1
|
Zhao X, Qiu Y, Liang L, Fu X. Interkingdom signaling between gastrointestinal hormones and the gut microbiome. Gut Microbes 2025; 17:2456592. [PMID: 39851261 PMCID: PMC11776477 DOI: 10.1080/19490976.2025.2456592] [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: 05/12/2024] [Revised: 08/12/2024] [Accepted: 01/02/2025] [Indexed: 01/26/2025] Open
Abstract
The interplay between the gut microbiota and gastrointestinal hormones plays a pivotal role in the health of the host and the development of diseases. As a vital component of the intestinal microecosystem, the gut microbiota influences the synthesis and release of many gastrointestinal hormones through mechanisms such as modulating the intestinal environment, producing metabolites, impacting mucosal barriers, generating immune and inflammatory responses, and releasing neurotransmitters. Conversely, gastrointestinal hormones exert feedback regulation on the gut microbiota by modulating the intestinal environment, nutrient absorption and utilization, and the bacterial biological behavior and composition. The distributions of the gut microbiota and gastrointestinal hormones are anatomically intertwined, and close interactions between the gut microbiota and gastrointestinal hormones are crucial for maintaining gastrointestinal homeostasis. Interventions leveraging the interplay between the gut microbiota and gastrointestinal hormones have been employed in the clinical management of metabolic diseases and inflammatory bowel diseases, such as bariatric surgery and fecal microbiota transplantation, offering promising targets for the treatment of dysbiosis-related diseases.
Collapse
Affiliation(s)
- Xinyu Zhao
- Department of Gastroenterology, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Ye Qiu
- Department of Gastroenterology, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Lanfan Liang
- Department of Gastroenterology, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Xiangsheng Fu
- Department of Gastroenterology, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| |
Collapse
|
2
|
Tarasiuk-Zawadzka A, Fichna J. Interaction between nutritional factors and the enteric nervous system in inflammatory bowel diseases. J Nutr Biochem 2025:109959. [PMID: 40354831 DOI: 10.1016/j.jnutbio.2025.109959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 01/30/2025] [Accepted: 05/09/2025] [Indexed: 05/14/2025]
Abstract
The enteric nervous system (ENS) is a highly conserved, yet complicated network of neurons and glial cells located throughout the gut wall that controls digestive processes and gastrointestinal (GI) homeostasis. The intestinal epithelium, the immune system, and the gut microbiota are just a few examples of the cellular networks that the ENS interacts with on a variety of levels to maintain GI function. The presence or absence of nutrients in the intestinal lumen may cause short- and/or long-term changes in neurotransmitter expression, excitability, and neuronal survival, which ultimately affect gut motility, secretion, and permeability. Hence, the ENS should be identified as a key factor in initiating coordinated responses to nutrients. In this review we summarize current knowledge on nutrient-dependent ENS activity and how ENS secondary to nutrition may affect likelihood of developing inflammatory bowel disease. Our findings highlight that nutrients interact with enteroendocrine cells in the gut, triggering hormone secretion that plays a crucial role in signaling food-related information to the brain and regulating metabolic processes such as feeding behavior, insulin secretion, and energy balance; however, the complex interactions between nutrients, the ENS, and the immune system require further research to understand their contributions to GI disorders and potential therapeutic applications in treating obesity and metabolic diseases. Lay Summary: The enteric nervous system (ENS) controls digestion and interacts with nutrients in the gut to regulate processes like gut movement and hormone release, affecting metabolism and overall gut health. This review highlights the need for further research on how nutrient-ENS interactions contribute to conditions like inflammatory bowel disease, obesity, and metabolic disorders.
Collapse
Affiliation(s)
| | - Jakub Fichna
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland
| |
Collapse
|
3
|
Hoornenborg CW, Somogyi E, Bruggink JE, Boyle CN, Lutz TA, Emous M, van Beek AP, van Dijk G. A Western-Style Diet Influences Ingestive Behavior and Glycemic Control in a Rat Model of Roux-en-Y Gastric Bypass Surgery. J Clin Med 2025; 14:2642. [PMID: 40283472 PMCID: PMC12027894 DOI: 10.3390/jcm14082642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 04/04/2025] [Accepted: 04/08/2025] [Indexed: 04/29/2025] Open
Abstract
Background: Roux-en-Y gastric bypass (RYGB) surgery results in weight reduction and decreased energy intake and can ameliorate type 2 diabetes. These beneficial effects are usually attributed to changes in hunger and satiety and relatively rapid improvements in glycemic control, but these effects may depend on dietary adherence. The aim of this study is to investigate the relatively early effects of RYGB surgery on weight reduction (by focusing on eating patterns) and glycemic control in rats subjected to a healthy maintenance diet or an unhealthy Western-style diet. Methods: Rats were fed a high-fat diet with added sucrose (HF/S) or a low-fat (LF) diet. Body weight, high-resolution tracking of meal-related parameters, and glucose regulation after overnight fasting and during a mixed meal tolerance test (MMTT; 2 mL sweet/condensed milk) were measured before and after RYGB (RYGB+) or sham surgery (RYGB-). Results: HF/S feeding led to an increased body weight just before RYGB surgery, but it also caused enhanced weight loss following RYGB, which led to similar body weights in the HF/S and LF diet groups twenty-four days post-operatively. RYGB surgery and diet dependently and independently influenced meal-related parameter outcomes, where both RYGB+ and HF/S feeding resulted in shorter meal duration (p < 0.01), higher ingestion rates (p < 0.001), and increased satiety ratio (p < 0.05), especially in the HF/S diet group subjected to RYGB. While RYGB surgery generally improved baseline glycemic parameters including HOMA-IR (p < 0.01), it often interacted with diet to affect MMTT-induced hyperglycemia (p < 0.05), beta-cell sensitivity (p < 0.01), and the insulinogenic index (p < 0.01), with the LF rats overall maintaining better glycemic control than the HF/S-fed rats. Conclusions: This study shows the importance of controlling diet after RYGB surgery, as diet type significantly influences ingestive behavior, post-prandial glucose regulation, beta-cell sensitivity, and glucose tolerance after RYGB.
Collapse
Affiliation(s)
- C. Warner Hoornenborg
- Department of Behavioral Neuroscience, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, 9747 AG Groningen, The Netherlands; (C.W.H.); (E.S.); (J.E.B.)
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands;
| | - Edit Somogyi
- Department of Behavioral Neuroscience, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, 9747 AG Groningen, The Netherlands; (C.W.H.); (E.S.); (J.E.B.)
- School of Kinesiology, Faculty of Health Sciences, University of Western Ontario, London, ON N6G 2V4, Canada
| | - Jan E. Bruggink
- Department of Behavioral Neuroscience, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, 9747 AG Groningen, The Netherlands; (C.W.H.); (E.S.); (J.E.B.)
| | - Christina N. Boyle
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, CH-8057 Zurich, Switzerland; (C.N.B.); (T.A.L.)
| | - Thomas A. Lutz
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, CH-8057 Zurich, Switzerland; (C.N.B.); (T.A.L.)
| | - Marloes Emous
- Department of Bariatric and Metabolic Surgery, Medical Center Leeuwarden, 8934 AD Leeuwarden, The Netherlands;
| | - André P. van Beek
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands;
| | - Gertjan van Dijk
- Department of Behavioral Neuroscience, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, 9747 AG Groningen, The Netherlands; (C.W.H.); (E.S.); (J.E.B.)
| |
Collapse
|
4
|
Ježek P. Physiological Fatty Acid-Stimulated Insulin Secretion and Redox Signaling Versus Lipotoxicity. Antioxid Redox Signal 2025; 42:566-622. [PMID: 39834189 DOI: 10.1089/ars.2024.0799] [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] [Indexed: 01/22/2025]
Abstract
Significance: Type 2 diabetes as a world-wide epidemic is characterized by the insulin resistance concomitant to a gradual impairment of β-cell mass and function (prominently declining insulin secretion) with dysregulated fatty acids (FAs) and lipids, all involved in multiple pathological development. Recent Advances: Recently, redox signaling was recognized to be essential for insulin secretion stimulated with glucose (GSIS), branched-chain keto-acids, and FAs. FA-stimulated insulin secretion (FASIS) is a normal physiological event upon postprandial incoming chylomicrons. This contrasts with the frequent lipotoxicity observed in rodents. Critical Issues: Overfeeding causes FASIS to overlap with GSIS providing repeating hyperinsulinemia, initiates prediabetic states by lipotoxic effects and low-grade inflammation. In contrast the protective effects of lipid droplets in human β-cells counteract excessive lipids. Insulin by FASIS allows FATP1 recruitment into adipocyte plasma membranes when postprandial chylomicrons come late at already low glycemia. Future Directions: Impaired states of pancreatic β-cells and peripheral organs at prediabetes and type 2 diabetes should be revealed, including the inter-organ crosstalk by extracellular vesicles. Details of FA/lipid molecular physiology are yet to be uncovered, such as complex phenomena of FA uptake into cells, postabsorptive inactivity of G-protein-coupled receptor 40, carnitine carrier substrate specificity, the role of carnitine-O-acetyltransferase in β-cells, and lipid droplet interactions with mitochondria. Antioxid. Redox Signal. 42, 566-622.
Collapse
Affiliation(s)
- Petr Ježek
- Department of Mitochondrial Physiology, No.75, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
5
|
Bitarafan V, Anjom-Shoae J, Rezaie P, Fitzgerald PCE, Lange K, Horowitz M, Feinle-Bisset C. Dose-related effects of intraduodenal quinine on plasma glucose, glucoregulatory hormones and gastric emptying of a nutrient drink, and energy intake, in men with type 2 diabetes: a double-blind, randomised, crossover study. Diabetologia 2025; 68:727-738. [PMID: 39690248 DOI: 10.1007/s00125-024-06344-9] [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: 09/09/2024] [Accepted: 11/11/2024] [Indexed: 12/19/2024]
Abstract
AIMS/HYPOTHESIS Quinine, when administered intraduodenally to activate bitter-taste receptors, in a dose of 600 mg, stimulates glucagon-like peptide-1 (GLP-1) and insulin, slows gastric emptying and lowers postprandial glucose in healthy people, with consequent implications for the management of type 2 diabetes; the effect of quinine on energy intake is uncertain. We have investigated the dose-related effects of quinine on postprandial blood glucose levels and energy intake in people with type 2 diabetes. METHODS Male participants with type 2 diabetes (age: 68±5 years; HbA1c: 49.0±5.0 mmol/mol [6.7±0.4%], BMI: 30±1 kg/m2) received in two study parts (A and B, n=12 each), on three separate occasions each, in randomised, crossover fashion, control, or 300 mg (QHCl-300) or 600 mg (QHCl-600) quinine hydrochloride, intraduodenally 30 min before a nutrient drink (2092 kJ, 74 g carbohydrate) (part A) or a standardised buffet-lunch (part B). Both the participants and investigators performing the study procedures were blinded to the treatments. In part A, plasma glucose, GLP-1, C-peptide and glucagon were measured at baseline, for 30 min after quinine alone and for 3 h post drink. Gastric emptying of the drink was measured with a 13C-acetate breath test. In part B, energy intake from the buffet-lunch was quantified. RESULTS Quinine alone had no effect. Post drink, both quinine doses reduced peak plasma glucose markedly (QHCl-600 by 2.8±0.6 mmol/l) and slowed gastric emptying (all p<0.05; n=12, except for gastric emptying, n=11). QHCl-600, but not QHCl-300, stimulated plasma GLP-1 and C-peptide modestly (both p<0.05). Quinine did not affect energy intake. CONCLUSIONS/INTERPRETATION In type 2 diabetes, acute intraduodenal administration of quinine markedly reduces the plasma glucose response to oral carbohydrate, but does not affect energy intake. These findings support the potential use of quinine to reduce postprandial blood glucose levels in type 2 diabetes. TRIAL REGISTRATION anzctr.org.au ACTRN12620000972921/ACTRN12621000218897 FUNDING: The study was funded by a Diabetes Australia Research Project Grant.
Collapse
Affiliation(s)
- Vida Bitarafan
- Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
- Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, SA, Australia
| | - Javad Anjom-Shoae
- Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
- Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, SA, Australia
| | - Peyman Rezaie
- Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
- Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, SA, Australia
- Canberra Hospital, Canberra, ACT, Australia
| | - Penelope C E Fitzgerald
- Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
- Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, SA, Australia
| | - Kylie Lange
- Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
- Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, SA, Australia
| | - Michael Horowitz
- Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
- Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, SA, Australia
- Endocrine and Metabolic Unit, Royal Adelaide Hospital, Adelaide, SA, Australia
| | - Christine Feinle-Bisset
- Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia.
- Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, SA, Australia.
| |
Collapse
|
6
|
Anjom-Shoae J, Hajishafiee M, Fitzgerald PC, Coleman R, Martin AM, Poppitt SD, Lee M, Higgs S, Rehfeld JF, Holst JJ, Veedfald S, Horowitz M, Feinle-Bisset C. Acute decrease in the plasma tryptophan-to-large-neutral-amino-acids ratio attenuates the effects of L-tryptophan on gut hormones and energy intake in healthy males: a randomized, cross-over, exploratory trial. Am J Clin Nutr 2025; 121:816-825. [PMID: 39978467 DOI: 10.1016/j.ajcnut.2025.02.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 02/09/2025] [Accepted: 02/13/2025] [Indexed: 02/22/2025] Open
Abstract
BACKGROUND L-tryptophan ("Trp") and L-leucine ("Leu"), when administered intraduodenally, increase plasma cholecystokinin (CCK) and glucagon-like peptide 1 (GLP-1) and stimulate pyloric pressures, which all slow gastric emptying and suppress subsequent energy intake. The circulating Trp-to-large-neutral-amino-acids ("Trp/LNAAs") ratio is also inversely related to energy intake. OBJECTIVES This exploratory study characterized the impact of standardized changes in the plasma Trp/LNAAs ratio, achieved by combining a fixed-load intraduodenal infusion of Trp with increasing loads of Leu, on the appetite-inhibitory effects of enteral Trp. METHODS Twelve males of normal weight [mean ± standard deviation; age: 23 ± 2 y; body mass index (in kg/m2: 23±1)], received on 4 separate occasions, 90-min iso-osmotic intraduodenal infusions of 1) isotonic 0.9% saline ("control"), 2) Trp (0.15 kcal/min; "Trp"), 3) Trp + Leu (0.22 kcal/min; "Trp+Leu-0.22"), or 4) Trp + Leu (0.45 kcal/min; "Trp+Leu-0.45"), in a randomized, double-blind, cross-over fashion. Immediately postinfusion ad-libitum energy intake was quantified. Plasma CCK, GLP-1, amino acid concentrations, and antropyloroduodenal pressures were measured throughout. RESULTS Although there was a transient stimulation of CCK and GLP-1 by Trp + Leu - 0.45 (at t = 15 min), only Trp led to a sustained increase in plasma CCK (P = 0.04) and GLP-1 (P = 0.009) from t = 60-90 min, and stimulated pyloric pressures (P = 0.01), compared with control. Only Trp reduced energy intake [kcal (mean ± standard error of the mean); control: 1085 ± 49, Trp: 881 ± 75, Trp + Leu - 0.22: 963 ± 57, Trp + Leu - 0.45: 932 ± 60] compared with control (P = 0.008). The Trp/LNAAs ratio was dose-dependently decreased by Trp + Leu - 0.22 and Trp + Leu - 0.45, compared with Trp (all P = 0.001), and energy intake correlated inversely with the Trp/LNAAs ratio (R = -0.38; P = 0.02). CONCLUSIONS Acute reduction in the Trp/LNAAs ratio appears to be associated with a diminished capacity of Trp to stimulate CCK and GLP-1 and suppress energy intake. Although these observations should be interpreted with caution given the exploratory nature of the study, they attest to the complexity of the relationships between pre and postabsorptive mechanisms underlying Trp's appetite-inhibitory effect. This trial was registered at the Australian New Zealand clinical trial registry as ACTRN12620001275954.
Collapse
Affiliation(s)
- Javad Anjom-Shoae
- Centre of Research Excellence in Translating Nutritional Science to Good Health, Adelaide Medical School, University of Adelaide, Adelaide, Australia
| | - Maryam Hajishafiee
- Centre of Research Excellence in Translating Nutritional Science to Good Health, Adelaide Medical School, University of Adelaide, Adelaide, Australia
| | - Penelope Ce Fitzgerald
- Centre of Research Excellence in Translating Nutritional Science to Good Health, Adelaide Medical School, University of Adelaide, Adelaide, Australia
| | - Rosie Coleman
- Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Adelaide, Australia
| | - Alyce M Martin
- Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Adelaide, Australia
| | - Sally D Poppitt
- Department of Medicine, Human Nutrition Unit, School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - Michelle Lee
- School of Psychology, Faculty of Medicine, Health and Life Science, Swansea University, Swansea, United Kingdom
| | - Suzanne Higgs
- School of Psychology, University of Birmingham, Birmingham, United Kingdom
| | - Jens F Rehfeld
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen, Denmark
| | - Jens J Holst
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Simon Veedfald
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Michael Horowitz
- Centre of Research Excellence in Translating Nutritional Science to Good Health, Adelaide Medical School, University of Adelaide, Adelaide, Australia; Endocrine and Metabolic Unit, Royal Adelaide Hospital, Adelaide, Australia
| | - Christine Feinle-Bisset
- Centre of Research Excellence in Translating Nutritional Science to Good Health, Adelaide Medical School, University of Adelaide, Adelaide, Australia.
| |
Collapse
|
7
|
Jones D, Morrison DJ, Gray SR, Ozanne SE, Celis-Morales C, Jain M, Mattin LR, Gittins M, Alkhedhairi SAA, Dorling JL, Burden S. Dietary intake in healthy older individuals is associated with lipopolysaccharide binding protein a biomarker of gut function: an exploratory cross-sectional study. FRONTIERS IN AGING 2025; 6:1572867. [PMID: 40231185 PMCID: PMC11994966 DOI: 10.3389/fragi.2025.1572867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Accepted: 03/12/2025] [Indexed: 04/16/2025]
Abstract
Diet, physical function and gut health are important modifiable factors in ageing. However, it is unclear how ageing affects various domains of gut function. Aims of this cross-sectional study were to explore relationships between nutrient intake, physical function, and biomarkers of gut function in older individuals. Healthy participants (n = 94, mean age 71.1 years SD 5.10, 56% female) were recruited to investigate the relationship between nutrient intake (protein, fibre, carbohydrate, fat), physical function (chair rise time, handgrip strength) and lipopolysaccharide (LPS) binding protein (LBP); a marker of gut permeability. Linear regression models, adjusted for age, fat mass/fat free mass ratio, weight and gender, reported LBP changed by; -161.9 ng/mL (95% CI -323.0, -0.8) for every 1 g increase in daily fibre/1,000 kilocalories; 80.5 ng/mL (6.7, 154.2) for 1% increase in daily energy intake as fat; and -88.1 ng/mL (-146.7, -29.6) for 1% increase in daily energy as carbohydrates. When further adjusted for C-reactive protein (CRP), a marker of inflammation, LBP decreased by an additional 6.9 ng/mL for fibre, increased by an additional 4.0 ng/mL for fat and decreased by an additional 3.7 ng/mL for carbohydrate. These findings suggest that in healthy older adults' nutrient intake is associated with LBP, and CRP appears to slightly modify these associations. There were no associations between LBP and handgrip strength or chair rise time. Results suggest that fibre, fat, and carbohydrates are important for maintaining gut function, potentially mediated by inflammation in older adults, although further research is needed to explore the implications for physical function and CRP as a mediator.
Collapse
Affiliation(s)
- Debra Jones
- School of Health Sciences, University of Manchester, Manchester, United Kingdom
| | - Douglas J. Morrison
- Scottish Universities Environmental Research Centre (SUERC), University of Glasgow, Glasgow, United Kingdom
| | - Stuart R. Gray
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, United Kingdom
- Institute of Sports Science and Innovation, Lithuanian Sports University, Kaunas, Lithuania
| | - Susan E. Ozanne
- Institute of Metabolic Science - Metabolic Research Laboratories and MRC Metabolic Diseases Unit, University of Cambridge, Addenbrookes Hospital Cambridge, Cambridge, United Kingdom
| | - Carlos Celis-Morales
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, United Kingdom
| | - Mahek Jain
- Scottish Universities Environmental Research Centre (SUERC), University of Glasgow, Glasgow, United Kingdom
| | - Lewis R. Mattin
- School of Life Sciences, University of Westminster, London, United Kingdom
| | - Matthew Gittins
- School of Health Sciences, University of Manchester, Manchester, United Kingdom
| | - Saleh A. A. Alkhedhairi
- Department of Medical Biosciences, College of Veterinary Medicine, Qassim University, Buraidah, Saudi Arabia
| | - James L. Dorling
- Human Nutrition, School of Medicine, Dentistry and Nursing, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Sorrel Burden
- School of Health Sciences, University of Manchester, Manchester, United Kingdom
- Salford Care Organisation, Northern Care Alliance NHS Trust, Salford, United Kingdom
| |
Collapse
|
8
|
Xu M, Zhou EY, Shi H. Tryptophan and Its Metabolite Serotonin Impact Metabolic and Mental Disorders via the Brain-Gut-Microbiome Axis: A Focus on Sex Differences. Cells 2025; 14:384. [PMID: 40072112 PMCID: PMC11899299 DOI: 10.3390/cells14050384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 02/27/2025] [Accepted: 03/04/2025] [Indexed: 03/15/2025] Open
Abstract
The crisis of metabolic and mental disorders continues to escalate worldwide. A growing body of research highlights the influence of tryptophan and its metabolites, such as serotonin, beyond their traditional roles in neural signaling. Serotonin acts as a key neurotransmitter within the brain-gut-microbiome axis, a critical bidirectional communication network affecting both metabolism and behavior. Emerging evidence suggests that the gut microbiome regulates brain function and behavior, particularly through microbial influences on tryptophan metabolism and the serotonergic system, both of which are essential for normal functioning. Additionally, sex differences exist in multiple aspects of serotonin-mediated modulation within the brain-gut-microbiome axis, affecting feeding and affective behaviors. This review summarizes the current knowledge from human and animal studies on the influence of tryptophan and its metabolite serotonin on metabolic and behavioral regulation involving the brain and gut microbiome, with a focus on sex differences and the role of sex hormones. We speculate that gut-derived tryptophan and serotonin play essential roles in the pathophysiology that modifies neural circuits, potentially contributing to eating and affective disorders. We propose the gut microbiome as an appealing therapeutic target for metabolic and affective disorders, emphasizing the importance of understanding sex differences in metabolic and behavioral regulation influenced by the brain-gut-microbiome axis. The therapeutic targeting of the gut microbiota and its metabolites may offer a viable strategy for treating serotonin-related disorders, such as eating and affective disorders, with potential differences in treatment efficacy between men and women. This review would promote research on sex differences in metabolic and behavioral regulation impacted by the brain-gut-microbiome axis.
Collapse
Affiliation(s)
- Mengyang Xu
- Program in Cell, Molecular, and Structural Biology, Miami University, Oxford, OH 45056, USA
| | - Ethan Y. Zhou
- Institute for the Environment and Sustainability, Miami University, Oxford, OH 45056, USA
| | - Haifei Shi
- Program in Cell, Molecular, and Structural Biology, Miami University, Oxford, OH 45056, USA
| |
Collapse
|
9
|
López-Almada G, Domínguez-Avila JA, Robles-Sánchez RM, Arauz-Cabrera J, Martínez-Coronilla G, González-Aguilar GA, Salazar-López NJ. Naringenin Decreases Retroperitoneal Adiposity and Improves Metabolic Parameters in a Rat Model of Western Diet-Induced Obesity. Metabolites 2025; 15:109. [PMID: 39997735 PMCID: PMC11857789 DOI: 10.3390/metabo15020109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 02/04/2025] [Accepted: 02/06/2025] [Indexed: 02/26/2025] Open
Abstract
Background: Obesity is a multifactorial disease with detrimental effects on health and quality of life; unregulated satiety plays a crucial role in food intake and obesity development. Naringenin (NAR) has shown beneficial effects on lipid and carbohydrate metabolism, although its impact on adiposity and satiety remains unclear. This study reports a Western diet (WD)-induced obesity model in rats, wherein 100 mg/kg of NAR was administered as an anti-obesity agent for 8 weeks; oxidative stress, lipid profile, and satiety biomarkers were then studied, as well as in silico interaction between NAR and cholecystokinin (CCK) and ghrelin receptors. Results: NAR supplementation resulted in a significant decrease in retroperitoneal adipose tissue and liver weight, as compared to the untreated WD group (p < 0.05), potentially associated with a decreased feed efficiency. NAR also inhibited the development of dyslipidemia, particularly by reducing serum triglycerides (p < 0.05). NAR supplementation increased CCK serum levels in the basal diet group, an effect that was abolished by the WD (p < 0.05); likewise, no changes were determined on ghrelin (p > 0.05). In silico data shows that NAR is capable of interacting with the CCK and ghrelin receptors, which suggests a potential for it to modulate hunger/satiety signaling by interacting with them. Conclusions: We conclude that NAR has anti-obesogenic effects and may regulate CCK serum levels, although further research is still needed.
Collapse
Affiliation(s)
- Gabriela López-Almada
- Facultad de Medicina de Mexicali, Universidad Autónoma de Baja California, Dr. Humberto Torres Sanginés, Centro Cívico, Mexicali 21000, BCN, Mexico; (G.L.-A.)
| | - J. Abraham Domínguez-Avila
- SECIHTI—Centro de Investigación en Alimentación y Desarrollo A.C., Carretera Gustavo Enrique Astiazarán Rosas No. 46, Col. La Victoria, Hermosillo 83304, SO, Mexico;
| | - Rosario Maribel Robles-Sánchez
- Departamento de Investigación y Posgrado en Alimentos, Universidad de Sonora, Blvd. Luis Encinas y Rosales, Col. Centro, Hermosillo 83000, SO, Mexico
| | - Jonathan Arauz-Cabrera
- Facultad de Medicina de Mexicali, Universidad Autónoma de Baja California, Dr. Humberto Torres Sanginés, Centro Cívico, Mexicali 21000, BCN, Mexico; (G.L.-A.)
| | - Gustavo Martínez-Coronilla
- Facultad de Medicina de Mexicali, Universidad Autónoma de Baja California, Dr. Humberto Torres Sanginés, Centro Cívico, Mexicali 21000, BCN, Mexico; (G.L.-A.)
| | - Gustavo A. González-Aguilar
- Centro de Investigación en Alimentación y Desarrollo A.C., Carretera Gustavo Enrique Astiazarán Rosas No. 46, Col. La Victoria, Hermosillo 83304, SO, Mexico
| | - Norma Julieta Salazar-López
- Facultad de Medicina de Mexicali, Universidad Autónoma de Baja California, Dr. Humberto Torres Sanginés, Centro Cívico, Mexicali 21000, BCN, Mexico; (G.L.-A.)
| |
Collapse
|
10
|
Yamamoto Y, Narumi K, Yamagishi N, Yonejima Y, Iseki K, Kobayashi M, Kanai Y. HYA ameliorated postprandial hyperglycemia in type 1 diabetes model rats with bolus insulin treatment. Acta Diabetol 2025:10.1007/s00592-025-02459-6. [PMID: 39899133 DOI: 10.1007/s00592-025-02459-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 01/24/2025] [Indexed: 02/04/2025]
Abstract
AIMS The oral administration of linoleic acid immediately before glucose tolerance test (OGTT) ameliorated postprandial hyperglycemia via GPR120 pathway in normal and type 1 diabetes (T1DM) rats. Linoleic acid could promote inflammatory mediators, but 10-hydroxy-cis-12-octadecenoic acid (HYA) converted from linoleic acid by Lactobacillus plantarum has higher GPR120 agonistic activity without promoting inflammatory mediators. This study examined whether the oral-administration of HYA immediately before OGTT also ameliorated the postprandial hyperglycemia in normal rats and T1DM rats injected with bolus insulin. METHODS Normal and T1DM male Sprague-Dawley rats received HYA immediately before OGTT. Other T1DM rats were given HYA and Humulin R immediately before OGTT. We measured the concentration of glucose, insulin, glucagon-like peptide 1 (GLP-1) and cholecystokinin in blood before and after OGTT. We also measured the amount of glucose in the gastric tract after OGTT, and the amount of uptake of methyl-α-D-glucopyranoside in CACO-2 cells. RESULTS Postprandial hyperglycemia was ameliorated by HYA in normal rats, and the postprandial blood glucose levels were slowly elevated by HYA in the T1DM model rats. HYA partially inhibited the uptake of methyl-α-D-glucopyranoside in CACO-2 cells. HYA slowed gastric motility and increased the plasma GLP-1 and cholecystokinin levels in normal rats. HYA also ameliorated the postprandial hyperglycemia in T1DM rats given bolus insulin. CONCLUSION Oral administration of HYA immediately before OGTT ameliorated postprandial hyperglycemia through inhibition of glucose absorption and slowing of gastric motility in normal rats. Furthermore, this beneficial effect of HYA was also revealed in T1DM rats injected with bolus insulin.
Collapse
Affiliation(s)
- Yuta Yamamoto
- Department of Anatomy and Cell Biology, Graduate School of Medical and Pharmaceutical Sciences, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8509, Japan.
| | - Katsuya Narumi
- Laboratory of Clinical Pharmaceutics and Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Naoko Yamagishi
- Department of Anatomy and Cell Biology, Graduate School of Medical and Pharmaceutical Sciences, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8509, Japan
| | | | - Ken Iseki
- Laboratory of Clinical Pharmaceutics and Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Masaki Kobayashi
- Laboratory of Clinical Pharmaceutics and Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Yoshimitsu Kanai
- Department of Anatomy and Cell Biology, Graduate School of Medical and Pharmaceutical Sciences, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8509, Japan
| |
Collapse
|
11
|
Aukan MI, Rehfeld JF, Holst JJ, Martins C. Plasma concentration of gastrointestinal hormones and subjective appetite ratings after diet or bariatric surgery: 1-year results from the DISGAP study. Int J Obes (Lond) 2025; 49:306-314. [PMID: 39572763 DOI: 10.1038/s41366-024-01658-5] [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: 03/04/2024] [Revised: 10/10/2024] [Accepted: 10/14/2024] [Indexed: 01/04/2025]
Abstract
OBJECTIVE Long-term weight loss outcomes are contrasting between bariatric surgery and dietary restriction alone. This is the first study to investigate changes in gastrointestinal (GI) hormones involved in appetite regulation, and subjective appetite feelings, at 1-year follow-up, after initial weight loss induced by a very-low energy (VLED) alone (controls), or with bariatric surgery. METHODS Patients scheduled for Sleeve Gastrectomy (SG) (n = 19) or Roux-en-Y gastric Bypass (RYGB) (n = 19), and controls (n = 16) were recruited. All groups underwent 10 weeks of a VLED (initial phase), followed by a 9-month maintenance phase. Body weight/composition, plasma concentrations of ghrelin, glucagon-like peptide 1 (GLP-1), peptide YY (PYY), cholecystokinin (CCK), and appetite ratings were measured before and after a meal, at baseline, week 11(W11), and 1Y follow-up. RESULTS Participants who completed all three follow ups were included in the analysis. Initial changes in body weight/composition were comparable across groups. SG (n = 11) and RYGB (n = 12) continued to lose weight from W11 to 1Y, whereas controls (n = 12) had regained weight. Postprandial GLP-1 increased over time post bariatric surgery and remained unchanged and lower in controls. Postprandial PYY increased in all groups, but greatest post-RYGB. Basal ghrelin decreased over time post-SG, while a small or marked increase was seen after RYGB and diet, respectively, with the control group exhibiting the greatest basal and postprandial concentrations at 1Y. A reduction in basal and postprandial CCK was seen in controls at 1Y, while no changes were observed post-bariatric surgery. Overall, small changes in subjective appetite ratings were seen over time. CONCLUSION Weight change at 1Y follow up after SG and RYGB is followed by a GI hormone profile favoring a lower drive to eat and increased satiety. The opposite is seen 1Y after WL induced by dietary restriction alone. CLINICAL TRIAL REGISTRATION clinicaltrials.gov NCT04051190.
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.
| | - Jens Frederik Rehfeld
- Department of Clinical Biochemistry, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Jens Juul Holst
- The Novo Nordisk Foundation Center for Basic Metabolic Research and Department of Biomedical Sciences, University of Copenhagen, The Panum Institute, Copenhagen, Denmark
| | - Catia Martins
- Department of Nutrition Sciences, University of Alabama at Birmingham (UAB), Birmingham, AL, USA
| |
Collapse
|
12
|
Huang Y, Xu B. Critical review on the intervention effects of flavonoids from cereal grains and food legumes on lipid metabolism. Food Chem 2025; 464:141790. [PMID: 39509881 DOI: 10.1016/j.foodchem.2024.141790] [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: 05/26/2024] [Revised: 09/22/2024] [Accepted: 10/24/2024] [Indexed: 11/15/2024]
Abstract
Obesity, often caused by disorders of lipid metabolism, is a global health concern. Flavonoids from staple grains and legumes are expected as a safer and more cost-effective alternative for the future development of dietary flavonoid-based anti-obesity dietary supplements or drugs. This review systematically summarized their content variation, metabolism in the human body, effects and molecular mechanisms on lipid metabolism. These flavonoids intervene in lipid metabolism by inhibiting lipogenesis, promoting lipolysis, enhancing energy metabolism, reducing appetite, suppressing inflammation, enhancing insulin sensitivity, and improving the composition of the gut microbial. Fermentation and sprouting techniques enhance flavonoid content and these beneficial effects. The multidirectional intervention of lipid metabolism is mainly through regulating AMPK signaling pathway. This study provides potential improvement for challenges of application, including addressing high extraction costs and improving bioavailability, ensuring safety, filling clinical study gaps, and investigating potential synergistic effects between flavonoids in grains and legumes, and other components.
Collapse
Affiliation(s)
- Yin Huang
- Food Science and Technology Program, Department of Life Sciences, BNU-HKBU United International College, Zhuhai, Guangdong 519087, China
| | - Baojun Xu
- Food Science and Technology Program, Department of Life Sciences, BNU-HKBU United International College, Zhuhai, Guangdong 519087, China.
| |
Collapse
|
13
|
Tack J, Raymenants K, Van de Bruaene C, Scarpellini E. Dumping syndrome: Update on pathophysiology, diagnosis, and management. Neurogastroenterol Motil 2025; 37:e14962. [PMID: 39529492 DOI: 10.1111/nmo.14962] [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: 04/15/2024] [Revised: 10/07/2024] [Accepted: 10/26/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Dumping syndrome is a complex of gastrointestinal symptoms originally studied in peptic ulcer surgery patients. At present, it is most prevalent in patients who underwent bariatric, upper gastrointestinal cancer or anti-reflux surgery. The symptom pattern comprises early and late dumping symptoms. Several management options have been reported including nutritional, pharmacological and surgical approaches. AIMS AND METHODS In this study, we aimed to review the current evidence on dumping syndrome definition, diagnosis and treatment, including preliminary data from newer pharmacological studies. RESULTS Current pathophysiological concepts and analyses of provocative tests has led to a clear definition of dumping syndrome, including both early and late dumping symptoms. The term postbariatric hypoglycemia represents a limited focus on late dumping only. The diagnosis relies on recognition of symptoms and signs in a patient with appropriate surgical history; and can be confirmed by provocative testing or registration of spontaneous hypoglycemia. The initial treatment focuses on dietary intervention, to which meal viscosity enhancers and/or the glycosidase inhibitor acarbose can be added. The most effective therapy is the use of short- or long-acting somatostatin analogues, which is however expensive and entails side effect issues. In case of refractory hypoglycemia, diazoxide or SGLT2 inhibitors can be considered, based on limited evidence. In refractory patients, continuous enteral feeding or (rarely) surgical reinterventions have been advocated, although not supported by solid evidence. Therapies under current evaluation include the broad-spectrum somatostatin analogue pasireotide, GLP-1 receptor antagonists, GLP-1 receptor agonists and administration of stable forms of glucagon are currently under study. CONCLUSIONS Dumping syndrome is a well-defined but probably under-diagnosed complication of upper gastrointestinal, especially bariatric, and surgeries. Diagnosis is confirmed by a provocative test and incremental therapies starting with diet, adding meal viscosity enhancers or glycosidase inhibitors and adding somatostatin analogues in refractory cases. A number of emerging therapies targeting intestinal propulsion, peptide hormone effects and hypoglycemic events are under evaluation.
Collapse
Affiliation(s)
- Jan Tack
- Translational Research Center for Gastrointestinal Diseases (TARGID), University of Leuven, Leuven, Belgium
- Division of Gastroenterology and Hepatology, Leuven University Hospitals, Leuven, Belgium
| | - Karlien Raymenants
- Translational Research Center for Gastrointestinal Diseases (TARGID), University of Leuven, Leuven, Belgium
| | - Cedric Van de Bruaene
- Translational Research Center for Gastrointestinal Diseases (TARGID), University of Leuven, Leuven, Belgium
| | - Emidio Scarpellini
- Translational Research Center for Gastrointestinal Diseases (TARGID), University of Leuven, Leuven, Belgium
| |
Collapse
|
14
|
Krieger JP, Daniels D, Lee S, Mastitskaya S, Langhans W. Glucagon-Like Peptide-1 Links Ingestion, Homeostasis, and the Heart. Compr Physiol 2025; 15:e7. [PMID: 39887844 PMCID: PMC11790259 DOI: 10.1002/cph4.7] [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: 11/02/2024] [Revised: 11/02/2024] [Accepted: 11/05/2024] [Indexed: 02/01/2025]
Abstract
Glucagon-like peptide-1 (GLP-1), a hormone released from enteroendocrine cells in the distal small and large intestines in response to nutrients and other stimuli, not only controls eating and insulin release, but is also involved in drinking control as well as renal and cardiovascular functions. Moreover, GLP-1 functions as a central nervous system peptide transmitter, produced by preproglucagon (PPG) neurons in the hindbrain. Intestinal GLP-1 inhibits eating by activating vagal sensory neurons directly, via GLP-1 receptors (GLP-1Rs), but presumably also indirectly, by triggering the release of serotonin from enterochromaffin cells. GLP-1 enhances glucose-dependent insulin release via a vago-vagal reflex and by direct action on beta cells. Finally, intestinal GLP-1 acts on the kidneys to modulate electrolyte and water movements, and on the heart, where it provides numerous benefits, including anti-inflammatory, antiatherogenic, and vasodilatory effects, as well as protection against ischemia/reperfusion injury and arrhythmias. Hindbrain PPG neurons receive multiple inputs and project to many GLP-1R-expressing brain areas involved in reward, autonomic functions, and stress. PPG neuron-derived GLP-1 is involved in the termination of large meals and is implicated in the inhibition of water intake. This review details GLP-1's roles in these interconnected systems, highlighting recent findings and unresolved issues, and integrating them to discuss the physiological and pathological relevance of endogenous GLP-1 in coordinating these functions. As eating poses significant threats to metabolic, fluid, and immune homeostasis, the body needs mechanisms to mitigate these challenges while sustaining essential nutrient intake. Endogenous GLP-1 plays a crucial role in this "ingestive homeostasis."
Collapse
Affiliation(s)
- Jean-Philippe Krieger
- Jean-Philippe Krieger, Institute of Veterinary Pharmacology and Toxicology, Vetsuisse Faculty, University of Zurich, Winterthurerstr. 260, 8057 Zurich
| | - Derek Daniels
- Department of Biological Sciences and the Center for Ingestive Behavior Research, University at Buffalo, the State University of New York, Buffalo NY 14260 USA
| | - Shin Lee
- Shin J. Lee, Neurimmune AG, Wagistrasse 18, 8952 Schlieren, Switzerland
| | - Svetlana Mastitskaya
- Department of Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Wolfgang Langhans
- Physiology and Behavior Laboratory, Dept. of Health Sciences and Technology, ETH Zurich, 8603 Schwerzenbach, Switzerland
| |
Collapse
|
15
|
Cencioni C, Malatesta S, Vigiano Benedetti V, Licursi V, Perfetto L, Conte F, Ranieri D, Bartolazzi A, Kunkl M, Tuosto L, Larghi A, Piro G, Agostini A, Tortora G, Corbo V, Carbone C, Spallotta F. The GLP-1R agonist semaglutide reshapes pancreatic cancer associated fibroblasts reducing collagen proline hydroxylation and favoring T lymphocyte infiltration. J Exp Clin Cancer Res 2025; 44:18. [PMID: 39828692 PMCID: PMC11744909 DOI: 10.1186/s13046-024-03263-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 12/19/2024] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND Metabolic syndrome represents a pancreatic ductal adenocarcinoma (PDAC) risk factor. Metabolic alterations favor PDAC onset, which occurs early upon dysmetabolism. Pancreatic neoplastic lesions evolve within a dense desmoplastic stroma, consisting in abundant extracellular matrix settled by cancer associated fibroblasts (CAFs). Hereby, dysmetabolism and PDAC association was analyzed focusing on CAF functions. METHODS PDAC development upon dysmetabolic conditions was investigated in: 1) high fat diet fed wild type immunocompetent syngeneic mice by orthotopic transplantation of pancreatic intraepithelial neoplasia (PanIN) organoids; and 2) primary pancreatic CAFs isolated from chemotherapy naïve PDAC patients with/without an history of metabolic syndrome. RESULTS The dysmetabolic-associated higher PDAC aggressiveness was paralleled by collagen fibril enrichment due to prolyl 4-hydroxylase subunit alpha 1 (P4HA1) increased function. Upon dysmetabolism, P4HA1 boosts collagen proline hydroxylation, intensifies collagen contraction strength, precluding PDAC infiltration. Noteworthy, semaglutide, an incretin agonist, prevents the higher dysmetabolism-dependent PDAC stromal deposition and allows T lymphocyte infiltration, reducing tumor development. CONCLUSIONS These results shed light on novel therapeutic options for PDAC patients with metabolic syndrome aimed at PDAC stroma reshape.
Collapse
Affiliation(s)
- Chiara Cencioni
- Institute of System Analysis and Informatics "Antonio Ruberti", National Research Council (IASI-CNR), 00185, Rome, Italy
| | - Silvia Malatesta
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University, 00185, Rome, Italy
- Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University, 00185, Rome, Italy
| | | | - Valerio Licursi
- Institute of Molecular Biology and Pathology, National Research Council (IBPM-CNR), 00185, Rome, Italy
| | - Livia Perfetto
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University, 00185, Rome, Italy
| | - Federica Conte
- Institute of System Analysis and Informatics "Antonio Ruberti", National Research Council (IASI-CNR), 00185, Rome, Italy
| | - Danilo Ranieri
- Dipartimento Di Scienze Della Vita, Della Salute E Delle Professioni Sanitarie. Università Degli Studi "Link Campus University", 00165, Rome, Italy
| | - Armando Bartolazzi
- Pathology Research Laboratory, Sant' Andrea University Hospital, 00189, Rome, Italy
| | - Martina Kunkl
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University, 00185, Rome, Italy
- Neuroimmunology Unit, IRCCS Santa Lucia Foundation, 00179, Rome, Italy
| | - Loretta Tuosto
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University, 00185, Rome, Italy
| | - Alberto Larghi
- Digestive Endoscopy Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168, Rome, Italy
- Center for Endoscopic Research Therapeutics and Training (CERTT), Catholic University, 00168, Rome, Italy
| | - Geny Piro
- Medical Oncology, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168, Rome, Italy
| | - Antonio Agostini
- Medical Oncology, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168, Rome, Italy
| | - Giampaolo Tortora
- Department of Translational Medicine, Catholic University of the Sacred Heart, 00168, Rome, Italy
- Medical Oncology, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168, Rome, Italy
| | - Vincenzo Corbo
- Department of Engineering for Innovation Medicine (DIMI), University and Hospital Trust of Verona, 37100, Verona, Italy
| | - Carmine Carbone
- Medical Oncology, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168, Rome, Italy
| | - Francesco Spallotta
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University, 00185, Rome, Italy.
- Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University, 00185, Rome, Italy.
| |
Collapse
|
16
|
Shi Y, Wu C, Liu T, Wang R, Ding B, Sun Q. Metabolics-Based Study on the Therapeutic Mechanism Behind the Effect of Shenhuang Plaster Applied to the Shenque Acupoint on Gastrointestinal Motility in POI Mice. Metabolites 2025; 15:65. [PMID: 39852407 PMCID: PMC11767606 DOI: 10.3390/metabo15010065] [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: 12/20/2024] [Revised: 01/06/2025] [Accepted: 01/14/2025] [Indexed: 01/26/2025] Open
Abstract
BACKGROUND Postoperative ileus (POI) is a common postoperative clinical complication that significantly affects postoperative rehabilitation and quality of life in patients and can even produce secondary complications, leading to serious consequences. External treatment using Shenhuang Plaster (SHP) (Shenque acupoint administration) has definite effects and unique advantages in the prevention and treatment of POI, but its mechanism is not completely clear. In this study, we investigated the therapeutic mechanism behind the effect of Shenhuang Plaster applied to the Shenque acupoint on gastrointestinal motility in POI mice based on metabolomics. MATERIALS AND METHODS C57BL/6 mice were divided into three groups: blank control (Ctrl), model (POI), and intervention (POI + SHP) groups. SHP treatment was started 3 days before modeling. We employed several behavioral tests and gastrointestinal transit function measurements and performed qRT-PCR analysis, 16S rRNA gene sequencing, and metabolomics analysis on serum metabolites. RESULTS We found that SHP could reduce the mRNA expression of inflammatory mediators in the smooth muscle tissue of the small intestine, regulate the structure and function of the intestinal microbiota, and modulate serum phenylalanine, carnitine, and glutamic acid levels. CONCLUSIONS POI mice had obvious intestinal flora disorders and metabolic disorders of amino acids and their derivatives, and there was a significant correlation between differential flora and differential metabolites. SHP could effectively regulate the concentration of intestinal flora and serum metabolites and the metabolic pathway related to amino acids in vivo and, ultimately, achieve a therapeutic purpose in POI. In this study, it was found, for the first time, that applying SHP to the Shenque acupoint could effectively regulate the serum metabolites of phenylalanine, carnitine, and glutamate, and improve postoperative intestinal motile disturbance through association with the intestinal flora.
Collapse
Affiliation(s)
- Yanan Shi
- The College of Nursing, Zhejiang Chinese Medical University, Hangzhou 310053, China; (Y.S.); (C.W.); (T.L.); (R.W.)
| | - Chenglei Wu
- The College of Nursing, Zhejiang Chinese Medical University, Hangzhou 310053, China; (Y.S.); (C.W.); (T.L.); (R.W.)
| | - Ting Liu
- The College of Nursing, Zhejiang Chinese Medical University, Hangzhou 310053, China; (Y.S.); (C.W.); (T.L.); (R.W.)
| | - Rongyun Wang
- The College of Nursing, Zhejiang Chinese Medical University, Hangzhou 310053, China; (Y.S.); (C.W.); (T.L.); (R.W.)
| | - Bin Ding
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Qiuhua Sun
- The College of Nursing, Zhejiang Chinese Medical University, Hangzhou 310053, China; (Y.S.); (C.W.); (T.L.); (R.W.)
| |
Collapse
|
17
|
Holliday A, Horner K, Johnson KO, Dagbasi A, Crabtree DR. Appetite-related Gut Hormone Responses to Feeding Across the Life Course. J Endocr Soc 2025; 9:bvae223. [PMID: 39777204 PMCID: PMC11702868 DOI: 10.1210/jendso/bvae223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Indexed: 01/11/2025] Open
Abstract
Appetite-related hormones are secreted from the gut, signaling the presence of nutrients. Such signaling allows for cross-talk between the gut and the appetite-control regions of the brain, influencing appetite and food intake. As nutritional requirements change throughout the life course, it is perhaps unsurprising that appetite and eating behavior are not constant. Changes in appetite-related gut hormones may underpin these alterations in appetite and eating. In this article, we review evidence of how the release of appetite-related gut hormones changes throughout the life course and how this impacts appetite and eating behaviour. We focus on hormones for which there is the strongest evidence of impact on appetite, food intake, and body weight: the anorexigenic glucagon like peptide-1, peptide tyrosine tyrosine, and cholecystokinin, and the orexigenic ghrelin. We consider hormone concentrations, particularly in response to feeding, from the very early days of life, through childhood and adolescence, where responses may reflect energy requirements to support growth and development. We discuss the period of adulthood and midlife, with a particular focus on sex differences and the effect of menstruation, pregnancy, and menopause, as well as the potential influence of appetite-related gut hormones on body composition and weight status. We then discuss recent advancements in our understanding of how unfavorable changes in appetite-related gut hormone responses to feeding in later life may contribute to undernutrition and a detrimental aging trajectory. Finally, we briefly highlight priorities for future research.
Collapse
Affiliation(s)
- Adrian Holliday
- School of Biomedical, Nutritional, and Sport Sciences, 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
| | - Katy Horner
- Institute of Sport and Health, University College Dublin, Belfield, Dublin D04 V1W8, Ireland
| | - Kelsie O Johnson
- Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, Liverpool L3 5RF, UK
| | - Aygul Dagbasi
- Section of Nutrition, Department of Metabolism Digestion and Reproduction, Imperial College London, Hammersmith Campus, London W12 0NN, UK
| | - Daniel R Crabtree
- The Rowett Institute, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen AB25 2ZD, UK
| |
Collapse
|
18
|
Soengas JL, Comesaña S, Blanco AM, Conde-Sieira M. Feed Intake Regulation in Fish: Implications for Aquaculture. REVIEWS IN FISHERIES SCIENCE & AQUACULTURE 2025; 33:8-60. [DOI: 10.1080/23308249.2024.2374259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Affiliation(s)
- José L. Soengas
- Departamento de Bioloxía Funcional e Ciencias da Saúde, Facultade de Bioloxía, Laboratorio de Fisioloxía Animal, Centro de Investigación Mariña, Universidade de Vigo, Vigo, Spain
| | - Sara Comesaña
- Departamento de Bioloxía Funcional e Ciencias da Saúde, Facultade de Bioloxía, Laboratorio de Fisioloxía Animal, Centro de Investigación Mariña, Universidade de Vigo, Vigo, Spain
| | - Ayelén M. Blanco
- Departamento de Bioloxía Funcional e Ciencias da Saúde, Facultade de Bioloxía, Laboratorio de Fisioloxía Animal, Centro de Investigación Mariña, Universidade de Vigo, Vigo, Spain
| | - Marta Conde-Sieira
- Departamento de Bioloxía Funcional e Ciencias da Saúde, Facultade de Bioloxía, Laboratorio de Fisioloxía Animal, Centro de Investigación Mariña, Universidade de Vigo, Vigo, Spain
| |
Collapse
|
19
|
Chao AM, Taylor S, Moore M, Amaro A, Wadden TA. Evolving Approaches for Pharmacological Therapy of Obesity. Annu Rev Pharmacol Toxicol 2025; 65:169-189. [PMID: 39348522 PMCID: PMC11770897 DOI: 10.1146/annurev-pharmtox-031124-101146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/02/2024]
Abstract
Obesity is a global health concern. Progress in understanding the physiology of obesity and weight reduction has provided new drug targets. Development and testing of new antiobesity medications (AOMs) has the potential to quickly expand options for treatment. In this review, we briefly summarize the physiology of obesity and weight reduction, as well as medications currently approved for weight management. We highlight the increasing use of incretin and nutrient-stimulated hormone-based therapies. We conclude with an overview of AOMs progressing through the pipeline and discuss their implications for the rapidly evolving field of obesity management.
Collapse
Affiliation(s)
- Ariana M Chao
- Johns Hopkins School of Nursing, Baltimore, Maryland, USA;
| | - Simeon Taylor
- Division of Endocrinology, Diabetes, and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Molly Moore
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA;
| | - Anastassia Amaro
- Division of Endocrinology, Diabetes and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Thomas A Wadden
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA;
| |
Collapse
|
20
|
Rösch SA, Wünsche L, Thiele C, Reinstaller T, Zähle T, Schag K, Giel KE, Plewnia C, Steiner J, Junne F. Enhancing the outcomes of bariatric surgery with inhibitory control training, electrical brain stimulation and psychosocial aftercare: a pilot study protocol. J Eat Disord 2024; 12:202. [PMID: 39654020 PMCID: PMC11626763 DOI: 10.1186/s40337-024-01160-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 11/22/2024] [Indexed: 12/12/2024] Open
Abstract
BACKGROUND Notwithstanding the documented short- and long-term weight loss and remission of physical and mental diseases following bariatric surgery, a significant proportion of patients fail to respond (fully) to treatment in terms of physical and mental health improvement. Mounting evidence links food-specific impulsivity, prefrontal cortex (PFC) hypoactivity and disrupted hormone secretion in bariatric surgery candidates to poorer post-surgical health outcomes. Neuromodulatory treatments like transcranial direct current stimulation (tDCS) uniquely target these neurobehavioral impairments. We present a pilot study protocol offering tDCS combined with an inhibitory control training and a structured psychosocial intervention to patients after bariatric surgery. METHODS A total of N = 20 patients are randomized to 6 sessions of verum or sham tDCS over the PFC, combined with an individualized food-specific inhibitory control training and a structured psychosocial intervention within 18 months after bariatric surgery (t0). Beyond acceptability, feasibility and satisfaction of the intervention, effects of verum versus sham tDCS on food-specific impulsivity and on secondary outcomes quality of life, general impulsivity and psychopathology, food-related cravings, eating disorder psychopathology, weight trajectory and endocrine markers are assessed 4 weeks (t1) and 3 months after the intervention (t2). DISCUSSION Results will provide information on the potential of combining tDCS with an inhibitory control training and a structured psychosocial intervention to enhance physical and mental outcomes after bariatric surgery. The present study may guide the development of future research with regard to tDCS as a brain-based intervention and of future post-surgical clinical programs, paving the way for randomized-controlled trials in larger samples. TRIAL REGISTRATION The trial was prospectively registered on July 8, 2024, under the registration number DRKS00034620 in the German Clinical Trials Register ( https://drks.de/search/de/trial/DRKS00034620 ).
Collapse
Affiliation(s)
- Sarah A Rösch
- University Clinic of Psychosomatic Medicine and Psychotherapy, University Hospital, Otto von Guericke University Magdeburg, Leipziger Str. 44, 39120, Magdeburg, Germany.
- German Center for Mental Health (DZPG), Partner Site Halle-Jena-Magdeburg, Magdeburg, Germany.
- Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C-I-R-C), Halle-Jena-Magdeburg, Magdeburg, Germany.
| | - Lennart Wünsche
- University Clinic of Psychosomatic Medicine and Psychotherapy, University Hospital, Otto von Guericke University Magdeburg, Leipziger Str. 44, 39120, Magdeburg, Germany
| | - Carsten Thiele
- Department of Neurology, University Hospital, Otto von Guericke University Magdeburg, Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
| | - Therese Reinstaller
- Visceral, Vascular and Transplant Surgery, University Clinic for General, University Hospital, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Tino Zähle
- Department of Neurology, University Hospital, Otto von Guericke University Magdeburg, Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
| | - Kathrin Schag
- German Center for Mental Health (DZPG), Partner Site Halle-Jena-Magdeburg, Magdeburg, Germany
- Department of Psychosomatic Medicine und Psychotherapy, Medical University Hospital Tübingen, Eberhard Karl University Tübingen, Tübingen, Germany
- Center of Excellence for Eating Disorders, Tübingen, Germany
| | - Katrin E Giel
- German Center for Mental Health (DZPG), Partner Site Halle-Jena-Magdeburg, Magdeburg, Germany
- Department of Psychosomatic Medicine und Psychotherapy, Medical University Hospital Tübingen, Eberhard Karl University Tübingen, Tübingen, Germany
- Center of Excellence for Eating Disorders, Tübingen, Germany
| | - Christian Plewnia
- German Center for Mental Health (DZPG), Partner Site Halle-Jena-Magdeburg, Magdeburg, Germany
- Department of Psychiatry and Psychotherapy, Medical University Hospital Tübingen, Eberhard Karl University Tübingen, Tübingen, Germany
| | - Johann Steiner
- German Center for Mental Health (DZPG), Partner Site Halle-Jena-Magdeburg, Magdeburg, Germany
- Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C-I-R-C), Halle-Jena-Magdeburg, Magdeburg, Germany
- Department of Psychiatry, University Hospital, Otto von Guericke University, Magdeburg, Germany
- Laboratory of Translational Psychiatry, University Hospital, Otto von Guericke University Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
- Center for Health and Medical Prevention (CHaMP), Magdeburg, Germany
| | - Florian Junne
- University Clinic of Psychosomatic Medicine and Psychotherapy, University Hospital, Otto von Guericke University Magdeburg, Leipziger Str. 44, 39120, Magdeburg, Germany
- German Center for Mental Health (DZPG), Partner Site Halle-Jena-Magdeburg, Magdeburg, Germany
- Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C-I-R-C), Halle-Jena-Magdeburg, Magdeburg, Germany
| |
Collapse
|
21
|
López-Almada G, Mejía-León ME, Salazar-López NJ. Probiotic, Postbiotic, and Paraprobiotic Effects of Lactobacillus rhamnosus as a Modulator of Obesity-Associated Factors. Foods 2024; 13:3529. [PMID: 39593945 PMCID: PMC11592899 DOI: 10.3390/foods13223529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 10/28/2024] [Accepted: 11/02/2024] [Indexed: 11/28/2024] Open
Abstract
Obesity is a pandemic currently affecting the world's population that decreases the quality of life and promotes the development of chronic non-communicable diseases. Lactobacillus rhamnosus is recognized for multiple positive effects on obesity and overall health. In fact, such effects may occur even when the microorganisms do not remain alive (paraprobiotic effects). This raises the need to elucidate the mechanisms by which obesity-associated factors can be modulated. This narrative review explores recent findings on the effects of L. rhamnosus, particularly, its postbiotic and paraprobiotic effects, on the modulation of adiposity, weight gain, oxidative stress, inflammation, adipokines, satiety, and maintenance of intestinal integrity, with the aim of providing a better understanding of its mechanisms of action in order to contribute to streamlining its clinical and therapeutic applications. The literature shows that L. rhamnosus can modulate obesity-associated factors when analyzed in vitro and in vivo. Moreover, its postbiotic and paraprobiotic effects may be comparable to the more studied probiotic actions. Some mechanisms involve regulation of gene expression, intracellular signaling, and enteroendocrine communication, among others. We conclude that the evidence is promising, although there are still multiple knowledge gaps that require further study in order to fully utilize L. rhamnosus to improve human health.
Collapse
Affiliation(s)
| | | | - Norma Julieta Salazar-López
- Facultad de Medicina de Mexicali, Universidad Autónoma de Baja California, Dr. Humberto Torres Sanginés, Centro Cívico, Mexicali 21000, BCN, Mexico
| |
Collapse
|
22
|
Cho H, Lim J. The emerging role of gut hormones. Mol Cells 2024; 47:100126. [PMID: 39426686 PMCID: PMC11577206 DOI: 10.1016/j.mocell.2024.100126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 10/13/2024] [Accepted: 10/13/2024] [Indexed: 10/21/2024] Open
Abstract
The gut is traditionally recognized as the central organ for the digestion and absorption of nutrients, however, it also functions as a significant endocrine organ, secreting a variety of hormones such as glucagon-like peptide 1, serotonin, somatostatin, and glucocorticoids. These gut hormones, produced by specialized intestinal epithelial cells, are crucial not only for digestive processes but also for the regulation of a wide range of physiological functions, including appetite, metabolism, and immune responses. While gut hormones can exert systemic effects, they also play a pivotal role in maintaining local homeostasis within the gut. This review discusses the role of the gut as an endocrine organ, emphasizing the stimuli, the newly discovered functions, and the clinical significance of gut-secreted hormones. Deciphering the emerging role of gut hormones will lead to a better understanding of gut homeostasis, innovative treatments for disorders in the gut, as well as systemic diseases.
Collapse
Affiliation(s)
- Hyeryeong Cho
- College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Jaechul Lim
- College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea; Research Institute for Veterinary Science, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|
23
|
Eftychidis V, Ellender TJ, Szymanski J, Minichiello L. Cholecystokinin-expressing neurons of the ventromedial hypothalamic nucleus control energy homeostasis. Front Cell Neurosci 2024; 18:1483368. [PMID: 39529694 PMCID: PMC11550940 DOI: 10.3389/fncel.2024.1483368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 10/11/2024] [Indexed: 11/16/2024] Open
Abstract
The hypothalamus is the primary center of the brain that regulates energy homeostasis. The ventromedial hypothalamus (VMH) plays a central role in maintaining energy balance by regulating food intake, energy expenditure, and glucose levels. However, the cellular and molecular mechanisms underlying its functions are still poorly understood. Cholecystokinin (CCK) is one of many genes expressed in this hypothalamic nucleus. Peripheral CCK regulates food intake, body weight, and glucose homeostasis. However, current research does not explain the function of CCK neurons in specific nuclei of the hypothalamus and their likely roles in network dynamics related to energy balance and food intake. This study uses genetic and pharmacological methods to examine the role of CCK-expressing neurons in the VMH (CCKVMH). Namely, using a previously generated BAC transgenic line expressing Cre recombinase under the CCK promoter, we performed targeted manipulations of CCKVMH neurons. Histological and transcriptomic database analysis revealed extensive distribution of these neurons in the VMH, with significant heterogeneity in gene expression related to energy balance, including co-expression with PACAP and somatostatin. Pharmacogenetic acute inhibition via Designer Receptors Exclusively Activated by Designer Drugs (DREADDs) resulted in increased food intake and altered meal patterns, characterized by higher meal frequency and shorter intermeal intervals. Furthermore, diphtheria toxin-mediated ablation of CCKVMH neurons led to significant weight gain and hyperphagia over time, increasing meal size and duration. These mice also exhibited impaired glucose tolerance, indicative of disrupted glucose homeostasis. Our findings underscore the integral role of CCKVMH neurons in modulating feeding behavior, energy homeostasis, and glucose regulation. This study enhances our understanding of the neurohormonal mechanisms underlying obesity and metabolic disorders, providing potential targets for therapeutic interventions.
Collapse
Affiliation(s)
| | - Tommas J Ellender
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Jacek Szymanski
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | | |
Collapse
|
24
|
Kuckuck S, Lengton R, Meeusen RE, van der Valk ES, Hillegers MH, Penninx BW, Kavousi M, Visser JA, Boon MR, van den Berg SA, van Rossum EF. Perceived Stress, Hair Cortisol, and Hair Cortisone in Relation to Appetite-Regulating Hormones in Patients with Obesity. Obes Facts 2024; 18:206-214. [PMID: 39433032 PMCID: PMC12017749 DOI: 10.1159/000542079] [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: 05/30/2024] [Accepted: 10/13/2024] [Indexed: 10/23/2024] Open
Abstract
INTRODUCTION Stress predicts unhealthy eating, obesity, and metabolic deterioration, likely mediated by altered levels of appetite-regulating hormones. Yet, evidence regarding the association between long-term stress and levels of appetite-regulating hormones in humans is lacking. METHODS We included 65 patients with obesity (44 women) to investigate the cross-sectional association of long-term biological stress (scalp hair cortisol and cortisone) and long-term psychological stress (Perceived Stress Scale) with overnight-fasted serum levels of the hormonal appetite regulators leptin, adiponectin, insulin, pancreatic polypeptide, gastric-inhibitory peptide, peptide tyrosine-tyrosine, cholecystokinin and agouti-related protein, adjusted for age, sex, and body mass index. RESULTS Hair cortisone and, in trend, hair cortisol were positively associated with cholecystokinin (p = 0.003 and p = 0.058, respectively). No other associations between stress measures and hormonal appetite regulators were observed. CONCLUSION Long-term biological stress, measured using scalp hair glucocorticoid levels, is associated with elevated levels of circulating cholecystokinin. More research is needed to pinpoint potential effects on appetite.
Collapse
Affiliation(s)
- Susanne Kuckuck
- Division of Endocrinology, Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Obesity Center CGG, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Robin Lengton
- Division of Endocrinology, Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Obesity Center CGG, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Renate E.H. Meeusen
- Division of Endocrinology, Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Obesity Center CGG, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Eline S. van der Valk
- Division of Endocrinology, Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Obesity Center CGG, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Manon H.J. Hillegers
- Department of Child and Adolescent Psychiatry and Psychology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Brenda W.J.H. Penninx
- Department of Psychiatry and Amsterdam Public Health, Vrije Universiteit, Amsterdam, The Netherlands
| | - Maryam Kavousi
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Jenny A. Visser
- Division of Endocrinology, Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Obesity Center CGG, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Mariëtte R. Boon
- Division of Endocrinology, Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Obesity Center CGG, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Sjoerd A.A. van den Berg
- Division of Endocrinology, Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Clinical Chemistry, Erasmus MC, Rotterdam, The Netherlands
| | - Elisabeth F.C. van Rossum
- Division of Endocrinology, Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Obesity Center CGG, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
25
|
Jalševac F, Descamps-Solà M, Grau-Bové C, Segú H, Auguet T, Avilés-Jurado FX, Balaguer F, Jorba R, Beltrán-Debón R, Blay MT, Terra Barbadora X, Pinent M, Ardévol A. Profiling bitter taste receptors (TAS2R) along the gastrointestinal tract and their influence on enterohormone secretion. Gender- and age-related effects in the colon. Front Endocrinol (Lausanne) 2024; 15:1436580. [PMID: 39512758 PMCID: PMC11541047 DOI: 10.3389/fendo.2024.1436580] [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: 05/22/2024] [Accepted: 09/25/2024] [Indexed: 11/15/2024] Open
Abstract
Extraoral bitter taste receptors offer intriguing potential for modulating metabolism and the gut-brain axis through dietary interventions. Our understanding of these receptors is limited, and data on their effects on ageing are scarce. The complexity conveyed by their high diversity, low expression levels and species-dependent variability challenges our comprehension. We used real-time PCR to examine the relative abundance of multiple TAS2R across different segments of gastrointestinal mucosa in four human cohorts and related them to enteroendocrine secretions at the colon site. TAS2R14 exhibited the highest expression levels in all analyzed tissues. In contrast, TAS2R39, -38 and -42 consistently exhibited lower expression levels. Ageing was found to upregulate TAS2R4, -5, -13, -20 and GLP-1 mRNA in the descending colon. Stimulating TAS2R14 in Hutu-80 cells induced GLP-1 secretion, while stimulating TAS2R5 modulated GLP-1 and PYY secretion. Given the modifications TAS2R agonists may undergo along the GIT, as well as the distinctive expression patterns and possible functional roles of TAS2R receptors along the intestinal tract, our findings suggest the viability of a targeted strategy aimed at enhancing specific functions to improve health outcomes. This study offers valuable insights into the intricate interplay between bitter taste receptors, gut physiology and potential dietary interventions.
Collapse
Affiliation(s)
- Florijan Jalševac
- Universitat Rovira i Virgili, Department of Biochemistry and Biotechnology, MoBioFood Research Group, Tarragona, Spain
| | - Maria Descamps-Solà
- Universitat Rovira i Virgili, Department of Biochemistry and Biotechnology, MoBioFood Research Group, Tarragona, Spain
| | - Carme Grau-Bové
- Universitat Rovira i Virgili, Department of Biochemistry and Biotechnology, MoBioFood Research Group, Tarragona, Spain
| | - Helena Segú
- Universitat Rovira i Virgili, Department of Biochemistry and Biotechnology, MoBioFood Research Group, Tarragona, Spain
| | - Teresa Auguet
- Institut d’Investigació Sanitària Pere Virgili (IISPV), Hospital Joan XXIII, GEMMAIR Research Group, Tarragona, Spain
| | - Francesc Xavier Avilés-Jurado
- Institut d’Investigació Sanitària Pere Virgili (IISPV), Hospital Joan XXIII, Tarragona, Spain
- Head Neck Tumors Unit, Hospital Clínic de Barcelona, Universitat de Barcelona, IDIBAPS, Barcelona, Spain
| | - Francesc Balaguer
- Department of Gastroenterology, Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), University of Barcelona, Barcelona, Spain
| | - Rosa Jorba
- Institut d’Investigació Sanitària Pere Virgili (IISPV), Hospital Joan XXIII, Tarragona, Spain
| | - Raúl Beltrán-Debón
- Universitat Rovira i Virgili, Department of Biochemistry and Biotechnology, MoBioFood Research Group, Tarragona, Spain
- Institut d’Investigació Sanitària Pere Virgili (IISPV), Hospital Joan XXIII, MoBioFood Research Group, Tarragona, Spain
| | - Maria Teresa Blay
- Universitat Rovira i Virgili, Department of Biochemistry and Biotechnology, MoBioFood Research Group, Tarragona, Spain
- Institut d’Investigació Sanitària Pere Virgili (IISPV), Hospital Joan XXIII, MoBioFood Research Group, Tarragona, Spain
| | - Ximena Terra Barbadora
- Universitat Rovira i Virgili, Department of Biochemistry and Biotechnology, MoBioFood Research Group, Tarragona, Spain
- Institut d’Investigació Sanitària Pere Virgili (IISPV), Hospital Joan XXIII, MoBioFood Research Group, Tarragona, Spain
| | - Montserrat Pinent
- Universitat Rovira i Virgili, Department of Biochemistry and Biotechnology, MoBioFood Research Group, Tarragona, Spain
- Institut d’Investigació Sanitària Pere Virgili (IISPV), Hospital Joan XXIII, MoBioFood Research Group, Tarragona, Spain
| | - Anna Ardévol
- Universitat Rovira i Virgili, Department of Biochemistry and Biotechnology, MoBioFood Research Group, Tarragona, Spain
- Institut d’Investigació Sanitària Pere Virgili (IISPV), Hospital Joan XXIII, MoBioFood Research Group, Tarragona, Spain
| |
Collapse
|
26
|
Zhang M, Zhu L, Zhang H, Wang X, Wu G. Pea protein hydrolysate stimulates GLP-1 secretion in NCI-H716 cells via simultaneously activating the sensing receptors CaSR and PepT1. Food Funct 2024; 15:10316-10322. [PMID: 39302035 DOI: 10.1039/d4fo01290a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/22/2024]
Abstract
Glucagon-like peptide-1 (GLP-1) plays a crucial role in regulating glucose homeostasis by stimulating insulin secretion and suppressing glucagon release. Our previous study observed that pea protein hydrolysate (PPH) exhibited the function of triggering GLP-1 secretion. However, the underlying mechanisms have not been revealed. Herein, the mechanisms of PPH-stimulated GLP-1 secretion were investigated in NCI-H716 cells. The PPH-induced GLP-1 secretion was reduced (p < 0.05) after adding the sensing receptor antagonists NPS-2143 and 4-AMBA, indicating that activation of both calcium-sensing receptor (CaSR) and peptide-transporter 1 (PepT1) was involved in PPH-triggered GLP-1 release. Moreover, the intracellular Ca2+ level increased by 2.01 times during the PPH-induced GLP-1 secretion. Similarly, the cAMP content also increased by 1.43 times after stimulation by PPH. The RT-qPCR results showed that PPH increased the gene expression of prohormone convertase 1/3 (PCSK-1) by 2.79-fold, which effectively promoted the conversion of proglucagon (GCG) to GLP-1. The specific pathway of PPH-induced GLP-1 secretion may involve both CaSR and PepT1 activation-induced Ca2+ influx and cAMP generation, which effectively enhanced the enzyme activity of prohormone convertase 1/3 (PCSK-1) and ultimately promoted GLP-1 secretion.
Collapse
Affiliation(s)
- Mingkai Zhang
- National Engineering Research Center for Functional Food, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, School of Food Science and Technology, Jiangnan University, 1800 Lihu Road, Wuxi, 214122, China.
| | - Ling Zhu
- National Engineering Research Center for Functional Food, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, School of Food Science and Technology, Jiangnan University, 1800 Lihu Road, Wuxi, 214122, China.
| | - Hui Zhang
- National Engineering Research Center for Functional Food, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, School of Food Science and Technology, Jiangnan University, 1800 Lihu Road, Wuxi, 214122, China.
| | - Xingguo Wang
- National Engineering Research Center for Functional Food, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, School of Food Science and Technology, Jiangnan University, 1800 Lihu Road, Wuxi, 214122, China.
| | - Gangcheng Wu
- National Engineering Research Center for Functional Food, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, School of Food Science and Technology, Jiangnan University, 1800 Lihu Road, Wuxi, 214122, China.
| |
Collapse
|
27
|
Osakabe N, Ohmoto M, Shimizu T, Iida N, Fushimi T, Fujii Y, Abe K, Calabrese V. Gastrointestinal hormone-mediated beneficial bioactivities of bitter polyphenols. FOOD BIOSCI 2024; 61:104550. [DOI: 10.1016/j.fbio.2024.104550] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
28
|
Forst T, De Block C, Del Prato S, Armani S, Frias J, Lautenbach A, Ludvik B, Marinez M, Mathieu C, Müller TD, Schnell O. The role of incretin receptor agonists in the treatment of obesity. Diabetes Obes Metab 2024; 26:4178-4196. [PMID: 39072877 DOI: 10.1111/dom.15796] [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: 05/03/2024] [Revised: 06/28/2024] [Accepted: 06/29/2024] [Indexed: 07/30/2024]
Abstract
INTRODRODUCTION Obesity and its associated metabolic conditions have become a significant global health problem in recent years, with many people living with obesity fulfilling criteria for pharmacological treatment. The development of the glucagon-like peptide-1 receptor agonists for chronic weight management has triggered new interest in the incretins and other hormones as targets for obesity, and investigations into dual and triple co-agonists. METHODS The objective of this narrative review was to summarize the available data on approved and emerging incretin-based agents for the treatment of obesity. RESULTS In clinical trials of currently available agents in people with overweight or obesity, weight loss of between 6% and 21% of baseline body weight has been observed, with between 23% and 94% of participants achieving 10% or higher weight loss, depending on the study and the agent used. Favourable outcomes have also been seen with regard to cardiovascular risk and outcomes, diabetes prevention, metabolic dysfunction-associated steatotic liver disease/steatohepatitis and prevention of weight regain after metabolic surgery. Limitations associated with these agents include high costs, the potential for weight regain once treatment is stopped, the potential loss of lean body mass and gastrointestinal adverse events; potential issues with respect to gallbladder and biliary diseases require further investigation. CONCLUSIONS Many dual and triple co-agonists are still in development, and more data are needed to assess the efficacy, safety and tolerability of these emerging therapies versus the established incretin-based therapies; however, data are promising, and further results are eagerly awaited.
Collapse
Affiliation(s)
- Thomas Forst
- CRS Clinical Research Services GmbH, Mannheim, Germany
| | | | - Stefano Del Prato
- Interdisciplinary Research Center "Health Science," Sant'Anna School of Advanced Studies, Pisa, Italy
| | - Sara Armani
- CRS Clinical Research Services GmbH, Mannheim, Germany
| | - Juan Frias
- Biomea Fusion, Redwood City, California, USA
| | - Anne Lautenbach
- University Medical-Center Hamburg-Eppendorf, Hamburg, Germany
| | - Bernhard Ludvik
- Landstrasse Clinic and Karl Landsteiner Institute for Obesity and Metabolic Disorders, Vienna, Austria
| | | | | | - Timo D Müller
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Munich, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Walther-Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-Universität München (LMU), Munich, Germany
| | - Oliver Schnell
- Forschergruppe Diabetes e.V. at the Helmholtz Center Munich, Munich, Germany
| |
Collapse
|
29
|
Baccari MC, Vannucchi MG, Idrizaj E. The Possible Involvement of Glucagon-like Peptide-2 in the Regulation of Food Intake through the Gut-Brain Axis. Nutrients 2024; 16:3069. [PMID: 39339669 PMCID: PMC11435434 DOI: 10.3390/nu16183069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 09/05/2024] [Accepted: 09/10/2024] [Indexed: 09/30/2024] Open
Abstract
Food intake regulation is a complex mechanism involving the interaction between central and peripheral structures. Among the latter, the gastrointestinal tract represents one of the main sources of both nervous and hormonal signals, which reach the central nervous system that integrates them and sends the resulting information downstream to effector organs involved in energy homeostasis. Gut hormones released by nutrient-sensing enteroendocrine cells can send signals to central structures involved in the regulation of food intake through more than one mechanism. One of these is through the modulation of gastric motor phenomena known to be a source of peripheral satiety signals. In the present review, our attention will be focused on the ability of the glucagon-like peptide 2 (GLP-2) hormone to modulate gastrointestinal motor activity and discuss how its effects could be related to peripheral satiety signals generated in the stomach and involved in the regulation of food intake through the gut-brain axis. A better understanding of the possible role of GLP-2 in regulating food intake through the gut-brain axis could represent a starting point for the development of new strategies to treat some pathological conditions, such as obesity.
Collapse
Affiliation(s)
- Maria Caterina Baccari
- Department of Experimental & Clinical Medicine, Section of Physiological Sciences, University of Florence, 50134 Florence, Italy;
| | - Maria Giuliana Vannucchi
- Department of Experimental & Clinical Medicine, Research Unit of Histology & Embryology, University of Florence, 50139 Florence, Italy;
| | - Eglantina Idrizaj
- Department of Experimental & Clinical Medicine, Section of Physiological Sciences, University of Florence, 50134 Florence, Italy;
| |
Collapse
|
30
|
Zago L, Pessoa HR, Rosado CP, da Silva AA, Pasqualone A, Koury JC. Acute Consumption of Cooked Green Banana Pulp Beverage (Musa cavendishii) Decreases Plasma Glucose in Healthy Women: A Cross-Sectional Controlled Study. PLANT FOODS FOR HUMAN NUTRITION (DORDRECHT, NETHERLANDS) 2024; 79:641-647. [PMID: 38951375 DOI: 10.1007/s11130-024-01202-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 06/11/2024] [Indexed: 07/03/2024]
Abstract
This study aimed at comparing the carbohydrate composition of three banana varieties (cv. Nanica, Nanicão, and Prata) and investigating the effect of a single dose of cooked green banana pulp beverage (GBPd) on plasma glycemic homeostasis indexes (glucose, PYY, GIP, insulin) and hunger and satiety sensation (visual analog scale-VAS). The bananas were classified according to the color scale. The fiber, total carbohydrate, and resistant starch (RS) were determined using validated methods. Glucose homeostasis indexes and hunger/satiety sensation were determined in ten healthy women in two stages before and after intake: (1) glucose solution (250 g/L); (2) one week later, consumption of the glucose solution plus 75 g/L of GBPd. Blood samples were collected twice in stage-1 and every 15 min for 2 h in stage-2. Cv. Nanicão was selected, because it presented a higher content in RS and dietary fiber on dry base than the other cultivars. Thus, it was used to test glycemic response. After 2 h of GBPd intake, no difference was observed in hunger/satiety sensation and plasma glycemic homeostasis indexes, except for a decrease in plasma glucose concentration (-15%, p = 0.0232) compared to stage-1. These results suggest that cv. Nanicão has a higher potential as a functional ingredient and can influence the reduction in the glycemic index of a meal compared to other cultivars. However, it had not a short-term effect on hormones GIP and PYY in healthy women. Further research is needed to understand the long-term effects and mechanisms of green banana on glycemic control and satiety.
Collapse
Affiliation(s)
- Lilia Zago
- Institute of Nutrition, State University of Rio de Janeiro (UERJ), Rua São Francisco Xavier, 524, 12º andar, sala 12006 D - Maracanã, Rio de Janeiro, RJ, Brazil.
| | - Heloisa Rodrigues Pessoa
- Institute of Nutrition, State University of Rio de Janeiro (UERJ), Rua São Francisco Xavier, 524, 12º andar, sala 12006 D - Maracanã, Rio de Janeiro, RJ, Brazil
| | - Carolyne Pimentel Rosado
- Institute of Nutrition, State University of Rio de Janeiro (UERJ), Rua São Francisco Xavier, 524, 12º andar, sala 12006 D - Maracanã, Rio de Janeiro, RJ, Brazil
| | - Andreia Ana da Silva
- Institute of Nutrition, State University of Rio de Janeiro (UERJ), Rua São Francisco Xavier, 524, 12º andar, sala 12006 D - Maracanã, Rio de Janeiro, RJ, Brazil
| | - Antonella Pasqualone
- Department of Soil, Plant and Food Science, University of Bari Aldo Moro, Bari, Italy
| | - Josely Correa Koury
- Institute of Nutrition, State University of Rio de Janeiro (UERJ), Rua São Francisco Xavier, 524, 12º andar, sala 12006 D - Maracanã, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
31
|
Anjom-Shoae J, Fitzgerald PC, Horowitz M, Mohammadpour Z, Hall GV, Holst JJ, Rehfeld JF, Veedfald S, Feinle-Bisset C. Intraduodenal calcium enhances the effects of L-tryptophan to stimulate gut hormone secretion and suppress energy intake in healthy males: a randomized, crossover, clinical trial. Am J Clin Nutr 2024; 120:528-539. [PMID: 38996913 DOI: 10.1016/j.ajcnut.2024.07.006] [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: 03/21/2024] [Revised: 06/05/2024] [Accepted: 07/08/2024] [Indexed: 07/14/2024] Open
Abstract
BACKGROUND In humans, intraduodenal infusion of L-tryptophan (Trp) increases plasma concentrations of gastrointestinal hormones and stimulates pyloric pressures, both key determinants of gastric emptying and associated with potent suppression of energy intake. The stimulation of gastrointestinal hormones by Trp has been shown, in preclinical studies, to be enhanced by extracellular calcium and mediated in part by the calcium-sensing receptor. OBJECTIVES This study aim was to determine whether intraduodenal calcium can enhance the effects of Trp to stimulate gastrointestinal hormones and pyloric pressures and, if so, whether it is associated with greater suppression of energy intake, in healthy males. METHODS Fifteen males with normal weight (mean ± standard deviation; age: 26 ± 7 years; body mass index: 22 ± 2 kg/m2), received on 3 separate occasions, 150-min intraduodenal infusions of 0, 500, or 1000 mg calcium (Ca), each combined with Trp (load: 0.1 kcal/min, with submaximal energy intake-suppressant effects) from t = 75-150 min, in a randomized, double-blind, crossover study. Plasma concentrations of GI hormones [gastrin, cholecystokinin, glucose-dependent insulinotropic polypeptide (GIP), glucagon-like peptide (GLP)-1, and peptide tyrosine-tyrosine (PYY)], and Trp and antropyloroduodenal pressures were measured throughout. Immediately postinfusions (t = 150-180 min), energy intake at a standardized buffet-style meal was quantified. RESULTS In response to calcium alone, both 500- and 1000-mg doses stimulated PYY, while only the 1000-mg dose stimulated GLP-1 and pyloric pressures (all P < 0.05). The 1000-mg dose also enhanced the effects of Trp to stimulate cholecystokinin and GLP-1, and both doses stimulated PYY but, surprisingly, reduced the stimulation of GIP (all P < 0.05). Both doses substantially and dose dependently enhanced the effects of Trp to suppress energy intake (Ca-0+Trp: 1108 ± 70 kcal; Ca-500+Trp: 961 ± 90 kcal; and Ca-1000+Trp: 922 ± 96 kcal; P < 0.05). CONCLUSIONS Intraduodenal administration of calcium enhances the effect of Trp to stimulate plasma cholecystokinin, GLP-1, and PYY and suppress energy intake in healthy males. These findings have potential implications for novel nutrient-based approaches to energy intake regulation in obesity. The trial was registered at the Australian New Zealand Clinical Trial Registry (www.anzctr.org.au) as ACTRN12620001294943).
Collapse
Affiliation(s)
- Javad Anjom-Shoae
- Adelaide Medical School and Center of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, Australia
| | - Penelope Ce Fitzgerald
- Adelaide Medical School and Center of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, Australia
| | - Michael Horowitz
- Adelaide Medical School and Center of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, Australia; Endocrine and Metabolic Unit, Royal Adelaide Hospital, Adelaide, Australia
| | - Zinat Mohammadpour
- South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Gerrit van Hall
- Department of Biomedical Sciences and the Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark; Department of Clinical Biochemistry, Rigshospitalet, Copenhagen, Denmark
| | - Jens J Holst
- Department of Biomedical Sciences and the Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Jens F Rehfeld
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen, Denmark
| | - Simon Veedfald
- Department of Biomedical Sciences and the Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Christine Feinle-Bisset
- Adelaide Medical School and Center of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, Australia.
| |
Collapse
|
32
|
Xu M, Chen B, Qiao K, Liu S, Su Y, Cai S, Liu Z, Li L, Li Q. Mechanism of Takifugu bimaculatus Skin Peptides in Alleviating Hyperglycemia in Rats with Type 2 Diabetic Mellitus Based on Microbiome and Metabolome Analyses. Mar Drugs 2024; 22:377. [PMID: 39195493 DOI: 10.3390/md22080377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 08/09/2024] [Accepted: 08/21/2024] [Indexed: 08/29/2024] Open
Abstract
In this study, we aimed to explore the hypoglycemic effects of a hydrolysate on Takifugu bimaculatus skin (TBSH). The effect of the dipeptidyl peptidase-IV (DPP-IV) inhibitory activities from different TBSH fractions was investigated on basic indexes, gut hormones, blood lipid indexes, viscera, and the gut microbiota and its metabolites in rats with type 2 diabetes mellitus (T2DM). The results showed that the <1 kDa peptide fraction from TBSH (TBP) exhibited a more potent DPP-IV inhibitory effect (IC50 = 0.45 ± 0.01 mg/mL). T2DM rats were induced with streptozocin, followed by the administration of TBP. The 200 mg/kg TBP mitigated weight loss, lowered fasting blood glucose levels, and increased insulin secretion by 20.47%, 25.23%, and 34.55%, respectively, rectified irregular hormonal fluctuations, lipid metabolism, and tissue injuries, and effectively remedied gut microbiota imbalance. In conclusion, TBP exerts a hypoglycemic effect in rats with T2DM. This study offers the potential to develop nutritional supplements to treat T2DM and further promote the high-value utilization of processing byproducts from T. bimaculatus. It will provide information for developing nutritional supplements to treat T2DM and further promote the high-value utilization of processing byproducts from T. bimaculatus.
Collapse
Affiliation(s)
- Min Xu
- College of Ocean Food and Bioengineering, Jimei University, Xiamen 361021, China
- Key Laboratory of Cultivation and High-Value Utilization of Marine Organisms in Fujian Province, Fisheries Research Institute of Fujian, Xiamen 361013, China
| | - Bei Chen
- Key Laboratory of Cultivation and High-Value Utilization of Marine Organisms in Fujian Province, Fisheries Research Institute of Fujian, Xiamen 361013, China
| | - Kun Qiao
- Key Laboratory of Cultivation and High-Value Utilization of Marine Organisms in Fujian Province, Fisheries Research Institute of Fujian, Xiamen 361013, China
| | - Shuji Liu
- Key Laboratory of Cultivation and High-Value Utilization of Marine Organisms in Fujian Province, Fisheries Research Institute of Fujian, Xiamen 361013, China
| | - Yongchang Su
- Key Laboratory of Cultivation and High-Value Utilization of Marine Organisms in Fujian Province, Fisheries Research Institute of Fujian, Xiamen 361013, China
| | - Shuilin Cai
- Key Laboratory of Cultivation and High-Value Utilization of Marine Organisms in Fujian Province, Fisheries Research Institute of Fujian, Xiamen 361013, China
| | - Zhiyu Liu
- Key Laboratory of Cultivation and High-Value Utilization of Marine Organisms in Fujian Province, Fisheries Research Institute of Fujian, Xiamen 361013, China
| | - Lijun Li
- College of Ocean Food and Bioengineering, Jimei University, Xiamen 361021, China
| | - Qingbiao Li
- College of Ocean Food and Bioengineering, Jimei University, Xiamen 361021, China
| |
Collapse
|
33
|
Driva S, Korkontzelou A, Tonstad S, Tentolouris N, Litsiou E, Vasileiou V, Vassiliou AG, Saltagianni V, Katsaounou P. Metabolic Changes Following Smoking Cessation in Patients with Type 2 Diabetes Mellitus. Biomedicines 2024; 12:1882. [PMID: 39200346 PMCID: PMC11352061 DOI: 10.3390/biomedicines12081882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 08/08/2024] [Accepted: 08/14/2024] [Indexed: 09/02/2024] Open
Abstract
BACKGROUND Smoking cessation is crucial for reducing complications of type 2 diabetes mellitus (T2DM), but associated weight gain can worsen glycemic control, discouraging quitting attempts. Varenicline, a partial agonist of α4β2 nicotinic receptors, aids smoking cessation. This study examines the effects of varenicline on body weight and metabolic parameters in patients with T2DM and prediabetes. METHODS Fifty-three patients were enrolled, of which 32 successfully quit smoking after a three-month course of varenicline and were examined after an additional month with no medication. Measurements taken at baseline, 2.5 months, and 4 months included body weight, blood pressure, resting metabolic rate (RMR), glycated hemoglobin (HbA1c), fasting glucose, blood lipids, C-reactive protein (CRP), appetite-related hormones, and physical activity. RESULTS Post-treatment, there were no significant changes in body weight, blood pressure, RMR, or glycemic control. Total (CHOL) and low-density lipoprotein (LDL-C) cholesterol decreased significantly at 4 months of the study (from 168 to 156 mg/dL, p = 0.013, and from 96 to 83 mg/dL, p = 0.013, respectively). Leptin levels increased (from 11 to 13.8 ng/dL, p = 0.004), as did glucagon-like peptide-1 (GLP-1) levels (from 39.6 to 45.8 pM, p = 0.016) at 4 months of follow-up. The percentage of participants who reported moderate-intensity activity increased from 28% to 56%, while those reporting high-intensity activity increased from 19% to 22%, respectively (p = 0.039). CONCLUSIONS Our study showed that smoking cessation with varenicline in smokers with T2DM and prediabetes led to significant improvements in lipid profile, significant increase in plasma leptin and GLP-1 levels, and increased physical activity, without significant weight gain. Thus, smoking cessation without weight gain or deteriorated glycemic control is feasible for these smokers, with added benefits to lipid profiles, GLP-1 regulation, and physical activity.
Collapse
Affiliation(s)
- Stamatina Driva
- Smoking Cessation Outpatient Clinic, Respiratory Department, First Intensive Care Unit, Evangelismos General Hospital, 10676 Athens, Greece (E.L.)
- Diabetes Centre, First Department of Propaedeutic Internal Medicine, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, 11527 Athens, Greece
- Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Aliki Korkontzelou
- Smoking Cessation Outpatient Clinic, Respiratory Department, First Intensive Care Unit, Evangelismos General Hospital, 10676 Athens, Greece (E.L.)
- Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Serena Tonstad
- Department of Preventive Cardiology, Oslo University Hospital, 0424 Oslo, Norway
| | - Nikolaos Tentolouris
- Diabetes Centre, First Department of Propaedeutic Internal Medicine, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, 11527 Athens, Greece
| | - Eleni Litsiou
- Smoking Cessation Outpatient Clinic, Respiratory Department, First Intensive Care Unit, Evangelismos General Hospital, 10676 Athens, Greece (E.L.)
| | - Vasiliki Vasileiou
- Endocrinology Department, Diabetes Center, Alexandra Hospital, 11528 Athens, Greece
| | - Alice G. Vassiliou
- Smoking Cessation Outpatient Clinic, Respiratory Department, First Intensive Care Unit, Evangelismos General Hospital, 10676 Athens, Greece (E.L.)
| | - Vassiliki Saltagianni
- Smoking Cessation Outpatient Clinic, Respiratory Department, First Intensive Care Unit, Evangelismos General Hospital, 10676 Athens, Greece (E.L.)
| | - Paraskevi Katsaounou
- Smoking Cessation Outpatient Clinic, Respiratory Department, First Intensive Care Unit, Evangelismos General Hospital, 10676 Athens, Greece (E.L.)
| |
Collapse
|
34
|
Pepin X, Arora S, Borges L, Cano-Vega M, Carducci T, Chatterjee P, Chen G, Cristofoletti R, Dallmann A, Delvadia P, Dressman J, Fotaki N, Gray E, Heimbach T, Holte Ø, Kijima S, Kotzagiorgis E, Lennernäs H, Lindahl A, Loebenberg R, Mackie C, Malamatari M, McAllister M, Mitra A, Moody R, Mudie D, Musuamba Tshinanu F, Polli JE, Rege B, Ren X, Rullo G, Scherholz M, Song I, Stillhart C, Suarez-Sharp S, Tannergren C, Tsakalozou E, Veerasingham S, Wagner C, Seo P. Parameterization of Physiologically Based Biopharmaceutics Models: Workshop Summary Report. Mol Pharm 2024; 21:3697-3731. [PMID: 38946085 PMCID: PMC11304397 DOI: 10.1021/acs.molpharmaceut.4c00526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 06/18/2024] [Accepted: 06/18/2024] [Indexed: 07/02/2024]
Abstract
This Article shares the proceedings from the August 29th, 2023 (day 1) workshop "Physiologically Based Biopharmaceutics Modeling (PBBM) Best Practices for Drug Product Quality: Regulatory and Industry Perspectives". The focus of the day was on model parametrization; regulatory authorities from Canada, the USA, Sweden, Belgium, and Norway presented their views on PBBM case studies submitted by industry members of the IQ consortium. The presentations shared key questions raised by regulators during the mock exercise, regarding the PBBM input parameters and their justification. These presentations also shed light on the regulatory assessment processes, content, and format requirements for future PBBM regulatory submissions. In addition, the day 1 breakout presentations and discussions gave the opportunity to share best practices around key questions faced by scientists when parametrizing PBBMs. Key questions included measurement and integration of drug substance solubility for crystalline vs amorphous drugs; impact of excipients on apparent drug solubility/supersaturation; modeling of acid-base reactions at the surface of the dissolving drug; choice of dissolution methods according to the formulation and drug properties with a view to predict the in vivo performance; mechanistic modeling of in vitro product dissolution data to predict in vivo dissolution for various patient populations/species; best practices for characterization of drug precipitation from simple or complex formulations and integration of the data in PBBM; incorporation of drug permeability into PBBM for various routes of uptake and prediction of permeability along the GI tract.
Collapse
Affiliation(s)
- Xavier Pepin
- Regulatory
Affairs, Simulations Plus Inc., 42505 10th Street West, Lancaster, California 93534-7059, United States
| | - Sumit Arora
- Janssen
Pharmaceutica NV, Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Luiza Borges
- ANVISA, SIA Trecho 5́, Guara, Brasília, Federal District 71205-050, Brazil
| | - Mario Cano-Vega
- Drug
Product Technologies, Amgen Inc., Thousand Oaks, California 91320-1799, United
States
| | - Tessa Carducci
- Analytical
Commercialization Technology, Merck & Co., Inc., 126 E. Lincoln Ave., Rahway, New Jersey 07065, United States
| | - Parnali Chatterjee
- Office
of
Pharmaceutical Quality (OPQ), Center for Drug Evaluation and Research
(CDER), Food and Drug Administration (FDA), Silver Spring, Maryland 20903-1058, United
States
| | - Grace Chen
- Takeda
Development Center Americas Inc., 300 Shire Way, Lexington, Massachusetts 02421, United States
| | - Rodrigo Cristofoletti
- College
of Pharmacy, University of Florida, 6550 Sanger Rd., Orlando, Florida 32827, United States
| | - André Dallmann
- Bayer
HealthCare SAS, 59000 Lille, France, on behalf of Bayer
AG, Pharmacometrics/Modeling and Simulation, Systems Pharmacology
& Medicine, PBPK, Leverkusen, Germany
| | - Poonam Delvadia
- Office
of Translational Science, Office of Clinical Pharmacology (OCP), Center
for Drug Evaluation and Research (CDER), Food and Drug Administration (FDA), Silver Spring, Maryland 20903-1058, United States
| | - Jennifer Dressman
- Fraunhofer Institute of Translational Medicine and Pharmacology, Frankfurt am Main 60596, Germany
| | - Nikoletta Fotaki
- University of Bath, Claverton Down, Bath BA2
7AY, United Kingdom
| | - Elizabeth Gray
- Office
of
Pharmaceutical Quality (OPQ), Center for Drug Evaluation and Research
(CDER), Food and Drug Administration (FDA), Silver Spring, Maryland 20903-1058, United
States
| | - Tycho Heimbach
- Pharmaceutical Sciences and Clinical Supply, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Øyvind Holte
- Norwegian Medical Products Agency, Oslo 0213, Norway
| | - Shinichi Kijima
- Office
of New Drug V, Pharmaceuticals and Medical
Devices Agency (PMDA), Tokyo 100-0013, Japan
| | - Evangelos Kotzagiorgis
- European Medicines Agency (EMA), Domenico Scarlattilaan 6, Amsterdam 1083 HS, The Netherlands
| | - Hans Lennernäs
- Translational
Drug Discovery and Development, Department of Pharmaceutical Bioscience, Uppsala University, Uppsala 751 05, Sweden
| | | | - Raimar Loebenberg
- Faculty
of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmontonton T6G 2E1, Canada
| | - Claire Mackie
- Janssen
Pharmaceutica NV, Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Maria Malamatari
- Medicines & Healthcare Products Regulatory Agency, 10 S Colonnade, London SW1W 9SZ, United Kingdom
| | - Mark McAllister
- Global
Biopharmaceutics, Drug Product Design, Pfizer, Sandwich CT13 9NJ, United Kingdom
| | - Amitava Mitra
- Clinical
Pharmacology, Kura Oncology Inc., Boston, Massachusetts 02210, United States
| | - Rebecca Moody
- Office
of
Pharmaceutical Quality (OPQ), Center for Drug Evaluation and Research
(CDER), Food and Drug Administration (FDA), Silver Spring, Maryland 20903-1058, United
States
| | - Deanna Mudie
- Global
Research and Development, Small Molecules, Lonza, 63045 NE Corporate
Pl., Bend, Oregon 97701, United States
| | - Flora Musuamba Tshinanu
- Belgian Federal Agency for Medicines and Health Products, Galileelaan 5/03, Brussel 1210, Belgium
| | - James E. Polli
- School
of Pharmacy, University of Maryland, Baltimore, Maryland 21201, United States
| | - Bhagwant Rege
- Office
of
Pharmaceutical Quality (OPQ), Center for Drug Evaluation and Research
(CDER), Food and Drug Administration (FDA), Silver Spring, Maryland 20903-1058, United
States
| | - Xiaojun Ren
- PK
Sciences/Translational Medicine, BioMedical Research, Novartis, One Health Plaza, East Hanover, New Jersey 07936, United States
| | - Gregory Rullo
- Regulatory
CMC, AstraZeneca, 1 Medimmune Way, Gaithersburg, Maryland 20878, United States
| | - Megerle Scherholz
- Pharmaceutical
Development, Bristol Myers Squibb, Route 206 & Province Line Road, Princeton, New Jersey 08543, United States
| | - Ivy Song
- Takeda
Development Center Americas Inc., 300 Shire Way, Lexington, Massachusetts 02421, United States
| | - Cordula Stillhart
- Pharmaceutical
R&D, F. Hoffmann-La Roche Ltd., Basel 4070, Switzerland
| | - Sandra Suarez-Sharp
- Regulatory
Affairs, Simulations Plus Inc., 42505 10th Street West, Lancaster, California 93534-7059, United States
| | - Christer Tannergren
- Biopharmaceutics
Science, New Modalities & Parenteral Product Development, Pharmaceutical
Technology & Development, Operations, AstraZeneca, Gothenburg 431 50, Sweden
| | - Eleftheria Tsakalozou
- Division
of Quantitative Methods and Modeling, Office of Research and Standards,
Office of Generic Drugs, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland 20903-1058, United
States
| | - Shereeni Veerasingham
- Pharmaceutical
Drugs Directorate (PDD), Health Canada, 1600 Scott St., Ottawa K1A 0K9, Canada
| | - Christian Wagner
- Global
Drug Product Development, Global CMC Development, the Healthcare Business of Merck KGaA, Darmstadt D-64293, Germany
| | - Paul Seo
- Office
of Translational Science, Office of Clinical Pharmacology (OCP), Center
for Drug Evaluation and Research (CDER), Food and Drug Administration (FDA), Silver Spring, Maryland 20903-1058, United States
| |
Collapse
|
35
|
Fotouhi Ardakani A, Anjom-Shoae J, Sadeghi O, Marathe CS, Feinle-Bisset C, Horowitz M. Association between total, animal, and plant protein intake and type 2 diabetes risk in adults: A systematic review and dose-response meta-analysis of prospective cohort studies. Clin Nutr 2024; 43:1941-1955. [PMID: 39032197 DOI: 10.1016/j.clnu.2024.07.001] [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: 12/26/2023] [Revised: 06/04/2024] [Accepted: 07/03/2024] [Indexed: 07/22/2024]
Abstract
BACKGROUND AND AIMS While clinical studies indicate that dietary protein may benefit glucose homeostasis in type 2 diabetes (T2D), the impact of dietary protein, including whether the protein is of animal or plant origin, on the risk of T2D is uncertain. We conducted a systematic review and meta-analysis to evaluate the associations of total, animal, and plant protein intakes with the risk of T2D. METHODS A systematic search was performed using multiple data sources, including PubMed/Medline, ISI Web of Science, Scopus, and Google Scholar, with the data cut-off in May 2023. Our selection criteria focused on prospective cohort studies that reported risk estimates for the association between protein intake and T2D risk. For data synthesis, we calculated summary relative risks and 95% confidence intervals for the highest versus lowest categories of protein intake using random-effects models. Furthermore, we conducted both linear and non-linear dose-response analyses to assess the dose-response associations between protein intake and T2D risk. RESULTS Sixteen prospective cohort studies, involving 615,125 participants and 52,342 T2D cases, were identified, of which eleven studies reported data on intake of both animal and plant protein. Intakes of total (pooled effect size: 1.14, 95% CI: 1.04-1.24) and animal (pooled effect size: 1.18, 95% CI: 1.09-1.27) protein were associated with an increased risk of T2D. These effects were dose-related - each 20-g increase in total or animal protein intake increased the risk of T2D by ∼3% and ∼7%, respectively. In contrast, there was no association between intake of plant protein and T2D risk (pooled effect size: 0.98, 95% CI: 0.89-1.08), while replacing animal with plant protein intake (per each 20 g) was associated with a reduced risk of T2D (pooled effect size: 0.80, 95% CI: 0.76-0.84). CONCLUSIONS Our findings indicate that long-term consumption of animal, but not plant, protein is associated with a significant and dose-dependent increase in the risk of T2D, with the implication that replacement of animal with plant protein intake may lower the risk of T2D.
Collapse
Affiliation(s)
- Amirmahdi Fotouhi Ardakani
- Student Research Committee, School of Public Health, Kerman University of Medical Sciences, Kerman, Iran; Student Research Committee, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Javad Anjom-Shoae
- Adelaide Medical School, University of Adelaide, Adelaide, Australia; Centre of Research Excellence in Translating Nutritional Sciences to Good Health, University of Adelaide, Adelaide, Australia
| | - Omid Sadeghi
- Nutrition and Food Security Research Centre and Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Chinmay S Marathe
- Adelaide Medical School, University of Adelaide, Adelaide, Australia; Centre of Research Excellence in Translating Nutritional Sciences to Good Health, University of Adelaide, Adelaide, Australia; Endocrine and Metabolic Unit, Royal Adelaide Hospital, Adelaide, Australia.
| | - Christine Feinle-Bisset
- Adelaide Medical School, University of Adelaide, Adelaide, Australia; Centre of Research Excellence in Translating Nutritional Sciences to Good Health, University of Adelaide, Adelaide, Australia
| | - Michael Horowitz
- Adelaide Medical School, University of Adelaide, Adelaide, Australia; Centre of Research Excellence in Translating Nutritional Sciences to Good Health, University of Adelaide, Adelaide, Australia; Endocrine and Metabolic Unit, Royal Adelaide Hospital, Adelaide, Australia
| |
Collapse
|
36
|
Wang R, Mijiti S, Xu Q, Liu Y, Deng C, Huang J, Yasheng A, Tian Y, Cao Y, Su Y. The Potential Mechanism of Remission in Type 2 Diabetes Mellitus After Vertical Sleeve Gastrectomy. Obes Surg 2024; 34:3071-3083. [PMID: 38951388 DOI: 10.1007/s11695-024-07378-z] [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: 03/03/2024] [Revised: 06/13/2024] [Accepted: 06/19/2024] [Indexed: 07/03/2024]
Abstract
In recent years, there has been a gradual increase in the prevalence of obesity and type 2 diabetes mellitus (T2DM), with bariatric surgery remaining the most effective treatment strategy for these conditions. Vertical sleeve gastrectomy (VSG) has emerged as the most popular surgical procedure for bariatric/metabolic surgeries, effectively promoting weight loss and improving or curing T2DM. The alterations in the gastrointestinal tract following VSG may improve insulin secretion and resistance by increasing incretin secretion (especially GLP-1), modifying the gut microbiota composition, and through mechanisms dependent on weight loss. This review focuses on the potential mechanisms through which the enhanced action of incretin and metabolic changes in the digestive system after VSG may contribute to the remission of T2DM.
Collapse
Affiliation(s)
- Rongfei Wang
- Department of Gastrointestinal Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, No.57 Mei Hua East Road, Xiang Zhou District, Zhuhai, 519000, Guangdong, China
| | - Salamu Mijiti
- Department of General Surgery, The First People's Hospital of Kashi, Autonomous Region, Kashi, 844000, Xinjiang Uygur, China
| | - Qilin Xu
- Department of General Surgery, The First People's Hospital of Kashi, Autonomous Region, Kashi, 844000, Xinjiang Uygur, China
| | - Yile Liu
- Department of Gastrointestinal Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, No.57 Mei Hua East Road, Xiang Zhou District, Zhuhai, 519000, Guangdong, China
| | - Chaolun Deng
- Department of Gastrointestinal Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, No.57 Mei Hua East Road, Xiang Zhou District, Zhuhai, 519000, Guangdong, China
| | - Jiangtao Huang
- Department of Gastrointestinal Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, No.57 Mei Hua East Road, Xiang Zhou District, Zhuhai, 519000, Guangdong, China
| | - Abudoukeyimu Yasheng
- Department of General Surgery, The First People's Hospital of Kashi, Autonomous Region, Kashi, 844000, Xinjiang Uygur, China
| | - Yunping Tian
- Department of General Surgery, The First People's Hospital of Kashi, Autonomous Region, Kashi, 844000, Xinjiang Uygur, China.
| | - Yanlong Cao
- Department of General Surgery, The First People's Hospital of Kashi, Autonomous Region, Kashi, 844000, Xinjiang Uygur, China.
| | - Yonghui Su
- Department of Gastrointestinal Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, No.57 Mei Hua East Road, Xiang Zhou District, Zhuhai, 519000, Guangdong, China.
| |
Collapse
|
37
|
Anjom-Shoae J, Feinle-Bisset C, Horowitz M. Impacts of dietary animal and plant protein on weight and glycemic control in health, obesity and type 2 diabetes: friend or foe? Front Endocrinol (Lausanne) 2024; 15:1412182. [PMID: 39145315 PMCID: PMC11321983 DOI: 10.3389/fendo.2024.1412182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 07/17/2024] [Indexed: 08/16/2024] Open
Abstract
It is well established that high-protein diets (i.e. ~25-30% of energy intake from protein) provide benefits for achieving weight loss, and subsequent weight maintenance, in individuals with obesity, and improve glycemic control in type 2 diabetes (T2D). These effects may be attributable to the superior satiating property of protein, at least in part, through stimulation of both gastrointestinal (GI) mechanisms by protein, involving GI hormone release and slowing of gastric emptying, as well as post-absorptive mechanisms facilitated by circulating amino acids. In contrast, there is evidence that the beneficial effects of greater protein intake on body weight and glycemia may only be sustained for 6-12 months. While both suboptimal dietary compliance and metabolic adaptation, as well as substantial limitations in the design of longer-term studies are all likely to contribute to this contradiction, the source of dietary protein (i.e. animal vs. plant) has received inappropriately little attention. This issue has been highlighted by outcomes of recent epidemiological studies indicating that long-term consumption of animal-based protein may have adverse effects in relation to the development of obesity and T2D, while plant-based protein showed either protective or neutral effects. This review examines information relating to the effects of dietary protein on appetite, energy intake and postprandial glycemia, and the relevant GI functions, as reported in acute, intermediate- and long-term studies in humans. We also evaluate knowledge relating to the relevance of the dietary protein source, specifically animal or plant, to the prevention, and management, of obesity and T2D.
Collapse
Affiliation(s)
- Javad Anjom-Shoae
- Adelaide Medical School and Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, SA, Australia
| | - Christine Feinle-Bisset
- Adelaide Medical School and Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, SA, Australia
| | - Michael Horowitz
- Adelaide Medical School and Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, SA, Australia
- Endocrine and Metabolic Unit, Royal Adelaide Hospital, Adelaide, SA, Australia
| |
Collapse
|
38
|
Lin Z, Sun L. Research advances in the therapy of metabolic syndrome. Front Pharmacol 2024; 15:1364881. [PMID: 39139641 PMCID: PMC11319131 DOI: 10.3389/fphar.2024.1364881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 07/08/2024] [Indexed: 08/15/2024] Open
Abstract
Metabolic syndrome refers to the pathological state of metabolic disorder of protein, fat, carbohydrate, and other substances in the human body. It is a syndrome composed of a group of complex metabolic disorders, whose pathogenesis includes multiple genetic and acquired entities falling under the category of insulin resistance and chronic low-grade inflammationand. It is a risk factor for increased prevalence and mortality from diabetes and cardiovascular disease. Cardiovascular diseases are the predominant cause of morbidity and mortality globally, thus it is imperative to investigate the impact of metabolic syndrome on alleviating this substantial disease burden. Despite the increasing number of scientists dedicating themselves to researching metabolic syndrome in recent decades, numerous aspects of this condition remain incompletely understood, leaving many questions unanswered. In this review, we present an epidemiological analysis of MetS, explore both traditional and novel pathogenesis, examine the pathophysiological repercussions of metabolic syndrome, summarize research advances, and elucidate the mechanisms underlying corresponding treatment approaches.
Collapse
Affiliation(s)
- Zitian Lin
- Edinburgh Medical School, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, United Kingdom
- Zhejiang University-University of Edinburgh Institute, International Campus, Zhejiang University, Haining, China
| | - Luning Sun
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, China
| |
Collapse
|
39
|
Mohammadhasani K, Fard MV, Yadegari M, Barati M, Bahari H, Nattagh-Eshtivani E, Rashidmayvan M. A Healthy Dietary Pattern May Have a Protective Effect Against Cardiovascular Disease Through Its Interaction With the MC4R Gene Polymorphism. Clin Nutr Res 2024; 13:214-225. [PMID: 39165291 PMCID: PMC11333145 DOI: 10.7762/cnr.2024.13.3.214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 07/19/2024] [Accepted: 07/23/2024] [Indexed: 08/22/2024] Open
Abstract
Polymorphisms in the melanocortin 4 receptor (MC4R) gene with occurrence and progression of chronic diseases such as obesity and cardiovascular disease (CVD) have long been addressed but there is a lack of evidence for complex interrelationships, including direct and indirect effects of these variables. This review specifically focuses on studying the effects of healthy diet interaction and MC4R polymorphisms on the development of CVD. The quantity and quality of carbohydrates and proteins consumed are related to obesity susceptibility and cardiometabolic risk factors. A healthy dietary pattern such as a Mediterranean dietary can modulate the association between MC4R polymorphisms (rs17782313) and the risk of CVDs. Also, the Nordic diet can reduce lipid profiles such as low-density lipoprotein cholesterol (LDL-C) and total cholesterol levels. On the other hand, MC4R interaction with the dietary inflammatory index decreases high-density lipoprotein cholesterol levels and increases LDL-C and triglyceride (TG) levels. Additionally, the DASH diet decreases TG, atherogenic index of plasma, systolic blood pressure, diastolic blood pressure, and serum glucose. The interaction between MC4R genes and diets plays an important role in the development of CVD. Adherence to healthy diets such as the Mediterranean, Nordic, Anti-inflammatory, and Dash diets might be an efficient strategy to prevent CVD. The potential for personalized diets to be developed for the treatment and prevention of CVD and its related comorbidities is expected to expand as this field develops.
Collapse
Affiliation(s)
- Kimia Mohammadhasani
- Department of Nutrition, Food Sciences and Clinical Biochemistry, School of Medicine, Social Determinants of Health Research Center, Gonabad University of Medical Sciences, Gonabad 9597118949, Iran
| | - Mohammad Vahedi Fard
- Department of Nutrition, Food Sciences and Clinical Biochemistry, School of Medicine, Social Determinants of Health Research Center, Gonabad University of Medical Sciences, Gonabad 9597118949, Iran
| | - Mehran Yadegari
- Department of Nutrition, Food Sciences and Clinical Biochemistry, School of Medicine, Social Determinants of Health Research Center, Gonabad University of Medical Sciences, Gonabad 9597118949, Iran
| | - Mehdi Barati
- Department of Pathobiology and Laboratory Sciences, North Khorasan University of Medical Sciences, Bojnurd 9414974877, Iran
| | - Hossein Bahari
- Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad 9177948959, Iran
| | - Elyas Nattagh-Eshtivani
- Department of Nutrition, Food Sciences and Clinical Biochemistry, School of Medicine, Social Determinants of Health Research Center, Gonabad University of Medical Sciences, Gonabad 9597118949, Iran
| | - Mohammad Rashidmayvan
- Department of Nutrition, Food Sciences and Clinical Biochemistry, School of Medicine, Social Determinants of Health Research Center, Gonabad University of Medical Sciences, Gonabad 9597118949, Iran
| |
Collapse
|
40
|
Şensoy E. Comparison of the effect of Sunset yellow on the stomach and small intestine of developmental period of mice. Heliyon 2024; 10:e31998. [PMID: 38882373 PMCID: PMC11176863 DOI: 10.1016/j.heliyon.2024.e31998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 05/24/2024] [Accepted: 05/27/2024] [Indexed: 06/18/2024] Open
Abstract
Sunset Yellow (SY), a synthetic food dye, is widely used in the food industry worldwide. The acceptable daily dosage for SY is 2.5 mg/kg/bw in humans. If SY is consumed in overdosage, it may cause histopathological effects in several organs. Studies in the literature about the effects of SY on growth and development in mammals are contradictory, and there are not enough of them. The investigation aims to determine SY's effects on the stomach and small intestine in different age groups of mice using histological methods. Control and treatment groups were created via mice aged 4, 8, and 10 weeks (n = 6). SY was administered by gavage at a level of 30 mg/kg/bw for 28 days to treatment groups. On the last day of the study, the mice were weighed and sacrificed by cervical dislocation. Stomach and small intestine tissues were removed from mice and transferred to 10 % formaldehyde. After passing through alcohol and xylene series and staining with Hematoxylin-Eosin, the tissues were evaluated under light and electron microscopy. The mean body weight (p = 0.01), mean stomach weight (p = 0.03), and mean small intestine weight were increased (p = 0.02) in treatment groups. In these groups, ruptures, fractures, and hemorrhage were detected in the small intestine tissue. In the stomach tissue, necrotic areas and hemorrhage were detected among the epithelial cells. The degenerations were more advanced in the weaning group. SY may be more harmful during weaning and puberty, but additional long-term studies are needed on the subject.
Collapse
Affiliation(s)
- Erhan Şensoy
- Karamanoglu Mehmetbey University Faculty of Health Science, Karaman, Turkey
| |
Collapse
|
41
|
Rodbard HW, Barnard-Kelly K, Pfeiffer AFH, Mauersberger C, Schnell O, Giorgino F. Practical strategies to manage obesity in type 2 diabetes. Diabetes Obes Metab 2024; 26:2029-2045. [PMID: 38514387 DOI: 10.1111/dom.15556] [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: 01/19/2024] [Revised: 02/20/2024] [Accepted: 02/28/2024] [Indexed: 03/23/2024]
Abstract
The rising phenomenon of obesity, a major risk factor for the development and progression of type 2 diabetes, is a complex and multifaceted issue that requires a comprehensive and coordinated approach to be prevented and managed. Although novel pharmacological measures to combat obesity have achieved unprecedented efficacy, a healthy lifestyle remains essential for the long-term success of any therapeutic intervention. However, this requires a high level of intrinsic motivation and continued behavioural changes in the face of multiple metabolic, psychological and environmental factors promoting weight gain, particularly in the context of type 2 diabetes. This review is intended to provide practical recommendations in the context of a holistic, person-centred approach to weight management, including evidence-based and expert recommendations addressing supportive communication, shared decision-making, as well as nutritional and pharmacological therapeutic approaches to achieve sustained weight loss.
Collapse
Affiliation(s)
| | - Katharine Barnard-Kelly
- Southern Health NHS Foundation Trust, Southampton, UK
- BHR Limited, Portsmouth, UK
- Spotlight Consultations, Fareham, UK
| | - Andreas F H Pfeiffer
- Department of Endocrinology, Diabetes and Nutrition, Campus Benjamin Franklin, Charité Universitätsmedizin Berlin, Berlin, Germany
| | | | - Oliver Schnell
- Sciarc GmbH, Baierbrunn, Germany
- Forschergruppe Diabetes eV at the Helmholtz Centre, Munich-Neuherberg, Germany
| | - Francesco Giorgino
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Bari, Italy
| |
Collapse
|
42
|
ŞENSOY E. Determination of the effects of sunset yellow on mouse liver and pancreas using histological methods. Toxicol Res (Camb) 2024; 13:tfae070. [PMID: 38737341 PMCID: PMC11084755 DOI: 10.1093/toxres/tfae070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 03/11/2024] [Accepted: 05/02/2024] [Indexed: 05/14/2024] Open
Abstract
Background Sunset Yellow (SY) is an azo synthetic food dye. Although the amount of SY consumed varies in different periods of life, it increases especially in children and adolescents. It may cause pathologic effects in organs at early ages. The aim of this study was to determine the effects of SY on the liver and pancreas of mice of different age groups using histological methods. Methods The study included Swiss albino mice that were divided into three treatment groups and three control groups based on age (4, 8, and 10 weeks old), with six mice in each group (n = 6/group). SY was administered at 30 mg/kg/bw/week orally for 28 days to treatment groups. The liver and pancreas tissues were kept in 10% formaldehyde, then passed through alcohol and xylene series and stained with Hematoxylin-Eosin. Results They were evaluated using light and electron microscopy. In SY groups, the mean body weight (p: 0.026) and the mean liver weight (p: 0.013) of the mice increased, and their mean pancreas weight decreased (p: 0.045).The numbers of degenerative cells in the liver tissues of the mice in the SY groups were high. Severe dilation in the sinusoids and haemorrhages focused around the Vena Cava were detected. In the pancreatic tissues of the SY groups, increases in fibroblasts and lymphocytic infiltration were observed. Conclusions Pathologies interpreted as chronic pancreatitis were more intense in the weaning group (4 weeks old). SY may be more harmful at an early age, and it may be beneficial to limit its use during this period.
Collapse
Affiliation(s)
- Erhan ŞENSOY
- Department of Midwifery, Faculty of Health Sciences, Karamanoglu Mehmetbey University, Ibrahim Okten Street, Campus, 70100 Karaman, Turkey
| |
Collapse
|
43
|
Zou H, Chen W, Hu B, Liu H, Zhao J. Testis–Gut-Reproduction Axis: The Key to Reproductive Health. Andrologia 2024; 2024:1-13. [DOI: 10.1155/2024/5020917] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025] Open
Abstract
Reproductive health is an important issue for humanity. In the context of the increasing incidence rate of male infertility, it is essential to find the factors that affect male reproductive health. Gastrointestinal health is closely related to reproductive health. Gastrointestinal hormones (GIH) and gut microbiota (GM), as important material foundations for gastrointestinal function, can promote or inhibit testicular reproductive function, including spermatogenesis, sperm maturation, androgen synthesis, and even broader male diseases such as sexual function, prostate cancer, etc. On the contrary, the functional health of the testes is also of great significance for the stability of gastrointestinal function. This review mainly discusses the important regulatory effects of GIH and GM on male reproductive function.
Collapse
Affiliation(s)
- Hede Zou
- Graduate School, China Academy of Chinese Medical Sciences, Beijing, China
| | - Wenkang Chen
- Graduate School, China Academy of Chinese Medical Sciences, Beijing, China
| | - Baofeng Hu
- Qian’an Hospital of Traditional Chinese Medicine, Tangshan, Hebei, China
| | - Hanfei Liu
- Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Jiayou Zhao
- Graduate School, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
44
|
Lee SH, Suh JH, Heo MJ, Choi JM, Yang Y, Jung HJ, Gao Z, Yu Y, Jung SY, Kolonin MG, Cox AR, Hartig SM, Eltzschig HK, Ju C, Moore DD, Kim KH. The Hepatokine Orosomucoid 2 Mediates Beneficial Metabolic Effects of Bile Acids. Diabetes 2024; 73:701-712. [PMID: 38320268 PMCID: PMC11043061 DOI: 10.2337/db23-0520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 01/28/2024] [Indexed: 02/08/2024]
Abstract
Bile acids (BAs) are pleiotropic regulators of metabolism. Elevated levels of hepatic and circulating BAs improve energy metabolism in peripheral organs, but the precise mechanisms underlying the metabolic benefits and harm still need to be fully understood. In the current study, we identified orosomucoid 2 (ORM2) as a liver-secreted hormone (i.e., hepatokine) induced by BAs and investigated its role in BA-induced metabolic improvements in mouse models of diet-induced obesity. Contrary to our expectation, under a high-fat diet (HFD), our Orm2 knockout (Orm2-KO) exhibited a lean phenotype compared with C57BL/6J control, partly due to the increased energy expenditure. However, when challenged with a HFD supplemented with cholic acid, Orm2-KO eliminated the antiobesity effect of BAs, indicating that ORM2 governs BA-induced metabolic improvements. Moreover, hepatic ORM2 overexpression partially replicated BA effects by enhancing insulin sensitivity. Mechanistically, ORM2 suppressed interferon-γ/STAT1 activities in inguinal white adipose tissue depots, forming the basis for anti-inflammatory effects of BAs and improving glucose homeostasis. In conclusion, our study provides new insights into the molecular mechanisms of BA-induced liver-adipose cross talk through ORM2 induction. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Sung Ho Lee
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Chonnam National University, Gwangju, Korea
| | - Ji Ho Suh
- Department of Anesthesiology, Critical Care and Pain Medicine and Center for Perioperative Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX
| | - Mi Jeong Heo
- Department of Anesthesiology, Critical Care and Pain Medicine and Center for Perioperative Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX
| | - Jong Min Choi
- Systems Onco-Immunology Laboratory, David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX
| | - Yang Yang
- Department of Anesthesiology, Critical Care and Pain Medicine and Center for Perioperative Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX
| | - Hyun-Jung Jung
- Department of Anesthesiology, Critical Care and Pain Medicine and Center for Perioperative Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX
| | - Zhanguo Gao
- The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center, Houston, TX
| | - Yongmei Yu
- The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center, Houston, TX
| | - Sung Yun Jung
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX
| | - Mikhail G. Kolonin
- The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center, Houston, TX
| | - Aaron R. Cox
- The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center, Houston, TX
| | - Sean M. Hartig
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Baylor College of Medicine, Houston, TX
| | - Holger K. Eltzschig
- Department of Anesthesiology, Critical Care and Pain Medicine and Center for Perioperative Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX
| | - Cynthia Ju
- Department of Anesthesiology, Critical Care and Pain Medicine and Center for Perioperative Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX
| | - David D. Moore
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA
| | - Kang Ho Kim
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX
- Department of Anesthesiology, Critical Care and Pain Medicine and Center for Perioperative Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX
| |
Collapse
|
45
|
Simoneau M, McKay B, Brooks E, Doucet É, Baillot A. Gut peptides before and following Roux-En-Y gastric bypass: A systematic review and meta-analysis. Obes Rev 2024; 25:e13702. [PMID: 38327045 DOI: 10.1111/obr.13702] [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: 11/22/2022] [Revised: 12/20/2023] [Accepted: 12/21/2023] [Indexed: 02/09/2024]
Abstract
A systematic search was conducted in Medline Ovid, Embase, Scopus, and Cochrane Central Register of Controlled Trials up until March 2021 following PRISMA guidelines. Studies included evaluated ghrelin, GLP-1, PYY or appetite sensation via visual analogue scales (VASs) before and after Roux-en-Y gastric bypass (RYGB) in adults. A multilevel model with random effects for study and follow-up time points nested in study was fit to the data. The model included kcal consumption as a covariate and time points as moderators. Among the 2559 articles identified, k = 47 were included, among which k = 19 evaluated ghrelin, k = 40 GLP-1, k = 22 PYY, and k = 8 appetite sensation. Our results indicate that fasting ghrelin levels are decreased 2 weeks post-RYGB (p = 0.005) but do not differ from baseline from 6 weeks to 1-year post-RYGB. Postprandial ghrelin and fasting GLP-1 levels were not different from pre-surgical values. Postprandial levels of GLP-1 increased significantly from 1 week (p < 0.001) to 2 years post-RYGB (p < 0.01) compared with pre-RYGB. Fasting PYY increased at 6 months (p = 0.034) and 1 year (p = 0.029) post-surgery; also, postprandial levels increased up to 1 year (p < 0.01). Insufficient data on appetite sensation were available to be meta-analyzed.
Collapse
Affiliation(s)
- Mylène Simoneau
- School of Human Kinetics, University of Ottawa, Ottawa, Ontario, Canada
| | - Brad McKay
- Department of kinesiology, University of McMaster, Hamilton, Ontario, Canada
| | - Emma Brooks
- School of Human Kinetics, University of Ottawa, Ottawa, Ontario, Canada
| | - Éric Doucet
- School of Human Kinetics, University of Ottawa, Ottawa, Ontario, Canada
| | - Aurélie Baillot
- Department of nursing, University of Québec en Outaouais, Gatineau, Quebec, Canada
| |
Collapse
|
46
|
Liu H, Xiao H, Lin S, Zhou H, Cheng Y, Xie B, Xu D. Effect of gut hormones on bone metabolism and their possible mechanisms in the treatment of osteoporosis. Front Pharmacol 2024; 15:1372399. [PMID: 38725663 PMCID: PMC11079205 DOI: 10.3389/fphar.2024.1372399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 03/25/2024] [Indexed: 05/12/2024] Open
Abstract
Bone is a highly dynamic organ that changes with the daily circadian rhythm. During the day, bone resorption is suppressed due to eating, while it increases at night. This circadian rhythm of the skeleton is regulated by gut hormones. Until now, gut hormones that have been found to affect skeletal homeostasis include glucagon-like peptide-1 (GLP-1), glucagon-like peptide-2 (GLP-2), glucose-dependent insulinotropic polypeptide (GIP), and peptide YY (PYY), which exerts its effects by binding to its cognate receptors (GLP-1R, GLP-2R, GIPR, and Y1R). Several studies have shown that GLP-1, GLP-2, and GIP all inhibit bone resorption, while GIP also promotes bone formation. Notably, PYY has a strong bone resorption-promoting effect. In addition, gut microbiota (GM) plays an important role in maintaining bone homeostasis. This review outlines the roles of GLP-1, GLP-2, GIP, and PYY in bone metabolism and discusses the roles of gut hormones and the GM in regulating bone homeostasis and their potential mechanisms.
Collapse
Affiliation(s)
- Hongyu Liu
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Dongguan Key Laboratory of Traditional Chinese Medicine and New Pharmaceutical Development, School of Pharmacy, Guangdong Medical University, Dongguan, China
- Institute of Traditional Chinese Medicine and New Pharmacy Development, Guangdong Medical University, Dongguan, China
| | - Huimin Xiao
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Dongguan Key Laboratory of Traditional Chinese Medicine and New Pharmaceutical Development, School of Pharmacy, Guangdong Medical University, Dongguan, China
- Institute of Traditional Chinese Medicine and New Pharmacy Development, Guangdong Medical University, Dongguan, China
| | - Sufen Lin
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Dongguan Key Laboratory of Traditional Chinese Medicine and New Pharmaceutical Development, School of Pharmacy, Guangdong Medical University, Dongguan, China
- Institute of Traditional Chinese Medicine and New Pharmacy Development, Guangdong Medical University, Dongguan, China
| | - Huan Zhou
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Dongguan Key Laboratory of Traditional Chinese Medicine and New Pharmaceutical Development, School of Pharmacy, Guangdong Medical University, Dongguan, China
- Institute of Traditional Chinese Medicine and New Pharmacy Development, Guangdong Medical University, Dongguan, China
| | - Yizhao Cheng
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Dongguan Key Laboratory of Traditional Chinese Medicine and New Pharmaceutical Development, School of Pharmacy, Guangdong Medical University, Dongguan, China
- Institute of Traditional Chinese Medicine and New Pharmacy Development, Guangdong Medical University, Dongguan, China
| | - Baocheng Xie
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Dongguan Key Laboratory of Traditional Chinese Medicine and New Pharmaceutical Development, School of Pharmacy, Guangdong Medical University, Dongguan, China
- Department of Pharmacy, The 10th Affiliated Hospital of Southern Medical University (Dongguan People’s Hospital), Dongguan, China
| | - Daohua Xu
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Dongguan Key Laboratory of Traditional Chinese Medicine and New Pharmaceutical Development, School of Pharmacy, Guangdong Medical University, Dongguan, China
- Institute of Traditional Chinese Medicine and New Pharmacy Development, Guangdong Medical University, Dongguan, China
| |
Collapse
|
47
|
Noguchi H, Ikenaga T, Ueno S, Kohashi C, Matsumura Y, Kakumoto Y, Kohda N, Hara H, Hira T. Effect of Single Oral Coingestion of GABA and Malic Acid on Postprandial GLP-1, Glucose, and Insulin Responses in Healthy Volunteers: A Randomized, Double-Blind, Placebo-Controlled, Crossover Study. Mol Nutr Food Res 2024; 68:e2300610. [PMID: 38487986 DOI: 10.1002/mnfr.202300610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 12/19/2023] [Indexed: 04/17/2024]
Abstract
SCOPE This study examines whether coingestion of γ-aminobutyric acid (GABA) and malic acid (MA) before meals enhances glucagon-like peptide-1 (GLP-1) secretion, and which affects subsequent insulin and glycemic responses in humans. METHODS AND RESULTS Initially, a murine enteroendocrine STC-1 cell line is used to verify coadministration of GABA and MA synergistically induces GLP-1 secretion. Next, 22 healthy adults are given water (50 mL) containing 400 mg GABA and 400 mg MA (Test), or only 400 mg citric acid (CA) (Placebo) 20 min before meal tolerance test (MTT). Interval blood samples are taken postprandially over 180 min to determine GLP-1, insulin, and glucose responses. By comparison to preload of Placebo, preload of Test significantly increases plasma GLP-1 (total/active) levels (incremental area under the curve by 1.2- and 1.6-fold), respectively. However, there are no significant differences in postprandial blood glucose and insulin. CONCLUSION Coingestion of GABA and MA before meals enhances postprandial GLP-1 secretion. Future studies should explore optimal dosage regimens to find the efficacy of the mixture on insulin and glycemic response.
Collapse
Affiliation(s)
- Hiroki Noguchi
- Graduate School of Agriculture, Hokkaido University, Sapporo, 060-8589, Japan
- Otsu Nutraceuticals Research Institute, Otsuka Pharmaceutical Co., Ltd, Otsu, 520-0002, Japan
| | - Takeshi Ikenaga
- Otsu Nutraceuticals Research Institute, Otsuka Pharmaceutical Co., Ltd, Otsu, 520-0002, Japan
| | - Shota Ueno
- Otsu Nutraceuticals Research Institute, Otsuka Pharmaceutical Co., Ltd, Otsu, 520-0002, Japan
| | - Chieko Kohashi
- Otsu Nutraceuticals Research Institute, Otsuka Pharmaceutical Co., Ltd, Otsu, 520-0002, Japan
| | - Yasuhiro Matsumura
- Faculty of Health and Nutrition, Bunkyo University, Chigasaki, 253-8550, Japan
| | - Yusuke Kakumoto
- Department of Lead Discovery Research, Otsuka Pharmaceutical Co., Ltd, Tokushima, 771-0192, Japan
| | - Noriyuki Kohda
- Otsu Nutraceuticals Research Institute, Otsuka Pharmaceutical Co., Ltd, Otsu, 520-0002, Japan
| | - Hiroshi Hara
- Department of Food Science and Human Nutrition, Fuji Women's University, Ishikari, 061-3204, Japan
| | - Tohru Hira
- Graduate School of Agriculture, Hokkaido University, Sapporo, 060-8589, Japan
- Research Faculty of Agriculture, Hokkaido University, Sapporo, 060-8589, Japan
| |
Collapse
|
48
|
Dafne VJ, Manuel MA, Rocio CV. Chronobiotics, satiety signaling, and clock gene expression interplay. J Nutr Biochem 2024; 126:109564. [PMID: 38176625 DOI: 10.1016/j.jnutbio.2023.109564] [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: 07/24/2023] [Revised: 11/21/2023] [Accepted: 12/31/2023] [Indexed: 01/06/2024]
Abstract
The biological clock regulates the way our body works throughout the day, including releasing hormones and food intake. Disruption of the biological clock (chronodisruption) may deregulate satiety, which is strictly regulated by hormones and neurotransmitters, leading to health problems like obesity. Nowadays, using bioactive compounds as a coadjutant for several pathologies is a common practice. Phenolic compounds and short-chain fatty acids, called "chronobiotics," can modulate diverse mechanisms along the body to exert beneficial effects, including satiety regulation and circadian clock resynchronization; however, the evidence of the interplay between those processes is limited. This review compiles the evidence of natural chronobiotics, mainly polyphenols and short-chain fatty acids that affect the circadian clock mechanism and process modifications in genes or proteins resulting in a signaling chain that modulates satiety hormones or hunger pathways.
Collapse
Affiliation(s)
- Velásquez-Jiménez Dafne
- Research and Graduate Studies in Food Science, School of Chemistry, Autonomous University of Queretaro, Queretaro, Mexico
| | - Miranda-Anaya Manuel
- Multidisciplinary Unit for Teaching and Research (UMDI), School of Sciences, Autonomous National University of Mexico, Queretaro, Mexico
| | - Campos-Vega Rocio
- Research and Graduate Studies in Food Science, School of Chemistry, Autonomous University of Queretaro, Queretaro, Mexico.
| |
Collapse
|
49
|
Parfenov AI. The value of increased intestinal permeability in the pathogenesis of internal diseases. TERAPEVT ARKH 2024; 96:85-90. [DOI: 10.26442/00403660.2024.02.202587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
In the process of evolution in the gastrointestinal tract, a system of protection against bacterial and food antigens from getting into the blood was formed. The causes of increased intestinal permeability (IIP) can be microbiota imbalance, use of antibiotics, non-steroidal anti-inflammatory drugs, stress, diet rich in fructose, glucose, sucrose and long-chain fatty acids. The appearance of IIP may be of paramount importance in the pathogenesis of autoimmune diseases. A diet low in fermentable oligodimonosaccharides and polyols, pre- and probiotics, polyphenols, vitamins, short-chain fatty acids, dietary fiber, glutamine contributes to the reduction of IIP. It has been established that the cytoprotector rebamipide strengthens the barrier function throughout the gastrointestinal tract, which is reflected in practical recommendations for its use in diseases accompanied by IIP. The study of this direction will contribute to the emergence of a new strategy for the treatment of internal diseases.
Collapse
|
50
|
Kloock S, Haerting N, Herzog G, Oertel M, Geiger N, Geier A, Sequeira V, Nickel A, Kohlhaas M, Fassnacht M, Dischinger U. Effects of NPY-2 Receptor Antagonists, Semaglutide, PYY 3-36, and Empagliflozin on Early MASLD in Diet-Induced Obese Rats. Nutrients 2024; 16:904. [PMID: 38542814 PMCID: PMC10974407 DOI: 10.3390/nu16060904] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 03/09/2024] [Accepted: 03/19/2024] [Indexed: 01/04/2025] Open
Abstract
(1) Background: Modulators of the Neuropeptide Y (NPY) system are involved in energy metabolism, but the effect of NPY receptor antagonists on metabolic-dysfunction-associated steatotic liver disease (MASLD), a common obesity-related comorbidity, are largely unknown. In this study, we report on the effects of antagonists of the NPY-2 receptor (Y2R) in comparison with empagliflozin and semaglutide, substances that are known to be beneficial in MASLD. (2) Methods: Diet-induced obese (DIO) male Wistar rats were randomized into the following treatment groups: empagliflozin, semaglutide ± PYY3-36, the Y2R antagonists JNJ 31020028 and a food-restricted group, as well as a control group. After a treatment period of 8 weeks, livers were weighed and histologically evaluated. QrtPCR was performed to investigate liver inflammation and de novo lipogenesis (in liver and adipose tissue). Serum samples were analysed for metabolic parameters. (3) Results: Semaglutide + PYY3-36 led to significant weight loss, reduced liver steatosis (p = 0.05), and decreased inflammation, insulin resistance, and leptin levels. JNJ-31020028 prevented steatosis (p = 0.03) without significant weight loss. Hepatic downregulation of de novo lipogenesis-regulating genes (SREBP1 and MLXIPL) was observed in JNJ-31020028-treated rats (p ≤ 0.0001). Food restriction also resulted in significantly reduced weight, steatosis, and hepatic de novo lipogenesis. (4) Conclusions: Body weight reduction (e.g., by food restriction or drugs like semaglutide ± PYY3-36) is effective in improving liver steatosis in DIO rats. Remarkably, the body-weight-neutral Y2R antagonists may be effective in preventing liver steatosis through a reduction in de novo lipogenesis, making this drug class a candidate for the treatment of (early) MASLD.
Collapse
Affiliation(s)
- Simon Kloock
- Department of Internal Medicine, Division of Endocrinology and Diabetes, University Hospital Wuerzburg, University of Wuerzburg, 97080 Wuerzburg, Germany; (S.K.); (N.H.); (M.O.); (N.G.); (M.F.)
| | - Niklas Haerting
- Department of Internal Medicine, Division of Endocrinology and Diabetes, University Hospital Wuerzburg, University of Wuerzburg, 97080 Wuerzburg, Germany; (S.K.); (N.H.); (M.O.); (N.G.); (M.F.)
| | - Gloria Herzog
- Institute of Pathology, University of Wuerzburg, 97070 Wuerzburg, Germany;
| | - Marie Oertel
- Department of Internal Medicine, Division of Endocrinology and Diabetes, University Hospital Wuerzburg, University of Wuerzburg, 97080 Wuerzburg, Germany; (S.K.); (N.H.); (M.O.); (N.G.); (M.F.)
| | - Niklas Geiger
- Department of Internal Medicine, Division of Endocrinology and Diabetes, University Hospital Wuerzburg, University of Wuerzburg, 97080 Wuerzburg, Germany; (S.K.); (N.H.); (M.O.); (N.G.); (M.F.)
| | - Andreas Geier
- Department of Internal Medicine, Division of Hepatology, University Hospital Wuerzburg, University of Wuerzburg, 97080 Wuerzburg, Germany;
| | - Vasco Sequeira
- Comprehensive Heart Failure Center, University Hospital Wuerzburg, University of Wuerzburg, 97080 Wuerzburg, Germany; (V.S.); (A.N.); (M.K.)
| | - Alexander Nickel
- Comprehensive Heart Failure Center, University Hospital Wuerzburg, University of Wuerzburg, 97080 Wuerzburg, Germany; (V.S.); (A.N.); (M.K.)
| | - Michael Kohlhaas
- Comprehensive Heart Failure Center, University Hospital Wuerzburg, University of Wuerzburg, 97080 Wuerzburg, Germany; (V.S.); (A.N.); (M.K.)
| | - Martin Fassnacht
- Department of Internal Medicine, Division of Endocrinology and Diabetes, University Hospital Wuerzburg, University of Wuerzburg, 97080 Wuerzburg, Germany; (S.K.); (N.H.); (M.O.); (N.G.); (M.F.)
| | - Ulrich Dischinger
- Department of Internal Medicine, Division of Endocrinology and Diabetes, University Hospital Wuerzburg, University of Wuerzburg, 97080 Wuerzburg, Germany; (S.K.); (N.H.); (M.O.); (N.G.); (M.F.)
- Comprehensive Heart Failure Center, University Hospital Wuerzburg, University of Wuerzburg, 97080 Wuerzburg, Germany; (V.S.); (A.N.); (M.K.)
| |
Collapse
|