1
|
Liu J, Lu W, Wu H, Yan Z, Liu Y, Tang C, Chen Y, Wang S, Tang W, Han J, Wei C, Jiang N. Rational design of dual-agonist peptides targeting GLP-1 and NPY2 receptors for regulating glucose homeostasis and body weight with minimal nausea and emesis. Eur J Med Chem 2025; 287:117320. [PMID: 39892093 DOI: 10.1016/j.ejmech.2025.117320] [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: 11/18/2024] [Revised: 01/01/2025] [Accepted: 01/21/2025] [Indexed: 02/03/2025]
Abstract
There is an urgent need for effective treatments targeting comorbidities of type 2 diabetes (T2DM) and obesity. Developing dual agonists of glucagon-like peptide 1 receptor (GLP-1R) and neuropeptide Y receptor type 2 (NPY2R) with combined PYY3-36 and GLP-1 bioactivity is promising. However, designing such dual agonists that effectively control glycemia and reduce weight while minimizing gastrointestinal side effects is challenging. In this study, we systematically evaluated the side effects induced by co-administering various GLP-1R agonists and PYY3-36 analogue. Our findings revealed that different GLP-1R agonist-PYY analogue combinations elicited gastrointestinal side effects of varying intensities. Among these, the co-administration of bullfrog GLP-1 analogue (bGLP-1) with PYY3-36 analogue resulted in lower gastrointestinal side effects. Thus, bGLP-1 was selected as the preferred candidate for designing dual GLP-1R/NPY2R agonists. Through stepwise structural design, optimization of linker arms, and durability enhancements, coupled with in vitro receptor screening, the novel peptide bGLP/PYY-19 emerged as the lead candidate. Notably, experimental results in mice and rats showed a significant reduction in emesis with bGLP/PYY-19 compared to semaglutide and bGLP-1 long-acting analogue (LAbGLP-1). Furthermore, bGLP/PYY-19 significantly outperformed semaglutide and LAbGLP-1 in reducing body weight in diet-induced obese (DIO) mice, without inducing nausea-associated behavior. These findings underscore the potential of dual-targeting single peptide conjugates as a promising strategy for developing glucoregulatory treatments that offer superior weight loss benefits and are better tolerated compared to treatments targeting GLP-1R alone.
Collapse
Affiliation(s)
- Jing Liu
- Department of Pharmacy, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi, PR China
| | - Weiwen Lu
- Department of Pharmacy, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi, PR China
| | - Han Wu
- School of Chemistry & Materials Science, Jiangsu Normal University, Xuzhou, 221116, Jiangsu, PR China
| | - Zhiming Yan
- Department of Pharmacy, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi, PR China
| | - Yun Liu
- Department of Pharmacy, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi, PR China
| | - Chunli Tang
- Department of Pharmacy, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi, PR China
| | - Yangxin Chen
- Department of Pharmacy, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi, PR China
| | - Shuang Wang
- School of Chemistry & Materials Science, Jiangsu Normal University, Xuzhou, 221116, Jiangsu, PR China
| | - Weizhong Tang
- Department of Pharmacy, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi, PR China
| | - Jing Han
- School of Chemistry & Materials Science, Jiangsu Normal University, Xuzhou, 221116, Jiangsu, PR China.
| | - Changhong Wei
- Department of Research & Clinical Laboratory, The Fifth Affiliated Hospital of Guangxi Medical University & the First People's Hospital of Nanning, Nanning, Guangxi, PR China.
| | - Neng Jiang
- Department of Pharmacy, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi, PR China.
| |
Collapse
|
2
|
Forst T, De Block C, Del Prato S, Frias J, Lautenbach A, Ludvik B, Marinez M, Mathieu C, Müller TD, Schnell O. Novel pharmacotherapies for weight loss: Understanding the role of incretins to enable weight loss and improved health outcomes. Diabetes Obes Metab 2025; 27 Suppl 2:48-65. [PMID: 39931897 DOI: 10.1111/dom.16247] [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: 11/26/2024] [Revised: 01/25/2025] [Accepted: 01/26/2025] [Indexed: 04/17/2025]
Abstract
Obesity and type 2 diabetes mellitus (T2D) are widespread diseases that significantly impact cardiovascular and renal morbidity and mortality. In the recent years, intensive research has been performed to assess the role of adipose tissue and body fat distribution in the development of metabolic and non-metabolic complications in individuals with obesity. In addition to lifestyle modifications, glucagon-like peptide-1 receptor agonists (GLP-1-RA) have become a meaningful treatment expansion for the management of both disorders. In addition to improving metabolic control and reducing body weight, treatment with GLP-1-RAs reduces cardiovascular and renal events in individuals with obesity with and without diabetes. These important benefits of GLP-1-RAs have triggered new interest in other enteroendocrine and enteropancreatic peptides for treating obesity and its metabolic and non-metabolic consequences. The first peptide dual-agonist targeting glucose-dependent insulinotropic polypeptide (GIP) and GLP-1 receptors has been approved for the treatment of T2D and obesity. GIP/GLP-1 dual-agonism appear to provide better metabolic control and greater weight reduction compared with GLP-1-R mono-agonism. Other peptide and non-peptide co-agonists are in clinical development for obesity, T2D, metabolic dysfunction-associated steatotic liver disease (MASLD) and other metabolic disorders. This narrative review aims to summarize the available data on approved and emerging enteroendocrine and enteropancreatic based treatment approaches for obesity and metabolic disorders. In addition to available clinical efficacy measures, side effects, limitations and open challenges will also be addressed.
Collapse
Affiliation(s)
- Thomas Forst
- CRS Clinical Research Services GmbH, Mannheim, Germany
| | - Christophe De Block
- Department of Endocrinology-Diabetology, Antwerp University Hospital and University of Antwerp, Belgium
| | - Stefano Del Prato
- Interdisciplinary Research Center "Health Science," Sant'Anna School of Advanced Studies, Pisa, Italy
| | - 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
|
3
|
Østergaard S, Jessen C, Paulsson JF, Kasimova MA, Conde-Frieboes KW, Straarup EM, Skyggebjerg RB, Ynddal L, Sanfridson A, Wulff BS, Chambers AP. Variant screening of PYY 3-36 leads to potent long-acting PYY analogs with superior Y 2 receptor selectivity. Sci Transl Med 2025; 17:eadq6392. [PMID: 40138456 DOI: 10.1126/scitranslmed.adq6392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 09/27/2024] [Accepted: 03/05/2025] [Indexed: 03/29/2025]
Abstract
Peptide YY (PYY3-36) has attracted attention in diabetes and obesity research because of its involvement in food intake regulation and glucose homeostasis. Native PYY3-36 maintains high potency on the Y2 receptor with a loss of potency on the Y1, Y4, and Y5 receptors. However, PYY3-36 has a relatively short half-life, and the selectivity displayed by the native peptide may not be optimal if a long-acting analog is to be developed. We performed variant screening of PYY3-36 to identify key canonical amino acids that are pivotal to Y2 receptor selectivity, potency, and peptide stability. In combination with fatty diacid derivatization, this afforded highly selective long-acting analogs against the Y2 receptor, which improved glucose metabolism in diabetic db/db mice. When combined with a long-acting glucagon-like peptide 1 (GLP-1) receptor agonist, these analogs showed superior blood glucose lowering in diabetic ZSF1 rats and greater body weight loss in a high-fat diet-induced mouse model of obesity compared with treatment with the GLP-1 analog alone. One of the tested analogs, PYY1875, has progressed into clinical trials for obesity. Together, our results demonstrate the power of variant screening combined with fatty diacid derivatization in the development of a long-acting, highly efficacious PYY clinical candidate.
Collapse
Affiliation(s)
- Søren Østergaard
- Global Research Technologies, Novo Nordisk A/S, Novo Research Park, 2760 Måløv, Denmark
| | - Carsten Jessen
- Global Research Technologies, Novo Nordisk A/S, Novo Research Park, 2760 Måløv, Denmark
| | - Johan F Paulsson
- Global Drug Discovery, Novo Nordisk A/S, Novo Research Park, 2760 Måløv, Denmark
| | - Marina A Kasimova
- Digital Science and Innovation, Novo Nordisk A/S, Novo Research Park, 2760 Måløv, Denmark
| | | | - Ellen Marie Straarup
- Global Drug Discovery, Novo Nordisk A/S, Novo Research Park, 2760 Måløv, Denmark
| | | | - Lars Ynddal
- Global Research Technologies, Novo Nordisk A/S, Novo Research Park, 2760 Måløv, Denmark
| | - Annika Sanfridson
- Global Drug Discovery, Novo Nordisk A/S, Novo Research Park, 2760 Måløv, Denmark
| | - Birgitte S Wulff
- Global Drug Discovery, Novo Nordisk A/S, Novo Research Park, 2760 Måløv, Denmark
| | - Adam P Chambers
- Global Drug Discovery, Novo Nordisk A/S, Novo Research Park, 2760 Måløv, Denmark
| |
Collapse
|
4
|
James-Okoro PP, Lewis JE, Gribble FM, Reimann F. The role of GIPR in food intake control. Front Endocrinol (Lausanne) 2025; 16:1532076. [PMID: 40166681 PMCID: PMC11955450 DOI: 10.3389/fendo.2025.1532076] [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: 11/21/2024] [Accepted: 02/25/2025] [Indexed: 04/02/2025] Open
Abstract
Glucose-dependent insulinotropic polypeptide (GIP) is one of two incretin hormones playing key roles in the control of food intake, nutrient assimilation, insulin secretion and whole-body metabolism. Recent pharmacological advances and clinical trials show that unimolecular co-agonists that target the receptors for the incretins - GIP and glucagon-like peptide 1 (GLP-1) - offer more effective treatment strategies for obesity and type 2 diabetes mellitus (T2D) compared with GLP-1 receptor (GLP1R) agonists alone, suggesting previously underappreciated roles of GIP in regulating food intake and body weight. The mechanisms by which GIP regulates energy balance remain controversial as both agonism and antagonism of the GIP receptor (GIPR) produce weight loss and improve metabolic outcomes in preclinical models. Recent studies have shown that GIPR signalling in the central nervous system (CNS), especially in regions of the brain that regulate energy balance, is essential for its action on appetite regulation. This finding has sparked interest in understanding the mechanisms by which GIP engages brain circuits to reduce food intake and body weight. In this review, we present key knowledge around the actions of GIP on food intake regulation and the potential mechanisms by which GIPR and GIPR/GLP1R agonists may regulate energy balance.
Collapse
Affiliation(s)
| | | | - Fiona Mary Gribble
- Institute of Metabolic-Science-Metabolic Research Laboratories and MRC-Metabolic Diseases Unit, University of Cambridge, Cambridge, United Kingdom
| | - Frank Reimann
- Institute of Metabolic-Science-Metabolic Research Laboratories and MRC-Metabolic Diseases Unit, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
5
|
Paprocki MP, Sørensen KK, Jensen KJ. pH Controlled Transient Cyclization of Peptides for Increased Stability towards Oral Delivery. Chemistry 2025; 31:e202403503. [PMID: 39630536 DOI: 10.1002/chem.202403503] [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: 09/19/2024] [Revised: 11/29/2024] [Accepted: 11/29/2024] [Indexed: 12/07/2024]
Abstract
Peptides are highly efficient for treatment of many diseases, especially in oncology and diabetes. Oral delivery of peptides is desirable, but is challenged by low bioavailability and new chemical methods to enable oral delivery are needed. Here, we developed pH responsive linearization as a strategy for transient protection of peptides to extend their half-life in model systems. Peptides were cyclized to increase their stability at the low pH in the stomach, while they linearize at neutral pH to form the active peptide. We developed ester based responsive linkers with a protonable amine for O-to-N acyl shift, which allowed linearization strategies based on pyroglutamoyl (pGlu) or diketopiperazine (DKP) formation. After coupling of the linker, peptides were cyclized by CuAAC. We studied the stability against simulated gastric fluid (SGF) at different pH and the ability of cyclic peptides to linearize. This led to PYY3-36 analogues with pH responsive linearization for increased stability.
Collapse
Affiliation(s)
- Maciej P Paprocki
- Department of Chemistry, University of Copenhagen, DK-1871, Frederiksberg, Denmark
- Novo Nordisk A/S, DK-2880, Bagsværd, Denmark
| | - Kasper K Sørensen
- Department of Chemistry, University of Copenhagen, DK-1871, Frederiksberg, Denmark
| | - Knud J Jensen
- Department of Chemistry, University of Copenhagen, DK-1871, Frederiksberg, Denmark
| |
Collapse
|
6
|
Simonds SE, Pryor JT, Lam BYH, Dowsett GK, Mustafa T, Munder A, Elysee K, Balland E, Cowley LO, Yeo GSH, Lawrence A, Spanswick DC, Cowley MA. The metabolic and cardiovascular effects of amphetamine are partially mediated by the central melanocortin system. Cell Rep Med 2025; 6:101936. [PMID: 39914386 DOI: 10.1016/j.xcrm.2025.101936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 09/02/2024] [Accepted: 01/09/2025] [Indexed: 02/21/2025]
Abstract
Amphetamine (AMPH) exerts metabolic and cardiovascular effects. The central melanocortin system is a key regulator of both metabolic and cardiovascular functions. Here, we show that the melanocortin system partially mediates AMPH-induced anorexia, energy expenditure, tachycardia, and hypertension. AMPH increased α-melanocyte stimulating hormone (αMSH) secretion from the hypothalamus, elevated blood pressure and heart rate (HR), increased brown adipose tissue (BAT) thermogenesis, and reduced both food intake (FI) and body weight (BW). In melanocortin 4 receptor-deficient (MC4R knockout [KO]) mice, metabolic and cardiovascular effects of AMPH were significantly attenuated. Antagonism of serotonergic and noradrenergic neurotransmitter systems attenuated AMPH-induced αMSH secretion as well as AMPH-induced metabolic and cardiovascular effects. We propose that AMPH increases serotonergic activation of proopiomelanocortin (POMC) neurons and reduces the noradrenergic inhibition of POMC neurons, thereby disinhibiting them. Together, these presynaptic mechanisms result in increased POMC activity, increased αMSH secretion, and increased activation of MC4R pathways that regulate both the metabolic and cardiovascular systems.
Collapse
Affiliation(s)
- Stephanie E Simonds
- Department of Physiology, Metabolism, Diabetes, and Obesity Programme, Monash Biomedicine Discovery Institute Monash University, Clayton, VIC, Australia.
| | - Jack T Pryor
- Department of Physiology, Metabolism, Diabetes, and Obesity Programme, Monash Biomedicine Discovery Institute Monash University, Clayton, VIC, Australia
| | - Brian Y H Lam
- MRC Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK
| | - Georgina K Dowsett
- MRC Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK
| | - Tomris Mustafa
- Department of Physiology, Metabolism, Diabetes, and Obesity Programme, Monash Biomedicine Discovery Institute Monash University, Clayton, VIC, Australia
| | - Astrid Munder
- Department of Physiology, Metabolism, Diabetes, and Obesity Programme, Monash Biomedicine Discovery Institute Monash University, Clayton, VIC, Australia
| | - Kayla Elysee
- Department of Physiology, Metabolism, Diabetes, and Obesity Programme, Monash Biomedicine Discovery Institute Monash University, Clayton, VIC, Australia
| | - Eglantine Balland
- Department of Physiology, Metabolism, Diabetes, and Obesity Programme, Monash Biomedicine Discovery Institute Monash University, Clayton, VIC, Australia
| | - Lachlan O Cowley
- Department of Physiology, Metabolism, Diabetes, and Obesity Programme, Monash Biomedicine Discovery Institute Monash University, Clayton, VIC, Australia
| | - Giles S H Yeo
- MRC Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK
| | - Andrew Lawrence
- Mental Health Theme, The Florey Institute of Neuroscience and Mental Health, Parkville, VIC 3052, Australia; Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3052, Australia
| | - David C Spanswick
- Department of Physiology, Metabolism, Diabetes, and Obesity Programme, Monash Biomedicine Discovery Institute Monash University, Clayton, VIC, Australia; Warwick Medical School, Division of Biomedical Sciences, University of Warwick, CV4 7AL Coventry, UK
| | - Michael A Cowley
- Department of Physiology, Metabolism, Diabetes, and Obesity Programme, Monash Biomedicine Discovery Institute Monash University, Clayton, VIC, Australia.
| |
Collapse
|
7
|
Sixten HJ, Rønnestad I, Bogevik AS, Aspevik T, Oterhals Å, Gomes AS, Lai F, Tolås I, Gelebard V, Hillestad M, Kousoulaki K. Side-Stream Based Marine Solubles From Atlantic Cod ( Gadus morhua) Modulate Appetite and Dietary Nutrient Utilization in Atlantic Salmon ( Salmo salar L.) and can Replace Fish Meal. AQUACULTURE NUTRITION 2025; 2025:4872889. [PMID: 39949357 PMCID: PMC11824393 DOI: 10.1155/anu/4872889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 12/26/2024] [Indexed: 02/16/2025]
Abstract
Whitefish fisheries' side-stream biomass is an abundant underutilized resource that can be valorized to benefit future aquaculture sustainability. Four novel ingredients based on side-streams from Atlantic cod (Gadus morhua) fileting were produced. FM-hb, a fish meal (FM), and FPH-hb, a fish protein hydrolysate based on heads (h) and backbones (b); FM-hbg, a FM based on heads, backbones, and viscera/guts (g); and FPC-g, a fish protein concentrate based on viscera preserved in formic acid. Four diets were prepared containing one of the ingredients replacing 50% of the dietary FM protein, in addition to a positive (FM10) and a negative (FM5) control. The six diets were fed to triplicate tanks with Atlantic salmon (Salmo salar L.; 113 ± 1 g) over 8 weeks. Besides general performance, gut and brain gene expression for selected hormones and key neuropeptides involved in the control of appetite and digestive processes were studied during feeding and postprandial, and possible reference levels for Atlantic salmon were established. All side-stream-added diets performed well, with no significant differences in performance and biometrics between the treatments. Some gene expression differences were observed, but no well-defined patterns emerged supporting clear dietary effects related to digestive performance or appetite. However, in the brain, a short-time upregulation of agouti-related protein-1 (agrp1), corresponded to higher cumulative feed intake (FI) for the FM10 diet supporting notions that this may be a candidate biomarker for appetite in salmon. Expression of stomach ghrelin-1 (ghrl1) was higher than ghrelin-2 (ghrl2) and membrane-bound O-acyltransferase domain-containing 4 (mboat4), and midgut peptide YYa-2 (pyya2) and glucagon-a (gcga) were higher than peptide YYb-1 (pyyb1). A comparison showed that midgut peptide YYa-1 (pyya1), pyya2, and gcga expressions were higher than in the hindgut, which is opposite of what is found in mammals. In conclusion, this study shows that sustainable side-stream raw materials with different characteristics can partly replace high-quality commercial FMs giving similar performance.
Collapse
Affiliation(s)
- Hanne Jorun Sixten
- Department of Research and Development, BioMar AS, Trondheim, Norway
- Department of Biological Sciences, University of Bergen, Bergen, Norway
| | - Ivar Rønnestad
- Department of Biological Sciences, University of Bergen, Bergen, Norway
| | - André S. Bogevik
- Department of Nutrition and Feed Technology, NOFIMA, Fyllingsdalen, Norway
| | - Tone Aspevik
- Department of Nutrition and Feed Technology, NOFIMA, Fyllingsdalen, Norway
| | - Åge Oterhals
- Department of Nutrition and Feed Technology, NOFIMA, Fyllingsdalen, Norway
| | - Ana S. Gomes
- Department of Biological Sciences, University of Bergen, Bergen, Norway
- Institute of Marine Research, Tromsø, Norway
| | - Floriana Lai
- Department of Biological Sciences, University of Bergen, Bergen, Norway
| | - Ingvill Tolås
- Department of Biological Sciences, University of Bergen, Bergen, Norway
- Department of Biological Sciences, NTNU Ålesund, Ålesund, Norway
| | - Virginie Gelebard
- Department of Biological Sciences, University of Bergen, Bergen, Norway
| | - Marie Hillestad
- Department of Research and Development, BioMar AS, Trondheim, Norway
| | | |
Collapse
|
8
|
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
|
9
|
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
|
10
|
Kalomoiri P, Mortensen JS, Christensen NJ, Sørensen KK, Nielsen HM, Jensen KJ, Thygesen MB. Neo-Glycolipid Oximes as Intestinal Permeation Enhancers for Peptide Hormone PYY 3-36. Chemistry 2024; 30:e202401887. [PMID: 39504118 DOI: 10.1002/chem.202401887] [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: 05/14/2024] [Indexed: 11/21/2024]
Abstract
Herein, we describe the design and synthesis of 16 neo-glycolipids that are potential permeation enhancers for oral drug delivery of peptide therapeutics. These amphiphilic neo-glycolipids are composed of fatty acids and various carbohydrates (d-glucose, lactose, cellobiose, maltose) via an oxime linker. The ability of the synthesized neo-glycolipids to enhance permeation of fluorescein-labelled dextran (4 kDa) or 3H-mannitol across intestinal epithelium was investigated in vitro using monolayers of human epithelial Caco-2 cells. Their effects were compared with (pre-)clinically known enhancers as reference compounds; sodium salts of octanoic, decanoic, and dodecanoic acid, and sodium salcaprozate (SNAC). Most neo-glycolipids increased the permeation of the model compounds, proving that neo-glycolipids, which possess vastly different properties from the reference compounds, e. g., in terms of clogD and polar surface area, are effective permeation enhancers. The neo-glycolipid based on decanoic acid and glucose was more potent than related compounds based on disaccharides. Significant differences in solubility and cellular compatibility were found for neo-glyolipids based on different carbohydrates. Finally, neo-glycolipids were evaluated as permeation enhancers for the peptide hormone PYY3-36. Glucose- and maltose-derived neo-glycolipids based on decanoic and dodecanoic acid showed promising enhancements in PYY3-36 permeation in vitro while maintaining good cellular compatibility, relevant for oral delivery of obesity treatments.
Collapse
Affiliation(s)
- Panagiota Kalomoiri
- Department of Chemistry, University of Copenhagen, Thorvaldsensvej 40, DK-1871, Frederiksberg, Denmark
- Center for Biopharmaceuticals and Biobarriers in Drug Delivery (BioDelivery)
| | - Janni S Mortensen
- Drug Delivery and Biophysics of Biopharmaceuticals, Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100, Copenhagen, Denmark
- Center for Biopharmaceuticals and Biobarriers in Drug Delivery (BioDelivery)
| | - Niels Johan Christensen
- Department of Chemistry, University of Copenhagen, Thorvaldsensvej 40, DK-1871, Frederiksberg, Denmark
- Center for Biopharmaceuticals and Biobarriers in Drug Delivery (BioDelivery)
| | - Kasper K Sørensen
- Department of Chemistry, University of Copenhagen, Thorvaldsensvej 40, DK-1871, Frederiksberg, Denmark
| | - Hanne Mørck Nielsen
- Drug Delivery and Biophysics of Biopharmaceuticals, Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100, Copenhagen, Denmark
- Center for Biopharmaceuticals and Biobarriers in Drug Delivery (BioDelivery)
| | - Knud J Jensen
- Department of Chemistry, University of Copenhagen, Thorvaldsensvej 40, DK-1871, Frederiksberg, Denmark
- Center for Biopharmaceuticals and Biobarriers in Drug Delivery (BioDelivery)
| | - Mikkel B Thygesen
- Department of Chemistry, University of Copenhagen, Thorvaldsensvej 40, DK-1871, Frederiksberg, Denmark
- Center for Biopharmaceuticals and Biobarriers in Drug Delivery (BioDelivery)
| |
Collapse
|
11
|
Jia Q, Young D, Zhang Q, Sieburth D. Endogenous hydrogen peroxide positively regulates secretion of a gut-derived peptide in neuroendocrine potentiation of the oxidative stress response in Caenorhabditis elegans. eLife 2024; 13:RP97503. [PMID: 39636673 PMCID: PMC11620748 DOI: 10.7554/elife.97503] [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: 12/07/2024] Open
Abstract
The gut-brain axis mediates bidirectional signaling between the intestine and the nervous system and is critical for organism-wide homeostasis. Here, we report the identification of a peptidergic endocrine circuit in which bidirectional signaling between neurons and the intestine potentiates the activation of the antioxidant response in Caenorhabditis elegans in the intestine. We identify an FMRF-amide-like peptide, FLP-2, whose release from the intestine is necessary and sufficient to activate the intestinal oxidative stress response by promoting the release of the antioxidant FLP-1 neuropeptide from neurons. FLP-2 secretion from the intestine is positively regulated by endogenous hydrogen peroxide (H2O2) produced in the mitochondrial matrix by sod-3/superoxide dismutase, and is negatively regulated by prdx-2/peroxiredoxin, which depletes H2O2 in both the mitochondria and cytosol. H2O2 promotes FLP-2 secretion through the DAG and calcium-dependent protein kinase C family member pkc-2 and by the SNAP25 family member aex-4 in the intestine. Together, our data demonstrate a role for intestinal H2O2 in promoting inter-tissue antioxidant signaling through regulated neuropeptide-like protein exocytosis in a gut-brain axis to activate the oxidative stress response.
Collapse
Affiliation(s)
- Qi Jia
- Development, Stem Cells and Regenerative Medicine PhD program, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
- Neuromedicine Graduate Program, University of Southern CaliforniaLos AngelesUnited States
| | - Drew Young
- Neuroscience Graduate Program, University of Southern CaliforniaLos AngelesUnited States
- Zilkha Neurogenetic Institute, University of Southern CaliforniaLos AngelesUnited States
| | - Qixin Zhang
- Neuromedicine Graduate Program, University of Southern CaliforniaLos AngelesUnited States
- Zilkha Neurogenetic Institute, University of Southern CaliforniaLos AngelesUnited States
| | - Derek Sieburth
- Zilkha Neurogenetic Institute, University of Southern CaliforniaLos AngelesUnited States
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| |
Collapse
|
12
|
Dagbasi A, Fuller A, Hanyaloglu AC, Carroll B, McLaughlin J, Frost G, Holliday A. The role of nutrient sensing dysregulation in anorexia of ageing: The little we know and the much we don't. Appetite 2024; 203:107718. [PMID: 39423861 DOI: 10.1016/j.appet.2024.107718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 08/01/2024] [Accepted: 10/15/2024] [Indexed: 10/21/2024]
Abstract
The age-related decline in appetite and food intake - termed "anorexia of ageing" - is implicated in undernutrition in later life and hence provides a public health challenge for our ageing population. Eating behaviour is controlled, in part, by homeostatic mechanisms which sense nutrient status and provide feedback to appetite control regions of the brain. Such feedback signals, propagated by episodic gut hormones, are dysregulated in some older adults. The secretory responses of appetite-related gut hormones to feeding are amplified, inducing a more anorexigenic signal which is associated with reduced appetite and food intake. Such an augmented response would indicate an increase in gut sensitivity to nutrients. Consequently, this review explores the role of gastrointestinal tract nutrient sensing in age-related appetite dysregulation. We review and synthesise evidence for age-related alterations in nutrient sensing which may explain the observed hormonal dysregulation. Drawing on what is known regarding elements of nutrient sensing pathways in animal models, in other tissues of the body, and in certain models of disease, we identify potential causal mechanisms including alterations in enteroendocrine cell number and distribution, dysregulation of cell signalling pathways, and changes in the gut milieu. From identified gaps in evidence, we highlight interesting and important avenues for future research.
Collapse
Affiliation(s)
- Aygul Dagbasi
- Section of Nutrition, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, 6th Floor Commonwealth Building, Hammersmith Hospital, London, W12 0NN, UK
| | - Amy Fuller
- Research Centre for Health and Life Sciences, Institute of Health and Wellbeing, Faculty of Health and Life Science, Coventry University, Coventry, CV1 5FB, UK
| | - Aylin C Hanyaloglu
- Institute of Reproductive and Developmental Biology (IRDB), Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, W12 0NN, UK
| | - Bernadette Carroll
- School of Biochemistry, University of Bristol, University Walk, Bristol, BS1 8TD, UK
| | - John McLaughlin
- Division of Diabetes, Endocrinology and Gastroenterology, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester and Manchester Academic Health Sciences Centre, Manchester, M13 9PT, UK
| | - Gary Frost
- Section of Nutrition, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, 6th Floor Commonwealth Building, Hammersmith Hospital, London, W12 0NN, UK
| | - Adrian Holliday
- School of Biomedical, Nutritional, and Sport Science, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, NE2 4HH, UK; Human Nutrition and Exercise Research Centre, Population Health Sciences Institute, Newcastle University, Newcastle Upon Tyne, NE2 4HH, UK.
| |
Collapse
|
13
|
Rosendo-Silva D, Lopes E, Monteiro-Alfredo T, Falcão-Pires I, Eickhoff H, Viana S, Reis F, Pires AS, Abrantes AM, Botelho MF, Seiça R, Matafome P. The adipose tissue melanocortin 3 receptor is targeted by ghrelin and leptin and may be a therapeutic target in obesity. Mol Cell Endocrinol 2024; 594:112367. [PMID: 39293775 DOI: 10.1016/j.mce.2024.112367] [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: 07/24/2024] [Revised: 09/03/2024] [Accepted: 09/07/2024] [Indexed: 09/20/2024]
Abstract
OBJECTIVE Obesity is linked to perturbations in energy balance mechanisms, including ghrelin and leptin actions at the hypothalamic circuitry of neuropeptide Y (NPY) and melanocortin. However, information about the regulation of this system in the periphery is still scarce. Our objective was to study the regulation of the NPY/melanocortin system in the adipose tissue (AT) and evaluate its therapeutic potential for obesity and type 2 diabetes. METHODS The expression of the NPY/melanocortin receptors' levels was assessed in the visceral AT of individuals with obesity and altered metabolism. Protein levels of these receptors were evaluated in cultured adipocytes incubated with ghrelin (30 and 100 ng/mL) and leptin (1 and 10 nM) and in the AT of an animal model with a mutation in the leptin receptor (ZSF1 rat), to understand their regulation by leptin and ghrelin. The vertical sleeve gastrectomy animal model was used to evaluate the putative therapeutic potential of the NPY/melanocortin system. RESULTS In this study, we unravelled that leptin (1 nM and 10 nM) selectively reduced the levels of NPY5R and MC3R but no other NPYR/MCRs in cultured adipocytes. In turn, acylated ghrelin (100 ng/mL) significantly increased NPY1R, but the inhibition of its receptor also abrogates MC3R levels. However, in the Lepr-deficient ZSF1 rat, both NPY5R and MC3R levels were reduced, along with other NPYRs and MCRs, suggesting that leptin resistance negatively affects NPY and melanocortin signalling. In human adipose tissue, we found a downregulation of genes encoding the NPY and melanocortin receptors in the visceral AT of individuals with obesity and insulin resistance, being correlated with genes regulating metabolic activity. Additionally, diabetic obese rats submitted to vertical sleeve gastrectomy showed increased levels of NPY, melanocortin, ghrelin, and leptin receptors in the AT, including MC3R, suggesting it may constitute a therapeutic target in obesity. CONCLUSIONS Our results suggest that the AT NPY/melanocortin system, particularly the MC3R, may be involved in the neuroendocrine regulation of adipocyte metabolism. Altogether, our work shows MC3R is under the control of the ghrelin/leptin duo, is reduced in patients with obesity and prediabetes, and may constitute a therapeutic target in obesity.
Collapse
Affiliation(s)
- Daniela Rosendo-Silva
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR) and Institute of Physiology, Faculty of Medicine, Coimbra, Portugal; University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal; Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal.
| | - Eduardo Lopes
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR) and Institute of Physiology, Faculty of Medicine, Coimbra, Portugal; University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
| | - Tamaeh Monteiro-Alfredo
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR) and Institute of Physiology, Faculty of Medicine, Coimbra, Portugal; University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
| | - Inês Falcão-Pires
- UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine of the University of Porto, Porto, Portugal
| | - Hans Eickhoff
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR) and Institute of Physiology, Faculty of Medicine, Coimbra, Portugal
| | - Sofia Viana
- University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal; Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal; University of Coimbra, iCBR and Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, Coimbra, Portugal; Polytechnic University of Coimbra, Coimbra Health School (ESTeSC), Coimbra, Portugal
| | - Flávio Reis
- University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal; Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal; University of Coimbra, iCBR and Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, Coimbra, Portugal
| | - Ana Salomé Pires
- University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal; Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal; University of Coimbra, iCBR Area of Environment Genetics and Oncobiology (CIMAGO), Institute of Biophysics, Faculty of Medicine, Coimbra, Portugal
| | - Ana Margarida Abrantes
- University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal; Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal; University of Coimbra, iCBR Area of Environment Genetics and Oncobiology (CIMAGO), Institute of Biophysics, Faculty of Medicine, Coimbra, Portugal
| | - Maria Filomena Botelho
- University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal; Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal; University of Coimbra, iCBR Area of Environment Genetics and Oncobiology (CIMAGO), Institute of Biophysics, Faculty of Medicine, Coimbra, Portugal
| | - Raquel Seiça
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR) and Institute of Physiology, Faculty of Medicine, Coimbra, Portugal
| | - Paulo Matafome
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR) and Institute of Physiology, Faculty of Medicine, Coimbra, Portugal; University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal; Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal; Polytechnic University of Coimbra, Coimbra Health School (ESTeSC), Coimbra, Portugal.
| |
Collapse
|
14
|
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
|
15
|
Nielipińska D, Rubiak D, Pietrzyk-Brzezińska AJ, Małolepsza J, Błażewska KM, Gendaszewska-Darmach E. Stapled peptides as potential therapeutics for diabetes and other metabolic diseases. Biomed Pharmacother 2024; 180:117496. [PMID: 39362065 DOI: 10.1016/j.biopha.2024.117496] [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: 07/04/2024] [Revised: 09/10/2024] [Accepted: 09/24/2024] [Indexed: 10/05/2024] Open
Abstract
The field of peptide drug research has experienced notable progress, with stapled peptides featuring stabilized α-helical conformation, emerging as a promising field. These peptides offer enhanced stability, cellular permeability, and binding affinity and exhibit potential in the treatment of diabetes and metabolic disorders. Stapled peptides, through the disruption of protein-protein interactions, present varied functionalities encompassing agonism, antagonism, and dual-agonism. This comprehensive review offers insight into the technology of peptide stapling and targeting of crucial molecular pathways associated with glucose metabolism, insulin secretion, and food intake. Additionally, we address the challenges in developing stapled peptides, including concerns pertaining to structural stability, peptide helicity, isomer mixture, and potential side effects.
Collapse
Affiliation(s)
- Dominika Nielipińska
- Institute of Molecular and Industrial Biotechnology, Faculty of Biotechnology and Food Sciences, Lodz University of Technology, Poland.
| | - Dominika Rubiak
- Institute of Organic Chemistry, Faculty of Chemistry, Lodz University of Technology, Poland
| | - Agnieszka J Pietrzyk-Brzezińska
- Institute of Molecular and Industrial Biotechnology, Faculty of Biotechnology and Food Sciences, Lodz University of Technology, Poland
| | - Joanna Małolepsza
- Institute of Organic Chemistry, Faculty of Chemistry, Lodz University of Technology, Poland
| | - Katarzyna M Błażewska
- Institute of Organic Chemistry, Faculty of Chemistry, Lodz University of Technology, Poland.
| | - Edyta Gendaszewska-Darmach
- Institute of Molecular and Industrial Biotechnology, Faculty of Biotechnology and Food Sciences, Lodz University of Technology, Poland.
| |
Collapse
|
16
|
Tanday N, Zhu W, Tarasov AI, Flatt PR, Irwin N. [P 3]PP, a stable, long-acting pancreatic polypeptide analogue, evokes weight lowering and pancreatic beta-cell-protective effects in obesity-associated diabetes. Diabetes Obes Metab 2024; 26:4945-4957. [PMID: 39192525 DOI: 10.1111/dom.15897] [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: 06/21/2024] [Revised: 08/07/2024] [Accepted: 08/07/2024] [Indexed: 08/29/2024]
Abstract
AIM To thoroughly investigate the impact of sustained neuropeptide Y4 receptor (NPY4R) activation in obesity-associated diabetes. METHODS Initially, the prolonged pharmacodynamic profile of the enzymatically stable pancreatic polypeptide (PP) analogue, [P3]PP, was confirmed in normal mice up to 24 h after injection. Subsequent to this, [P3]PP was administered twice daily (25 nmol/kg) for 28 days to high-fat-fed mice with streptozotocin-induced insulin deficiency, known as HFF/STZ mice. RESULTS Treatment with [P3]PP for 28 days reduced energy intake and was associated with notable weight loss. In addition, circulating glucose was returned to values of approximately 8 mmol/L in [P3]PP-treated mice, with significantly increased plasma insulin and decreased glucagon concentrations. Glucose tolerance and glucose-stimulated insulin secretion were improved in [P3]PP-treated HFF/STZ mice, with no obvious effect on peripheral insulin sensitivity. Benefits on insulin secretion were associated with elevated pancreatic insulin content as well as islet and beta-cell areas. Positive effects on islet architecture were linked to increased beta-cell proliferation and decreased apoptosis. Treatment intervention also decreased islet alpha-cell area, but pancreatic glucagon content remained unaffected. In addition, [P3]PP-treated HFF/STZ mice presented with reduced plasma alanine transaminase and aspartate transaminase levels, with no change in circulating amylase concentrations. In terms of plasma lipid profile, triglyceride and cholesterol levels were significantly decreased by [P3]PP treatment, when compared to saline controls. CONCLUSION Collectively, these data highlight for the first time the potential of enzymatically stable PP analogues for the treatment of obesity and related diabetes.
Collapse
Affiliation(s)
- Neil Tanday
- Diabetes Research Centre, Ulster University, Coleraine, UK
| | - Wuyun Zhu
- Diabetes Research Centre, Ulster University, Coleraine, UK
| | | | - Peter R Flatt
- Diabetes Research Centre, Ulster University, Coleraine, UK
| | - Nigel Irwin
- Diabetes Research Centre, Ulster University, Coleraine, UK
| |
Collapse
|
17
|
Cao S, Pierson JT, Bond AH, Zhang S, Gold A, Zhang H, Zamary KM, Moats P, Teegarden MD, Peterson DG, Mo X, Zhu J, Bruno RS. Intestinal-level anti-inflammatory bioactivities of whole wheat: Rationale, design, and methods of a randomized, controlled, crossover dietary trial in adults with prediabetes. Nutr Res 2024; 131:83-95. [PMID: 39378659 DOI: 10.1016/j.nutres.2024.09.010] [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: 05/29/2024] [Revised: 09/12/2024] [Accepted: 09/13/2024] [Indexed: 10/10/2024]
Abstract
Randomized controlled trials (RCT) demonstrate that whole wheat consumption improves glycemia. However, substantial inter-individual variation is often observed, highlighting that dietary whole grain recommendations may not support the health of all persons. The objective of this report is to describe the rationale and design of a planned RCT aimed at establishing the gut microbiota and metabolome signatures that predict whole wheat-mediated improvements in glucose tolerance in adults with prediabetes. It is hypothesized that a controlled diet containing wheat bread (WHEAT; 160 g/day) compared with refined bread (WHITE) will improve glucose tolerance in a gut microbiota-mediated manner. Biospecimens will be collected before and after each 2-week study arm. Testing for oral glucose tolerance and gastrointestinal permeability will be performed post-intervention. Assessments will include oral glucose tolerance (primary outcome) and secondary outcomes including gut microbiota, targeted and untargeted metabolomics of fecal and plasma samples, intestinal and host inflammatory responses, and intestinal permeability. WHEAT is predicted to alleviate glucose intolerance by shifting microbiota composition to increase short-chain fatty acid-producing bacteria while reducing populations implicated in intestinal inflammation, barrier dysfunction, and systemic endotoxemia. Further, benefits from WHEAT are anticipated to correlate with gut-level and systemic metabolomic responses that can help to explain the expected inter-individual variability in glucose tolerance. Thus, knowledge gained from integrating multi-omic responses associating with glucose tolerance could help to establish a precision nutrition-based framework that can alleviate cardiometabolic risk. This framework could inform novel dietary whole grain recommendations by enhancing our understanding of inter-individual responsiveness to whole grain consumption.
Collapse
Affiliation(s)
- Sisi Cao
- Human Nutrition Program, The Ohio State University, Columbus, OH, USA
| | - Jillian T Pierson
- Human Nutrition Program, The Ohio State University, Columbus, OH, USA
| | - Ariana H Bond
- Human Nutrition Program, The Ohio State University, Columbus, OH, USA
| | - Shiqi Zhang
- Human Nutrition Program, The Ohio State University, Columbus, OH, USA; James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Andrew Gold
- Human Nutrition Program, The Ohio State University, Columbus, OH, USA; James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Huan Zhang
- Human Nutrition Program, The Ohio State University, Columbus, OH, USA; James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Kaitlyn M Zamary
- Human Nutrition Program, The Ohio State University, Columbus, OH, USA
| | - Palmer Moats
- Human Nutrition Program, The Ohio State University, Columbus, OH, USA
| | - Matthew D Teegarden
- Foods for Health Research Initiative, The Ohio State University, Columbus, OH, USA
| | - Devin G Peterson
- Department of Food Science and Technology, The Ohio State University, Columbus, OH, USA
| | - Xiaokui Mo
- Center for Biostatistics, Department of Biomedical Informatics, The Ohio State University, Columbus, OH, USA
| | - Jiangjiang Zhu
- Human Nutrition Program, The Ohio State University, Columbus, OH, USA; James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Richard S Bruno
- Human Nutrition Program, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
18
|
Siddique AB, Shaw B, Dwyer J, Fields DA, Fontaine K, Hand D, Schekman R, Alberts J, Locher J, Allison DB. Hidden: A Baker's Dozen Ways in Which Research Reporting is Less Transparent than it Could be and Suggestions for Implementing Einstein's Dictum. SCIENCE AND ENGINEERING ETHICS 2024; 30:48. [PMID: 39412686 PMCID: PMC11485062 DOI: 10.1007/s11948-024-00517-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 09/10/2024] [Indexed: 10/19/2024]
Abstract
The tutelage of our mentors as scientists included the analogy that writing a good scientific paper was an exercise in storytelling that omitted unessential details that did not move the story forward or that detracted from the overall message. However, the advice to not get lost in the details had an important flaw. In science, it is the many details of the data themselves and the methods used to generate and analyze them that give conclusions their probative meaning. Facts may sometimes slow or distract from the clarity, tidiness, intrigue, or flow of the narrative, but nevertheless they are important for the assessment of what was done, the trustworthiness of the science, and the meaning of the findings. Nevertheless, many critical elements and facts about research studies may be omitted from the narrative and become hidden from scholarly scrutiny. We describe a "baker's dozen" shortfalls in which such elements that are pertinent to evaluating the validity of scientific studies are sometimes hidden in reports of the work. Such shortfalls may be intentional or unintentional or lie somewhere in between. Additionally, shortfalls may occur at the level of the individual or an institution or of the entire system itself. We conclude by proposing countermeasures to these shortfalls.
Collapse
Affiliation(s)
- Abu Bakkar Siddique
- School of Public Administration, Florida Atlantic University, Boca Raton, FL, USA
| | - Brian Shaw
- Department of Epidemiology and Biostatistics, School of Public Health, Indiana University Bloomington, 1025 E 7 St, PH 111, Bloomington, IN, 47405, USA
| | - Johanna Dwyer
- School of Medicine, Friedman School of Nutrition Science and Policy, Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Medford, MA, USA
| | - David A Fields
- Department of Pediatrics, The University of Oklahoma Health Sciences, Oklahoma City, OK, USA
| | - Kevin Fontaine
- Department of Health Behavior, School of Public Health, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - David Hand
- Department of Mathematics, Imperial College, London, UK
| | - Randy Schekman
- Department of Molecular & Cell Biology, University of California, Berkeley, CA, USA
| | - Jeffrey Alberts
- Department of Psychological and Brain Sciences, Indiana University Bloomington, Bloomington, IN, USA
| | - Julie Locher
- Department of Epidemiology and Biostatistics, School of Public Health, Indiana University Bloomington, 1025 E 7 St, PH 111, Bloomington, IN, 47405, USA
- Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - David B Allison
- Department of Epidemiology and Biostatistics, School of Public Health, Indiana University Bloomington, 1025 E 7 St, PH 111, Bloomington, IN, 47405, USA.
| |
Collapse
|
19
|
Reddy YNV, Melenovsky V, Asokan AK, Haluzik M, Carter RE, Nair S, Jensen MD, Borlaug BA. Dapagliflozin, peptide YY, and weight loss in heart failure with preserved ejection fraction. Eur Heart J 2024; 45:3889-3891. [PMID: 39167062 PMCID: PMC11452744 DOI: 10.1093/eurheartj/ehae534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 07/05/2024] [Accepted: 08/04/2024] [Indexed: 08/23/2024] Open
Affiliation(s)
- Yogesh N V Reddy
- The Department of Cardiovascular Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN, USA
| | - Vojtech Melenovsky
- Institute for Clinical and Experimental Medicine (IKEM), Prague, Czech Republic
| | - Aneesh K Asokan
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Mayo Clinic, Rochester, MN, USA
| | - Martin Haluzik
- Institute for Clinical and Experimental Medicine (IKEM), Prague, Czech Republic
| | - Rickey E Carter
- Department of Quantitative Health Sciences, Division of Clinical Trials & Biostatistics, Mayo Clinic, Jacksonville, FL, USA
| | - Sreekumaran Nair
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Mayo Clinic, Rochester, MN, USA
| | - Michael D Jensen
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Mayo Clinic, Rochester, MN, USA
| | - Barry A Borlaug
- The Department of Cardiovascular Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN, USA
| |
Collapse
|
20
|
Jia Q, Young D, Zhang Q, Sieburth D. Endogenous hydrogen peroxide positively regulates secretion of a gut-derived peptide in neuroendocrine potentiation of the oxidative stress response in C. elegans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.03.587937. [PMID: 39345448 PMCID: PMC11429608 DOI: 10.1101/2024.04.03.587937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
The gut-brain axis mediates bidirectional signaling between the intestine and the nervous system and is critical for organism-wide homeostasis. Here we report the identification of a peptidergic endocrine circuit in which bidirectional signaling between neurons and the intestine potentiates the activation of the antioxidant response in C. elegans in the intestine. We identify a FMRF-amide-like peptide, FLP-2, whose release from the intestine is necessary and sufficient to activate the intestinal oxidative stress response by promoting the release of the antioxidant FLP-1 neuropeptide from neurons. FLP-2 secretion from the intestine is positively regulated by endogenous hydrogen peroxide (H2O2) produced in the mitochondrial matrix by sod-3/superoxide dismutase, and is negatively regulated by prdx-2/peroxiredoxin, which depletes H2O2 in both the mitochondria and cytosol. H2O2 promotes FLP-2 secretion through the DAG and calciumdependent protein kinase C family member pkc-2 and by the SNAP25 family member aex-4 in the intestine. Together, our data demonstrate a role for intestinal H2O2 in promoting inter-tissue antioxidant signaling through regulated neuropeptide-like protein exocytosis in a gut-brain axis to activate the oxidative stress response.
Collapse
Affiliation(s)
- Qi Jia
- Development, Stem Cells and Regenerative Medicine PhD program, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033
- Neuromedicine Graduate Program, University of Southern California, Los Angeles, CA 90089
| | - Drew Young
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA 90089
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA 90033
| | - Qixin Zhang
- Neuromedicine Graduate Program, University of Southern California, Los Angeles, CA 90089
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA 90033
| | - Derek Sieburth
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA 90033
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033
| |
Collapse
|
21
|
Jensen O, Trujillo E, Hanson L, Ost KS. Controlling Candida: immune regulation of commensal fungi in the gut. Infect Immun 2024; 92:e0051623. [PMID: 38647290 PMCID: PMC11385159 DOI: 10.1128/iai.00516-23] [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: 04/25/2024] Open
Abstract
The intestinal microbiome harbors fungi that pose a significant risk to human health as opportunistic pathogens and drivers of inflammation. Inflammatory and autoimmune diseases are associated with dysbiotic fungal communities and the expansion of potentially pathogenic fungi. The gut is also the main reservoir for disseminated fungal infections. Immune interactions are critical for preventing commensal fungi from becoming pathogenic. Significant strides have been made in defining innate and adaptive immune pathways that regulate intestinal fungi, and these discoveries have coincided with advancements in our understanding of the fungal molecular pathways and effectors involved in both commensal colonization and pathogenesis within the gut. In this review, we will discuss immune interactions important for regulating commensal fungi, with a focus on how specific cell types and effectors interact with fungi to limit their colonization or pathogenic potential. This will include how innate and adaptive immune pathways target fungi and orchestrate antifungal immune responses, in addition to how secreted immune effectors, such as mucus and antimicrobial peptides, regulate fungal colonization and inhibit pathogenic potential. These immune interactions will be framed around our current understanding of the fungal effectors and pathways regulating colonization and pathogenesis within this niche. Finally, we highlight important unexplored mechanisms by which the immune system regulates commensal fungi in the gut.
Collapse
Affiliation(s)
- Owen Jensen
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Emma Trujillo
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Luke Hanson
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Kyla S. Ost
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
22
|
Lytvyak E, Zarrinpar A, Ore CD, Lee E, Yazdani-Boset K, Horgan S, Grunvald E. Stronger control of eating 3 months after sleeve gastrectomy predicts successful weight loss outcomes at one year. OBESITY PILLARS 2024; 11:100111. [PMID: 38770521 PMCID: PMC11103426 DOI: 10.1016/j.obpill.2024.100111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 05/01/2024] [Accepted: 05/02/2024] [Indexed: 05/22/2024]
Abstract
Background Weight loss response to sleeve gastrectomy (SG) is variable and predicting the effectiveness of surgery is challenging and elusive. The aim of our study was to assess and quantify the association between eating control and weight loss outcomes and identify the control of eating (CoE) attributes during the early postoperative period that might predict good vs. poor response to SG at one year. Methods A prospective longitudinal cohort study using the Control of Eating Questionnaire (CoEQ) was designed as a series before and at 3-, 6-, and 12-months post-SG. Primary outcomes were changes in CoE attributes and percent of total weight loss (%TWL) 12-months post-surgery. Subjects were categorized based on %TWL as good (GR, ≥25 %) or poor responders (PR, <25 %). A receiver operating characteristic and logistic regression analyses were performed. Results We included 41 participants (80.5% females, 51.2% Hispanic, mean age 41.7±10.6, median baseline body mass index (BMI) 43.6 kg/m2 [range 35.2-66.3]) who completed the CoEQ at all four timepoints. The "Difficulty to control eating" score at 3 months revealed the highest area under the curve (AUC) (AUC 0.711; 95%CI 0.524-0.898; p=0.032). In a trade-off between a high Youden index and high sensitivity, the "Difficulty to control eating" score of 7 at 3 months was identified as the optimal cut-off for distinguishing between GRs and PRs. Score ≤7 at 3 months was strongly independently associated with a successful weight loss target of 25%TWL at one-year post-SG (Relative Risk 4.43; 95%CI 1.06-18.54; p=0.042). Conclusion "Difficulty to control eating" score at 3 months post-SG is an independent early predictor of optimal response (achieving a successful TWL target of ≥25 % at one-year post-SG). Our results support the utility of this easy-to-administer validated tool for predicting the effectiveness of SG and may assist in identifying individuals with suboptimal response early and helping them with interventions to attain optimal weight loss targets.
Collapse
Affiliation(s)
- Ellina Lytvyak
- Division of Preventive Medicine, Department of Medicine, University of Alberta, 5-30 University Terrace, 8303 112 Street, Edmonton, Alberta, T6G 2T4, Canada
| | - Amir Zarrinpar
- Division of Gastroenterology, University of California San Diego, La Jolla, CA, USA
- School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Cecilia Dalle Ore
- School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Euyhyun Lee
- Altman Clinical and Translational Research Institute, University of California San Diego, La Jolla, CA, USA
| | | | - Santiago Horgan
- Bariatric and Metabolic Institute, Division of Minimally Invasive Surgery, University of California San Diego, 4303 La Jolla Village Drive, Suite 2110, San Diego, CA, 92122, USA
| | - Eduardo Grunvald
- School of Medicine, University of California San Diego, La Jolla, CA, USA
- Division of General Internal Medicine, University of California San Diego, La Jolla, CA, USA
- Bariatric and Metabolic Institute, Division of Minimally Invasive Surgery, University of California San Diego, 4303 La Jolla Village Drive, Suite 2110, San Diego, CA, 92122, USA
| |
Collapse
|
23
|
Lafferty RA, Flatt PR, Irwin N. NPYR modulation: Potential for the next major advance in obesity and type 2 diabetes management? Peptides 2024; 179:171256. [PMID: 38825012 DOI: 10.1016/j.peptides.2024.171256] [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: 03/15/2024] [Revised: 05/13/2024] [Accepted: 05/30/2024] [Indexed: 06/04/2024]
Abstract
The approval of the glucagon-like peptide 1 (GLP-1) mimetics semaglutide and liraglutide for management of obesity, independent of type 2 diabetes (T2DM), has initiated a resurgence of interest in gut-hormone derived peptide therapies for the management of metabolic diseases, but side-effect profile is a concern for these medicines. However, the recent approval of tirzepatide for obesity and T2DM, a glucose-dependent insulinotropic polypeptide (GIP), GLP-1 receptor co-agonist peptide therapy, may provide a somewhat more tolerable option. Despite this, an increasing number of non-incretin alternative peptides are in development for obesity, and it stands to reason that other hormones will take to the limelight in the coming years, such as peptides from the neuropeptide Y family. This narrative review outlines the therapeutic promise of the neuropeptide Y family of peptides, comprising of the 36 amino acid polypeptides neuropeptide Y (NPY), peptide tyrosine-tyrosine (PYY) and pancreatic polypeptide (PP), as well as their derivatives. This family of peptides exerts a number of metabolically relevant effects such as appetite regulation and can influence pancreatic beta-cell survival. Although some of these actions still require full translation to the human setting, potential therapeutic application in obesity and type 2 diabetes is conceivable. However, like GLP-1 and GIP, the endogenous NPY, PYY and PP peptide forms are subject to rapid in vivo degradation and inactivation by the serine peptidase, dipeptidyl-peptidase 4 (DPP-4), and hence require structural modification to prolong circulating half-life. Numerous protective modification strategies are discussed in this regard herein, alongside related impact on biological activity profile and therapeutic promise.
Collapse
Affiliation(s)
- Ryan A Lafferty
- Diabetes Research Centre, Ulster University, Coleraine, Northern Ireland BT52 1SA, UK.
| | - Peter R Flatt
- Diabetes Research Centre, Ulster University, Coleraine, Northern Ireland BT52 1SA, UK
| | - Nigel Irwin
- Diabetes Research Centre, Ulster University, Coleraine, Northern Ireland BT52 1SA, UK
| |
Collapse
|
24
|
Liu CC, Khan A, Seban N, Littlejohn N, Shah A, Srinivasan S. A homeostatic gut-to-brain insulin antagonist restrains neuronally stimulated fat loss. Nat Commun 2024; 15:6869. [PMID: 39127676 PMCID: PMC11316803 DOI: 10.1038/s41467-024-51077-3] [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: 09/29/2023] [Accepted: 07/29/2024] [Indexed: 08/12/2024] Open
Abstract
In C. elegans mechanisms by which peripheral organs relay internal state information to the nervous system remain unknown, although strong evidence suggests that such signals do exist. Here we report the discovery of a peptide of the ancestral insulin superfamily called INS-7 that functions as an enteroendocrine peptide and is secreted from specialized cells of the intestine. INS-7 secretion is stimulated by food withdrawal, increases during fasting and acts as a bona fide gut-to-brain peptide that attenuates the release of a neuropeptide that drives fat loss in the periphery. Thus, INS-7 functions as a homeostatic signal from the intestine that gates the neuronal drive to stimulate fat loss during food shortage. Mechanistically, INS-7 functions as an antagonist at the canonical DAF-2 receptor and functions via FOXO and AMPK signaling in ASI neurons. Phylogenetic analysis suggests that INS-7 bears greater resemblance to members of the broad insulin/relaxin superfamily than to conventional mammalian insulin and IGF peptides. The discovery of an endogenous insulin antagonist secreted by specialized intestinal cells with enteroendocrine functions suggests unexpected and important properties of the intestine and its role in directing neuronal functions.
Collapse
Affiliation(s)
- Chung-Chih Liu
- Department of Neuroscience and Dorris Neuroscience Center, The Scripps Research Institute, San Diego, CA, USA
- The Skaggs Graduate School of Chemical and Biological Sciences, The Scripps Research Institute, San Diego, CA, USA
| | - Ayub Khan
- Department of Neuroscience and Dorris Neuroscience Center, The Scripps Research Institute, San Diego, CA, USA
| | - Nicolas Seban
- Department of Neuroscience and Dorris Neuroscience Center, The Scripps Research Institute, San Diego, CA, USA
| | - Nicole Littlejohn
- Department of Neuroscience and Dorris Neuroscience Center, The Scripps Research Institute, San Diego, CA, USA
| | - Aayushi Shah
- Department of Neuroscience and Dorris Neuroscience Center, The Scripps Research Institute, San Diego, CA, USA
| | - Supriya Srinivasan
- Department of Neuroscience and Dorris Neuroscience Center, The Scripps Research Institute, San Diego, CA, USA.
| |
Collapse
|
25
|
Farooqi IS, Xu Y. Translational potential of mouse models of human metabolic disease. Cell 2024; 187:4129-4143. [PMID: 39067442 DOI: 10.1016/j.cell.2024.07.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 07/05/2024] [Accepted: 07/05/2024] [Indexed: 07/30/2024]
Abstract
Obesity causes significant morbidity and mortality globally. Research in the last three decades has delivered a step-change in our understanding of the fundamental mechanisms that regulate energy homeostasis, building on foundational discoveries in mouse models of metabolic disease. However, not all findings made in rodents have translated to humans, hampering drug discovery in this field. Here, we review how studies in mice and humans have informed our current framework for understanding energy homeostasis, discuss their challenges and limitations, and offer a perspective on how human studies may play an increasingly important role in the discovery of disease mechanisms and identification of therapeutic targets in the future.
Collapse
Affiliation(s)
- I Sadaf Farooqi
- University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science and NIHR Cambridge Biomedical Research Centre, Cambridge, UK.
| | - Yong Xu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Department of Molecular and Cellular Biology and Department of Medicine, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
26
|
Rubinić I, Kurtov M, Likić R. Novel Pharmaceuticals in Appetite Regulation: Exploring emerging gut peptides and their pharmacological prospects. Pharmacol Res Perspect 2024; 12:e1243. [PMID: 39016695 PMCID: PMC11253306 DOI: 10.1002/prp2.1243] [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: 12/12/2023] [Revised: 03/23/2024] [Accepted: 06/22/2024] [Indexed: 07/18/2024] Open
Abstract
Obesity, a global health challenge, necessitates innovative approaches for effective management. Targeting gut peptides in the development of anti-obesity pharmaceuticals has already demonstrated significant efficacy. Ghrelin, peptide YY (PYY), cholecystokinin (CCK), and amylin are crucial in appetite regulation offering promising targets for pharmacological interventions in obesity treatment using both peptide-based and small molecule-based pharmaceuticals. Ghrelin, a sole orexigenic gut peptide, has a potential for anti-obesity therapies through various approaches, including endogenous ghrelin neutralization, ghrelin receptor antagonists, ghrelin O-acyltransferase, and functional inhibitors. Anorexigenic gut peptides, peptide YY, cholecystokinin, and amylin, have exhibited appetite-reducing effects in animal models and humans. Overcoming substantial obstacles is imperative for translating these findings into clinically effective pharmaceuticals. Peptide YY and cholecystokinin analogues, characterized by prolonged half-life and resistance to proteolytic enzymes, present viable options. Positive allosteric modulators emerge as a novel approach for modulating the cholecystokinin pathway. Amylin is currently the most promising, with both amylin analogues and dual amylin and calcitonin receptor agonists (DACRAs) progressing to advanced stages of clinical trials. Despite persistent challenges, innovative pharmaceutical strategies provide a glimpse into the future of anti-obesity therapies.
Collapse
Affiliation(s)
- Igor Rubinić
- Department of Basic and Clinical Pharmacology and Toxicology, Faculty of MedicineUniversity of RijekaRijekaCroatia
- Clinical Pharmacology unitClinical Hospital Center RijekaRijekaCroatia
| | - Marija Kurtov
- Division of Clinical Pharmacology and Toxicology, Department of Internal MedicineUniversity Hospital “Sveti Duh”ZagrebCroatia
| | - Robert Likić
- Department of Internal MedicineSchool of Medicine University of ZagrebZagrebCroatia
| |
Collapse
|
27
|
Elizazu J, Artetxe-Zurutuza A, Otaegi-Ugartemendia M, Moncho-Amor V, Moreno-Valladares M, Matheu A, Carrasco-Garcia E. Identification of a novel gene signature related to prognosis and metastasis in gastric cancer. Cell Oncol (Dordr) 2024; 47:1355-1373. [PMID: 38480611 PMCID: PMC11322236 DOI: 10.1007/s13402-024-00932-y] [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] [Accepted: 03/02/2024] [Indexed: 08/16/2024] Open
Abstract
BACKGROUND Gastric Cancer (GC) presents poor outcome, which is consequence of the high incidence of recurrence and metastasis at early stages. GC patients presenting recurrent or metastatic disease display a median life expectancy of only 8 months. The mechanisms underlying GC progression remain poorly understood. METHODS We took advantage of public available GC datasets from TCGA using GEPIA, and identified the matched genes among the 100 genes most significantly associated with overall survival (OS) and disease free survival (DFS). Results were confirmed in ACRG cohort and in over 2000 GC cases obtained from several cohorts integrated using our own analysis pipeline. The Kaplan-Meier method and multivariate Cox regression analyses were used for prognostic significance and linear modelling and correlation analyses for association with clinic-pathological parameters and biological hallmarks. In vitro and in vivo functional studies were performed in GC cells with candidate genes and the related molecular pathways were studied by RNA sequencing. RESULTS High expression of ANKRD6, ITIH3, SORCS3, NPY1R and CCDC178 individually and as a signature was associated with poor prognosis and recurrent disease in GC. Moreover, the expression of ANKRD6 and ITIH3 was significantly higher in metastasis and their levels associated to Epithelial to Mesenchymal Transition (EMT) and stemness markers. In line with this, RNAseq analysis revealed genes involved in EMT differentially expressed in ANKRD6 silencing cells. Finally, ANKRD6 silencing in GC metastatic cells showed impairment in GC tumorigenic and metastatic traits in vitro and in vivo. CONCLUSIONS Our study identified a novel signature involved in GC malignancy and prognosis, and revealed a novel pro-metastatic role of ANKRD6 in GC.
Collapse
Affiliation(s)
- Joseba Elizazu
- Cellular Oncology Group, Biodonostia Health Research Institute, Paseo Dr. Beguiristain s/n, San Sebastian, 20014, Spain
| | - Aizpea Artetxe-Zurutuza
- Cellular Oncology Group, Biodonostia Health Research Institute, Paseo Dr. Beguiristain s/n, San Sebastian, 20014, Spain
| | - Maddalen Otaegi-Ugartemendia
- Cellular Oncology Group, Biodonostia Health Research Institute, Paseo Dr. Beguiristain s/n, San Sebastian, 20014, Spain
| | - Veronica Moncho-Amor
- Cellular Oncology Group, Biodonostia Health Research Institute, Paseo Dr. Beguiristain s/n, San Sebastian, 20014, Spain
- CIBER de Fragilidad y Envejecimiento Saludable (CIBERfes), Madrid, 28029, Spain
| | - Manuel Moreno-Valladares
- Cellular Oncology Group, Biodonostia Health Research Institute, Paseo Dr. Beguiristain s/n, San Sebastian, 20014, Spain
- CIBER de Fragilidad y Envejecimiento Saludable (CIBERfes), Madrid, 28029, Spain
- Pathology Department, Donostia University Hospital, San Sebastian, Spain
| | - Ander Matheu
- Cellular Oncology Group, Biodonostia Health Research Institute, Paseo Dr. Beguiristain s/n, San Sebastian, 20014, Spain.
- CIBER de Fragilidad y Envejecimiento Saludable (CIBERfes), Madrid, 28029, Spain.
- IKERBASQUE, Basque Foundation for Science, Bilbao, 48009, Spain.
| | - Estefania Carrasco-Garcia
- Cellular Oncology Group, Biodonostia Health Research Institute, Paseo Dr. Beguiristain s/n, San Sebastian, 20014, Spain.
- CIBER de Fragilidad y Envejecimiento Saludable (CIBERfes), Madrid, 28029, Spain.
| |
Collapse
|
28
|
Catalbas K, Pattnaik T, Congdon S, Nelson C, Villano LC, Sweeney P. Hypothalamic AgRP neurons regulate the hyperphagia of lactation. Mol Metab 2024; 86:101975. [PMID: 38925247 PMCID: PMC11268337 DOI: 10.1016/j.molmet.2024.101975] [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: 04/18/2024] [Revised: 06/19/2024] [Accepted: 06/21/2024] [Indexed: 06/28/2024] Open
Abstract
OBJECTIVE The lactational period is associated with profound hyperphagia to accommodate the energy demands of nursing. These changes are important for the long-term metabolic health of the mother and children as altered feeding during lactation increases the risk of mothers and offspring developing metabolic disorders later in life. However, the specific behavioral mechanisms and neural circuitry mediating the hyperphagia of lactation are incompletely understood. METHODS Here, we utilized home cage feeding devices to characterize the dynamics of feeding behavior in lactating mice. A combination of pharmacological and behavioral assays were utilized to determine how lactation alters meal structure, circadian aspects of feeding, hedonic feeding, and sensitivity to hunger and satiety signals in lactating mice. Finally, we utilized chemogenetic, immunohistochemical, and in vivo imaging approaches to characterize the role of hypothalamic agouti-related peptide (AgRP) neurons in lactational-hyperphagia. RESULTS The lactational period is associated with increased meal size, altered circadian patterns of feeding, reduced sensitivity to gut-brain satiety signals, and enhanced sensitivity to negative energy balance. Hypothalamic AgRP neurons display increased sensitivity to negative energy balance and altered in vivo activity during the lactational state. Further, using in vivo imaging approaches we demonstrate that AgRP neurons are directly activated by lactation. Chemogenetic inhibition of AgRP neurons acutely reduces feeding in lactating mice, demonstrating an important role for these neurons in lactational-hyperphagia. CONCLUSIONS Together, these results show that lactation collectively alters multiple components of feeding behavior and position AgRP neurons as an important cellular substrate mediating the hyperphagia of lactation.
Collapse
Affiliation(s)
- Kerem Catalbas
- University of Illinois Urbana-Champaign, Department of Molecular and Integrative Physiology, USA; University of Illinois Urbana-Champaign Neuroscience Program, USA
| | - Tanya Pattnaik
- University of Illinois Urbana-Champaign, Department of Molecular and Integrative Physiology, USA
| | - Samuel Congdon
- University of Illinois Urbana-Champaign, Department of Molecular and Integrative Physiology, USA
| | - Christina Nelson
- University of Illinois Urbana-Champaign, Department of Molecular and Integrative Physiology, USA
| | - Lara C Villano
- University of Illinois Urbana-Champaign, Department of Molecular and Integrative Physiology, USA
| | - Patrick Sweeney
- University of Illinois Urbana-Champaign, Department of Molecular and Integrative Physiology, USA; University of Illinois Urbana-Champaign Neuroscience Program, USA.
| |
Collapse
|
29
|
Canali S, Fischer AW, Nguyen M, Anderson K, Wu L, Graham AR, Hsiao CJ, Bankar C, Dussault N, Ritchie V, Goodridge M, Sparrow T, Pannoni A, Tse SW, Woo V, Klovdahl K, Iacovelli J, Huang E. Lipid-encapsulated mRNA encoding an extended serum half-life interleukin-22 ameliorates metabolic disease in mice. Mol Metab 2024; 86:101965. [PMID: 38871178 PMCID: PMC11296054 DOI: 10.1016/j.molmet.2024.101965] [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: 03/28/2024] [Revised: 05/21/2024] [Accepted: 05/31/2024] [Indexed: 06/15/2024] Open
Abstract
OBJECTIVE Interleukin (IL)-22 is a potential therapeutic protein for the treatment of metabolic diseases such as obesity, type 2 diabetes, and metabolic dysfunction-associated steatotic liver disease due to its involvement in multiple cellular pathways and observed hepatoprotective effects. The short serum half-life of IL-22 has previously limited its use in clinical applications; however, the development of mRNA-lipid nanoparticle (LNP) technology offers a novel therapeutic approach that uses a host-generated IL-22 fusion protein. In the present study, the effects of administration of an mRNA-LNP encoding IL-22 on metabolic disease parameters was investigated in various mouse models. METHODS C57BL/6NCrl mice were used to confirm mouse serum albumin (MSA)-IL-22 protein expression prior to assessments in C57BL/6NTac and CETP/ApoB transgenic mouse models of metabolic disease. Mice were fed either regular chow or a modified amylin liver nonalcoholic steatohepatitis-inducing diet prior to receiving either LNP-encapsulated MSA-IL-22 or MSA mRNA via intravenous or intramuscular injection. Metabolic markers were monitored for the duration of the experiments, and postmortem histology assessment and analysis of metabolic gene expression pathways were performed. RESULTS MSA-IL-22 was detectable for ≥8 days following administration. Improvements in body weight, lipid metabolism, glucose metabolism, and lipogenic and fibrotic marker gene expression in the liver were observed in the MSA-IL-22-treated mice, and these effects were shown to be durable. CONCLUSIONS These results support the application of mRNA-encoded IL-22 as a promising treatment strategy for metabolic syndrome and associated comorbidities in human populations.
Collapse
Affiliation(s)
- Susanna Canali
- Moderna, Inc., 325 Binney Street, Cambridge, MA 02142, USA.
| | | | - Mychael Nguyen
- Moderna, Inc., 325 Binney Street, Cambridge, MA 02142, USA.
| | - Karl Anderson
- Moderna, Inc., 325 Binney Street, Cambridge, MA 02142, USA.
| | - Lorna Wu
- Moderna, Inc., 325 Binney Street, Cambridge, MA 02142, USA.
| | | | | | | | - Nancy Dussault
- Moderna, Inc., 325 Binney Street, Cambridge, MA 02142, USA.
| | | | | | - Todd Sparrow
- Moderna, Inc., 325 Binney Street, Cambridge, MA 02142, USA.
| | | | - Sze-Wah Tse
- Moderna, Inc., 325 Binney Street, Cambridge, MA 02142, USA.
| | - Vivienne Woo
- Moderna, Inc., 325 Binney Street, Cambridge, MA 02142, USA.
| | | | | | - Eric Huang
- Moderna, Inc., 325 Binney Street, Cambridge, MA 02142, USA.
| |
Collapse
|
30
|
Yanagida B, Yamamoto T, Suzuki H. Amylin-like immunoreactivity in the extra-islet peptide YY-producing and glucagon-immunoreactive cells in Japanese quail pancreas. Anat Histol Embryol 2024; 53:e13074. [PMID: 38864153 DOI: 10.1111/ahe.13074] [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: 02/05/2024] [Revised: 05/21/2024] [Accepted: 05/31/2024] [Indexed: 06/13/2024]
Abstract
In this study, we investigated amylin-like substance distribution in the pancreas of Japanese quail (Coturnix japonica) using a specific anti-rat amylin serum. We detected amylin-immunoreactive cells dispersed in the pancreatic extra-islet region but not in the islet region. The synthetic rat amylin-containing serum pre-absorption abolished the staining profile. Almost all amylin-immunoreactive cells were immuno-positive for peptide YY (PYY). In addition, certain amylin-immunoreactive cells stained immuno-positive for glucagon. Amylin and PYY co-secreted from the extra-islet cells might participate in the insulin and glucagon release regulation in the pancreas and food intake modulation through the central nervous system.
Collapse
Affiliation(s)
- Bonten Yanagida
- Department of Biology, University of Teacher Education Fukuoka, Munakata, Fukuoka, Japan
| | - Toshiharu Yamamoto
- Department of Physical Therapy, Faculty of Medical Science, Nagoya Women's University, Nagoya, Aichi, Japan
- Brain Functions and Neuroscience Unit, Department of Oral Science, Graduate School of Dentistry, Kanagawa Dental University, Yokosuka, Kanagawa, Japan
| | - Hirohumi Suzuki
- Department of Biology, University of Teacher Education Fukuoka, Munakata, Fukuoka, Japan
| |
Collapse
|
31
|
Wulff BS, Kuhre RE, Selvaraj M, Rehfeld JF, Niss K, Fels JJ, Anna S, Raun K, Gerstenberg MK. Improved leptin sensitivity and increased soluble leptin receptor concentrations may underlie the additive effects of combining PYY [, , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , ] and exendin-4 on body weight lowering in diet-induced obese mice. Heliyon 2024; 10:e32009. [PMID: 39183855 PMCID: PMC11341243 DOI: 10.1016/j.heliyon.2024.e32009] [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: 09/18/2023] [Revised: 05/27/2024] [Accepted: 05/27/2024] [Indexed: 08/27/2024] Open
Abstract
Objective Co-treatment with long acting PYY and the GLP-1 receptor agonists has potential as an efficient obesity treatment. This study investigates whether the mechanisms behind additive reduction of food intake and weight loss depends on complementary effects in brain areas regulating food intake and if restoration of leptin sensitivity is involved. Methods Diet-induced obese (DIO) mice were co-treated with PYY(3-36) and exendin-4 (Ex4, GLP-1R agonist) for 14 days using minipumps. Leptin responsiveness was evaluated by measuring food intake and body weight after leptin injection, and gene expression profile was investigated in various of brain regions and liver. Results We show that weight loss associated with co-treatment of PYY(3-36) and Ex4 and Ex4 mono-treatment in DIO mice increased expression of several genes in area postrema (AP) known to be involved in appetite regulation and Cart, Pdyn, Bdnf and Klb were synergistically upregulated by the co-treatment. The upregulations were independent of weight loss, as shown by inclusion of a weight matched control. Moreover, PYY(3-36) and Ex4 co-treatment resulted in synergistically upregulated plasma concentrations of soluble leptin receptor (SLR) and improved sensitivity to exogenous leptin demonstrated by food intake lowering. Conclusion The study results suggest that synergistic upregulation of appetite-regulating genes in AP and improved leptin sensitivity are important mediators for the additive weight loss resulting from PYY and Ex4 co-treatment.
Collapse
Affiliation(s)
| | | | - Madhan Selvaraj
- Translational Research, Global Translation, Novo Nordisk A/S, 2760 Måløv, Denmark
| | - Jens F. Rehfeld
- Department of Clinical Biochemistry, Rigshospitalet, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Kristoffer Niss
- Biomarker Discovery, R&ED Digital Science and Innovation, Novo Nordisk A/S, 2760 Måløv, Denmark
| | - Johannes J. Fels
- Research Bioanalysis, Global Research Technologies, Novo Nordisk A/S, 2760 Måløv, Denmark
| | - Secher Anna
- Global Drug Discovery, Novo Nordisk A/S, 2760, Måløv, Denmark
| | - Kirsten Raun
- Global Drug Discovery, Novo Nordisk A/S, 2760, Måløv, Denmark
| | | |
Collapse
|
32
|
Podgórski R, Galiniak S, Mazur A, Domin A, Podgórska D. Serum levels of leptin, ghrelin putative peptide YY-3 in patients with fetal alcohol spectrum disorders. Sci Rep 2024; 14:14971. [PMID: 38951515 PMCID: PMC11217397 DOI: 10.1038/s41598-024-66052-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 06/26/2024] [Indexed: 07/03/2024] Open
Abstract
Fetal alcohol spectrum disorders (FASD) are a severe developmental condition resulting from exposure to alcohol during pregnancy. The aim of this study was to examine the concentrations of hormones involved in appetite regulation-ghrelin, leptin, and putative peptide YY-3 (PYY)-in the serum of individuals with FASD. Additionally, we investigated the relationship between these hormone levels and clinical indicators. We conducted an enzyme-linked immunosorbent assay on samples collected from 62 FASD patients and 23 individuals without the condition. Our results revealed a significant decrease in leptin levels among FASD patients compared to the control group (5.124 vs. 6.838 ng/mL, p = 0.002). We revealed no statistically significant differences in the levels of other hormones studied (ghrelin and PYY). Comparisons of hormone levels were also conducted in three subgroups: FAS, neurobehavioral disorders associated with prenatal alcohol exposure and FASD risk, as well as by sex. Assignment to FASD subgroups indicated changes only for leptin. Sex had no effect on the levels of hormones. Moreover, the levels of leptin showed a negative correlation with cortisol levels and a positive correlation with BMI and proopiomelanocortin. Alterations in appetite regulation can contribute to the improper development of children with FASD, which might be another factor that should be taken into consideration in the proper treatment of patients.
Collapse
Affiliation(s)
- Rafał Podgórski
- Department of Biochemistry, Institute of Medical Sciences, Medical College of Rzeszow University, Warzywna 1a, 35-310, Rzeszow, Poland.
| | - Sabina Galiniak
- Department of Biochemistry, Institute of Medical Sciences, Medical College of Rzeszow University, Warzywna 1a, 35-310, Rzeszow, Poland
| | - Artur Mazur
- Department of Pediatric, Institute of Medical Sciences, Medical College of Rzeszow University, 35-310, Rzeszow, Poland
| | - Agnieszka Domin
- Department of Pediatric, Institute of Medical Sciences, Medical College of Rzeszow University, 35-310, Rzeszow, Poland
| | - Dominika Podgórska
- Department of Rheumatology, Institute of Medical Sciences, Medical College of Rzeszow University, 35-310, Rzeszow, Poland
| |
Collapse
|
33
|
Gala K, Ghusn W, Abu Dayyeh BK. Gut motility and hormone changes after bariatric procedures. Curr Opin Endocrinol Diabetes Obes 2024; 31:131-137. [PMID: 38533785 DOI: 10.1097/med.0000000000000860] [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] [Indexed: 03/28/2024]
Abstract
PURPOSE OF REVIEW Metabolic and bariatric surgery (MBS) and endoscopic bariatric therapies (EBT) are being increasingly utilized for the management of obesity. They work through multiple mechanisms, including restriction, malabsorption, and changes in the gastrointestinal hormonal and motility. RECENT FINDINGS Roux-en-Y gastric bypass (RYGB) and laparoscopic sleeve gastrectomy (LSG) cause decrease in leptin, increase in GLP-1 and PYY, and variable changes in ghrelin (generally thought to decrease). RYGB and LSG lead to rapid gastric emptying, increase in small bowel motility, and possible decrease in colonic motility. Endoscopic sleeve gastroplasty (ESG) causes decrease in leptin and increase in GLP-1, ghrelin, and PYY; and delayed gastric motility. SUMMARY Understanding mechanisms of action for MBS and EBT is critical for optimal care of patients and will help in further refinement of these interventions.
Collapse
Affiliation(s)
- Khushboo Gala
- Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Wissam Ghusn
- Department of Internal Medicine, Boston University Medical Center, Boston, Massachusetts, USA
| | - Barham K Abu Dayyeh
- Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
34
|
Hong SH, Choi KM. Gut hormones and appetite regulation. Curr Opin Endocrinol Diabetes Obes 2024; 31:115-121. [PMID: 38511400 DOI: 10.1097/med.0000000000000859] [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] [Indexed: 03/22/2024]
Abstract
PURPOSE OF REVIEW Various gut hormones interact with the brain through delicate communication, thereby influencing appetite and subsequent changes in body weight. This review summarizes the effects of gut hormones on appetite, with a focus on recent research. RECENT FINDINGS Ghrelin is known as an orexigenic hormone, whereas glucagon-like peptide-1 (GLP-1), glucose-dependent insulinotropic polypeptide (GIP), cholecystokinin (CCK), postprandial peptide YY (PYY), and oxyntomodulin (OXM) are known as anorexigenic hormones. Recent human studies have revealed that gut hormones act differently in various systems, including adipose tissue, beyond appetite and energy intake, and even involve in high-order thinking. Environmental factors including meal schedule, food contents and quality, type of exercise, and sleep deprivation also play a role in the influence of gut hormone on appetite, weight change, and obesity. Recently published studies have shown that retatrutide, a triple-agonist of GLP-1, GIP, and glucagon receptor, and orforglipron, a GLP-1 receptor partial agonist, are effective in weight loss and improving various metabolic parameters associated with obesity. SUMMARY Various gut hormones influence appetite, and several drugs targeting these receptors have been reported to exert positive effects on weight loss in humans. Given that diverse dietary and environmental factors affect the actions of gut hormones and appetite, there is a need for integrated and largescale long-term studies in this field.
Collapse
Affiliation(s)
- So-Hyeon Hong
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Ewha Womans University College of Medicine, Seoul, South Korea
| | - Kyung Mook Choi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, South Korea
| |
Collapse
|
35
|
Dumiaty Y, Underwood BM, Phy-Lim J, Chee MJ. Neurocircuitry underlying the actions of glucagon-like peptide 1 and peptide YY 3-36 in the suppression of food, drug-seeking, and anxiogenesis. Neuropeptides 2024; 105:102427. [PMID: 38579490 DOI: 10.1016/j.npep.2024.102427] [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: 01/23/2024] [Revised: 03/27/2024] [Accepted: 03/29/2024] [Indexed: 04/07/2024]
Abstract
Obesity is a critical health condition worldwide that increases the risks of comorbid chronic diseases, but it can be managed with weight loss. However, conventional interventions relying on diet and exercise are inadequate for achieving and maintaining weight loss, thus there is significant market interest for pharmaceutical anti-obesity agents. For decades, receptor agonists for the gut peptide glucagon-like peptide 1 (GLP-1) featured prominently in anti-obesity medications by suppressing appetite and food reward to elicit rapid weight loss. As the neurocircuitry underlying food motivation overlaps with that for drugs of abuse, GLP-1 receptor agonism has also been shown to decrease substance use and relapse, thus its therapeutic potential may extend beyond weight management to treat addictions. However, as prolonged use of anti-obesity drugs may increase the risk of mood-related disorders like anxiety and depression, and individuals taking GLP-1-based medication commonly report feeling demotivated, the long-term safety of such drugs is an ongoing concern. Interestingly, current research now focuses on dual agonist approaches that include GLP-1 receptor agonism to enable synergistic effects on weight loss or associated functions. GLP-1 is secreted from the same intestinal cells as the anorectic gut peptide, Peptide YY3-36 (PYY3-36), thus this review assessed the therapeutic potential and underlying neural circuits targeted by PYY3-36 when administered independently or in combination with GLP-1 to curb the appetite for food or drugs of abuse like opiates, alcohol, and nicotine. Additionally, we also reviewed animal and human studies to assess the impact, if any, for GLP-1 and/or PYY3-36 on mood-related behaviors in relation to anxiety and depression. As dual agonists targeting GLP-1 and PYY3-36 may produce synergistic effects, they can be effective at lower doses and offer an alternative approach for therapeutic benefits while mitigating undesirable side effects.
Collapse
Affiliation(s)
- Yasmina Dumiaty
- Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, ON K1S 5B6, Canada.
| | - Brett M Underwood
- Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, ON K1S 5B6, Canada.
| | - Jenny Phy-Lim
- Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, ON K1S 5B6, Canada.
| | - Melissa J Chee
- Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, ON K1S 5B6, Canada.
| |
Collapse
|
36
|
Watkins JD, Smith HA, Hengist A, Nielsen SB, Mikkelsen UR, Saunders J, Koumanov F, Betts JA, Gonzalez JT. Effects of physical form of β-lactoglobulin and calcium ingestion on GLP-1 secretion, gastric emptying and energy intake in humans: a randomised crossover trial. Br J Nutr 2024; 131:1730-1739. [PMID: 38287700 PMCID: PMC11063665 DOI: 10.1017/s0007114524000321] [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: 07/07/2023] [Revised: 01/03/2024] [Accepted: 01/25/2024] [Indexed: 01/31/2024]
Abstract
The aim of this study was to assess whether adding Ca2+ to aggregate or native forms of β-lactoglobulin alters gut hormone secretion, gastric emptying rates and energy intake in healthy men and women. Fifteen healthy adults (mean ± sd: 9M/6F, age: 24 ± 5 years) completed four trials in a randomised, double-blind, crossover design. Participants consumed test drinks consisting of 30 g of β-lactoglobulin in a native form with (NATIVE + MINERALS) and without (NATIVE) a Ca2+-rich mineral supplement and in an aggregated form both with (AGGREG + MINERALS) and without the mineral supplement (AGGREG). Arterialised blood was sampled for 120 min postprandially to determine gut hormone concentrations. Gastric emptying was determined using 13C-acetate and 13C-octanoate, and energy intake was assessed with an ad libitum meal at 120 min. A protein × mineral interaction effect was observed for total glucagon-like peptide-1 (GLP-1TOTAL) incremental AUC (iAUC; P < 0·01), whereby MINERALS + AGGREG increased GLP-1TOTAL iAUC to a greater extent than AGGREG (1882 ± 603 v. 1550 ± 456 pmol·l-1·120 min, P < 0·01), but MINERALS + NATIVE did not meaningfully alter the GLP-1 iAUC compared with NATIVE (1669 ± 547 v. 1844 ± 550 pmol·l-1·120 min, P = 0·09). A protein × minerals interaction effect was also observed for gastric emptying half-life (P < 0·01) whereby MINERALS + NATIVE increased gastric emptying half-life compared with NATIVE (83 ± 14 v. 71 ± 8 min, P < 0·01), whereas no meaningful differences were observed between MINERALS + AGGREG v. AGGREG (P = 0·70). These did not result in any meaningful changes in energy intake (protein × minerals interaction, P = 0·06). These data suggest that the potential for Ca2+ to stimulate GLP-1 secretion at moderate protein doses may depend on protein form. This study was registered at clinicaltrials.gov (NCT04659902).
Collapse
Affiliation(s)
- Jonathan D. Watkins
- Centre for Nutrition, Exercise and Metabolism, Department for Health, University of Bath, Bath, UK
| | - Harry A. Smith
- Centre for Nutrition, Exercise and Metabolism, Department for Health, University of Bath, Bath, UK
| | - Aaron Hengist
- Centre for Nutrition, Exercise and Metabolism, Department for Health, University of Bath, Bath, UK
| | | | | | | | - Francoise Koumanov
- Centre for Nutrition, Exercise and Metabolism, Department for Health, University of Bath, Bath, UK
| | - James A. Betts
- Centre for Nutrition, Exercise and Metabolism, Department for Health, University of Bath, Bath, UK
| | - Javier T. Gonzalez
- Centre for Nutrition, Exercise and Metabolism, Department for Health, University of Bath, Bath, UK
| |
Collapse
|
37
|
Gan HW, Cerbone M, Dattani MT. Appetite- and Weight-Regulating Neuroendocrine Circuitry in Hypothalamic Obesity. Endocr Rev 2024; 45:309-342. [PMID: 38019584 PMCID: PMC11074800 DOI: 10.1210/endrev/bnad033] [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: 02/04/2023] [Revised: 10/25/2023] [Accepted: 11/27/2023] [Indexed: 11/30/2023]
Abstract
Since hypothalamic obesity (HyOb) was first described over 120 years ago by Joseph Babinski and Alfred Fröhlich, advances in molecular genetic laboratory techniques have allowed us to elucidate various components of the intricate neurocircuitry governing appetite and weight regulation connecting the hypothalamus, pituitary gland, brainstem, adipose tissue, pancreas, and gastrointestinal tract. On a background of an increasing prevalence of population-level common obesity, the number of survivors of congenital (eg, septo-optic dysplasia, Prader-Willi syndrome) and acquired (eg, central nervous system tumors) hypothalamic disorders is increasing, thanks to earlier diagnosis and management as well as better oncological therapies. Although to date the discovery of several appetite-regulating peptides has led to the development of a range of targeted molecular therapies for monogenic obesity syndromes, outside of these disorders these discoveries have not translated into the development of efficacious treatments for other forms of HyOb. This review aims to summarize our current understanding of the neuroendocrine physiology of appetite and weight regulation, and explore our current understanding of the pathophysiology of HyOb.
Collapse
Affiliation(s)
- Hoong-Wei Gan
- Department of Endocrinology, Great Ormond Street Hospital for Children NHS Foundation Trust, Great Ormond Street, London WC1N 3JH, UK
- Genetics & Genomic Medicine Research & Teaching Department, University College London Great Ormond Street Institute for Child Health, 30 Guilford Street, London WC1N 1EH, UK
| | - Manuela Cerbone
- Department of Endocrinology, Great Ormond Street Hospital for Children NHS Foundation Trust, Great Ormond Street, London WC1N 3JH, UK
- Genetics & Genomic Medicine Research & Teaching Department, University College London Great Ormond Street Institute for Child Health, 30 Guilford Street, London WC1N 1EH, UK
| | - Mehul Tulsidas Dattani
- Department of Endocrinology, Great Ormond Street Hospital for Children NHS Foundation Trust, Great Ormond Street, London WC1N 3JH, UK
- Genetics & Genomic Medicine Research & Teaching Department, University College London Great Ormond Street Institute for Child Health, 30 Guilford Street, London WC1N 1EH, UK
| |
Collapse
|
38
|
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
|
39
|
Gao J, Zhang S, Deng P, Wu Z, Lemaitre B, Zhai Z, Guo Z. Dietary L-Glu sensing by enteroendocrine cells adjusts food intake via modulating gut PYY/NPF secretion. Nat Commun 2024; 15:3514. [PMID: 38664401 PMCID: PMC11045819 DOI: 10.1038/s41467-024-47465-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 03/28/2024] [Indexed: 04/28/2024] Open
Abstract
Amino acid availability is monitored by animals to adapt to their nutritional environment. Beyond gustatory receptors and systemic amino acid sensors, enteroendocrine cells (EECs) are believed to directly percept dietary amino acids and secrete regulatory peptides. However, the cellular machinery underlying amino acid-sensing by EECs and how EEC-derived hormones modulate feeding behavior remain elusive. Here, by developing tools to specifically manipulate EECs, we find that Drosophila neuropeptide F (NPF) from mated female EECs inhibits feeding, similar to human PYY. Mechanistically, dietary L-Glutamate acts through the metabotropic glutamate receptor mGluR to decelerate calcium oscillations in EECs, thereby causing reduced NPF secretion via dense-core vesicles. Furthermore, two dopaminergic enteric neurons expressing NPFR perceive EEC-derived NPF and relay an anorexigenic signal to the brain. Thus, our findings provide mechanistic insights into how EECs assess food quality and identify a conserved mode of action that explains how gut NPF/PYY modulates food intake.
Collapse
Affiliation(s)
- Junjun Gao
- Department of Medical Genetics, School of Basic Medicine, Institute for Brain Research, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Song Zhang
- Department of Medical Genetics, School of Basic Medicine, Institute for Brain Research, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pan Deng
- State Key Laboratory of Digital Manufacturing Equipment and Technology, Huazhong University of Science and Technology, Wuhan, PR China
- Department of Mechanical Engineering, University of British Columbia, Vancouver, British Columbia, Canada
| | - Zhigang Wu
- State Key Laboratory of Digital Manufacturing Equipment and Technology, Huazhong University of Science and Technology, Wuhan, PR China
| | - Bruno Lemaitre
- Global Health Institute, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Zongzhao Zhai
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, Hunan, PR China.
| | - Zheng Guo
- Department of Medical Genetics, School of Basic Medicine, Institute for Brain Research, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Cell Architecture Research Center, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
40
|
Konitz C, Schwensfeier L, Predel HG, Brinkmann C. The Influence of Acute and Chronic Exercise on Appetite and Appetite Regulation in Patients with Prediabetes or Type 2 Diabetes Mellitus-A Systematic Review. Nutrients 2024; 16:1126. [PMID: 38674817 PMCID: PMC11054589 DOI: 10.3390/nu16081126] [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: 03/13/2024] [Revised: 04/01/2024] [Accepted: 04/06/2024] [Indexed: 04/28/2024] Open
Abstract
This systematic review aims to analyze the effects of acute and chronic exercise on appetite and appetite regulation in patients with abnormal glycemic control. PubMed, Web of Science, and the Cochrane Central Register of Controlled Trials were searched for eligible studies. The included studies had to report assessments of appetite (primary outcome). Levels of appetite-regulating hormones were analyzed as secondary outcomes (considered, if additionally reported). Seven studies with a total number of 211 patients with prediabetes or type 2 diabetes mellitus (T2DM) met the inclusion criteria. Ratings of hunger, satiety, fullness, prospective food consumption, nausea, and desire to eat, as well as levels of (des-)acylated ghrelin, glucagon-like peptide 1, glucose-dependent insulinotropic peptide, pancreatic polypeptide, peptide tyrosine tyrosine, leptin, and spexin were considered. Following acute exercise, the effects on appetite (measured up to one day post-exercise) varied, while there were either no changes or a decrease in appetite ratings following chronic exercise, both compared to control conditions (without exercise). These results were accompanied by inconsistent changes in appetite-regulating hormone levels. The overall risk of bias was low. The present results provide more evidence for an appetite-reducing rather than an appetite-increasing effect of (chronic) exercise on patients with prediabetes or T2DM. PROSPERO ID: CRD42023459322.
Collapse
Affiliation(s)
- Christoph Konitz
- Institute of Cardiovascular Research and Sport Medicine, German Sport University Cologne, 50933 Cologne, Germany; (L.S.); (H.-G.P.)
| | - Leon Schwensfeier
- Institute of Cardiovascular Research and Sport Medicine, German Sport University Cologne, 50933 Cologne, Germany; (L.S.); (H.-G.P.)
| | - Hans-Georg Predel
- Institute of Cardiovascular Research and Sport Medicine, German Sport University Cologne, 50933 Cologne, Germany; (L.S.); (H.-G.P.)
| | - Christian Brinkmann
- Institute of Cardiovascular Research and Sport Medicine, German Sport University Cologne, 50933 Cologne, Germany; (L.S.); (H.-G.P.)
- Department of Fitness and Health, IST University of Applied Sciences, 40233 Düsseldorf, Germany
| |
Collapse
|
41
|
Hamamah S, Hajnal A, Covasa M. Influence of Bariatric Surgery on Gut Microbiota Composition and Its Implication on Brain and Peripheral Targets. Nutrients 2024; 16:1071. [PMID: 38613104 PMCID: PMC11013759 DOI: 10.3390/nu16071071] [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: 03/14/2024] [Revised: 03/29/2024] [Accepted: 04/02/2024] [Indexed: 04/14/2024] Open
Abstract
Obesity remains a significant global health challenge, with bariatric surgery remaining as one of the most effective treatments for severe obesity and its related comorbidities. This review highlights the multifaceted impact of bariatric surgery beyond mere physical restriction or nutrient malabsorption, underscoring the importance of the gut microbiome and neurohormonal signals in mediating the profound effects on weight loss and behavior modification. The various bariatric surgery procedures, such as Roux-en-Y gastric bypass (RYGB) and sleeve gastrectomy (SG), act through distinct mechanisms to alter the gut microbiome, subsequently impacting metabolic health, energy balance, and food reward behaviors. Emerging evidence has shown that bariatric surgery induces profound changes in the composition of the gut microbiome, notably altering the Firmicutes/Bacteroidetes ratio and enhancing populations of beneficial bacteria such as Akkermansia. These microbiota shifts have far-reaching effects beyond gut health, influencing dopamine-mediated reward pathways in the brain and modulating the secretion and action of key gut hormones including ghrelin, leptin, GLP-1, PYY, and CCK. The resultant changes in dopamine signaling and hormone levels contribute to reduced hedonic eating, enhanced satiety, and improved metabolic outcomes. Further, post-bariatric surgical effects on satiation targets are in part mediated by metabolic byproducts of gut microbiota like short-chain fatty acids (SCFAs) and bile acids, which play a pivotal role in modulating metabolism and energy expenditure and reducing obesity-associated inflammation, as well as influencing food reward pathways, potentially contributing to the regulation of body weight and reduction in hedonic eating behaviors. Overall, a better understanding of these mechanisms opens the door to developing non-surgical interventions that replicate the beneficial effects of bariatric surgery on the gut microbiome, dopamine signaling, and gut hormone regulation, offering new avenues for obesity treatment.
Collapse
Affiliation(s)
- Sevag Hamamah
- Department of Basic Medical Sciences, College of Osteopathic Medicine, Western University of Health Sciences, Pomona, CA 9176, USA;
| | - Andras Hajnal
- Department of Neural and Behavioral Sciences, College of Medicine, The Pennsylvania State University, Hershey, PA 17033, USA;
| | - Mihai Covasa
- Department of Basic Medical Sciences, College of Osteopathic Medicine, Western University of Health Sciences, Pomona, CA 9176, USA;
- Department of Biomedical Sciences, College of Medicine and Biological Science, University of Suceava, 7200229 Suceava, Romania
| |
Collapse
|
42
|
Zheng W, Duan H, Cao L, Mao S, Shen J. Acid-base properties of non-protein nitrogen affect nutrients intake, rumen fermentation and antioxidant capacity of fattening Hu sheep. Front Vet Sci 2024; 11:1381871. [PMID: 38596467 PMCID: PMC11002212 DOI: 10.3389/fvets.2024.1381871] [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: 02/04/2024] [Accepted: 03/14/2024] [Indexed: 04/11/2024] Open
Abstract
This study conducted a comparison of the effects of non-protein nitrogen with different acid-base properties on feed intake, rumen fermentation, nutrient digestion and antioxidant capacity in fattening Hu sheep. Sixteen fattening male sheep (31.43 ± 2.41 kg) with permanent rumen cannulas were randomly assigned to two dietary treatments: 1% urea and 1.78% ammonium chloride (NH4Cl, AC). A 42 days experimental period was conducted, with 14 days for adaptation and 28 days for treatment. Daily feed intake was recorded and various samples including feed, feces, rumen fluid, and blood were collected at different time points during the final week. The results indicated that the urea group had significantly higher dry matter intake, average daily gain, and gain efficiency in comparison to the AC group (p < 0.01). There was no difference in rumen pH and concentration of ammonia nitrogen between different groups (p > 0.05), but the rumen pH of urea group was higher than that of the AC group at 1 and 3 h after feeding (p < 0.05). The urea group exhibited higher concentrations of total volatile fatty acids (VFA) and individual VFAs compared to the AC group at all-time points (p < 0.01). Compared to the urea group, the intake of all nutrients decreased in the AC group (p < 0.01), but the digestibility of dry matter and organic matter increased significantly (p < 0.01), and the digestibility of CP had an increasing trend (p = 0.06) in the AC group. Additionally, the urea group had lower levels of serum glucagon-like peptide-1, peptide YY, Cl, total protein and globulin than the AC group (p < 0.05). The overall levels of HCO3-, superoxide dismutase, glutathione peroxidase, catalase, albumin/globulin, blood urea nitrogen and total cholesterol in the urea group increased significantly compared to the AC group (p < 0.05). It was concluded that adding urea to the high-concentrate diet resulted in increased rumen pH and improved rumen fermentation and growth performance in fattening sheep compared to NH4Cl addition. Furthermore, urea addition improved sheep's antioxidant capacity and maintained their acid-base balance more effectively as compared to NH4Cl.
Collapse
Affiliation(s)
- Wenjin Zheng
- Laboratory of Gastrointestinal Microbiology, National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, China
- Ruminant Nutrition and Feed Engineering Technology Research Center, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Hongwei Duan
- Laboratory of Gastrointestinal Microbiology, National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, China
- Ruminant Nutrition and Feed Engineering Technology Research Center, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Liwen Cao
- Laboratory of Gastrointestinal Microbiology, National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, China
- Ruminant Nutrition and Feed Engineering Technology Research Center, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Shengyong Mao
- Laboratory of Gastrointestinal Microbiology, National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, China
- Ruminant Nutrition and Feed Engineering Technology Research Center, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Junshi Shen
- Laboratory of Gastrointestinal Microbiology, National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, China
- Ruminant Nutrition and Feed Engineering Technology Research Center, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
43
|
Salvadori M, Rosso G. Update on the reciprocal interference between immunosuppressive therapy and gut microbiota after kidney transplantation. World J Transplant 2024; 14:90194. [PMID: 38576749 PMCID: PMC10989467 DOI: 10.5500/wjt.v14.i1.90194] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 12/22/2023] [Accepted: 12/29/2023] [Indexed: 03/15/2024] Open
Abstract
Gut microbiota is often modified after kidney transplantation. This principally happens in the first period after transplantation. Antibiotics and, most of all, immunosuppressive drugs are the main responsible. The relationship between immunosuppressive drugs and the gut microbiota is bilateral. From one side immunosuppressive drugs modify the gut microbiota, often generating dysbiosis; from the other side microbiota may interfere with the immunosuppressant pharmacokinetics, producing products more or less active with respect to the original drug. These phenomena have influence over the graft outcomes and clinical consequences as rejections, infections, diarrhea may be caused by the dysbiotic condition. Corticosteroids, calcineurin inhibitors such as tacrolimus and cyclosporine, mycophenolate mofetil and mTOR inhibitors are the immunosuppressive drugs whose effect on the gut microbiota is better known. In contrast is well known how the gut microbiota may interfere with glucocorticoids, which may be transformed into androgens. Tacrolimus may be transformed by micro biota into a product called M1 that is 15-fold less active with respect to tacrolimus. The pro-drug mycophenolate mofetil is normally transformed in mycophenolic acid that according the presence or not of microbes producing the enzyme glu curonidase, may be transformed into the inactive product.
Collapse
Affiliation(s)
- Maurizio Salvadori
- Department of Renal Transplantation, Careggi University Hospital, Florence 50139, Tuscany, Italy
| | - Giuseppina Rosso
- Division of Nephrology, San Giovanni di Dio Hospital, Florence 50143, Toscana, Italy
| |
Collapse
|
44
|
Alonso AM, Cork SC, Phuah P, Hansen B, Norton M, Cheng S, Xu X, Suba K, Ma Y, Dowsett GK, Tadross JA, Lam BY, Yeo GS, Herzog H, Bloom SR, Arnold M, Distaso W, Murphy KG, Salem V. The vagus nerve mediates the physiological but not pharmacological effects of PYY 3-36 on food intake. Mol Metab 2024; 81:101895. [PMID: 38340808 PMCID: PMC10877939 DOI: 10.1016/j.molmet.2024.101895] [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: 11/26/2023] [Revised: 01/25/2024] [Accepted: 02/02/2024] [Indexed: 02/12/2024] Open
Abstract
Peptide YY (PYY3-36) is a post-prandially released gut hormone with potent appetite-reducing activity, the mechanism of action of which is not fully understood. Unravelling how this system physiologically regulates food intake may help unlock its therapeutic potential, whilst minimising unwanted effects. Here we demonstrate that germline and post-natal targeted knockdown of the PYY3-36 preferring receptor (neuropeptide Y (NPY) Y2 receptor (Y2R)) in the afferent vagus nerve is required for the appetite inhibitory effects of physiologically-released PYY3-36, but not peripherally administered pharmacological doses. Post-natal knockdown of the Y2R results in a transient body weight phenotype that is not evident in the germline model. Loss of vagal Y2R signalling also results in altered meal patterning associated with accelerated gastric emptying. These results are important for the design of PYY-based anti-obesity agents.
Collapse
Affiliation(s)
- Aldara Martin Alonso
- Section of Investigative Medicine and Endocrinology, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Simon C Cork
- Section of Investigative Medicine and Endocrinology, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom; School of Medicine, Faculty of Health, Education, Medicine & Social Care, Anglia Ruskin University, Chelmsford, CM1 1SQ, United Kingdom
| | - Phyllis Phuah
- Section of Investigative Medicine and Endocrinology, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Benjamin Hansen
- Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Mariana Norton
- Section of Investigative Medicine and Endocrinology, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Sijing Cheng
- Section of Investigative Medicine and Endocrinology, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Xiang Xu
- Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Kinga Suba
- Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Yue Ma
- Section of Investigative Medicine and Endocrinology, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Georgina Kc Dowsett
- Medical Research Council Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, United Kingdom
| | - John A Tadross
- Medical Research Council Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, United Kingdom
| | - Brian Yh Lam
- Medical Research Council Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, United Kingdom
| | - Giles Sh Yeo
- Medical Research Council Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, United Kingdom
| | - Herbert Herzog
- Neuroscience Division, Garvan Institute of Medical Research, Darlinghurst, Australia
| | - Stephen R Bloom
- Section of Investigative Medicine and Endocrinology, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Myrtha Arnold
- Department of Health Sciences and Technology, ETH Zurich, Schwerzenbach, Switzerland
| | - Walter Distaso
- Imperial College Business School, Imperial College London, United Kingdom
| | - Kevin G Murphy
- Section of Investigative Medicine and Endocrinology, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Victoria Salem
- Section of Investigative Medicine and Endocrinology, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom; Department of Bioengineering, Imperial College London, London, United Kingdom.
| |
Collapse
|
45
|
Shankar K, Ramborger J, Bonnet-Zahedi S, Carrette LLG, George O. Acute nicotine intake increases feeding behavior through decreasing glucagon signaling in dependent male and female rats. Horm Behav 2024; 159:105447. [PMID: 37926623 PMCID: PMC11384237 DOI: 10.1016/j.yhbeh.2023.105447] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 09/26/2023] [Accepted: 10/20/2023] [Indexed: 11/07/2023]
Abstract
Chronic use of nicotine is known to dysregulate metabolic signaling through altering circulating levels of feeding-related hormones, contributing to the onset of disorders like type 2 diabetes. However, little is known about the acute effects of nicotine on hormonal signaling. We previously identified an acute increase in food intake following acute nicotine, and we sought to determine whether this behavior was due to a change in hormone levels. We first identified that acute nicotine injection produces an increase in feeding behavior in dependent rats, but not nondependent rats. We confirmed that chronic nicotine use increases circulating levels of insulin, leptin, and ghrelin, and these correlate with rats' body weight and food intake. Acute nicotine injection in dependent animals decreased circulating GLP-1 and glucagon levels, and administration of glucagon prior to acute nicotine injection prevented the acute increase in feeding behavior. Thus, acute nicotine injection increases feeding behavior in dependent rats by decreasing glucagon signaling.
Collapse
Affiliation(s)
- Kokila Shankar
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093, USA; The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Jarryd Ramborger
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093, USA
| | - Sélène Bonnet-Zahedi
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093, USA; Institut de Neurosciences de la Timone, Aix-Marseille Université, Marseille 13005, France
| | - Lieselot L G Carrette
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093, USA
| | - Olivier George
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
46
|
Prajapati A, Rana D, Rangra S, Jindal AB, Benival D. Current Status of Therapeutic Peptides for the Management of Diabetes Mellitus. Int J Pept Res Ther 2024; 30:13. [DOI: 10.1007/s10989-024-10590-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/14/2024] [Indexed: 01/04/2025]
|
47
|
Browne N, Horgan K. The Impact of a Proprietary Blend of Yeast Cell Wall, Short-Chain Fatty Acids, and Zinc Proteinate on Growth, Nutrient Utilisation, and Endocrine Hormone Secretion in Intestinal Cell Models. Animals (Basel) 2024; 14:238. [PMID: 38254407 PMCID: PMC10812779 DOI: 10.3390/ani14020238] [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: 11/06/2023] [Revised: 01/08/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
In piglets, it is observed that early weaning can lead to poor weight gain due to an underdeveloped gastrointestinal (GI) tract, which is unsuitable for an efficient absorption of nutrients. Short-chain fatty acids (SCFAs) such as butyrate have demonstrated their ability to improve intestinal development by increasing cell proliferation, which is vital during this transition period when the small and large intestinal tracts are rapidly growing. Previous reports on butyrate inclusion in feed demonstrated significantly increased feed intakes (FIs) and average daily gains (ADGs) during piglet weaning. Similar benefits in piglet performance have been observed with the inclusion of yeast cell wall in diets. A proprietary mix of yeast cell wall, SCFAs, and zinc proteinate (YSM) was assessed here in vitro to determine its impact on cellular growth, metabolism and appetite-associated hormones in ex vivo small intestinal pig cells and STC-1 mouse intestinal neuroendocrine cells. Intestinal cells demonstrated greater cell densities with the addition of YSM (150 ppm) compared to the control and butyrate (150 ppm) at 24 h. This coincided with the higher utilisation of both protein and glucose from the media of intestinal cells receiving YSM. Ghrelin (an appetite-inducing hormone) demonstrated elevated levels in the YSM-treated cells on a protein and gene expression level compared to the cells receiving butyrate and the control, while satiety hormone peptide YY protein levels were lower in the cells receiving YSM compared to the control and butyrate-treated cells across each time point. Higher levels of ghrelin and lower PYY secretion in cells receiving YSM may drive the uptake of protein and glucose, which is potentially facilitated by elevated gene transporters for protein and glucose. Greater ghrelin levels observed with the inclusion of YSM may contribute to higher cell densities that could support pig performance to a greater extent than butyrate alone.
Collapse
Affiliation(s)
- Niall Browne
- Alltech Biotechnology Centre, Sarney, Summerhill Road, Dunboyne, A86 X006 Co. Meath, Ireland
| | - Karina Horgan
- Alltech Biotechnology Centre, Sarney, Summerhill Road, Dunboyne, A86 X006 Co. Meath, Ireland
| |
Collapse
|
48
|
Li Y, Tataka Y, Sakazaki M, Kamemoto K, Nagayama C, Yoshikawa Y, Yamada Y, Miyashita M. Acute effects of exercise intensity on butyrylcholinesterase and ghrelin in young men: A randomized controlled study. J Exerc Sci Fit 2024; 22:39-50. [PMID: 38033619 PMCID: PMC10687701 DOI: 10.1016/j.jesf.2023.11.001] [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: 08/06/2023] [Revised: 11/16/2023] [Accepted: 11/17/2023] [Indexed: 12/02/2023] Open
Abstract
Background/objectives Butyrylcholinesterase (BChE), a liver-derived enzyme that hydrolyzes acylated ghrelin to des-acylated ghrelin, may trigger a potential mechanism responsible for the acute exercise-induced suppression of acylated ghrelin. However, studies examining the effects of an acute bout of high-intensity exercise on BChE and acylated ghrelin have yielded inconsistent findings. This study aimed to examine the acute effects of exercise intensity on BChE, acylated ghrelin and des-acylated ghrelin concentrations in humans. Methods Fifteen young men (aged 22.7 ± 1.8 years, mean ± standard deviation) completed three, half-day laboratory-based trials (i.e., high-intensity exercise, low-intensity exercise and control), in a random order. In the exercise trials, the participants ran for 60 min (from 09:30 to 10:30) at a speed eliciting 70 % (high-intensity) or 40 % (low-intensity) of their maximum oxygen uptake and then rested for 90 min. In the control trial, participants sat on a chair for the entire trial (from 09:30 to 12:00). Venous blood samples were collected at 09:30, 10:00, 10:30, 11:00, 11:30 and 12:00. Results The BChE concentration was not altered over time among the three trials. Total acylated and des-acylated ghrelin area under the curve during the first 60 min (i.e., from 0 min to 60 min) of the main trial were lower in the high-intensity exercise trial than in the control (acylated ghrelin, mean difference: 62.6 pg/mL, p < 0.001; des-acylated ghrelin, mean difference: 31.4 pg/mL, p = 0.035) and the low-intensity exercise trial (acylated ghrelin, mean difference: 87.7 pg/mL, p < 0.001; des-acylated ghrelin, mean difference: 43.0 pg/mL, p = 0.042). Conclusion The findings suggest that BChE may not be involved in the modulation of ghrelin even though lowered acylated ghrelin concentration was observed after high-intensity exercise.
Collapse
Affiliation(s)
- Yibin Li
- Graduate School of Sport Sciences, Waseda University, Saitama, Japan
| | - Yusei Tataka
- Graduate School of Sport Sciences, Waseda University, Saitama, Japan
| | - Miki Sakazaki
- Graduate School of Sport Sciences, Waseda University, Saitama, Japan
| | - Kayoko Kamemoto
- Waseda Institute for Sport Sciences, Waseda University, Saitama, Japan
| | - Chihiro Nagayama
- Graduate School of Sport Sciences, Waseda University, Saitama, Japan
| | - Yoshie Yoshikawa
- Graduate School of Sport Sciences, Waseda University, Saitama, Japan
| | - Yoshiki Yamada
- Graduate School of Sport Sciences, Waseda University, Saitama, Japan
| | - Masashi Miyashita
- Faculty of Sport Sciences, Waseda University, Saitama, Japan
- School of Sport, Exercise and Health Sciences, Loughborough University, Leicestershire, United Kingdom
- Department of Sports Science and Physical Education, The Chinese University of Hong Kong, Shatin, Hong Kong
| |
Collapse
|
49
|
Kim TY, Schafer AL. Bariatric surgery, vitamin D, and bone loss. FELDMAN AND PIKE'S VITAMIN D 2024:161-184. [DOI: 10.1016/b978-0-323-91338-6.00009-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
50
|
Yu M, Yu B, Chen D. The effects of gut microbiota on appetite regulation and the underlying mechanisms. Gut Microbes 2024; 16:2414796. [PMID: 39501848 PMCID: PMC11542600 DOI: 10.1080/19490976.2024.2414796] [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: 08/07/2024] [Revised: 09/23/2024] [Accepted: 09/30/2024] [Indexed: 11/09/2024] Open
Abstract
Appetite, a crucial aspect regulated by both the central nervous system and peripheral hormones, is influenced by the composition and dynamics of the intestinal microbiota, as evidenced by recent research. This review highlights the role of intestinal microbiota in appetite regulation, elucidating the involvement of various pathways. Notably, the metabolites generated by intestinal microorganisms, including short-chain fatty acids, bile acids, and amino acid derivatives, play a pivotal role in this intricate process. Furthermore, intestinal microorganisms contribute to appetite regulation by modulating nutritional perception, neural signal transmission, and hormone secretion within the digestive system. Consequently, manipulating and modulating the intestinal microbiota represent innovative strategies for ameliorating appetite-related disorders. This paper provides a comprehensive review of the effects of gut microbes and their metabolites on the central nervous system and host appetite. By exploring their potential regulatory pathways and mechanisms, this study aims to enhance our understanding of how gut microbes influence appetite regulation in the host.
Collapse
Affiliation(s)
- Miao Yu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan Province, China
- DadHank(Chengdu)Biotech Corp, Chengdu, Sichuan Province, China
| | - Bing Yu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan Province, China
| | - Daiwen Chen
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan Province, China
| |
Collapse
|