polymorphism of CD36 and TG in allelic (p < 0.001), recessive (p = 0.001) and homozygous (p = 0.006) models. A relationship between this polymorphism and HDL-C and FBG level was observed in the recessive genetic model. In the subgroup analysis, the A allele was associated with impaired lipid profiles (TC, LDL-C and HDL-C) in the Asian population. The influences of health status, design of the study, confounders, and other sources of heterogeneity should be considered when interpreting present findings. Cohort studies with large sample size and in different ethnicities are needed to confirm the relationship between rs1761667 SNP and cardiometabolic risk factors.
Collapse
Affiliation(s)
- Zeinab Yazdanpanah
- Department of Nutrition, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, PO Code 8915173160, Iran
- Nutrition and Food Security Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Hassan Mozaffari-Khosravi
- Department of Nutrition, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, PO Code 8915173160, Iran
- Nutrition and Food Security Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Yazd Diabetic Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Masoud Mirzaei
- Yazd Cardiovascular Research Centre, Non-Communicable Research Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Mohammad Hasan Sheikhha
- Department of Medical Genetics, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Abortion Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Amin Salehi-Abargouei
- Department of Nutrition, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, PO Code 8915173160, Iran.
- Nutrition and Food Security Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| |
Collapse
|
14
|
Sinding C, Aveline C, Brindisi MC, Thomas-Danguin T. Flaveur et obésité. CAHIERS DE NUTRITION ET DE DIÉTÉTIQUE 2022. [DOI: 10.1016/j.cnd.2022.02.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
15
|
Brondel L, Quilliot D, Mouillot T, Khan NA, Bastable P, Boggio V, Leloup C, Pénicaud L. Taste of Fat and Obesity: Different Hypotheses and Our Point of View. Nutrients 2022; 14:nu14030555. [PMID: 35276921 PMCID: PMC8838004 DOI: 10.3390/nu14030555] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/20/2022] [Accepted: 01/21/2022] [Indexed: 01/09/2023] Open
Abstract
Obesity results from a temporary or prolonged positive energy balance due to an alteration in the homeostatic feedback of energy balance. Food, with its discriminative and hedonic qualities, is a key element of reward-based energy intake. An alteration in the brain reward system for highly palatable energy-rich foods, comprised of fat and carbohydrates, could be one of the main factors involved in the development of obesity by increasing the attractiveness and consumption of fat-rich foods. This would induce, in turn, a decrease in the taste of fat. A better understanding of the altered reward system in obesity may open the door to a new era for the diagnosis, management and treatment of this disease.
Collapse
Affiliation(s)
- Laurent Brondel
- Centre for Taste and Feeding Behaviour, UMR 6265 CNRS, 1324 INRAE, University of Burgundy, Franche-Comté, 21000 Dijon, France; (T.M.); (C.L.)
- Correspondence: ; Tel.: +33-3-80681677 or +33-6-43213100
| | - Didier Quilliot
- Unité Multidisciplinaire de la Chirurgie de L’obésité, University Hospital Nancy-Brabois, 54500 Vandoeuvre-les-Nancy, France;
| | - Thomas Mouillot
- Centre for Taste and Feeding Behaviour, UMR 6265 CNRS, 1324 INRAE, University of Burgundy, Franche-Comté, 21000 Dijon, France; (T.M.); (C.L.)
- Department of Hepato-Gastro-Enterology, University Hospital, 21000 Dijon, France
| | - Naim Akhtar Khan
- Physiologie de Nutrition & Toxicologie (NUTox), UMR/UB/AgroSup 1231, University of Burgundy, Franche-Comté, 21000 Dijon, France;
| | | | | | - Corinne Leloup
- Centre for Taste and Feeding Behaviour, UMR 6265 CNRS, 1324 INRAE, University of Burgundy, Franche-Comté, 21000 Dijon, France; (T.M.); (C.L.)
| | - Luc Pénicaud
- Institut RESTORE, Toulouse University, CNRS U-5070, EFS, ENVT, Inserm U1301 Toulouse, 31432 Toulouse, France;
| |
Collapse
|
16
|
OZTURK EE, DIKMEN D. Association between fat taste sensitivity and diet quality in healthy male Turkish adults. FOOD SCIENCE AND TECHNOLOGY 2022. [DOI: 10.1590/fst.66820] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
17
|
Igarashi M, Iwasa K, Hayakawa T, Tsuduki T, Kimura I, Maruyama K, Yoshikawa K. Dietary oleic acid contributes to the regulation of food intake through the synthesis of intestinal oleoylethanolamide. Front Endocrinol (Lausanne) 2022; 13:1056116. [PMID: 36733808 PMCID: PMC9886573 DOI: 10.3389/fendo.2022.1056116] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 12/07/2022] [Indexed: 01/18/2023] Open
Abstract
INTRODUCTION Among the fatty acid ethanolamides (FAEs), oleoylethanolamide (OEA), linoleoylethanolamide (LEA), and palmitoylethanolamide (PEA) are reported to be involved in feeding regulation. In particular, OEA is well characterized as a satiety signal. Following food consumption, OEA is synthesized from oleic acid (OA) via an N-acyl phosphatidylethanolamine-specific phospholipase D-dependent pathway in the gastroenterocytes, and OEA induces satiety by recruiting sensory fibers. Thus, we hypothesized that dietary OA is an important satiety-inducing molecule. However, there has been no direct demonstration of the effect of dietary OA on satiety induction without the influence of the endogenous biosynthesis of OA from stearic acid (SA) or other FAEs. METHODS In this study, we used two experimental diets to test our hypothesis: (i) an OA diet (OAD; 38.4 mg of OA/g and 7.2 mg of SA/g) and (ii) a low OA diet (LOAD; 3.1 mg of OA/g and 42.4 mg of SA/g). RESULTS Relative to mice fed the OAD, mice fed the LOAD for two weeks exhibited reduced levels of jejunal OEA but not jejunal LEA and PEA. The LOAD-fed mice showed an increase in food intake and body weight gain. Moreover, LOAD-induced increase in food intake was immediately observed after the switch from the OAD, whereas these effects were diminished by the switch back to the OAD. Furthermore, treatment with OA and OEA diminished the effects of LOAD on food intake. CONCLUSION Collectively, these results show that dietary OA is a key factor in the reduction of food intake and increase in satiety mediated by OEA signaling.
Collapse
Affiliation(s)
- Miki Igarashi
- Advanced Clinical Research Center, Institute of Neurological Disorders, Kawasaki, Japan
- Department of Pharmacology, Faculty of Medicine, Saitama Medical University, Saitama, Japan
- Department of Applied Biological Science, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Tokyo, Japan
- *Correspondence: Miki Igarashi,
| | - Kensuke Iwasa
- Department of Pharmacology, Faculty of Medicine, Saitama Medical University, Saitama, Japan
| | - Tetsuhiko Hayakawa
- Department of Applied Biological Science, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Tsuyoshi Tsuduki
- Department of Bioscience and Biotechnology for Future Bioindustries, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Ikuo Kimura
- Department of Applied Biological Science, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Tokyo, Japan
- Laboratory of Molecular Neurobiology, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Kei Maruyama
- Department of Pharmacology, Faculty of Medicine, Saitama Medical University, Saitama, Japan
| | - Keisuke Yoshikawa
- Department of Pharmacology, Faculty of Medicine, Saitama Medical University, Saitama, Japan
| |
Collapse
|
18
|
Hajishafiee M, Ullrich SS, Fitzgerald PC, Horowitz M, Lange K, Poppitt SD, Feinle-Bisset C. Suppression of Energy Intake by Intragastric l-Tryptophan in Lean and Obese Men: Relations with Appetite Perceptions and Circulating Cholecystokinin and Tryptophan. J Nutr 2021; 151:2932-2941. [PMID: 34255069 DOI: 10.1093/jn/nxab218] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/16/2021] [Accepted: 06/11/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND l-Tryptophan reduces energy intake in healthy men. The underlying mechanisms, including appetite, plasma cholecystokinin (CCK), tryptophan (Trp), and the ratio of Trp to large neutral amino acids (Trp:LNAAs ratio), and whether responses differ in lean and obese individuals, are uncertain. OBJECTIVES We evaluated the effects of intragastric Trp on energy intake (primary outcome) and their potential mechanisms, pre- and postmeal, in lean men and those with obesity. METHODS Twelve lean men [mean ± SD age: 30 ± 3 y; BMI (in kg/m2): 23 ± 1] and 13 men with obesity (mean ± SD age: 31 ± 3 y; BMI: 33 ± 1) received, on 3 separate occasions, in double-blind, randomized order, 3 g ("Trp-3") or 1.5 g ("Trp-1.5") Trp, or control ("C"), intragastrically, 30 min before a buffet-meal. Energy intake from the buffet-meal, hunger, fullness, and plasma CCK and amino acid concentrations were measured in response to Trp alone and for 2 h postmeal. Data were analyzed using maximum likelihood mixed-effects models, with treatment, group, and treatment-by-group interaction as fixed effects. RESULTS Trp alone increased plasma CCK, Trp, and the Trp:LNAAs ratio (all P < 0.001), with no difference between groups. Trp suppressed energy intake (P < 0.001), with no difference between groups (lean, C: 1085 ± 102 kcal, Trp-1.5: 1009 ± 92 kcal, Trp-3: 868 ± 104 kcal; obese, C: 1249 ± 98 kcal, Trp-1.5: 1217 ± 90 kcal, Trp-3: 1012 ± 100 kcal). Postmeal, fullness was greater after Trp-3 than after C and Trp-1.5 (all P < 0.05), and in men with obesity than in lean men (P < 0.05). Plasma Trp and the Trp:LNAAs ratio were greater after Trp-3 and Trp-1.5 than after C (all P < 0.001), and tended to be less in men with obesity than in the lean (P = 0.07) (Trp:LNAAs ratio: lean, C: 1.5 ± 0.2, Trp-1.5: 6.9 ± 0.7, Trp-3: 10.7 ± 1.4; obese, C: 1.4 ± 0.1, Trp-1.5: 4.6 ± 0.7, Trp-3: 7.8 ± 1.3). There were inverse correlations of energy intake with plasma Trp and the Trp:LNAAs ratio in both groups (lean, both r = -0.50, P < 0.01; obese, both r = -0.40, P < 0.05). CONCLUSIONS Intragastric Trp has potent energy intake-suppressant effects, in both lean men and those with obesity, apparently related to the Trp:LNAAs ratio.
Collapse
Affiliation(s)
- Maryam Hajishafiee
- Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
- Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, South Australia, Australia
| | | | - Penelope Ce Fitzgerald
- Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
- Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, South Australia, Australia
| | - Michael Horowitz
- Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
- Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, South Australia, Australia
- Endocrine and Metabolic Unit, Royal Adelaide Hospital, Adelaide, South Australia, Australia
| | - Kylie Lange
- Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
- Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, South Australia, Australia
| | - Sally D Poppitt
- Human Nutrition Unit, School of Biological Sciences, Department of Medicine, University of Auckland, Auckland, New Zealand
| | - Christine Feinle-Bisset
- Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
- Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
19
|
Smith KR, Moran TH. Gastrointestinal peptides in eating-related disorders. Physiol Behav 2021; 238:113456. [PMID: 33989649 PMCID: PMC8462672 DOI: 10.1016/j.physbeh.2021.113456] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 04/19/2021] [Accepted: 05/04/2021] [Indexed: 12/13/2022]
Abstract
Food intake is tightly controlled by homeostatic signals sensitive to metabolic need for the regulation of body weight. This review focuses on the peripherally-secreted gastrointestinal peptides (i.e., ghrelin, cholecystokinin, glucagon-like peptide 1, and peptide tyrosine tyrosine) that contribute to the control of appetite and discusses how these peptides or the signals arising from their release are disrupted in eating-related disorders across the weight spectrum, namely anorexia nervosa, bulimia nervosa, and obesity, and whether they are normalized following weight restoration or weight loss treatment. Further, the role of gut peptides in the pathogenesis and treatment response in human weight conditions as identified by rodent models are discussed. Lastly, we review the incretin- and hormone-based pharmacotherapies available for the treatment of obesity and eating-related disorders.
Collapse
Affiliation(s)
- Kimberly R Smith
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States.
| | - Timothy H Moran
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
| |
Collapse
|
20
|
Obesity-induced taste dysfunction, and its implications for dietary intake. Int J Obes (Lond) 2021; 45:1644-1655. [PMID: 34031530 DOI: 10.1038/s41366-021-00855-w] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 04/21/2021] [Accepted: 04/30/2021] [Indexed: 02/06/2023]
Abstract
The incidence of obesity has dramatically increased in recent years, and poses a public health challenge for which an effective and scalable intervention strategy is yet to be found. Our food choices are one of the primary drivers of obesity, where the overconsumption of energy from foods high in fat and sugar can be particularly problematic. Unfortunately, these same foods also tend to be highly palatable. We select foods more on their sensory properties than on any other factor, such as price, convenience, or healthfulness. Previous evidence from human sensory studies has suggested a depressed sense of taste in panelists with obesity. Evidence from animal models also demonstrates a clear deficiency in taste buds occurring with obesity, suggesting that damage to the taste system may result from an obese state. In this review only taste, as opposed to smell, will be examined. Here we seek to bring together evidence from a diverse array of human and animal studies into taste response, dietary intake, and physiology, to better understand changes in taste with obesity, with the goal of understanding whether taste may provide a novel target for intervention in the treatment of obesity.
Collapse
|
21
|
The Function of Gastrointestinal Hormones in Obesity-Implications for the Regulation of Energy Intake. Nutrients 2021; 13:nu13061839. [PMID: 34072172 PMCID: PMC8226753 DOI: 10.3390/nu13061839] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 05/20/2021] [Accepted: 05/24/2021] [Indexed: 02/07/2023] Open
Abstract
The global burden of obesity and the challenges of prevention prompted researchers to investigate the mechanisms that control food intake. Food ingestion triggers several physiological responses in the digestive system, including the release of gastrointestinal hormones from enteroendocrine cells that are involved in appetite signalling. Disturbed regulation of gut hormone release may affect energy homeostasis and contribute to obesity. In this review, we summarize the changes that occur in the gut hormone balance during the pre- and postprandial state in obesity and the alterations in the diurnal dynamics of their plasma levels. We further discuss how obesity may affect nutrient sensors on enteroendocrine cells that sense the luminal content and provoke alterations in their secretory profile. Gastric bypass surgery elicits one of the most favorable metabolic outcomes in obese patients. We summarize the effect of different strategies to induce weight loss on gut enteroendocrine function. Although the mechanisms underlying obesity are not fully understood, restoring the gut hormone balance in obesity by targeting nutrient sensors or by combination therapy with gut peptide mimetics represents a novel strategy to ameliorate obesity.
Collapse
|
22
|
Ozturk EE, Dikmen D. Is sonic hedgehog expression in saliva related to taste sensitivity in adults? Physiol Behav 2021; 236:113412. [PMID: 33823177 DOI: 10.1016/j.physbeh.2021.113412] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 03/16/2021] [Accepted: 03/29/2021] [Indexed: 10/21/2022]
Abstract
Recent studies have demonstrated the critical role of the sonic hedgehog in the sensory perception of taste. Salivary sonic hedgehog stimulates taste bud stem cells, also acts as a taste bud growth factor, and contributes to the receptors' development and longevity in the taste buds. This study's objective was to investigate the possible relationship between taste sensitivity and sonic hedgehog expression in saliva in normal-weight and pre-obese/obese participants (totally 46 male (47.8%) and female (52.2%) aged 19-44 were recruited). Thresholds for the six tastes (sweet, salty, bitter, sour, umami and fat) were investigated using 3-Alternate Forced Choice Methodology. Saliva was collected with the draining method for sonic hedgehog expression analysis. The results indicated significantly higher thresholds for sweet, bitter and fat in pre-obese /obese adults than normal-weight adults (p<0.05). The sonic hedgehog expression was also lower in pre-obese /obese participants than normal-weight participants (p<0.05). Furthermore, sonic hedgehog expression was higher in hypersensitive participants than hyposensitive participants (p<0.05) only among normal-weight individuals, with no significant difference in sonic hedgehog expression level by taste sensitivity in pre-obese/obese individuals (p>0.05). Sonic hedgehog expression level was negatively correlated with body mass index, sweet, salty, bitter and fat taste threshold. The study results indicated that sonic hedgehog expression level acted on taste sensitivity in normal-weight participants, with greater expression in hypersensitive individuals than hyposensitive individuals.
Collapse
Affiliation(s)
- Elif Esra Ozturk
- Hacettepe University, Faculty of Health Sciences, Department of Nutrition and Dietetics, Ankara, Turkey.
| | - Derya Dikmen
- Hacettepe University, Faculty of Health Sciences, Department of Nutrition and Dietetics, Ankara, Turkey.
| |
Collapse
|
23
|
Schalla MA, Taché Y, Stengel A. Neuroendocrine Peptides of the Gut and Their Role in the Regulation of Food Intake. Compr Physiol 2021; 11:1679-1730. [PMID: 33792904 DOI: 10.1002/cphy.c200007] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The regulation of food intake encompasses complex interplays between the gut and the brain. Among them, the gastrointestinal tract releases different peptides that communicate the metabolic state to specific nuclei in the hindbrain and the hypothalamus. The present overview gives emphasis on seven peptides that are produced by and secreted from specialized enteroendocrine cells along the gastrointestinal tract in relation with the nutritional status. These established modulators of feeding are ghrelin and nesfatin-1 secreted from gastric X/A-like cells, cholecystokinin (CCK) secreted from duodenal I-cells, glucagon-like peptide 1 (GLP-1), oxyntomodulin, and peptide YY (PYY) secreted from intestinal L-cells and uroguanylin (UGN) released from enterochromaffin (EC) cells. © 2021 American Physiological Society. Compr Physiol 11:1679-1730, 2021.
Collapse
Affiliation(s)
- Martha A Schalla
- Charité Center for Internal Medicine and Dermatology, Department for Psychosomatic Medicine, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Yvette Taché
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, CURE: Digestive Diseases Research Center, David Geffen School of Medicine, UCLA, Los Angeles, California, USA.,VA Greater Los Angeles Healthcare System, Los Angeles, California, USA
| | - Andreas Stengel
- Charité Center for Internal Medicine and Dermatology, Department for Psychosomatic Medicine, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany.,Department of Psychosomatic Medicine and Psychotherapy, Medical University Hospital Tübingen, Tübingen, Germany
| |
Collapse
|
24
|
Oral Signals of Short and Long Chain Fatty Acids: Parallel Taste Pathways to Identify Microbes and Triglycerides. CURRENT OPINION IN PHYSIOLOGY 2021; 20:126-133. [PMID: 33738372 DOI: 10.1016/j.cophys.2021.01.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Both short chain fatty acids (SCFAs) and long chain fatty acids (LCFAs) rely on free fatty acid receptors to signal their presence to the body, but their individual detection and putative reward systems are different. These separate, yet parallel, taste signaling pathways allow us to distinguish microbe-produced from triglyceride-based fatty acids. Free SCFAs indicate that the food has been fermented and may still contain living, probiotic microbes that can colonize the gut. Free LCFAs indicate the presence of calorie-rich triglycerides in foods. By contrast, LCFAs stimulate endocannabinoids, which reinforce overconsumption of triglycerides. Here we examine the separate oral detection and putative reward systems for both LCFA and SCFAs, and introduce a novel dietary LC:SC ratio as a guideline to improve metabolism and health.
Collapse
|
25
|
Ricardo-Silgado ML, McRae A, Acosta A. Role of Enteroendocrine Hormones in Appetite and Glycemia. ACTA ACUST UNITED AC 2021; 23. [PMID: 34179564 DOI: 10.1016/j.obmed.2021.100332] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Enteroendocrine cells (EECs) are specialized cells that are widely distributed throughout the gastrointestinal tract. EECs sense luminal content and release hormones, such as: ghrelin, cholecystokinin, glucagon like peptide 1, peptide YY, insulin like peptide 5, and oxyntomodulin. These hormones can enter the circulation to act on distant targets or act locally on neighboring cells and neuronal pathways to modulate food digestion, food intake, energy balance and body weight. Obesity, insulin resistance and diabetes are associated with alterations in the levels of enteroendocrine hormones. Evidence also suggests that modified regulation and release of gut hormones are the result of compensatory mechanisms in states of excess adipose tissue and hyperglycemia. This review collects the evidence available detailing pathophysiological alterations in enteroendocrine hormones and their association with appetite, obesity and glycemic control.
Collapse
Affiliation(s)
- Maria Laura Ricardo-Silgado
- Precision Medicine for Obesity Program, and Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN
| | - Alison McRae
- Precision Medicine for Obesity Program, and Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN
| | - Andres Acosta
- Precision Medicine for Obesity Program, and Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN
| |
Collapse
|
26
|
Macronutrient Sensing in the Oral Cavity and Gastrointestinal Tract: Alimentary Tastes. Nutrients 2021; 13:nu13020667. [PMID: 33669584 PMCID: PMC7922037 DOI: 10.3390/nu13020667] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 02/09/2021] [Accepted: 02/16/2021] [Indexed: 02/07/2023] Open
Abstract
There are numerous and diverse factors enabling the overconsumption of foods, with the sense of taste being one of these factors. There are four well established basic tastes: sweet, sour, salty, and bitter; all with perceptual independence, salience, and hedonic responses to encourage or discourage consumption. More recently, additional tastes have been added to the basic taste list including umami and fat, but they lack the perceptual independence and salience of the basics. There is also emerging evidence of taste responses to kokumi and carbohydrate. One interesting aspect is the link with the new and emerging tastes to macronutrients, with each macronutrient having two distinct perceptual qualities that, perhaps in combination, provide a holistic perception for each macronutrient: fat has fat taste and mouthfeel; protein has umami and kokumi; carbohydrate has sweet and carbohydrate tastes. These new tastes can be sensed in the oral cavity, but they have more influence post- than pre-ingestion. Umami, fat, kokumi, and carbohydrate tastes have been suggested as an independent category named alimentary. This narrative review will present and discuss evidence for macronutrient sensing throughout the alimentary canal and evidence of how each of the alimentary tastes may influence the consumption of foods.
Collapse
|
27
|
Duca FA, Waise TMZ, Peppler WT, Lam TKT. The metabolic impact of small intestinal nutrient sensing. Nat Commun 2021; 12:903. [PMID: 33568676 PMCID: PMC7876101 DOI: 10.1038/s41467-021-21235-y] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 01/19/2021] [Indexed: 12/12/2022] Open
Abstract
The gastrointestinal tract maintains energy and glucose homeostasis, in part through nutrient-sensing and subsequent signaling to the brain and other tissues. In this review, we highlight the role of small intestinal nutrient-sensing in metabolic homeostasis, and link high-fat feeding, obesity, and diabetes with perturbations in these gut-brain signaling pathways. We identify how lipids, carbohydrates, and proteins, initiate gut peptide release from the enteroendocrine cells through small intestinal sensing pathways, and how these peptides regulate food intake, glucose tolerance, and hepatic glucose production. Lastly, we highlight how the gut microbiota impact small intestinal nutrient-sensing in normal physiology, and in disease, pharmacological and surgical settings. Emerging evidence indicates that the molecular mechanisms of small intestinal nutrient sensing in metabolic homeostasis have physiological and pathological impact as well as therapeutic potential in obesity and diabetes. The gastrointestinal tract participates in maintaining metabolic homeostasis in part through nutrient-sensing and subsequent gut-brain signalling. Here the authors review the role of small intestinal nutrient-sensing in regulation of energy intake and systemic glucose metabolism, and link high-fat diet, obesity and diabetes with perturbations in these pathways.
Collapse
Affiliation(s)
- Frank A Duca
- BIO5 Institute, University of Arizona, Tucson, AZ, USA. .,School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ, USA.
| | - T M Zaved Waise
- Toronto General Hospital Research Institute, UHN, Toronto, Canada
| | - Willem T Peppler
- Toronto General Hospital Research Institute, UHN, Toronto, Canada
| | - Tony K T Lam
- Toronto General Hospital Research Institute, UHN, Toronto, Canada. .,Department of Physiology, University of Toronto, Toronto, Canada. .,Department of Medicine, University of Toronto, Toronto, Canada. .,Banting and Best Diabetes Centre, University of Toronto, Toronto, Canada.
| |
Collapse
|
28
|
Smith K, Taylor GS, Allerton DM, Brunsgaard LH, Bowden Davies KA, Stevenson EJ, West DJ. The Postprandial Glycaemic and Hormonal Responses Following the Ingestion of a Novel, Ready-to-Drink Shot Containing a Low Dose of Whey Protein in Centrally Obese and Lean Adult Males: A Randomised Controlled Trial. Front Endocrinol (Lausanne) 2021; 12:696977. [PMID: 34220720 PMCID: PMC8253223 DOI: 10.3389/fendo.2021.696977] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 05/19/2021] [Indexed: 11/13/2022] Open
Abstract
PURPOSE Elevated postprandial glycaemia [PPG] increases the risk of cardiometabolic complications in insulin-resistant, centrally obese individuals. Therefore, strategies that improve PPG are of importance for this population. Consuming large doses of whey protein [WP] before meals reduces PPG by delaying gastric emptying and stimulating the secretion of the incretin peptides, glucose-dependent insulinotropic polypeptide [GIP] and glucagon-like peptide 1 [GLP-1]. It is unclear if these effects are observed after smaller amounts of WP and what impact central adiposity has on these gastrointestinal processes. METHODS In a randomised-crossover design, 12 lean and 12 centrally obese adult males performed two 240 min mixed-meal tests, ~5-10 d apart. After an overnight fast, participants consumed a novel, ready-to-drink WP shot (15 g) or volume-matched water (100 ml; PLA) 10 min before a mixed-nutrient meal. Gastric emptying was estimated by oral acetaminophen absorbance. Interval blood samples were collected to measure glucose, insulin, GIP, GLP-1, and acetaminophen. RESULTS WP reduced PPG area under the curve [AUC0-60] by 13 and 18.2% in the centrally obese and lean cohorts, respectively (both p <0.001). In both groups, the reduction in PPG was accompanied by a two-three-fold increase in GLP-1 and delayed gastric emptying. Despite similar GLP-1 responses during PLA, GLP-1 secretion during the WP trial was ~27% lower in centrally obese individuals compared to lean (p = 0.001). In lean participants, WP increased the GLP-1ACTIVE/TOTAL ratio comparative to PLA (p = 0.004), indicative of reduced GLP-1 degradation. Conversely, no treatment effects for GLP-1ACTIVE/TOTAL were seen in obese subjects. CONCLUSION Pre-meal ingestion of a novel, ready-to-drink WP shot containing just 15 g of dietary protein reduced PPG in lean and centrally obese males. However, an attenuated GLP-1 response to mealtime WP and increased incretin degradation might impact the efficacy of nutritional strategies utilising the actions of GLP-1 to regulate PPG in centrally obese populations. Whether these defects are caused by an individual's insulin resistance, their obese state, or other obesity-related ailments needs further investigation. CLINICAL TRIAL REGISTRATION ISRCTN.com, identifier [ISRCTN95281775]. https://www.isrctn.com/.
Collapse
Affiliation(s)
- Kieran Smith
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Guy S. Taylor
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Dean M. Allerton
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Lise Hoej Brunsgaard
- Health and Performance Nutrition, Arla Foods Ingredients Group P/S, Viby J., Denmark
| | - Kelly A. Bowden Davies
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
- School of Sport and Exercise Sciences, Manchester Metropolitan University, Manchester, United Kingdom
| | - Emma J. Stevenson
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Daniel J. West
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
- *Correspondence: Daniel J. West,
| |
Collapse
|
29
|
Single-nucleotide polymorphism rs1761667 in the CD36 gene is associated with orosensory perception of a fatty acid in obese and normal-weight Moroccan subjects. J Nutr Sci 2020; 9:e24. [PMID: 32685140 PMCID: PMC7329752 DOI: 10.1017/jns.2020.18] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 05/26/2020] [Indexed: 01/29/2023] Open
Abstract
Obese subjects have shown a preference for dietary lipids. A recent collection of evidence has proposed that a variant in the CD36 gene plays a significant role in this pathway. We assessed the association between the orosensory detection of a long-chain fatty acid, i.e. oleic acid (OA), and genetic polymorphism of the lipid taste sensor CD36 in obese and normal-weight subjects. Adult participants were recruited in the fasting condition. They were invited to fat taste perception sessions, using emulsions containing OA and according to the three-alternative forced-choice (3-AFC) method. Genomic DNA was used to determine the polymorphism (SNP rs 1761667) of the CD36 gene. Obese (n 50; BMI 34⋅97 (sd 4⋅02) kg/m2) exhibited a significantly higher oral detection threshold for OA (3⋅056 (sd 3⋅53) mmol/l) than did the normal-weight (n 50; BMI 22⋅16 (sd 1⋅81) kg/m2) participants (1⋅20 (sd 3⋅23) mmol/l; P = 0⋅007). There was a positive correlation between OA detection thresholds and BMI in all subjects; evenly with body fat percentage (BF%). AA genotype was more frequent in the obese group than normal-weight group. OA detection thresholds were much higher for AA and AG genotypes in obese subjects compared with normal-weight participants. Higher oral detection thresholds for fatty acid taste are related to BMI, BF% and not always to CD36 genotype.
Collapse
|
30
|
Associations between sweet taste function, oral complex carbohydrate sensitivity, liking and consumption of ad libitum sweet and non-sweet carbohydrate milkshakes among female adults. Br J Nutr 2020; 122:829-840. [PMID: 31309910 DOI: 10.1017/s0007114519001703] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Excess energy intake is recognised as a strong contributing factor to the global rise of being overweight and obese. The aim of this paper was to investigate if oral sensitivity to complex carbohydrate relates to ad libitum consumption of complex carbohydrate foods in a sample group of female adults. Participants' ((n 51 females): age 23·0 (sd 0·6) years (range 20·0-41·0 years); excluding restrained eaters) sensitivity towards maltodextrin (oral complex carbohydrate) and glucose (sweet taste) was assessed by measuring detection threshold (DT) and suprathreshold intensity perception (ST). A crossover design was used to assess consumption of two different iso-energetic preload milkshakes and ad libitum milkshakes - (1) glucose-based milkshake, (2) maltodextrin-based milkshake. Ad libitum intake (primary outcome) and eating rate, liking, hunger, fullness and prospective consumption ratings were measured. Participants who were more sensitive towards complex carbohydrate (maltodextrin DT) consumed significantly more maltodextrin-based milkshake in comparison with less-sensitive participants (P = 0·01) and this was independent of liking. Participants who had higher liking for glucose-based milkshake consumed significantly more glucose-based milkshake in comparison with participants with lower hedonic ratings (P = 0·049). The results provide support regarding the role of the oral system sensitivity (potentially taste) to complex carbohydrate and the prospective to overconsume complex carbohydrate-based milkshake in a single sitting.
Collapse
|
31
|
Preference for dietary fat: From detection to disease. Prog Lipid Res 2020; 78:101032. [PMID: 32343988 DOI: 10.1016/j.plipres.2020.101032] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 03/22/2020] [Accepted: 03/25/2020] [Indexed: 12/11/2022]
Abstract
Recent advances in the field of taste physiology have clarified the role of different basic taste modalities and their implications in health and disease and proposed emphatically that there might be a distinct cue for oro-sensory detection of dietary long-chain fatty acids (LCFAs). Hence, fat taste can be categorized as a taste modality. During mastication, LCFAs activate tongue lipid sensors like CD36 and GPR120 triggering identical signaling pathways as the basic taste qualities do; however, the physico-chemical perception of fat is not as distinct as sweet or bitter or other taste sensations. The question arises whether "fat taste" is a basic or "alimentary" taste. There is compelling evidence that fat-rich dietary intervention modulates fat taste perception where an increase or a decrease in lipid contents in the diet results, respectively, in downregulation or upregulation of fat taste sensitivity. Evidently, a decrease in oro-sensory detection of LCFAs leads to high fat intake and, consequently, to obesity. In this article, we discuss recent relevant advances made in the field of fat taste physiology with regard to dietary fat preference and lipid sensors that can be the target of anti-obesity strategies.
Collapse
|
32
|
Costanzo A, Russell CG, Lewin S, Keast R. A Fatty Acid Mouth Rinse Decreases Self-Reported Hunger and Increases Self-Reported Fullness in Healthy Australian Adults: A Randomized Cross-Over Trial. Nutrients 2020; 12:nu12030678. [PMID: 32131518 PMCID: PMC7146224 DOI: 10.3390/nu12030678] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 02/27/2020] [Accepted: 02/28/2020] [Indexed: 12/21/2022] Open
Abstract
Fatty acid (FA) chemoreception in the oral cavity, known as fat taste, may trigger a satiety response that is homologous to FA chemoreception in the gastrointestinal tract. In addition, individuals with an impaired fat taste sensitivity are more likely to have an impaired satiety response. This study aimed to assess the effect of an FA mouth rinse on self-reported appetite, and to determine if the effect is modified by fat taste sensitivity. Thirty-one participants (age, 32.0 ± 8.4 y; body mass index (BMI), 26.1 ± 8.1 kg/m2) were studied on four separate days to evaluate the effect of a 20 mM oleic acid (OA) mouth rinse (in duplicate) compared to a control (in duplicate) on self-reported appetite by using a visual analogue scale (VAS) every 30 min for three hours following a standardized low-fat breakfast. The area under the curve ratings for fullness were greater (p = 0.003), and those for hunger were lower (p = 0.002) following the OA rinse compared to the control. The effect of the OA rinse was greater in individuals who were hypersensitive to fat taste compared to moderately sensitive and hyposensitive individuals for fullness (p < 0.010) and hunger (p < 0.010) ratings. In summary, an OA mouth rinse decreases self-reported hunger and increases self-reported fullness, particularly in those who are more sensitive to fat taste. FA receptors in the oral cavity may be potential targets to regulate appetite.
Collapse
|
33
|
Bitarafan V, Fitzgerald PCE, Little TJ, Meyerhof W, Jones KL, Wu T, Horowitz M, Feinle-Bisset C. Intragastric administration of the bitter tastant quinine lowers the glycemic response to a nutrient drink without slowing gastric emptying in healthy men. Am J Physiol Regul Integr Comp Physiol 2020; 318:R263-R273. [PMID: 31774306 DOI: 10.1152/ajpregu.00294.2019] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The rate of gastric emptying and the release of gastrointestinal (GI) hormones are major determinants of postprandial blood-glucose concentrations and energy intake. Preclinical studies suggest that activation of GI bitter-taste receptors potently stimulates GI hormones, including glucagon-like peptide-1 (GLP-1), and thus may reduce postprandial glucose and energy intake. We evaluated the effects of intragastric quinine on the glycemic response to, and the gastric emptying of, a mixed-nutrient drink and the effects on subsequent energy intake in healthy men. The study consisted of 2 parts: part A included 15 lean men, and part B included 12 lean men (aged 26 ± 2 yr). In each part, participants received, on 3 separate occasions, in double-blind, randomized fashion, intragastric quinine (275 or 600 mg) or control, 30 min before a mixed-nutrient drink (part A) or before a buffet meal (part B). In part A, plasma glucose, insulin, glucagon, and GLP-1 concentrations were measured at baseline, after quinine alone, and for 2 h following the drink. Gastric emptying of the drink was also measured. In part B, energy intake at the buffet meal was quantified. Quinine in 600 mg (Q600) and 275 mg (Q275) doses alone stimulated insulin modestly (P < 0.05). After the drink, Q600 and Q275 reduced plasma glucose and stimulated insulin (P < 0.05), Q275 stimulated GLP-1 (P < 0.05), and Q600 tended to stimulate GLP-1 (P = 0.066) and glucagon (P = 0.073) compared with control. Quinine did not affect gastric emptying of the drink or energy intake. In conclusion, in healthy men, intragastric quinine reduces postprandial blood glucose and stimulates insulin and GLP-1 but does not slow gastric emptying or reduce energy intake under our experimental conditions.
Collapse
Affiliation(s)
- Vida Bitarafan
- Adelaide Medical School and Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, SA, Australia
| | - Penelope C E Fitzgerald
- Adelaide Medical School and Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, SA, Australia
| | - Tanya J Little
- Adelaide Medical School and Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, SA, Australia
| | - Wolfgang Meyerhof
- Center for Integrative Physiology and Molecular Medicine, Saarland University, Homburg, Germany
| | - Karen L Jones
- Adelaide Medical School and Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, SA, Australia
- Endocrine and Metabolic Unit, Royal Adelaide Hospital, Adelaide, SA, Australia
| | - Tongzhi Wu
- Adelaide Medical School and Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, SA, Australia
- Endocrine and Metabolic Unit, Royal Adelaide Hospital, Adelaide, SA, Australia
| | - Michael Horowitz
- Adelaide Medical School and Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, SA, Australia
- Endocrine and Metabolic Unit, Royal Adelaide Hospital, Adelaide, SA, Australia
| | - Christine Feinle-Bisset
- Adelaide Medical School and Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, SA, Australia
| |
Collapse
|
34
|
Flechtner-Mors M, Thoma U, Wittmann R, Boehm BO, Mors M, Steinacker JM, Schumann U. The Effect of Potato Protease Inhibitor II on Gastrointestinal Hormones and Satiety in Humans During Weight Reduction. Diabetes Metab Syndr Obes 2020; 13:521-534. [PMID: 32161479 PMCID: PMC7049780 DOI: 10.2147/dmso.s201853] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 12/07/2019] [Indexed: 12/16/2022] Open
Abstract
CONTEXT It is questioned whether the potato protein protease inhibitor II (PI2) reduces appetite and exerts effects on the satiety hormone cholecystokinin (CCK). OBJECTIVE To investigate PI2 impact on gastrointestinal hormones and appetite measures during weight reduction. DESIGN In a randomized, placebo-controlled trial over 20 weeks, fifty-two overweight/obese participants (BMI 25.2-38.0 kg/m2) received a protein-rich diet (30%) adjusted to 500 kcal below their individual daily needs. Subjects ingested a capsule containing either PI2 (150 mg) or placebo twice daily 1 hr before lunch and dinner. At week 0 and week 10 participants joined breakfast test meals to determine CCK, GLP-1, ghrelin, leptin, glucose and insulin concentrations in a time course experimental manner. Appetite sensations were measured on test meal days and in week 4, 9, 14 and 19 using visual analogue scales. RESULTS Weight loss at week 10 and 20 in the PI2 group was 4.3±3.1 kg and 5.6±4.1 kg, in the control group: 4.7±4.0 kg and 6.8±3.7 kg. A significant effect of PI2 on circulating CCK levels was observed at week 10. The other hormones were unaffected by PI2. At week 10, PI2 group subjects showed higher satiety and decreased desire to eat compared to placebo. During study duration, PI2 showed a significant impact on appetite ratings prior to lunch, one hour before dinner and just before dinner. CONCLUSION PI2 increased circulating CCK plasma levels during the diet intervention. Likewise, PI2 modulated appetite sensation from week 4 to 20. The study demonstrated that the PI2 can modulate a key satiety signal.
Collapse
Affiliation(s)
- Marion Flechtner-Mors
- University Medical Center Ulm, Division of Sports and Rehabilitation Medicine, University Hospital Ulm, Ulm, Germany
- Correspondence: Marion Flechtner-Mors Department of Internal Medicine II, Division of Sports and Rehabilitation Medicine, University Hospital Ulm, Leimgrubenweg 14, Ulm89075, GermanyTel + 49 731 50045330Fax + 49 731 50045333 Email
| | - Ulrike Thoma
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | - Regina Wittmann
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | - Bernhard O Boehm
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore
- Imperial College London, London, UK
| | - Mona Mors
- University Medical Center Ulm, Division of Sports and Rehabilitation Medicine, University Hospital Ulm, Ulm, Germany
| | - Jürgen M Steinacker
- University Medical Center Ulm, Division of Sports and Rehabilitation Medicine, University Hospital Ulm, Ulm, Germany
| | - Uwe Schumann
- University Medical Center Ulm, Division of Sports and Rehabilitation Medicine, University Hospital Ulm, Ulm, Germany
| |
Collapse
|
35
|
Emerenziani S, Pier Luca Guarino M, Trillo Asensio LM, Altomare A, Ribolsi M, Balestrieri P, Cicala M. Role of Overweight and Obesity in Gastrointestinal Disease. Nutrients 2019; 12:111. [PMID: 31906216 PMCID: PMC7019431 DOI: 10.3390/nu12010111] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 11/28/2019] [Accepted: 12/20/2019] [Indexed: 02/07/2023] Open
Abstract
The prevalence of obesity is increasing worldwide, leading to a severe impairment of overall health. Actually, obesity has been associated with several pathological conditions, causing an excess overall mortality. In particular, overweight and obesity are well known risk factors for a variety of gastrointestinal (GI) disorders i.e., functional GI disorders as well as, inflammatory bowel disease (IBD), pancreatitis, and GI cancer. The aim of the present review is to summarize the potential role of overweight and obesity in GI disease with particular focus on plausible biological mechanisms that could explain the association between obesity and GI disease based on the most recent evidence in the literature.
Collapse
Affiliation(s)
- Sara Emerenziani
- Gastroenterology Unit, University Campus Bio-Medico di Roma, via Álvaro del Portillo 21, 00128 Rome, Italy; (M.P.L.G.); (A.A.); (M.R.); (P.B.); (M.C.)
| | | | - Laura Maria Trillo Asensio
- Gastroenterology Unit, University Campus Bio-Medico di Roma, via Álvaro del Portillo 21, 00128 Rome, Italy; (M.P.L.G.); (A.A.); (M.R.); (P.B.); (M.C.)
| | | | | | | | | |
Collapse
|
36
|
Walther B, Lett AM, Bordoni A, Tomás‐Cobos L, Nieto JA, Dupont D, Danesi F, Shahar DR, Echaniz A, Re R, Fernandez AS, Deglaire A, Gille D, Schmid A, Vergères G. GutSelf: Interindividual Variability in the Processing of Dietary Compounds by the Human Gastrointestinal Tract. Mol Nutr Food Res 2019; 63:e1900677. [PMID: 31483113 PMCID: PMC6900003 DOI: 10.1002/mnfr.201900677] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 07/25/2019] [Indexed: 12/19/2022]
Abstract
Nutritional research is currently entering the field of personalized nutrition, to a large extent driven by major technological breakthroughs in analytical sciences and biocomputing. An efficient launching of the personalized approach depends on the ability of researchers to comprehensively monitor and characterize interindividual variability in the activity of the human gastrointestinal tract. This information is currently not available in such a form. This review therefore aims at identifying and discussing published data, providing evidence on interindividual variability in the processing of the major nutrients, i.e., protein, fat, carbohydrates, vitamins, and minerals, along the gastrointestinal tract, including oral processing, intestinal digestion, and absorption. Although interindividual variability is not a primary endpoint of most studies identified, a significant number of publications provides a wealth of information on this topic for each category of nutrients. This knowledge remains fragmented, however, and understanding the clinical relevance of most of the interindividual responses to food ingestion described in this review remains unclear. In that regard, this review has identified a gap and sets the base for future research addressing the issue of the interindividual variability in the response of the human organism to the ingestion of foods.
Collapse
Affiliation(s)
- Barbara Walther
- AgroscopeFederal Department of Economic AffairsEducation and Research EAER3003BerneSwitzerland
| | - Aaron M. Lett
- Section for Nutrition ResearchDepartment of MedicineImperial College LondonLondonUK
| | - Alessandra Bordoni
- Department of Agri‐Food Sciences and TechnologiesUniversity of Bologna47521CesenaItaly
| | | | | | - Didier Dupont
- UMR 1253Science et Technologie du Lait et de l'ŒufINRA35000RennesFrance
| | - Francesca Danesi
- Department of Agri‐Food Sciences and TechnologiesUniversity of Bologna47521CesenaItaly
| | - Danit R. Shahar
- Department of Public HealthThe S. Daniel Abraham International Center for Health and NutritionBen‐Gurion University of the Negev84105Beer‐ShevaIsrael
| | - Ana Echaniz
- Cambridge Food Science LtdCB23 5ABCambridgeUK
| | - Roberta Re
- Cambridge Food Science LtdCB23 5ABCambridgeUK
| | | | - Amélie Deglaire
- UMR 1253Science et Technologie du Lait et de l'ŒufINRA35000RennesFrance
| | - Doreen Gille
- AgroscopeFederal Department of Economic AffairsEducation and Research EAER3003BerneSwitzerland
| | - Alexandra Schmid
- AgroscopeFederal Department of Economic AffairsEducation and Research EAER3003BerneSwitzerland
| | - Guy Vergères
- AgroscopeFederal Department of Economic AffairsEducation and Research EAER3003BerneSwitzerland
| |
Collapse
|
37
|
Argueta DA, Perez PA, Makriyannis A, DiPatrizio NV. Cannabinoid CB 1 Receptors Inhibit Gut-Brain Satiation Signaling in Diet-Induced Obesity. Front Physiol 2019; 10:704. [PMID: 31281260 PMCID: PMC6597959 DOI: 10.3389/fphys.2019.00704] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Accepted: 05/20/2019] [Indexed: 01/08/2023] Open
Abstract
Gut-brain signaling controls feeding behavior and energy homeostasis; however, the underlying molecular mechanisms and impact of diet-induced obesity (DIO) on these pathways are poorly defined. We tested the hypothesis that elevated endocannabinoid activity at cannabinoid CB1 receptor (CB1Rs) in the gut of mice rendered DIO by chronic access to a high fat and sucrose diet for 60 days inhibits nutrient-induced release of satiation peptides and promotes overeating. Immunoreactivity for CB1Rs was present in enteroendocrine cells in the mouse’s upper small-intestinal epithelium that produce and secrete the satiation peptide, cholecystokinin (CCK), and expression of mRNA for CB1Rs was greater in these cells when compared to non-CCK producing cells. Oral gavage of corn oil increased levels of bioactive CCK (CCK-8) in plasma from mice fed a low fat no-sucrose diet. Pretreatment with the cannabinoid receptor agonist, WIN55,212-2, blocked this response, which was reversed by co-administration with the peripherally-restricted CB1R neutral antagonist, AM6545. Furthermore, monoacylglycerol metabolic enzyme function was dysregulated in the upper small-intestinal epithelium from DIO mice, which was met with increased levels of a variety of monoacylglycerols including the endocannabinoid, 2-arachidonoyl-sn-glycerol. Corn oil failed to affect levels of CCK in DIO mouse plasma; however, pretreatment with AM6545 restored the ability for corn oil to stimulate increases in levels of CCK, which suggests that elevated endocannabinoid signaling at small intestinal CB1Rs in DIO mice inhibits nutrient-induced CCK release. Moreover, the hypophagic effect of AM6545 in DIO mice was reversed by co-administration with the CCKA receptor antagonist, devazepide. Collectively, these results provide evidence that hyperphagia associated with DIO is driven by a mechanism that includes CB1R-mediated inhibition of gut-brain satiation signaling.
Collapse
Affiliation(s)
- Donovan A Argueta
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, United States
| | - Pedro A Perez
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, United States
| | | | - Nicholas V DiPatrizio
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, United States
| |
Collapse
|
38
|
Gastrointestinal Sensing of Meal-Related Signals in Humans, and Dysregulations in Eating-Related Disorders. Nutrients 2019; 11:nu11061298. [PMID: 31181734 PMCID: PMC6627312 DOI: 10.3390/nu11061298] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 05/29/2019] [Accepted: 06/05/2019] [Indexed: 12/13/2022] Open
Abstract
The upper gastrointestinal (GI) tract plays a critical role in sensing the arrival of a meal, including its volume as well as nutrient and non-nutrient contents. The presence of the meal in the stomach generates a mechanical distension signal, and, as gastric emptying progresses, nutrients increasingly interact with receptors on enteroendocrine cells, triggering the release of gut hormones, with lipid and protein being particularly potent. Collectively, these signals are transmitted to the brain to regulate appetite and energy intake, or in a feedback loop relayed back to the upper GI tract to further adjust GI functions, including gastric emptying. The research in this area to date has provided important insights into how sensing of intraluminal meal-related stimuli acutely regulates appetite and energy intake in humans. However, disturbances in the detection of these stimuli have been described in a number of eating-related disorders. This paper will review the GI sensing of meal-related stimuli and the relationship with appetite and energy intake, and examine changes in GI responses to luminal stimuli in obesity, functional dyspepsia and anorexia of ageing, as examples of eating-related disorders. A much better understanding of the mechanisms underlying these dysregulations is still required to assist in the development of effective management and treatment strategies in the future.
Collapse
|
39
|
McVeay C, Fitzgerald PCE, Ullrich SS, Steinert RE, Horowitz M, Feinle-Bisset C. Effects of intraduodenal administration of lauric acid and L-tryptophan, alone and combined, on gut hormones, pyloric pressures, and energy intake in healthy men. Am J Clin Nutr 2019; 109:1335-1343. [PMID: 31051504 DOI: 10.1093/ajcn/nqz020] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 01/25/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The fatty acid, lauric acid ('C12'), and the amino acid, L-tryptophan ('Trp'), modulate gastrointestinal functions including gut hormones and pyloric pressures, which are important for the regulation of energy intake, and both potently suppress energy intake. OBJECTIVE We hypothesized that the intraduodenal administration of C12 and Trp, at loads that do not affect energy intake individually, when combined will reduce energy intake, which is associated with greater modulation of gut hormones and pyloric pressures. DESIGN Sixteen healthy, lean males (age: 24 ± 1.5 y) received 90-min intraduodenal infusions of saline (control), C12 (0.3 kcal/min), Trp (0.1 kcal/min), or C12 + Trp (0.4 kcal/min), in a randomized, double-blind, cross-over study. Antropyloroduodenal pressures were measured continuously, and plasma cholecystokinin (CCK), ghrelin, and glucagon-like peptide-1 (GLP-1) concentrations, appetite perceptions, and gastrointestinal symptoms at 15-min intervals. Immediately after the infusions, energy intake from a standardized buffet meal was quantified. RESULTS C12 + Trp markedly reduced energy intake (kcal; control: 1,232 ± 72, C12: 1,180 ± 82, Trp: 1,269 ± 73, C12 + Trp: 1,056 ± 106), stimulated plasma CCK (AUC(area under the curve)0-90 min, pmol/L*min; control: 21 ± 8; C12: 129 ± 15; Trp: 97 ± 16; C12 + Trp: 229 ± 22) and GLP-1 (AUC0-90 min, pmol/L*min; control: 102 ± 41; C12: 522 ± 102; Trp: 198 ± 63; C12 + Trp: 545 ± 138), and suppressed ghrelin (AUC0-90 min, pg/mL*min; control: -3,433 ± 2,647; C12: -11,825 ± 3,521; Trp: -8,417 ± 3,734; C12 + Trp: -18,188 ± 4,165) concentrations, but did not stimulate tonic, or phasic, pyloric pressures, compared with the control (all P < 0.05), or have adverse effects. C12 and Trp each stimulated CCK (P < 0.05), but to a lesser degree than C12 + Trp, and did not suppress energy intake or ghrelin. C12, but not Trp, stimulated GLP-1 (P < 0.05) and phasic pyloric pressures (P < 0.05), compared with the control. CONCLUSION The combined intraduodenal administration of C12 and Trp, at loads that individually do not affect energy intake, substantially reduces energy intake, which is associated with a marked stimulation of CCK and suppression of ghrelin. The study was registered as a clinical trial at the Australian and New Zealand Clinical Trial Registry (www.anzctr.org.au,) as 12613000899741.
Collapse
Affiliation(s)
- Christina McVeay
- Adelaide Medical School and National Health and Medical Research Council of Australia Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, Australia
| | - Penelope C E Fitzgerald
- Adelaide Medical School and National Health and Medical Research Council of Australia Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, Australia
| | - Sina S Ullrich
- Adelaide Medical School and National Health and Medical Research Council of Australia Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, Australia
| | - Robert E Steinert
- Adelaide Medical School and National Health and Medical Research Council of Australia Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, Australia
| | - Michael Horowitz
- Adelaide Medical School and National Health and Medical Research Council of Australia Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, Australia
| | - Christine Feinle-Bisset
- Adelaide Medical School and National Health and Medical Research Council of Australia Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, Australia
| |
Collapse
|
40
|
Encapsulation of lipids as emulsion-alginate beads reduces food intake: a randomized placebo-controlled cross-over human trial in overweight adults. Nutr Res 2019; 63:86-94. [DOI: 10.1016/j.nutres.2018.12.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 10/03/2018] [Accepted: 12/05/2018] [Indexed: 01/09/2023]
|
41
|
Dirksen C, Graff J, Fuglsang S, Rehfeld JF, Holst JJ, Madsen JL. Energy intake, gastrointestinal transit, and gut hormone release in response to oral triglycerides and fatty acids in men with and without severe obesity. Am J Physiol Gastrointest Liver Physiol 2019; 316:G332-G337. [PMID: 30520691 DOI: 10.1152/ajpgi.00310.2018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Dietary fat, and particularly fatty acids (FAs) from hydrolyzed triglycerides (TGs), reduces appetite, whereas paradoxically, a high-fat diet leads to excess calorie intake. We therefore hypothesized that the appetite-regulating effects of FAs are perturbed in obesity. Ten men with severe obesity [median body mass index (BMI) of 51.0 kg/m2 (range of 47.9-69.0)] and 10 men without obesity [BMI of 24.6 kg/m2 (range of 21.7-26.8)] were recruited for a double-blind randomized crossover study. On two occasions, participants were given isocaloric (2,660 kJ) and isovolemic (80 ml) loads of either oleic acid (long-chain FA) or olive oil (TG) containing radiolabeled lipid and water markers. Postload scintigraphy, blood sampling, and assessment of appetite were performed for 10 h, after which an ad libitum meal was served. Compared with olive oil, oleic acid slowed gastric mean emptying time (GMET) for lipids ( P < 0.001), accelerated orocoecal transit time (OCTT; P = 0.005), increased postload cholecystokinin section ( P < 0.001), and suppressed ad libitum energy intake ( P = 0.028) in men with severe obesity, and similar effects were seen in the nonobese group (no group × lipid interactions). However, independent of lipid loads, GMET and OCTT were slower (GMETlipid P = 0.046; GMETwater P = 0.003; OCTT P = 0.001), and basal and postload secretion of glucagon-like peptide-1 (GLP-1) was attenuated ( P = 0.045 and P = 0.048, respectively) in men with severe obesity compared with men without obesity. We conclude that the more potent appetite-regulating effects of oleic acid versus olive oil are unimpaired in men with severe obesity. However, regardless of lipid formulations, severe obesity is associated with slowed gastrointestinal transit and attenuated GLP-1 secretion. NEW & NOTEWORTHY Orally ingested fatty acids more efficiently reduce appetite and energy intake than triglycerides also in men with severe obesity. Men with severe obesity have delayed gastrointestinal transit and attenuated early gut hormone responses after an oral lipid load compared with men without obesity.
Collapse
Affiliation(s)
- Carsten Dirksen
- Department of Endocrinology, Hvidovre Hospital, Hvidovre, Denmark.,Novo Nordisk Foundation Centre for Basic Metabolic Research and Department of Biomedical Sciences, University of Copenhagen , Copenhagen , Denmark
| | - Jesper Graff
- Department of Clinical Physiology and Nuclear Medicine, Centre for Functional Imaging and Research, Hvidovre Hospital, Hvidovre, Denmark
| | - Stefan Fuglsang
- Department of Clinical Physiology and Nuclear Medicine, Centre for Functional Imaging and Research, Hvidovre Hospital, Hvidovre, Denmark
| | - Jens F Rehfeld
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen , Denmark
| | - Jens J Holst
- Novo Nordisk Foundation Centre for Basic Metabolic Research and Department of Biomedical Sciences, University of Copenhagen , Copenhagen , Denmark
| | - Jan L Madsen
- Department of Clinical Physiology and Nuclear Medicine, Centre for Functional Imaging and Research, Hvidovre Hospital, Hvidovre, Denmark
| |
Collapse
|
42
|
Obesity is associated with altered gene expression in human tastebuds. Int J Obes (Lond) 2019; 43:1475-1484. [PMID: 30696932 DOI: 10.1038/s41366-018-0303-y] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 10/04/2018] [Accepted: 10/08/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND The role of taste perception in the development and persistence of obesity is currently unclear due to conflicting results from psychophysical and other studies. No study to date has assessed whether there is an underlying fundamental difference in the physiology of taste tissue between lean and obese individuals. METHOD/SUBJECTS We analysed the transcriptomic profile (RNA-seq) of human fungiform taste papillae biopsied from lean (n = 23) and obese (n = 13) Caucasian females (age range 18-55) to identify differences in gene expression. RESULTS Obesity status was the major contributor to variance in global gene expression between individuals. A total of 62 genes had significantly different gene expression levels between lean and obese (P < 0.0002), with the specific taste associated genes phospholipase C beta 2 (PLCβ2) and sonic hedge-hog (SHH) having significantly reduced expression in obese group. Genes associated with inflammation and immune response were the top enriched biological pathways differing between the lean and the obese groups. Analysis of a broader gene set having a twofold change in expression (2619 genes) identified three enriched theme groups (sensory perception, cell and synaptic signalling, and immune response). Further, analysis of taste associated genes identified a consistent reduction in the expression of taste-related genes (in particular reduced type II taste cell genes) in the obese compared to the lean group. CONCLUSION The findings show obesity is associated with altered gene expression in tastebuds. Furthermore, the results suggest the tastebud microenvironment is distinctly different between lean and obese persons and, that changes in sensory gene expression contribute to this altered microenvironment. This research provides new evidence of a link between obesity and altered taste and in the future may help design strategies to combat obesity.
Collapse
|
43
|
Heruc GA, Little TJ, Kohn M, Madden S, Clarke S, Horowitz M, Feinle-Bisset C. Appetite Perceptions, Gastrointestinal Symptoms, Ghrelin, Peptide YY and State Anxiety Are Disturbed in Adolescent Females with Anorexia Nervosa and Only Partially Restored with Short-Term Refeeding. Nutrients 2018; 11:nu11010059. [PMID: 30597915 PMCID: PMC6356798 DOI: 10.3390/nu11010059] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 12/23/2018] [Accepted: 12/25/2018] [Indexed: 02/07/2023] Open
Abstract
Factors underlying disturbed appetite perception in anorexia nervosa (AN) are poorly characterized. We examined in patients with AN whether fasting and postprandial appetite perceptions, gastrointestinal (GI) hormones, GI symptoms and state anxiety (i) differed from healthy controls (HCs) and (ii) were modified by two weeks of refeeding. 22 female adolescent inpatients with restricting AN, studied on hospital admission once medically stable (Wk0), and after one (Wk1) and two (Wk2) weeks of high-calorie refeeding, were compared with 17 age-matched HCs. After a 4 h fast, appetite perceptions, GI symptoms, state anxiety, and plasma acyl-ghrelin, cholecystokinin (CCK), peptide tyrosine tyrosine (PYY) and pancreatic polypeptide (PP) concentrations were assessed at baseline and in response to a mixed-nutrient test-meal (479 kcal). Compared with HCs, in patients with AN at Wk0, baseline ghrelin, PYY, fullness, bloating and anxiety were higher, and hunger less, and in response to the meal, ghrelin, bloating and anxiety were greater, and hunger less (all p < 0.05). After two weeks of refeeding, there was no change in baseline or postprandial ghrelin or bloating, or postprandial anxiety, but baseline PYY, fullness and anxiety decreased, and baseline and postprandial hunger increased (p < 0.05). We conclude that in AN, refeeding for 2 weeks was associated with improvements in PYY, appetite and baseline anxiety, while increased ghrelin, bloating and postprandial anxiety persisted.
Collapse
Affiliation(s)
- Gabriella A Heruc
- Adelaide Medical School and National Health and Medical Research Council of Australia Centre of Research Excellence in Translating Nutritional Science to Good Health, Level 5 Adelaide Health and Medical Sciences Building, Corner North Terrace and George Street, Adelaide 5005, Australia.
| | - Tanya J Little
- Adelaide Medical School and National Health and Medical Research Council of Australia Centre of Research Excellence in Translating Nutritional Science to Good Health, Level 5 Adelaide Health and Medical Sciences Building, Corner North Terrace and George Street, Adelaide 5005, Australia.
| | - Michael Kohn
- The Children's Hospital at Westmead, Sydney 2145, Australia.
- Adolescent and Young Adult Medicine Department, Westmead Hospital, Sydney 2145, Australia.
| | - Sloane Madden
- The Children's Hospital at Westmead, Sydney 2145, Australia.
| | - Simon Clarke
- Adolescent and Young Adult Medicine Department, Westmead Hospital, Sydney 2145, Australia.
| | - Michael Horowitz
- Adelaide Medical School and National Health and Medical Research Council of Australia Centre of Research Excellence in Translating Nutritional Science to Good Health, Level 5 Adelaide Health and Medical Sciences Building, Corner North Terrace and George Street, Adelaide 5005, Australia.
| | - Christine Feinle-Bisset
- Adelaide Medical School and National Health and Medical Research Council of Australia Centre of Research Excellence in Translating Nutritional Science to Good Health, Level 5 Adelaide Health and Medical Sciences Building, Corner North Terrace and George Street, Adelaide 5005, Australia.
| |
Collapse
|
44
|
The role of fatty acids and their endocannabinoid-like derivatives in the molecular regulation of appetite. Mol Aspects Med 2018; 64:45-67. [DOI: 10.1016/j.mam.2018.01.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 01/05/2018] [Accepted: 01/07/2018] [Indexed: 02/07/2023]
|
45
|
Alleleyn AME, van Avesaat M, Ripken D, Bleiel SB, Keszthelyi D, Wilms E, Troost FJ, Hendriks HFJ, Masclee AAM. The Effect of an Encapsulated Nutrient Mixture on Food Intake and Satiety: A Double-Blind Randomized Cross-Over Proof of Concept Study. Nutrients 2018; 10:nu10111787. [PMID: 30453597 PMCID: PMC6265922 DOI: 10.3390/nu10111787] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 11/10/2018] [Accepted: 11/14/2018] [Indexed: 02/07/2023] Open
Abstract
Activation of the intestinal brake by infusing nutrients into the distal small intestine with catheters inhibits food intake and enhances satiety. Encapsulation of macronutrients, which protects against digestion in the proximal gastrointestinal tract, can be a non-invasive alternative to activate this brake. In this study, we investigate the effect of oral ingestion of an encapsulated casein and sucrose mixture (active) targeting the distal small intestine versus a control product designed to be released in the stomach on food intake, satiety, and plasma glucose concentrations. Fifty-nine volunteers received the active and control product on two separate test days. Food intake was determined during an ad libitum meal 90 min after ingestion of the test product. Visual analogue scale scores for satiety and blood samples for glucose analysis were collected at regular intervals. Ingestion of the active product decreased food intake compared to the control product (655 kcal compared with 699 kcal, respectively, p < 0.05). The area under the curve (AUC) for hunger was decreased (p < 0.05) and AUC for satiety was increased (p < 0.01) after ingestion of the active product compared to the control product. Ingestion of an encapsulated protein-carbohydrate mixture resulted in inhibition of food intake compared to a non-encapsulated control product.
Collapse
Affiliation(s)
- Annick M E Alleleyn
- Top Institute of Food and Nutrition, P.O. Box 557, 6700 AN Wageningen, The Netherlands.
- Division of Gastroenterology-Hepatology, Department of Internal Medicine, School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands.
| | - Mark van Avesaat
- Top Institute of Food and Nutrition, P.O. Box 557, 6700 AN Wageningen, The Netherlands.
- Division of Gastroenterology-Hepatology, Department of Internal Medicine, School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands.
| | - Dina Ripken
- Top Institute of Food and Nutrition, P.O. Box 557, 6700 AN Wageningen, The Netherlands.
- The Netherlands Organization for Applied Scientific Research, TNO, P.O. Box 360, 3700 AJ Zeist, The Netherlands.
- Division of Human Nutrition, Wageningen University, P.O. Box 17, 6700 AA Wageningen, The Netherlands.
| | - Sinéad B Bleiel
- AnaBio Technologies LTD., Innovation Centre, Carrigtwohill, T45 RW24 Cork, Ireland.
| | - Daniel Keszthelyi
- Division of Gastroenterology-Hepatology, Department of Internal Medicine, School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands.
| | - Ellen Wilms
- Division of Gastroenterology-Hepatology, Department of Internal Medicine, School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands.
| | - Freddy J Troost
- Top Institute of Food and Nutrition, P.O. Box 557, 6700 AN Wageningen, The Netherlands.
- Division of Gastroenterology-Hepatology, Department of Internal Medicine, School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands.
- Food Innovation and Health, Centre of Healthy Eating and Food Innovation, Maastricht University, 5911 AA Venlo, The Netherlands.
| | - Henk F J Hendriks
- Top Institute of Food and Nutrition, P.O. Box 557, 6700 AN Wageningen, The Netherlands.
| | - Adrian A M Masclee
- Top Institute of Food and Nutrition, P.O. Box 557, 6700 AN Wageningen, The Netherlands.
- Division of Gastroenterology-Hepatology, Department of Internal Medicine, School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands.
| |
Collapse
|
46
|
Feron G. Unstimulated saliva: Background noise in taste molecules. J Texture Stud 2018; 50:6-18. [PMID: 30246386 DOI: 10.1111/jtxs.12369] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 09/10/2018] [Accepted: 09/11/2018] [Indexed: 12/16/2022]
Abstract
Saliva is a highly complex bodily fluid composed of many proteins, peptides, small organic molecules, and ions. Saliva is produced and secreted by the major and minor salivary glands to protect the mouth and to participate in digestion. Generally, a distinction is made between unstimulated saliva that is a result of autonomic stimulation and stimulated saliva that is produced during chewing and taste stimulation. The link between saliva and sensory perception can thus be regarded in two ways: the role of unstimulated saliva as a background taste and the mechanistic role of stimulated saliva during eating. Indeed, unstimulated saliva (and its components) is continuously bathing our oral cavity and as such stimulates our taste receptors, thus playing a role in taste sensitivity. However, the role of unstimulated salivary components in mediating taste has been studied only in very few substances. To explore this question, this review attempts to compare data from the literature on unstimulated salivary composition with those on taste sensitivity. The main conclusion centres around the concept that the gustatory self-adaptation phenomenon may be relevant for only a few salivary compounds. Further studies at the level of the salivary Von Ebner glands and salivary pellicle are necessary before arriving at definitive conclusions on this subject. PRACTICAL APPLICATION: Unstimulated saliva contains taste substances that can influence sensory perception through taste adaptation. However, large inter-individual variability exists in unstimulated salivary composition both qualitatively and quantitatively. These differences may explain the variability in taste perception and thus the food choices and behaviors of an individual. Thus, in the context of providing personalized food and nutrition to the consumer, variability of unstimulated saliva should be considered for specific formulation of food products.
Collapse
Affiliation(s)
- Gilles Feron
- Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, CNRS, INRA, Université Bourgogne Franche-Comté, Dijon, France
| |
Collapse
|
47
|
Expression of the candidate fat taste receptors in human fungiform papillae and the association with fat taste function. Br J Nutr 2018; 120:64-73. [DOI: 10.1017/s0007114518001265] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
AbstractSignificant experimental evidence supports fat as a taste modality; however, the associated peripheral mechanisms are not well established. Several candidate taste receptors have been identified, but their expression pattern and potential functions in human fungiform papillae remain unknown. The aim of this study is to identify the fat taste candidate receptors and ion channels that were expressed in human fungiform taste buds and their association with oral sensory of fatty acids. For the expression analysis, quantitative RT-PCR (qRT-PCR) from RNA extracted from human fungiform papillae samples was used to determine the expression of candidate fatty acid receptors and ion channels. Western blotting analysis was used to confirm the presence of the proteins in fungiform papillae. Immunohistochemistry analysis was used to localise the expressed receptors or ion channels in the taste buds of fungiform papillae. The correlation study was analysed between the expression level of the expressed fat taste receptors or ion channels indicated by qRT-PCR and fat taste threshold, liking of fatty food and fat intake. As a result, qRT-PCR and western blotting indicated that mRNA and protein of CD36, FFAR4, FFAR2, GPR84 and delayed rectifying K+ channels are expressed in human fungiform taste buds. The expression level of CD36 was associated with the liking difference score (R −0·567, β=−0·04, P=0·04) between high-fat and low-fat food and FFAR2 was associated with total fat intake (ρ=−0·535, β=−0·01, P=0·003) and saturated fat intake (ρ=−0·641, β=−0·02, P=0·008).
Collapse
|
48
|
Costanzo A, Nowson C, Orellana L, Bolhuis D, Duesing K, Keast R. Effect of dietary fat intake and genetics on fat taste sensitivity: a co-twin randomized controlled trial. Am J Clin Nutr 2018; 107:683-694. [DOI: 10.1093/ajcn/nqy022] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Accepted: 01/24/2018] [Indexed: 12/21/2022] Open
Affiliation(s)
| | - Caryl Nowson
- Institute of Physical Activity and Nutrition, School of Exercise and Nutrition Sciences
| | - Liliana Orellana
- Biostatistics Unit, Faculty of Health, Deakin University, Geelong, Australia
| | - Dieuwerke Bolhuis
- Food Quality and Design, Wageningen University and Research, Wageningen, the Netherlands
| | | | | |
Collapse
|
49
|
Kershaw JC, Mattes RD. Nutrition and taste and smell dysfunction. World J Otorhinolaryngol Head Neck Surg 2018; 4:3-10. [PMID: 30035256 PMCID: PMC6051307 DOI: 10.1016/j.wjorl.2018.02.006] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 02/27/2018] [Indexed: 12/01/2022] Open
Abstract
Food selection plays a pivotal role in maintaining adequate nutrient intake, thus elucidating drivers of food choice is a meaningful strategy to maintain health and manage disease. Taste and smell are key determinants of food choice and warrant careful consideration. In this review, we first discuss how sensory stimulation influences food selection and metabolism. We then review the evidence regarding the relationship between taste and smell dysfunction and food preferences and selection, with attention given to contexts of certain chronic diseases. We conclude with brief recommendations for the management of chemosensory disorders. While sensory abilities influence food selection, the effect of taste and smell dysfunction on long-term consumption patterns and health status must be considered in light of environment, exposure, and culture.
Collapse
Affiliation(s)
- Jonathan C. Kershaw
- Department of Nutrition Science, 700 W State St, Purdue University, West Lafayette, IN, USA
- Department of Food Science, 745 Agriculture Mall, Purdue University, West Lafayette, IN, USA
| | - Richard D. Mattes
- Department of Nutrition Science, 700 W State St, Purdue University, West Lafayette, IN, USA
- Department of Food Science, 745 Agriculture Mall, Purdue University, West Lafayette, IN, USA
| |
Collapse
|
50
|
Abstract
The gastrointestinal tract represents the largest interface between the human body and the external environment. It must continuously monitor and discriminate between nutrients that need to be assimilated and harmful substances that need to be expelled. The different cells of the gut epithelium are therefore equipped with a subtle chemosensory system that communicates the sensory information to several effector systems involved in the regulation of appetite, immune responses, and gastrointestinal motility. Disturbances or adaptations in the communication of this sensory information may contribute to the development or maintenance of disease. This is a new emerging research field in which perception of taste can be considered as a novel key player participating in the regulation of gut function. Specific diets or agonists that target these chemosensory signaling pathways may be considered as new therapeutic targets to tune adequate physiological processes in the gut in health and disease.
Collapse
Affiliation(s)
- S Steensels
- Translational Research Center for Gastrointestinal Disorders, KU Leuven, 3000 Leuven, Belgium;
| | - I Depoortere
- Translational Research Center for Gastrointestinal Disorders, KU Leuven, 3000 Leuven, Belgium;
| |
Collapse
|