1
|
Dagbasi A, Fuller A, Hanyaloglu AC, Carroll B, McLaughlin J, Frost G, Holliday A. The role of nutrient sensing dysregulation in anorexia of ageing: The little we know and the much we don't. Appetite 2024; 203:107718. [PMID: 39423861 DOI: 10.1016/j.appet.2024.107718] [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: 04/08/2024] [Revised: 08/01/2024] [Accepted: 10/15/2024] [Indexed: 10/21/2024]
Abstract
The age-related decline in appetite and food intake - termed "anorexia of ageing" - is implicated in undernutrition in later life and hence provides a public health challenge for our ageing population. Eating behaviour is controlled, in part, by homeostatic mechanisms which sense nutrient status and provide feedback to appetite control regions of the brain. Such feedback signals, propagated by episodic gut hormones, are dysregulated in some older adults. The secretory responses of appetite-related gut hormones to feeding are amplified, inducing a more anorexigenic signal which is associated with reduced appetite and food intake. Such an augmented response would indicate an increase in gut sensitivity to nutrients. Consequently, this review explores the role of gastrointestinal tract nutrient sensing in age-related appetite dysregulation. We review and synthesise evidence for age-related alterations in nutrient sensing which may explain the observed hormonal dysregulation. Drawing on what is known regarding elements of nutrient sensing pathways in animal models, in other tissues of the body, and in certain models of disease, we identify potential causal mechanisms including alterations in enteroendocrine cell number and distribution, dysregulation of cell signalling pathways, and changes in the gut milieu. From identified gaps in evidence, we highlight interesting and important avenues for future research.
Collapse
Affiliation(s)
- Aygul Dagbasi
- Section of Nutrition, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, 6th Floor Commonwealth Building, Hammersmith Hospital, London, W12 0NN, UK
| | - Amy Fuller
- Research Centre for Health and Life Sciences, Institute of Health and Wellbeing, Faculty of Health and Life Science, Coventry University, Coventry, CV1 5FB, UK
| | - Aylin C Hanyaloglu
- Institute of Reproductive and Developmental Biology (IRDB), Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, W12 0NN, UK
| | - Bernadette Carroll
- School of Biochemistry, University of Bristol, University Walk, Bristol, BS1 8TD, UK
| | - John McLaughlin
- Division of Diabetes, Endocrinology and Gastroenterology, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester and Manchester Academic Health Sciences Centre, Manchester, M13 9PT, UK
| | - Gary Frost
- Section of Nutrition, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, 6th Floor Commonwealth Building, Hammersmith Hospital, London, W12 0NN, UK
| | - Adrian Holliday
- School of Biomedical, Nutritional, and Sport Science, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, NE2 4HH, UK; Human Nutrition and Exercise Research Centre, Population Health Sciences Institute, Newcastle University, Newcastle Upon Tyne, NE2 4HH, UK.
| |
Collapse
|
2
|
Dagbasi A, Byrne C, Blunt D, Serrano-Contreras JI, Becker GF, Blanco JM, Camuzeaux S, Chambers E, Danckert N, Edwards C, Bernal A, Garcia MV, Hanyaloglu A, Holmes E, Ma Y, Marchesi J, Martinez-Gili L, Mendoza L, Tashkova M, Perez-Moral N, Garcia-Perez I, Robles AC, Sands C, Wist J, Murphy KG, Frost G. Diet shapes the metabolite profile in the intact human ileum, which affects PYY release. Sci Transl Med 2024; 16:eadm8132. [PMID: 38896603 DOI: 10.1126/scitranslmed.adm8132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 05/24/2024] [Indexed: 06/21/2024]
Abstract
The human ileum contains a high density of enteroendocrine L-cells, which release the appetite-suppressing hormones glucagon-like peptide-1 (GLP-1) and peptide tyrosine tyrosine (PYY) in response to food intake. Recent evidence highlighted the potential role of food structures in PYY release, but the link between food structures, ileal metabolites, and appetite hormone release remains unclear owing to limited access to intact human ileum. In a randomized crossover trial (ISRCTN11327221; isrctn.com), we investigated the role of human ileum in GLP-1 and PYY release by giving healthy volunteers diets differing in fiber and food structure: high-fiber (intact or disrupted food structures) or low-fiber disrupted food structures. We used nasoenteric tubes to sample chyme from the intact distal ileum lumina of humans in the fasted state and every 60 min for 480 min postprandially. We demonstrate the highly dynamic, wide-ranging molecular environment of the ileum over time, with a substantial decrease in ileum bacterial numbers and bacterial metabolites after food intake. We also show that high-fiber diets, independent of food structure, increased PYY release compared with a low-fiber diet during 0 to 240 min postprandially. High-fiber diets also increased ileal stachyose, and a disrupted high-fiber diet increased certain ileal amino acids. Treatment of human ileal organoids with ileal fluids or an amino acid and stachyose mixture stimulated PYY expression in a similar profile to blood PYY concentrations, confirming the role of ileal metabolites in PYY release. Our study demonstrates the diet-induced changes over time in the metabolite environment of intact human ileum, which play a role in PYY release.
Collapse
Affiliation(s)
- Aygul Dagbasi
- Section of Nutrition, Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, Imperial College London, 6th Floor Commonwealth Building, Hammersmith Hospital, London W12 0NN, UK
| | - Claire Byrne
- Section of Nutrition, Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, Imperial College London, 6th Floor Commonwealth Building, Hammersmith Hospital, London W12 0NN, UK
| | - Dominic Blunt
- Department of Imaging, Charing Cross Hospital, Imperial NHS Trust, London W6 8RF, UK
| | - Jose Ivan Serrano-Contreras
- Section of Nutrition, Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, Imperial College London, 6th Floor Commonwealth Building, Hammersmith Hospital, London W12 0NN, UK
| | - Georgia Franco Becker
- Section of Nutrition, Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, Imperial College London, 6th Floor Commonwealth Building, Hammersmith Hospital, London W12 0NN, UK
| | - Jesus Miguens Blanco
- Division of Digestive Diseases, Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, 6th Floor Commonwealth Building, Hammersmith Hospital, London W12 0NN, UK
| | - Stephane Camuzeaux
- National Phenome Centre, Imperial College London, Hammersmith Hospital Campus, London W12 0HS, UK
| | - Edward Chambers
- Section of Nutrition, Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, Imperial College London, 6th Floor Commonwealth Building, Hammersmith Hospital, London W12 0NN, UK
| | - Nathan Danckert
- Division of Digestive Diseases, Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, 6th Floor Commonwealth Building, Hammersmith Hospital, London W12 0NN, UK
| | | | - Andres Bernal
- Centre for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Murdoch, WA 6150, Australia
| | - Maria Valdivia Garcia
- Section of Nutrition, Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, Imperial College London, 6th Floor Commonwealth Building, Hammersmith Hospital, London W12 0NN, UK
| | - Aylin Hanyaloglu
- Institute of Reproductive and Development Biology (IRDB), Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, Hammersmith Hospital, London W12 0NN, UK
| | - Elaine Holmes
- Section of Nutrition, Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, Imperial College London, 6th Floor Commonwealth Building, Hammersmith Hospital, London W12 0NN, UK
- Centre for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Murdoch, WA 6150, Australia
| | - Yue Ma
- Section of Nutrition, Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, Imperial College London, 6th Floor Commonwealth Building, Hammersmith Hospital, London W12 0NN, UK
| | - Julian Marchesi
- Division of Digestive Diseases, Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, 6th Floor Commonwealth Building, Hammersmith Hospital, London W12 0NN, UK
| | - Laura Martinez-Gili
- Division of Digestive Diseases, Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, 6th Floor Commonwealth Building, Hammersmith Hospital, London W12 0NN, UK
- Section of Bioinformatics, Division of Systems Medicine, Department of Metabolism, Digestion, and Reproduction, Imperial College London, London W12 0NN, UK
| | - Lilian Mendoza
- Section of Nutrition, Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, Imperial College London, 6th Floor Commonwealth Building, Hammersmith Hospital, London W12 0NN, UK
| | - Martina Tashkova
- Section of Nutrition, Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, Imperial College London, 6th Floor Commonwealth Building, Hammersmith Hospital, London W12 0NN, UK
| | | | - Isabel Garcia-Perez
- Section of Nutrition, Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, Imperial College London, 6th Floor Commonwealth Building, Hammersmith Hospital, London W12 0NN, UK
| | - Andres Castillo Robles
- Centre for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Murdoch, WA 6150, Australia
| | - Caroline Sands
- National Phenome Centre, Imperial College London, Hammersmith Hospital Campus, London W12 0HS, UK
| | - Julien Wist
- Section of Nutrition, Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, Imperial College London, 6th Floor Commonwealth Building, Hammersmith Hospital, London W12 0NN, UK
- Centre for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Murdoch, WA 6150, Australia
- Chemistry Department, Universidad del Valle, Cali 76001, Colombia
| | - Kevin G Murphy
- Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, 6th Floor Commonwealth Building, Hammersmith Hospital, London W12 0NN, UK
| | - Gary Frost
- Section of Nutrition, Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, Imperial College London, 6th Floor Commonwealth Building, Hammersmith Hospital, London W12 0NN, UK
| |
Collapse
|
3
|
Li RJW, Barros DR, Kuah R, Lim YM, Gao A, Beaudry JL, Zhang SY, Lam TKT. Small intestinal CaSR-dependent and CaSR-independent protein sensing regulates feeding and glucose tolerance in rats. Nat Metab 2024; 6:39-49. [PMID: 38167726 DOI: 10.1038/s42255-023-00942-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 11/08/2023] [Indexed: 01/05/2024]
Abstract
Proteins activate small intestinal calcium sensing receptor (CaSR) and/or peptide transporter 1 (PepT1) to increase hormone secretion1-8, but the effect of small intestinal protein sensing and the mechanistic potential of CaSR and/or PepT1 in feeding and glucose regulation remain inconclusive. Here we show that, in male rats, CaSR in the upper small intestine is required for casein infusion to increase glucose tolerance and GLP1 and GIP secretion, which was also dependent on PepT1 (ref. 9). PepT1, but not CaSR, is required for casein infusion to lower feeding. Upper small intestine casein sensing fails to regulate feeding, but not glucose tolerance, in high-fat-fed rats with decreased PepT1 but increased CaSR expression. In the ileum, a CaSR-dependent but PepT1-independent pathway is required for casein infusion to lower feeding and increase glucose tolerance in chow-fed rats, in parallel with increased PYY and GLP1 release, respectively. High fat decreases ileal CaSR expression and disrupts casein sensing on feeding but not on glucose control, suggesting an ileal CaSR-independent, glucose-regulatory pathway. In summary, we discover small intestinal CaSR- and PepT1-dependent and -independent protein sensing mechanisms that regulate gut hormone release, feeding and glucose tolerance. Our findings highlight the potential of targeting small intestinal CaSR and/or PepT1 to regulate feeding and glucose tolerance.
Collapse
Affiliation(s)
- Rosa J W Li
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, UHN, Toronto, Ontario, Canada
| | - Daniel R Barros
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, UHN, Toronto, Ontario, Canada
| | - Rachel Kuah
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, UHN, Toronto, Ontario, Canada
| | - Yu-Mi Lim
- Toronto General Hospital Research Institute, UHN, Toronto, Ontario, Canada
- Medical Research Institute, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Anna Gao
- Toronto General Hospital Research Institute, UHN, Toronto, Ontario, Canada
| | - Jacqueline L Beaudry
- Department of Nutritional Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Song-Yang Zhang
- Toronto General Hospital Research Institute, UHN, Toronto, Ontario, Canada
| | - Tony K T Lam
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada.
- Toronto General Hospital Research Institute, UHN, Toronto, Ontario, Canada.
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada.
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada.
- Banting and Best Diabetes Centre, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
4
|
Rezaie P, Bitarafan V, Rose BD, Lange K, Mohammadpour Z, Rehfeld JF, Horowitz M, Feinle-Bisset C. Effects of Quinine on the Glycaemic Response to, and Gastric Emptying of, a Mixed-Nutrient Drink in Females and Males. Nutrients 2023; 15:3584. [PMID: 37630774 PMCID: PMC10459881 DOI: 10.3390/nu15163584] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 08/09/2023] [Accepted: 08/14/2023] [Indexed: 08/27/2023] Open
Abstract
Intraduodenal quinine, in the dose of 600 mg, stimulates glucagon-like peptide-1 (GLP-1), cholecystokinin and insulin; slows gastric emptying (GE); and lowers post-meal glucose in men. Oral sensitivity to bitter substances may be greater in women than men. We, accordingly, evaluated the dose-related effects of quinine on GE, and the glycaemic responses to, a mixed-nutrient drink in females, and compared the effects of the higher dose with those in males. A total of 13 female and 13 male healthy volunteers received quinine-hydrochloride (600 mg ('QHCl-600') or 300 mg ('QHCl-300', females only) or control ('C'), intraduodenally (10 mL bolus) 30 min before a drink (500 kcal, 74 g carbohydrates). Plasma glucose, insulin, C-peptide, GLP-1, glucose-dependent insulinotropic polypeptide (GIP) and cholecystokinin were measured at baseline, for 30 min after quinine alone, and then for 2 h post-drink. GE was measured by 13C-acetate breath-test. QHCl-600 alone stimulated insulin, C-peptide and GLP-1 secretion compared to C. Post-drink, QHCl-600 reduced plasma glucose, stimulated C-peptide and GLP-1, and increased the C-peptide/glucose ratio and oral disposition index, while cholecystokinin and GIP were less, in females and males. QHCl-600 also slowed GE compared to C in males and compared to QHCl-300 in females (p < 0.05). QHCl-300 reduced post-meal glucose concentrations and increased the C-peptide/glucose ratio, compared to C (p < 0.05). Magnitudes of glucose lowering and increase in C-peptide/glucose ratio by QHCl-600 were greater in females than males (p < 0.05). We conclude that quinine modulates glucoregulatory functions, associated with glucose lowering in healthy males and females. However, glucose lowering appears to be greater in females than males, without apparent differential effects on GI functions.
Collapse
Affiliation(s)
- Peyman Rezaie
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia
- Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, SA 5005, Australia
| | - Vida Bitarafan
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia
- Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, SA 5005, Australia
| | - Braden David Rose
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia
- Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, SA 5005, Australia
- Nutrition, Diabetes and Gut Health, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, SA 5001, Australia
| | - Kylie Lange
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia
- Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, SA 5005, Australia
| | - Zinat Mohammadpour
- Nutrition, Diabetes and Gut Health, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, SA 5001, Australia
- School of Biomedicine, University of Adelaide, Adelaide, SA 5005, Australia
| | - Jens Frederik Rehfeld
- Department of Clinical Biochemistry, Rigshospitalet, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Michael Horowitz
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia
- 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
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia
- Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, SA 5005, Australia
| |
Collapse
|
5
|
PH van Trijp M, Wilms E, Ríos-Morales M, Masclee AA, Brummer RJ, Witteman BJ, Troost FJ, Hooiveld GJ. Using naso- and oro-intestinal catheters in physiological research for intestinal delivery and sampling in vivo: practical and technical aspects to be considered. Am J Clin Nutr 2021; 114:843-861. [PMID: 34036315 PMCID: PMC8408849 DOI: 10.1093/ajcn/nqab149] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 04/09/2021] [Indexed: 01/19/2023] Open
Abstract
Intestinal catheters have been used for decades in human nutrition, physiology, pharmacokinetics, and gut microbiome research, facilitating the delivery of compounds directly into the intestinal lumen or the aspiration of intestinal fluids in human subjects. Such research provides insights about (local) dynamic metabolic and other intestinal luminal processes, but working with catheters might pose challenges to biomedical researchers and clinicians. Here, we provide an overview of practical and technical aspects of applying naso- and oro-intestinal catheters for delivery of compounds and sampling luminal fluids from the jejunum, ileum, and colon in vivo. The recent literature was extensively reviewed, and combined with experiences and insights we gained through our own clinical trials. We included 60 studies that involved a total of 720 healthy subjects and 42 patients. Most of the studies investigated multiple intestinal regions (24 studies), followed by studies investigating only the jejunum (21 studies), ileum (13 studies), or colon (2 studies). The ileum and colon used to be relatively inaccessible regions in vivo. Custom-made state-of-the-art catheters are available with numerous options for the design, such as multiple lumina, side holes, and inflatable balloons for catheter progression or isolation of intestinal segments. These allow for multiple controlled sampling and compound delivery options in different intestinal regions. Intestinal catheters were often used for delivery (23 studies), sampling (10 studies), or both (27 studies). Sampling speed decreased with increasing distance from the sampling syringe to the specific intestinal segment (i.e., speed highest in duodenum, lowest in ileum/colon). No serious adverse events were reported in the literature, and a dropout rate of around 10% was found for these types of studies. This review is highly relevant for researchers who are active in various research areas and want to expand their research with the use of intestinal catheters in humans in vivo.
Collapse
Affiliation(s)
- Mara PH van Trijp
- Division of Human Nutrition and Health, Wageningen University, Wageningen, The Netherlands
| | - Ellen Wilms
- Division Gastroenterology-Hepatology, Department of Internal Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
| | - Melany Ríos-Morales
- Laboratory of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Ad Am Masclee
- Division Gastroenterology-Hepatology, Department of Internal Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
| | - Robert Jan Brummer
- Nutrition-Gut-Brain Interactions Research Centre, School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Ben Jm Witteman
- Division of Human Nutrition and Health, Wageningen University, Wageningen, The Netherlands,Hospital Gelderse Vallei, Department of Gastroenterology and Hepatology, Ede, The Netherlands
| | - Freddy J Troost
- Division Gastroenterology-Hepatology, Department of Internal Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands,Food Innovation and Health, Centre for Healthy Eating and Food Innovation, Maastricht University, Maastricht, The Netherlands
| | | |
Collapse
|
6
|
Wilbrink J, Masclee G, Klaassen T, van Avesaat M, Keszthelyi D, Masclee A. Review on the Regional Effects of Gastrointestinal Luminal Stimulation on Appetite and Energy Intake: (Pre)clinical Observations. Nutrients 2021; 13:nu13051601. [PMID: 34064724 PMCID: PMC8151500 DOI: 10.3390/nu13051601] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 04/22/2021] [Accepted: 05/05/2021] [Indexed: 02/06/2023] Open
Abstract
Macronutrients in the gastrointestinal (GI) lumen are able to activate “intestinal brakes”, feedback mechanisms on proximal GI motility and secretion including appetite and energy intake. In this review, we provide a detailed overview of the current evidence with respect to four questions: (1) are regional differences (duodenum, jejunum, ileum) present in the intestinal luminal nutrient modulation of appetite and energy intake? (2) is this “intestinal brake” effect macronutrient specific? (3) is this “intestinal brake” effect maintained during repetitive activation? (4) can the “intestinal brake” effect be activated via non-caloric tastants? Recent evidence indicates that: (1) regional differences exist in the intestinal modulation of appetite and energy intake with a proximal to distal gradient for inhibition of energy intake: ileum and jejunum > duodenum at low but not at high caloric infusion rates. (2) the “intestinal brake” effect on appetite and energy appears not to be macronutrient specific. At equi-caloric amounts, the inhibition on energy intake and appetite is in the same range for fat, protein and carbohydrate. (3) data on repetitive ileal brake activation are scarce because of the need for prolonged intestinal intubation. During repetitive activation of the ileal brake for up to 4 days, no adaptation was observed but overall the inhibitory effect on energy intake was small. (4) the concept of influencing energy intake by intra-intestinal delivery of non-caloric tastants is intriguing. Among tastants, the bitter compounds appear to be more effective in influencing energy intake. Energy intake decreases modestly after post-oral delivery of bitter tastants or a combination of tastants (bitter, sweet and umami). Intestinal brake activation provides an interesting concept for preventive and therapeutic approaches in weight management strategies.
Collapse
Affiliation(s)
- Jennifer Wilbrink
- Division of Gastroenterology-Hepatology, Maastricht University Medical Center, 6229 HX Maastricht, The Netherlands; (J.W.); (G.M.); (T.K.); (M.v.A.); (D.K.)
| | - Gwen Masclee
- Division of Gastroenterology-Hepatology, Maastricht University Medical Center, 6229 HX Maastricht, The Netherlands; (J.W.); (G.M.); (T.K.); (M.v.A.); (D.K.)
| | - Tim Klaassen
- Division of Gastroenterology-Hepatology, Maastricht University Medical Center, 6229 HX Maastricht, The Netherlands; (J.W.); (G.M.); (T.K.); (M.v.A.); (D.K.)
| | - Mark van Avesaat
- Division of Gastroenterology-Hepatology, Maastricht University Medical Center, 6229 HX Maastricht, The Netherlands; (J.W.); (G.M.); (T.K.); (M.v.A.); (D.K.)
| | - Daniel Keszthelyi
- Division of Gastroenterology-Hepatology, Maastricht University Medical Center, 6229 HX Maastricht, The Netherlands; (J.W.); (G.M.); (T.K.); (M.v.A.); (D.K.)
- NUTRIM School of Nutrition and Translational Research in Metabolism, 6229 ER Maastricht, The Netherlands
| | - Adrian Masclee
- Division of Gastroenterology-Hepatology, Maastricht University Medical Center, 6229 HX Maastricht, The Netherlands; (J.W.); (G.M.); (T.K.); (M.v.A.); (D.K.)
- NUTRIM School of Nutrition and Translational Research in Metabolism, 6229 ER Maastricht, The Netherlands
- Correspondence: ; Tel.: +31-43-3875021
| |
Collapse
|
7
|
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: 106] [Impact Index Per Article: 26.5] [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
|
8
|
Wackers G, Putzeys T, Peeters M, Van de Cauter L, Cornelis P, Wübbenhorst M, Tack J, Troost F, Verhaert N, Doll T, Wagner P. Towards a catheter-based impedimetric sensor for the assessment of intestinal histamine levels in IBS patients. Biosens Bioelectron 2020; 158:112152. [PMID: 32275205 DOI: 10.1016/j.bios.2020.112152] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 03/09/2020] [Accepted: 03/12/2020] [Indexed: 12/18/2022]
Abstract
In this work, we report on the development of a catheter-based sensor designed for measuring the concentration of histamine in the human duodenum. Certain gut disorders, such as the irritable bowel syndrome (IBS), are associated with elevated levels of intestinal histamine due to chronic immune activation. As it is still impossible to determine histamine concentrations in vivo, a nasointestinal catheter with histamine-sensing capabilities has the potential to become a valuable diagnostic instrument. Regarding the sensing principle, we selected impedance spectroscopy using voltages that are compatible with intra-body applications with molecularly imprinted polymers (MIPs) as recognition elements. MIPs are synthetic receptors that offer the advantages of robustness, high specificity and selectivity for histamine as a target. In this specific case, the MIPs were synthesized from acryclic acid monomers, which guarantees a uniform binding capacity within the pH range of intestinal fluid. We have validated the catheter sensor on human intestinal liquids spiked with histamine in a testing setup that mimics the environment inside the duodenum. The dose-response curves show an analytical range between 5 and 200 nM of histamine, corresponding to physiologically normal conditions while higher concentrations correlate with disease. The key output signal of the sensor is the resistive component of the MIP-functionalized titanium electrodes as derived from the equivalent-circuit modelling of full-range impedance spectra. Future applications could be catheters tailored to cardiovascular, urological, gastrointestinal, and neurovascular applications. This, in combination with the versatility of the MIPs, will make this sensor platform a versatile diagnostic tool.
Collapse
Affiliation(s)
- Gideon Wackers
- KU Leuven, Laboratory for Soft Matter and Biophysics, Celestijnenlaan 200 D, B-3001, Leuven, Belgium.
| | - Tristan Putzeys
- KU Leuven, Laboratory for Soft Matter and Biophysics, Celestijnenlaan 200 D, B-3001, Leuven, Belgium; KU Leuven, Research Group Experimental Oto-rhino-laryngology, O&N II, Herestraat 49, B-3001, Leuven, Belgium
| | - Marloes Peeters
- Newcastle University, School of Engineering, Newcastle NE1 7RU, United Kingdom
| | - Lori Van de Cauter
- KU Leuven, Laboratory for Soft Matter and Biophysics, Celestijnenlaan 200 D, B-3001, Leuven, Belgium
| | - Peter Cornelis
- KU Leuven, Laboratory for Soft Matter and Biophysics, Celestijnenlaan 200 D, B-3001, Leuven, Belgium
| | - Michael Wübbenhorst
- KU Leuven, Laboratory for Soft Matter and Biophysics, Celestijnenlaan 200 D, B-3001, Leuven, Belgium
| | - Jan Tack
- KU Leuven, Translational Research in Gastrointestinal Disorders, O&N I, Herestraat 49, B-3001, Leuven, Belgium
| | - Freddy Troost
- Food Innovation and Health, Centre for Healthy Eating and Food Innovation, Maastricht University, NUTRIM School of Nutrition and Translational Research in Metabolism, Universiteitssingel 40, NL-6229 ER, Maastricht, the Netherlands
| | - Nicolas Verhaert
- KU Leuven, Research Group Experimental Oto-rhino-laryngology, O&N II, Herestraat 49, B-3001, Leuven, Belgium
| | - Theodor Doll
- Hannover Medical School, Institute of AudioNeuroTechnology VIANNA, Stadtfelddamm 34, D-30625, Hannover, Germany
| | - Patrick Wagner
- KU Leuven, Laboratory for Soft Matter and Biophysics, Celestijnenlaan 200 D, B-3001, Leuven, Belgium
| |
Collapse
|
9
|
How Satiating Are the 'Satiety' Peptides: A Problem of Pharmacology versus Physiology in the Development of Novel Foods for Regulation of Food Intake. Nutrients 2019; 11:nu11071517. [PMID: 31277416 PMCID: PMC6682889 DOI: 10.3390/nu11071517] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 06/27/2019] [Accepted: 06/28/2019] [Indexed: 12/15/2022] Open
Abstract
Developing novel foods to suppress energy intake and promote negative energy balance and weight loss has been a long-term but commonly unsuccessful challenge. Targeting regulation of appetite is of interest to public health researchers and industry in the quest to develop ‘functional’ foods, but poor understanding of the underpinning mechanisms regulating food intake has hampered progress. The gastrointestinal (GI) or ‘satiety’ peptides including cholecystokinin (CCK), glucagon-like peptide 1 (GLP-1) and peptide YY (PYY) secreted following a meal, have long been purported as predictive biomarkers of appetite response, including food intake. Whilst peptide infusion drives a clear change in hunger/fullness and eating behaviour, inducing GI-peptide secretion through diet may not, possibly due to modest effects of single meals on peptide levels. We conducted a review of 70 dietary preload (DIET) and peptide infusion (INFUSION) studies in lean healthy adults that reported outcomes of CCK, GLP-1 and PYY. DIET studies were acute preload interventions. INFUSION studies showed that minimum increase required to suppress ad libitum energy intake for CCK, GLP-1 and PYY was 3.6-, 4.0- and 3.1-fold, respectively, achieved through DIET in only 29%, 0% and 8% of interventions. Whether circulating ‘thresholds’ of peptide concentration likely required for behavioural change can be achieved through diet is questionable. As yet, no individual or group of peptides can be measured in blood to reliably predict feelings of hunger and food intake. Developing foods that successfully target enhanced secretion of GI-origin ‘satiety’ peptides for weight loss remains a significant challenge.
Collapse
|
10
|
Klaassen T, Alleleyn AME, van Avesaat M, Troost FJ, Keszthelyi D, Masclee AAM. Intraintestinal Delivery of Tastants Using a Naso-Duodenal-Ileal Catheter Does Not Influence Food Intake or Satiety. Nutrients 2019; 11:nu11020472. [PMID: 30813412 PMCID: PMC6412712 DOI: 10.3390/nu11020472] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 02/20/2019] [Accepted: 02/20/2019] [Indexed: 12/21/2022] Open
Abstract
Intraduodenal activity of taste receptors reduces food intake. Taste receptors are expressed throughout the entire gastrointestinal tract. Currently, there are no data available on the effects of distal taste receptor activation. In this study, we investigate the effect of intraduodenal and/or intraileal activation of taste receptors on food intake and satiety. In a single-blind randomized crossover trial, fourteen participants were intubated with a naso-duodenal-ileal catheter and received four infusion regimens: duodenal placebo and ileal placebo (DPIP), duodenal tastants and ileal placebo (DTIP), duodenal placebo and ileal tastants (DPIT), duodenal tastants and ileal tastants (DTIT). Fifteen minutes after cessation of infusion, subjects received an ad libitum meal to measure food intake. Visual analog scale scores for satiety feelings were collected at regular intervals. No differences in food intake were observed between the various interventions (DPIP: 786.6 ± 79.2 Kcal, DTIP: 803.3 ± 69.0 Kcal, DPIT: 814.7 ± 77.3 Kcal, DTIT: 834.8 ± 59.2 Kcal, p = 0.59). No differences in satiety feelings were observed. Intestinal infusion of tastants using a naso-duodenal-ileal catheter did not influence food intake or satiety feelings. Possibly, the burden of the four-day naso-duodenal-ileal intubation masked a small effect that tastants might have on food intake and satiety.
Collapse
Affiliation(s)
- Tim Klaassen
- 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, Center for Healthy Eating and Food Innovation, Maastricht University, 5911 AA Venlo, The Netherlands.
| | - Annick M E Alleleyn
- 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
- 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
- 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, Center for Healthy Eating and Food Innovation, Maastricht University, 5911 AA Venlo, The Netherlands.
| | - 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.
| | - Adrian A M Masclee
- 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
|
11
|
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
|