1
|
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
|
2
|
Beetz N, Kalsch B, Forst T, Schmid B, Schultz A, Hennige AM. A randomized phase I study of BI 1820237, a novel neuropeptide Y receptor type 2 agonist, alone or in combination with low-dose liraglutide in otherwise healthy men with overweight or obesity. Diabetes Obes Metab 2024. [PMID: 39373311 DOI: 10.1111/dom.15984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 09/06/2024] [Accepted: 09/13/2024] [Indexed: 10/08/2024]
Abstract
AIMS Pharmacotherapeutic options for obesity treatment include glucagon-like peptide-1 receptor (GLP-1R) agonists, for example, liraglutide. However, an unmet need remains, particularly in patients with a high body mass index (BMI), as GLP-1R agonists are associated with gastrointestinal adverse events (AEs) and some patients do not respond to treatment. Neuropeptide Y (NPY) and peptide YY bind G-protein-coupled Y receptors and represent attractive targets for modulating bodyweight. MATERIALS AND METHODS This first-in-human, three-part, partially blinded phase I study (NCT04903509) investigated the safety, tolerability, pharmacokinetics (PK) and pharmacodynamics (PD) of single ascending doses of the peptidic NPY2R agonist BI 1820237, with/without low-dose liraglutide: part 1 (participants randomized to receive BI 1820237: 0.075-2.4 mg or placebo), part 2 (BI 1820237: 1.2 mg or placebo) and part 3 (BI 1820237: 0.025-1.2 mg + liraglutide 0.6 mg or placebo + liraglutide 0.6 mg). Primary endpoint is the proportion of participants with drug-related AEs. Secondary endpoints are tolerability, PK and PD. RESULTS In total, 95 otherwise healthy men with increased BMI (25.0-34.9 kg/m2) were randomized. Drug-related AEs, mainly gastrointestinal events, were reported by 39.0% of participants (n = 23) in parts 1 + 2 and 30.6% of participants (n = 11) in part 3; one drug-related AE (11.1%, part 3) was reported in a participant receiving placebo with liraglutide. Post-dose paracetamol PK suggested that BI 1820237 and low-dose liraglutide exhibited additive effects on gastric emptying. CONCLUSIONS BI 1820237 treatment was associated with transient nausea and vomiting at higher doses. No differences in tolerability were observed when combined with liraglutide; effects on gastric emptying appeared additive.
Collapse
Affiliation(s)
- Nadine Beetz
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß, Germany
| | - Brigitte Kalsch
- CRS Clinical Research Services Mannheim GmbH, Mannheim, Germany
| | - Thomas Forst
- CRS Clinical Research Services Mannheim GmbH, Mannheim, Germany
| | - Bernhard Schmid
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß, Germany
| | - Armin Schultz
- CRS Clinical Research Services Mannheim GmbH, Mannheim, Germany
| | - Anita M Hennige
- Boehringer Ingelheim International GmbH, Biberach an der Riß, Germany
| |
Collapse
|
3
|
Warner J, Stocker R, Brandt K, Crabtree DR, Ormond L, Stevenson E, Holliday A. Appetite, food intake, and gut hormone responses to glycomacropeptide protein ingestion in older adults: A feasibility, acceptability, and pilot study. Appetite 2024; 200:107509. [PMID: 38795943 DOI: 10.1016/j.appet.2024.107509] [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: 02/07/2024] [Revised: 05/20/2024] [Accepted: 05/21/2024] [Indexed: 05/28/2024]
Abstract
Glycomacropeptide (GMP) has a unique amino acid profile which may make less satiating than other dietary proteins. This study assessed the feasibility and likely acceptability of a leucine-enriched GMP drink and determined appetite response in older adults (OA). Thirteen OA (11f; 70 ± 4 years) were recruited for sensory assessments of a leucine-enriched GMP drink when mixed with water and with fruit smoothie, compared with whey protein isolate (WHEY). Participants also partook in a single focus group exploring acceptability to protein and supplementation. Separately, a counterbalanced, double-blind study with twelve OA (8f; 69 ± 3 years) was conducted to determine appetite and gut hormone responses. Fasting subjective appetite was recorded using visual analogue scales and a fasted venous blood sample was collected (to measures acyl-ghrelin, PYY, GLP-1, and CCK) before participants consumed either: GMP protein (27g + 3g leucine, 350 mL water), WHEY (30g, 350 mL water), or water. Participants rested for 240 min, with appetite measures and blood sampling throughout. An ad libitum pasta-based meal was then consumed. Sensory testing revealed low pleasantness rating for GMP in water vs. WHEY (16 ± 14 vs 31 ± 24, p = 0.016). GMP addition to smoothie reduced pleasantness (26 ± 21 vs. 61 ± 29, p = 0.009) and worsened the aroma (46 ± 15 vs. 69 ± 28, p = 0.014). The focus group revealed uncertainty of protein needs and a scepticism of supplements, with preference for food. Gut hormone response did not differ between GMP and WHEY (nAUC for all gut hormones p > 0.05). There was no difference between conditions for lunch ad libitum intake (549 ± 171 kcal, 512 ± 238 kcal, 460 ± 199 kcal for GMP, WHEY, and water, p = 0.175), or for subjective appetite response. Leucine-enriched GMP was not less satiating than WHEY, and low palatability and scepticism of supplements question the likely acceptability of GMP supplementation. Providing trusted nutritional advice and food enrichment/fortification may be preferred strategies for increasing protein intake in OA.
Collapse
Affiliation(s)
- Jordan Warner
- School of Biomedical, Nutritional, and Sport Sciences, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
| | - Rachel Stocker
- School of Biomedical, Nutritional, and Sport Sciences, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
| | - Kirsten Brandt
- Population Health Sciences Institute, Newcastle University, Newcastle Upon Tyne, UK; Human Nutrition and Exercise Research Centre, Population Health Sciences Institute, Newcastle University, Newcastle Upon Tyne, UK
| | | | | | - Emma Stevenson
- School of Biomedical, Nutritional, and Sport Sciences, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK; Human Nutrition and Exercise Research Centre, Population Health Sciences Institute, Newcastle University, Newcastle Upon Tyne, UK
| | - Adrian Holliday
- School of Biomedical, Nutritional, and Sport Sciences, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK; Human Nutrition and Exercise Research Centre, Population Health Sciences Institute, Newcastle University, Newcastle Upon Tyne, UK.
| |
Collapse
|
4
|
Ansari S, Khoo B, Tan T. Targeting the incretin system in obesity and type 2 diabetes mellitus. Nat Rev Endocrinol 2024; 20:447-459. [PMID: 38632474 DOI: 10.1038/s41574-024-00979-9] [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] [Accepted: 03/12/2024] [Indexed: 04/19/2024]
Abstract
Obesity and type 2 diabetes mellitus (T2DM) are widespread, non-communicable diseases that are responsible for considerable levels of morbidity and mortality globally, primarily in the form of cardiovascular disease (CVD). Changes to lifestyle and behaviour have insufficient long-term efficacy in most patients with these diseases; metabolic surgery, although effective, is not practically deliverable on the scale that is required. Over the past two decades, therapies based on incretin hormones, spearheaded by glucagon-like peptide 1 (GLP1) receptor agonists (GLP1RAs), have become the treatment of choice for obesity and T2DM, and clinical evidence now suggests that these agents have benefits for CVD. We review the latest advances in incretin-based pharmacotherapy. These include 'GLP1 plus' agents, which combine the known advantages of GLP1RAs with the activity of additional hormones, such as glucose-dependent insulinotropic peptide, glucagon and amylin, to achieve desired therapeutic goals. Second-generation non-peptidic oral GLP1RAs promise to extend the benefits of GLP1 therapy to those who do not want, or cannot have, subcutaneous injection therapy. We conclude with a discussion of the knowledge gaps that must be addressed before incretin-based therapies can be properly deployed for maximum benefit in the treatment of obesity and T2DM.
Collapse
Affiliation(s)
- Saleem Ansari
- Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, UK
| | - Bernard Khoo
- Department of Endocrinology, Division of Medicine, Royal Free Campus, University College London, London, UK
| | - Tricia Tan
- Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, UK.
| |
Collapse
|
5
|
Kowalka AM, Alexiadou K, Cuenco J, Clarke RE, Minnion J, Williams EL, Bech P, Purkayastha S, Ahmed AR, Takats Z, Whitwell HJ, Romero MG, Bloom SR, Camuzeaux S, Lewis MR, Khoo B, Tan TM. The postprandial secretion of peptide YY 1-36 and 3-36 in obesity is differentially increased after gastric bypass versus sleeve gastrectomy. Clin Endocrinol (Oxf) 2023; 99:272-284. [PMID: 36345253 PMCID: PMC10952770 DOI: 10.1111/cen.14846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 10/13/2022] [Accepted: 11/04/2022] [Indexed: 11/09/2022]
Abstract
OBJECTIVES Peptide tyrosine tyrosine (PYY) exists as two species, PYY1-36 and PYY3-36 , with distinct effects on insulin secretion and appetite regulation. The detailed effects of bariatric surgery on PYY1-36 and PYY3-36 secretion are not known as previous studies have used nonspecific immunoassays to measure total PYY. Our objective was to characterize the effect of sleeve gastrectomy (SG) and Roux-en-Y gastric bypass (RYGB) on fasting and postprandial PYY1-36 and PYY3-36 secretion using a newly developed liquid chromatography-tandem mass spectrometry (LC-MS/MS) assay. DESIGN AND SUBJECTS Observational study in 10 healthy nonobese volunteers and 30 participants with obesity who underwent RYGB (n = 24) or SG (n = 6) at the Imperial Weight Centre [NCT01945840]. Participants were studied using a standardized mixed meal test (MMT) before and 1 year after surgery. The outcome measures were PYY1-36 and PYY3-36 concentrations. RESULTS Presurgery, the fasting and postprandial levels of PYY1-36 and PYY3-36 were low, with minimal responses to the MMT, and these did not differ from healthy nonobese volunteers. The postprandial secretion of both PYY1-36 and PYY3-36 at 1 year was amplified after RYGB, but not SG, with the response being significantly higher in RYGB compared with SG. CONCLUSIONS There appears to be no difference in PYY secretion between nonobese and obese volunteers at baseline. At 1 year after surgery, RYGB, but not SG, is associated with increased postprandial secretion of PYY1-36 and PYY3-36 , which may account for long-term differences in efficacy and adverse effects between the two types of surgery.
Collapse
Affiliation(s)
- Anna M. Kowalka
- Section of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and ReproductionImperial College LondonLondonUK
| | - Kleopatra Alexiadou
- Section of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and ReproductionImperial College LondonLondonUK
| | - Joyceline Cuenco
- Section of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and ReproductionImperial College LondonLondonUK
| | | | - James Minnion
- Section of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and ReproductionImperial College LondonLondonUK
| | - Emma L. Williams
- Department of Clinical Biochemistry, North West London PathologyCharing Cross HospitalLondonUK
| | - Paul Bech
- Section of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and ReproductionImperial College LondonLondonUK
| | - Sanjay Purkayastha
- Department of Surgery and CancerImperial College Healthcare NHS TrustLondonUK
| | - Ahmed R. Ahmed
- Department of Surgery and CancerImperial College Healthcare NHS TrustLondonUK
| | - Zoltan Takats
- Section of Bioanalytical Chemistry, Department of Metabolism, Digestion and ReproductionImperial College LondonLondonUK
- National Phenome CentreImperial College LondonLondonUK
| | - Harry J. Whitwell
- Section of Bioanalytical Chemistry, Department of Metabolism, Digestion and ReproductionImperial College LondonLondonUK
- National Phenome CentreImperial College LondonLondonUK
| | - Maria Gomez Romero
- Section of Bioanalytical Chemistry, Department of Metabolism, Digestion and ReproductionImperial College LondonLondonUK
- National Phenome CentreImperial College LondonLondonUK
| | - Stephen R. Bloom
- Section of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and ReproductionImperial College LondonLondonUK
| | - Stephane Camuzeaux
- Section of Bioanalytical Chemistry, Department of Metabolism, Digestion and ReproductionImperial College LondonLondonUK
- National Phenome CentreImperial College LondonLondonUK
| | - Matthew R. Lewis
- Section of Bioanalytical Chemistry, Department of Metabolism, Digestion and ReproductionImperial College LondonLondonUK
- National Phenome CentreImperial College LondonLondonUK
| | - Bernard Khoo
- Endocrinology, Division of MedicineUniversity College LondonLondonUK
| | - Tricia M.‐M. Tan
- Section of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and ReproductionImperial College LondonLondonUK
| |
Collapse
|
6
|
Wu Q, Burley G, Li L, Lin S, Shi Y. The role of dietary salt in metabolism and energy balance: Insights beyond cardiovascular disease. Diabetes Obes Metab 2023; 25:1147-1161. [PMID: 36655379 PMCID: PMC10946535 DOI: 10.1111/dom.14980] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 01/14/2023] [Accepted: 01/18/2023] [Indexed: 01/20/2023]
Abstract
Dietary salt (NaCl) is essential to an organism's survival. However, today's diets are dominated by excessive salt intake, which significantly impacts individual and population health. High salt intake is closely linked to cardiovascular disease (CVD), especially hypertension, through a number of well-studied mechanisms. Emerging evidence indicates that salt overconsumption may also be associated with metabolic disorders. In this review, we first summarize recent updates on the mechanisms of salt-induced CVD, the effects of salt reduction and the use of salt substitution as a therapy. Next, we focus on how high salt intake can impact metabolism and energy balance, describing the mechanisms through which this occurs, including leptin resistance, the overproduction of fructose and ghrelin, insulin resistance and altered hormonal factors. A further influence on metabolism worth noting is the reported role of salt in inducing thermogenesis and increasing body temperature, leading to an increase in energy expenditure. While this result could be viewed as a positive metabolic effect because it promotes a negative energy balance to combat obesity, caution must be taken with this frame of thinking given the deleterious consequences of chronic high salt intake on cardiovascular health. Nevertheless, this review highlights the importance of salt as a noncaloric nutrient in regulating whole-body energy homeostasis. Through this review, we hope to provide a scientific framework for future studies to systematically address the metabolic impacts of dietary salt and salt replacement treatments. In addition, we hope to form a foundation for future clinical trials to explore how these salt-induced metabolic changes impact obesity development and progression, and to elucidate the regulatory mechanisms that drive these changes, with the aim of developing novel therapeutics for obesity and CVD.
Collapse
Affiliation(s)
- Qi Wu
- Obesity and Metabolic Disease Research GroupGarvan Institute of Medical ResearchSydneyNew South WalesAustralia
- Centre of Neurological and Metabolic Researchthe Second Affiliated Hospital of Fujian Medical UniversityQuanzhouChina
| | - George Burley
- Obesity and Metabolic Disease Research GroupGarvan Institute of Medical ResearchSydneyNew South WalesAustralia
| | - Li‐Cheng Li
- Centre of Neurological and Metabolic Researchthe Second Affiliated Hospital of Fujian Medical UniversityQuanzhouChina
| | - Shu Lin
- Obesity and Metabolic Disease Research GroupGarvan Institute of Medical ResearchSydneyNew South WalesAustralia
- Centre of Neurological and Metabolic Researchthe Second Affiliated Hospital of Fujian Medical UniversityQuanzhouChina
| | - Yan‐Chuan Shi
- Obesity and Metabolic Disease Research GroupGarvan Institute of Medical ResearchSydneyNew South WalesAustralia
- Centre of Neurological and Metabolic Researchthe Second Affiliated Hospital of Fujian Medical UniversityQuanzhouChina
- School of Clinical Medicine, St Vincent's Clinical CampusFaculty of Medicine and HealthSydneyNew South WalesAustralia
| |
Collapse
|
7
|
Unlu Y, Vinales KL, Hollstein T, Chang D, Cabeza de Baca T, Walter M, Krakoff J, Piaggi P. The association between gut hormones and diet-induced metabolic flexibility in metabolically healthy adults. Obesity (Silver Spring) 2023; 31:139-149. [PMID: 36471908 PMCID: PMC9780166 DOI: 10.1002/oby.23584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 08/11/2022] [Accepted: 08/15/2022] [Indexed: 12/12/2022]
Abstract
OBJECTIVE This study investigated whether interindividual variance in diet-induced metabolic flexibility is explained by differences in gut hormone concentrations. METHODS A total of 69 healthy volunteers with normal glucose regulation underwent 24-hour assessments of respiratory quotient (RQ) in a whole-room indirect calorimeter during eucaloric feeding (EBL; 50% carbohydrate, 30% fat) and then, in a crossover design, during 24-hour fasting and three normal-protein (20%) overfeeding diets (200% energy requirements). Metabolic flexibility was defined as the change in 24-hour RQ from EBL during standard (50% carbohydrate), high-fat (60%), and high-carbohydrate (75%) overfeeding diets. Plasma concentrations of glucagon-like peptide-1 (GLP-1) and peptide YY (PYY) after an overnight fast were measured prior to and after each diet. RESULTS Compared with EBL, on average, 24-hour RQ decreased by ~4% during high-fat overfeeding, whereas it increased by ~4% during standard overfeeding and by ~9% during high-carbohydrate overfeeding. During high-carbohydrate overfeeding, but not during any other overfeeding diet or fasting, increased GLP-1 concentration was associated with increased RQ (r = 0.44, p < 0.001), higher/lower carbohydrate/lipid oxidation rates (r = 0.34 and r = -0.51, both p < 0.01), respectively, and increased plasma insulin concentration (r = 0.38, p = 0.02). CONCLUSIONS Increased GLP-1 concentration following high-carbohydrate overfeeding associated with a greater shift to carbohydrate oxidation, suggesting that GLP-1 may be implicated in diet-induced metabolic flexibility to carbohydrate overload.
Collapse
Affiliation(s)
- Yigit Unlu
- Obesity and Diabetes Clinical Research Section, Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Department of Health and Human Services, Phoenix, AZ
| | - Karyne L. Vinales
- Obesity and Diabetes Clinical Research Section, Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Department of Health and Human Services, Phoenix, AZ
- Endocrinology Division, Medicine Department, Phoenix VA Health Care System, Phoenix, AZ, USA
| | - Tim Hollstein
- Obesity and Diabetes Clinical Research Section, Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Department of Health and Human Services, Phoenix, AZ
- Division of Endocrinology, Diabetology and Clinical Nutrition, Department of Internal Medicine 1, University of Kiel, Arnold-Heller-Straße 3, Kiel 24105, Germany
| | - Douglas Chang
- Obesity and Diabetes Clinical Research Section, Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Department of Health and Human Services, Phoenix, AZ
| | - Tomás Cabeza de Baca
- Obesity and Diabetes Clinical Research Section, Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Department of Health and Human Services, Phoenix, AZ
| | - Mary Walter
- Clinical Core Laboratory, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jonathan Krakoff
- Obesity and Diabetes Clinical Research Section, Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Department of Health and Human Services, Phoenix, AZ
| | - Paolo Piaggi
- Obesity and Diabetes Clinical Research Section, Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Department of Health and Human Services, Phoenix, AZ
- Department of Information Engineering, University of Pisa, Pisa, Italy
| |
Collapse
|
8
|
Song C, Wen H, Liu G, Ma X, Lv G, Wu N, Chen J, Xue M, Li H, Xu P. Gut Microbes Reveal Pseudomonas Medicates Ingestion Preference via Protein Utilization and Cellular Homeostasis Under Feed Domestication in Freshwater Drum, Aplodinotus grunniens. Front Microbiol 2022; 13:861705. [PMID: 35722333 PMCID: PMC9204248 DOI: 10.3389/fmicb.2022.861705] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 05/02/2022] [Indexed: 12/15/2022] Open
Abstract
With strong demand for aquatic products, as well as a rapid decrease in global fishery resources and capture fisheries, domesticating animals to provide more high-quality proteins is meaningful for humans. Freshwater drum (Aplodinotus grunniens) is widely distributed in the wild habitats of North America. However, the research on A. grunniens and the feed domestication with diets composed of artificial compounds remains unclear. In this study, a 4-month feeding domestication experiment was conducted with A. grunniens larvae to evaluate the underlying mechanism and molecular targets responsible for alternations in the ingestion performance. The results indicated that a significant increase in the final body weight was exhibited by the feed domesticated group (DOM, 114.8 g) when compared to the group that did not ingest the feed (WT, 5.3 g) as the latest version we raised From the result, the final body weight exhibited significant increase between unfavorable with the feed (WT, 5.3 g) and feed domesticated group (DOM, 114.8 g). In addition, the enzyme activity of digestive enzymes like amylase, lipase, and trypsin was increased in DOM. Genes related to appetite and perception, such as NPY4R, PYY, and LEPR, were activated in DOM. 16s rRNA gene sequencing analysis revealed that Pseudomonas sp. increased from 58.74% to 89.77% in DOM, which accounts for the dominant upregulated microbial community at the genus level, followed by Plesiomonas. Analogously, Mycobacterium, Methylocystis, and Romboutsia also accounted for the down-regulated microbes in the diversity. Transcriptome and RT-PCR analysis revealed that feed domestication significantly improved protein digestion and absorption, inhibited apoptosis by AGE-RAGE signaling, and activated extracellular matrix remodeling by relaxin signaling. Integrated analysis of the microbiome and host transcriptome revealed that Pseudomonas-mediated ingestion capacity, protein utilization, and cellular homeostasis might be the underlying mechanism under feed domestication. These results indicate Pseudomonas and its key genes relating to food ingestion and digestion could serve as the molecular targets for feed domestication and sustainable development in A. grunniens.
Collapse
Affiliation(s)
- Changyou Song
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, China
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi, China
| | - Haibo Wen
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, China
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi, China
| | - Guangxiang Liu
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi, China
| | - Xueyan Ma
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, China
| | - Guohua Lv
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, China
| | - Ningyuan Wu
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi, China
| | - Jianxiang Chen
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi, China
| | - Miaomiao Xue
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi, China
| | - Hongxia Li
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, China
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi, China
| | - Pao Xu
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, China
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi, China
| |
Collapse
|
9
|
Holst JJ, Jepsen SL, Modvig I. GLP-1 – Incretin and pleiotropic hormone with pharmacotherapy potential. Increasing secretion of endogenous GLP-1 for diabetes and obesity therapy. Curr Opin Pharmacol 2022; 63:102189. [DOI: 10.1016/j.coph.2022.102189] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 12/22/2021] [Accepted: 01/24/2022] [Indexed: 02/09/2023]
|
10
|
Blonde GD, Mathes CM, Inui T, Hamel EA, Price RK, Livingstone MBE, Le Roux CW, Spector AC. Oromotor and somatic taste reactivity during sucrose meals reveals internal state and stimulus palatability after gastric bypass in rats. Am J Physiol Regul Integr Comp Physiol 2022; 322:R204-R218. [PMID: 35043683 PMCID: PMC8858674 DOI: 10.1152/ajpregu.00285.2021] [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: 11/19/2021] [Revised: 01/07/2022] [Accepted: 01/12/2022] [Indexed: 11/22/2022]
Abstract
After Roux-en-Y gastric bypass (RYGB), rats consume less high-energy foods and fluids, though whether this reflects a concomitant change in palatability remains unclear. By measuring behavior during intraorally delivered liquid meals across days (1 water, 8 sucrose sessions), we showed that RYGB rats (RYGB, n = 8/sex) consumed less 1.0 M sucrose than their sham surgery counterparts (SHAM, n = 8 males, n = 11 females) but displayed similarly high levels of ingestive taste reactivity responses at the start of infusions. Relative to water, both groups increased intake of sucrose, and ingestive responses were dominated by tongue protrusions rather than mouth movements. Thus, RYGB animals still found sucrose palatable despite consuming less than the SHAM group. As the intraoral infusion progressed but before meal termination, aversive behavior remained low and both RYGB and SHAM animals showed fewer ingestive responses, predominantly mouth movements as opposed to tongue protrusions. This shift in responsiveness unrelated to surgical manipulation suggests negative alliesthesia, or a decreased palatability, as rats approach satiation. Notably, only in RYGB rats, across sessions, there was a striking emergence of aversive behavior immediately after the sucrose meal. Thus, although lower intake in RYGB rats seems independent of the hedonic taste properties of sucrose, taste reactivity behavior in these animals immediately after termination of a liquid meal appears to be influenced by postoral events and reflects a state of nimiety or excessive consumption. Measurement of taste reactivity behaviors during an intraorally delivered meal represents a promising way to make inferences about internal state in nonverbal preclinical models.
Collapse
Affiliation(s)
- Ginger D Blonde
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, Florida
| | - Clare M Mathes
- Department of Neuroscience, Baldwin Wallace University, Berea, Ohio
| | - Tadashi Inui
- Department of Oral Physiology, Graduate School of Dental Medicine, Hokkaido University, Sapporo, Japan
| | - Elizabeth A Hamel
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, Florida
| | - Ruth K Price
- Nutrition Innovation Centre for Food and Health (NICHE), School of Biomedical Sciences, Ulster University, Coleraine, United Kingdom
| | - M Barbara E Livingstone
- Nutrition Innovation Centre for Food and Health (NICHE), School of Biomedical Sciences, Ulster University, Coleraine, United Kingdom
| | - Carel W Le Roux
- Diabetes Complications Research Centre, Conway Institute, School of Medicine, University College Dublin, Dublin, Ireland
| | - Alan C Spector
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, Florida
| |
Collapse
|
11
|
Woodward ORM, Gribble FM, Reimann F, Lewis JE. Gut peptide regulation of food intake - evidence for the modulation of hedonic feeding. J Physiol 2022; 600:1053-1078. [PMID: 34152020 DOI: 10.1113/jp280581] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 05/17/2021] [Indexed: 12/13/2022] Open
Abstract
The number of people living with obesity has tripled worldwide since 1975 with serious implications for public health, as obesity is linked to a significantly higher chance of early death from associated comorbidities (metabolic syndrome, type 2 diabetes, cardiovascular disease and cancer). As obesity is a consequence of food intake exceeding the demands of energy expenditure, efforts are being made to better understand the homeostatic and hedonic mechanisms governing food intake. Gastrointestinal peptides are secreted from enteroendocrine cells in response to nutrient and energy intake, and modulate food intake either via afferent nerves, including the vagus nerve, or directly within the central nervous system, predominantly gaining access at circumventricular organs. Enteroendocrine hormones modulate homeostatic control centres at hypothalamic nuclei and the dorso-vagal complex. Additional roles of these peptides in modulating hedonic food intake and/or preference via the neural systems of reward are starting to be elucidated, with both peripheral and central peptide sources potentially contributing to central receptor activation. Pharmacological interventions and gastric bypass surgery for the treatment of type 2 diabetes and obesity elevate enteroendocrine hormone levels and also alter food preference. Hence, understanding of the hedonic mechanisms mediated by gut peptide action could advance development of potential therapeutic strategies for the treatment of obesity and its comorbidities.
Collapse
Affiliation(s)
- Orla R M Woodward
- Wellcome Trust - MRC Institute of Metabolic Science Metabolic Research Laboratories, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 0QQ, UK
| | - Fiona M Gribble
- Wellcome Trust - MRC Institute of Metabolic Science Metabolic Research Laboratories, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 0QQ, UK
| | - Frank Reimann
- Wellcome Trust - MRC Institute of Metabolic Science Metabolic Research Laboratories, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 0QQ, UK
| | - Jo E Lewis
- Wellcome Trust - MRC Institute of Metabolic Science Metabolic Research Laboratories, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 0QQ, UK
| |
Collapse
|
12
|
Blonde GD, Price RK, le Roux CW, Spector AC. Meal Patterns and Food Choices of Female Rats Fed a Cafeteria-Style Diet Are Altered by Gastric Bypass Surgery. Nutrients 2021; 13:3856. [PMID: 34836110 PMCID: PMC8623594 DOI: 10.3390/nu13113856] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/21/2021] [Accepted: 10/26/2021] [Indexed: 12/28/2022] Open
Abstract
After Roux-en-Y gastric bypass surgery (RYGB), rats tend to reduce consumption of high-sugar and/or high-fat foods over time. Here, we sought to investigate the behavioral mechanisms underlying these intake outcomes. Adult female rats were provided a cafeteria diet comprised of five palatable foodstuffs varying in sugar and fat content and intake was monitored continuously. Rats were then assigned to either RYGB, or one of two control (CTL) groups: sham surgery or a nonsurgical control group receiving the same prophylactic iron treatments as RYGB rats. Post-sur-gically, all rats consumed a large first meal of the cafeteria diet. After the first meal, RYGB rats reduced intake primarily by decreasing the meal sizes relative to CTL rats, ate meals more slowly, and displayed altered nycthemeral timing of intake yielding more daytime meals and fewer nighttime meals. Collectively, these meal patterns indicate that despite being motivated to consume a cafeteria diet after RYGB, rats rapidly learn to modify eating behaviors to consume foods more slowly across the entire day. RYGB rats also altered food preferences, but more slowly than the changes in meal patterns, and ate proportionally more energy from complex carbohydrates and protein and proportionally less fat. Overall, the pattern of results suggests that after RYGB rats quickly learn to adjust their size, eating rate, and distribution of meals without altering meal number and to shift their macronutrient intake away from fat; these changes appear to be more related to postingestive events than to a fundamental decline in the palatability of food choices.
Collapse
Affiliation(s)
- Ginger D. Blonde
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL 32306, USA;
| | - Ruth K. Price
- Nutrition Innovation Centre for Food and Health (NICHE), School of Biomedical Sciences, Ulster University, Coleraine BT52 1SA, UK;
| | - Carel W. le Roux
- Diabetes Complications Research Centre, Conway Institute, School of Medicine, University College Dublin, D04 V1W8 Dublin, Ireland;
| | - Alan C. Spector
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL 32306, USA;
| |
Collapse
|
13
|
Smith KR, Moran TH. Gastrointestinal peptides in eating-related disorders. Physiol Behav 2021; 238:113456. [PMID: 33989649 PMCID: PMC8462672 DOI: 10.1016/j.physbeh.2021.113456] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 04/19/2021] [Accepted: 05/04/2021] [Indexed: 12/13/2022]
Abstract
Food intake is tightly controlled by homeostatic signals sensitive to metabolic need for the regulation of body weight. This review focuses on the peripherally-secreted gastrointestinal peptides (i.e., ghrelin, cholecystokinin, glucagon-like peptide 1, and peptide tyrosine tyrosine) that contribute to the control of appetite and discusses how these peptides or the signals arising from their release are disrupted in eating-related disorders across the weight spectrum, namely anorexia nervosa, bulimia nervosa, and obesity, and whether they are normalized following weight restoration or weight loss treatment. Further, the role of gut peptides in the pathogenesis and treatment response in human weight conditions as identified by rodent models are discussed. Lastly, we review the incretin- and hormone-based pharmacotherapies available for the treatment of obesity and eating-related disorders.
Collapse
Affiliation(s)
- Kimberly R Smith
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States.
| | - Timothy H Moran
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
| |
Collapse
|
14
|
Henquin JC. Non-glucose modulators of insulin secretion in healthy humans: (dis)similarities between islet and in vivo studies. Metabolism 2021; 122:154821. [PMID: 34174327 DOI: 10.1016/j.metabol.2021.154821] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 06/10/2021] [Accepted: 06/18/2021] [Indexed: 12/17/2022]
Abstract
Optimal metabolic homeostasis requires precise temporal and quantitative control of insulin secretion. Both in vivo and in vitro studies have often focused on the regulation by glucose although many additional factors including other nutrients, neurotransmitters, hormones and drugs, modulate the secretory function of pancreatic β-cells. This review is based on the analysis of clinical investigations characterizing the effects of non-glucose modulators of insulin secretion in healthy subjects, and of experimental studies testing the same modulators in islets isolated from normal human donors. The aim was to determine whether the information gathered in vitro can reliably be translated to the in vivo situation. The comparison evidenced both convincing similarities and areas of discordance. The lack of coherence generally stems from the use of exceedingly high concentrations of test agents at too high or too low glucose concentrations in vitro, which casts doubts on the physiological relevance of a number of observations made in isolated islets. Future projects resorting to human islets should avoid extreme experimental conditions, such as oversized stimulations or inhibitions of β-cells, which are unlikely to throw light on normal insulin secretion and contribute to the elucidation of its defects.
Collapse
Affiliation(s)
- Jean-Claude Henquin
- Unit of Endocrinology and Metabolism, Faculty of Medicine, University of Louvain, Brussels, Belgium.
| |
Collapse
|
15
|
Nogueiras R. MECHANISMS IN ENDOCRINOLOGY: The gut-brain axis: regulating energy balance independent of food intake. Eur J Endocrinol 2021; 185:R75-R91. [PMID: 34260412 PMCID: PMC8345901 DOI: 10.1530/eje-21-0277] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 07/14/2021] [Indexed: 12/15/2022]
Abstract
Obesity is a global pandemic with a large health and economic burden worldwide. Bodyweight is regulated by the ability of the CNS, and especially the hypothalamus, to orchestrate the function of peripheral organs that play a key role in metabolism. Gut hormones play a fundamental role in the regulation of energy balance, as they modulate not only feeding behavior but also energy expenditure and nutrient partitioning. This review examines the recent discoveries about hormones produced in the stomach and gut, which have been reported to regulate food intake and energy expenditure in preclinical models. Some of these hormones act on the hypothalamus to modulate thermogenesis and adiposity in a food intake-independent fashion. Finally, the association of these gut hormones to eating, energy expenditure, and weight loss after bariatric surgery in humans is discussed.
Collapse
Affiliation(s)
- Ruben Nogueiras
- Department of Physiology, CIMUS, USC, CIBER Fisiopatología Obesidad y Nutrición (CiberOBN), Instituto Salud Carlos III, Galician Agency of Innovation, Xunta de Galicia, Santiago de Compostela, Spain
| |
Collapse
|
16
|
Deoxynivalenol (Vomitoxin)-Induced Anorexia Is Induced by the Release of Intestinal Hormones in Mice. Toxins (Basel) 2021; 13:toxins13080512. [PMID: 34437383 PMCID: PMC8402572 DOI: 10.3390/toxins13080512] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 07/07/2021] [Accepted: 07/15/2021] [Indexed: 12/25/2022] Open
Abstract
Deoxynivalenol (DON), also known as vomitoxin, is a mycotoxin that can cause antifeeding and vomiting in animals. However, the mechanism of DON inducing anorexia is complicated. Studies have shown that intestinal hormones play a significant part in the anorexia caused by DON. We adopted the “modeling of acute antifeeding in mice” as the basic experimental model, and used two methods of gavage and intraperitoneal injection to explore the effect of intestinal hormones on the antifeedant response induced by DON in mice. We found that 1 and 2.5 mg/kg·bw of DON can acutely induce anorexia and increase the plasma intestinal hormones CCK, PYY, GIP, and GLP-1 in mice within 3 h. Direct injection of exogenous intestinal hormones CCK, PYY, GIP, and GLP-1 can trigger anorexia behavior in mice. Furthermore, the PYY receptor antagonist JNJ-31020028, GLP-1 receptor antagonist Exendin(9-39), CCK receptor antagonist Proglumide, GIP receptor antagonist GIP(3-30)NH2 attenuated both intestinal hormone and DON-induced anorectic responses. These results indicate that intestinal hormones play a critical role in the anorexia response induced by DON.
Collapse
|
17
|
Lewgood J, Oliveira B, Korzepa M, Forbes SC, Little JP, Breen L, Bailie R, Candow DG. Efficacy of Dietary and Supplementation Interventions for Individuals with Type 2 Diabetes. Nutrients 2021; 13:2378. [PMID: 34371888 PMCID: PMC8308746 DOI: 10.3390/nu13072378] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 07/07/2021] [Accepted: 07/08/2021] [Indexed: 02/06/2023] Open
Abstract
The prevalence of Type 2 diabetes (T2D) is increasing, which creates a large economic burden. Diet is a critical factor in the treatment and management of T2D; however, there are a large number of dietary approaches and a general lack of consensus regarding the efficacy of each. Therefore, the purpose of this narrative review is twofold: (1) to critically evaluate the effects of various dietary strategies on diabetes management and treatment, such as Mediterranean diet, plant-based diet, low-calorie and very low-calorie diets, intermittent fasting, low-carbohydrate and very low-carbohydrate diets, and low glycemic diets and (2) to examine several purported supplements, such as protein, branched-chain amino acids, creatine, and vitamin D to improve glucose control and body composition. This review can serve as a resource for those wanting to evaluate the evidence supporting the various dietary strategies and supplements that may help manage T2D.
Collapse
Affiliation(s)
- Jessica Lewgood
- Faculty of Kinesiology and Health Studies, University of Regina, Regina, SK S4S0A2, Canada; (J.L.); (R.B.)
| | - Barbara Oliveira
- Okanagan Campus, School of Health and Exercise Sciences, University of British Columbia, Kelowna, BC V1V1V7, Canada; (B.O.); (J.P.L.)
| | - Marie Korzepa
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK; (M.K.); (L.B.)
| | - Scott C. Forbes
- Department of Physical Education Studies, Faculty of Education, Brandon University, Brandon, MB R7A6A9, Canada;
| | - Jonathan P. Little
- Okanagan Campus, School of Health and Exercise Sciences, University of British Columbia, Kelowna, BC V1V1V7, Canada; (B.O.); (J.P.L.)
| | - Leigh Breen
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK; (M.K.); (L.B.)
| | - Robert Bailie
- Faculty of Kinesiology and Health Studies, University of Regina, Regina, SK S4S0A2, Canada; (J.L.); (R.B.)
| | - Darren G. Candow
- Faculty of Kinesiology and Health Studies, University of Regina, Regina, SK S4S0A2, Canada; (J.L.); (R.B.)
| |
Collapse
|
18
|
Abstract
PURPOSE OF REVIEW The prevalence of obesity is increasing in all age groups. Following its success in adults, and with limited success using conservative therapies, metabolic and bariatric surgery (MBS) is increasingly being utilized in adolescents. This review highlights the current evidence and guidelines supporting its use. RECENT FINDINGS Safety and efficacy mirror results seen in adults. The most recent evidence, as outcomes enter the long term, suggests that comorbidity resolution, including diabetes and hypertension, can even outperform that of adults. Mental health problems persist despite good weight loss. Overall, the positive early weight and comorbidity outcomes are well sustained into the long term. There is a growing need to prevent and treat adolescent obesity. Current evidence supports the use of MBS in adolescents. Ongoing and future studies will provide 10-year outcomes and assist in the refinement of multimodal pathways incorporating MBS for the treatment of severe childhood obesity.
Collapse
Affiliation(s)
| | | | - Andrew J Beamish
- Department of Gastrosurgical Research, Institute of Clinical Sciences, Gothenburg University, Gothenburg, Sweden.
- Swansea University Medical School, Swansea University, Swansea, UK.
- Department of GastroSurgical Research and Education, Sahlgrenska Universitetsjukhuset, Institute of Clinical Sciences, Gothenburg University, Gothenburg, 41431, UK.
| |
Collapse
|
19
|
Hansen HH, Grønlund RV, Baader-Pagler T, Haebel P, Tammen H, Larsen LK, Jelsing J, Vrang N, Klein T. Characterization of combined linagliptin and Y2R agonist treatment in diet-induced obese mice. Sci Rep 2021; 11:8060. [PMID: 33850212 PMCID: PMC8044192 DOI: 10.1038/s41598-021-87539-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 03/31/2021] [Indexed: 02/01/2023] Open
Abstract
Dipeptidyl peptidase IV (DPP-IV) inhibitors improve glycemic control by prolonging the action of glucagon-like peptide-1 (GLP-1). In contrast to GLP-1 analogues, DPP-IV inhibitors are weight-neutral. DPP-IV cleavage of PYY and NPY gives rise to PYY3-36 and NPY3-36 which exert potent anorectic action by stimulating Y2 receptor (Y2R) function. This invites the possibility that DPP-IV inhibitors could be weight-neutral by preventing conversion of PYY/NPY to Y2R-selective peptide agonists. We therefore investigated whether co-administration of an Y2R-selective agonist could unmask potential weight lowering effects of the DDP-IV inhibitor linagliptin. Male diet-induced obese (DIO) mice received once daily subcutaneous treatment with linagliptin (3 mg/kg), a Y2R-selective PYY3-36 analogue (3 or 30 nmol/kg) or combination therapy for 14 days. While linagliptin promoted marginal weight loss without influencing food intake, the PYY3-36 analogue induced significant weight loss and transient suppression of food intake. Both compounds significantly improved oral glucose tolerance. Because combination treatment did not further improve weight loss and glucose tolerance in DIO mice, this suggests that potential negative modulatory effects of DPP-IV inhibitors on endogenous Y2R peptide agonist activity is likely insufficient to influence weight homeostasis. Weight-neutrality of DPP-IV inhibitors may therefore not be explained by counter-regulatory effects on PYY/NPY responses.
Collapse
Affiliation(s)
| | | | - Tamara Baader-Pagler
- Department of Cardiometabolic Diseases Research, Boehringer Ingelheim Pharma GmbH & Co., Biberach, Germany
| | - Peter Haebel
- Department of Cardiometabolic Diseases Research, Boehringer Ingelheim Pharma GmbH & Co., Biberach, Germany
| | | | | | - Jacob Jelsing
- Gubra, Hørsholm Kongevej 11B, 2970, Hørsholm, Denmark
| | - Niels Vrang
- Gubra, Hørsholm Kongevej 11B, 2970, Hørsholm, Denmark
| | - Thomas Klein
- Department of Cardiometabolic Diseases Research, Boehringer Ingelheim Pharma GmbH & Co., Biberach, Germany
| |
Collapse
|
20
|
Schalla MA, Taché Y, Stengel A. Neuroendocrine Peptides of the Gut and Their Role in the Regulation of Food Intake. Compr Physiol 2021; 11:1679-1730. [PMID: 33792904 DOI: 10.1002/cphy.c200007] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The regulation of food intake encompasses complex interplays between the gut and the brain. Among them, the gastrointestinal tract releases different peptides that communicate the metabolic state to specific nuclei in the hindbrain and the hypothalamus. The present overview gives emphasis on seven peptides that are produced by and secreted from specialized enteroendocrine cells along the gastrointestinal tract in relation with the nutritional status. These established modulators of feeding are ghrelin and nesfatin-1 secreted from gastric X/A-like cells, cholecystokinin (CCK) secreted from duodenal I-cells, glucagon-like peptide 1 (GLP-1), oxyntomodulin, and peptide YY (PYY) secreted from intestinal L-cells and uroguanylin (UGN) released from enterochromaffin (EC) cells. © 2021 American Physiological Society. Compr Physiol 11:1679-1730, 2021.
Collapse
Affiliation(s)
- Martha A Schalla
- Charité Center for Internal Medicine and Dermatology, Department for Psychosomatic Medicine, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Yvette Taché
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, CURE: Digestive Diseases Research Center, David Geffen School of Medicine, UCLA, Los Angeles, California, USA.,VA Greater Los Angeles Healthcare System, Los Angeles, California, USA
| | - Andreas Stengel
- Charité Center for Internal Medicine and Dermatology, Department for Psychosomatic Medicine, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany.,Department of Psychosomatic Medicine and Psychotherapy, Medical University Hospital Tübingen, Tübingen, Germany
| |
Collapse
|
21
|
Grunddal KV, Diep TA, Petersen N, Tough IR, Skov LJ, Liu L, Buijink JA, Mende F, Jin C, Jepsen SL, Sørensen LME, Achiam MP, Strandby RB, Bach A, Hartmann B, Frimurer TM, Hjorth SA, Bouvier M, Cox H, Holst B. Selective release of gastrointestinal hormones induced by an orally active GPR39 agonist. Mol Metab 2021; 49:101207. [PMID: 33711555 PMCID: PMC8042403 DOI: 10.1016/j.molmet.2021.101207] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 03/03/2021] [Accepted: 03/04/2021] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVES Obesity is a complex disease associated with a high risk of comorbidities. Gastric bypass surgery, an invasive procedure with low patient eligibility, is currently the most effective intervention that achieves sustained weight loss. This beneficial effect is attributed to alterations in gut hormone signaling. An attractive alternative is to pharmacologically mimic the effects of bariatric surgery by targeting several gut hormonal axes. The G protein-coupled receptor 39 (GPR39) expressed in the gastrointestinal tract has been shown to mediate ghrelin signaling and control appetite, food intake, and energy homeostasis, but the broader effect on gut hormones is largely unknown. A potent and efficacious GPR39 agonist (Cpd1324) was recently discovered, but the in vivo function was not addressed. Herein we studied the efficacy of the GPR39 agonist, Cpd1324, on metabolism and gut hormone secretion. METHODS Body weight, food intake, and energy expenditure in GPR39 agonist-treated mice and GPR39 KO mice were studied in calorimetric cages. Plasma ghrelin, glucose-dependent insulinotropic polypeptide (GIP), glucagon-like peptide-1 (GLP-1), and peptide YY (PYY) levels were measured. Organoids generated from murine and human small intestine and mouse colon were used to study GLP-1 and PYY release. Upon GPR39 agonist administration, dynamic changes in intracellular GLP-1 content were studied via immunostaining and changes in ion transport across colonic mucosa were monitored in Ussing chambers. The G protein activation underlying GPR39-mediated selective release of gut hormones was studied using bioluminescence resonance energy transfer biosensors. RESULTS The GPR39 KO mice displayed a significantly increased food intake without corresponding increases in respiratory exchange ratios or energy expenditure. Oral administration of a GPR39 agonist induced an acute decrease in food intake and subsequent weight loss in high-fat diet (HFD)-fed mice without affecting their energy expenditure. The tool compound, Cpd1324, increased GLP-1 secretion in the mice as well as in mouse and human intestinal organoids, but not in GPR39 KO mouse organoids. In contrast, the GPR39 agonist had no effect on PYY or GIP secretion. Transepithelial ion transport was acutely affected by GPR39 agonism in a GLP-1- and calcitonin gene-related peptide (CGRP)-dependent manner. Analysis of Cpd1324 signaling properties showed activation of Gαq and Gαi/o signaling pathways in L cells, but not Gαs signaling. CONCLUSIONS The GPR39 agonist described in this study can potentially be used by oral administration as a weight-lowering agent due to its stimulatory effect on GLP-1 secretion, which is most likely mediated through a unique activation of Gα subunits. Thus, GPR39 agonism may represent a novel approach to effectively treat obesity through selective modulation of gastrointestinal hormonal axes.
Collapse
Affiliation(s)
- Kaare V Grunddal
- Department of Medicine, University of California San Diego, La Jolla, CA 92037, USA
| | - Thi A Diep
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark; Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark
| | - Natalia Petersen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark
| | - Iain R Tough
- Wolfson Center for Age-Related Diseases, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, SE1 1UL, UK
| | - Louise J Skov
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark; Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark
| | - Lingzhi Liu
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark; Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark
| | - Jesse A Buijink
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark
| | - Franziska Mende
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark
| | - Chunyu Jin
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark
| | - Sara L Jepsen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark; Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark
| | - Louis M E Sørensen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2100, Denmark
| | - Michael P Achiam
- Department of Surgical Gastroenterology, Rigshospitalet, University of Copenhagen, Denmark
| | - Rune B Strandby
- Department of Surgical Gastroenterology, Rigshospitalet, University of Copenhagen, Denmark
| | - Anders Bach
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2100, Denmark
| | - Bolette Hartmann
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark
| | - Thomas M Frimurer
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark
| | - Siv A Hjorth
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark
| | - Michel Bouvier
- Department of Biochemistry and Molecular Medicine, Institute for Research in Immunology and Cancer, Molecular Pharmacology Research Unit, University of Montréal, Marcelle-Coutu Bureau Pavilion 1306-3, Montréal, QC H3T 1J4, Canada
| | - Helen Cox
- Wolfson Center for Age-Related Diseases, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, SE1 1UL, UK
| | - Birgitte Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark; Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark.
| |
Collapse
|
22
|
Milliken BT, Elfers C, Chepurny OG, Chichura KS, Sweet IR, Borner T, Hayes MR, De Jonghe BC, Holz GG, Roth CL, Doyle RP. Design and Evaluation of Peptide Dual-Agonists of GLP-1 and NPY2 Receptors for Glucoregulation and Weight Loss with Mitigated Nausea and Emesis. J Med Chem 2021; 64:1127-1138. [PMID: 33449689 PMCID: PMC7956155 DOI: 10.1021/acs.jmedchem.0c01783] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
![]()
There is a critical unmet need for
therapeutics to treat the epidemic
of comorbidities associated with obesity and type 2 diabetes, ideally
devoid of nausea/emesis. This study developed monomeric peptide agonists
of glucagon-like peptide 1 receptor (GLP-1R) and neuropeptide Y2 receptor
(Y2-R) based on exendin-4 (Ex-4) and PYY3–36. A
novel peptide, GEP44, was obtained via in vitro receptor
screens, insulin secretion in islets, stability assays, and in vivo rat and shrew studies of glucoregulation, weight
loss, nausea, and emesis. GEP44 in lean and diet-induced obese rats
produced greater reduction in body weight compared to Ex-4 without
triggering nausea associated behavior. Studies in the shrew demonstrated
a near absence of emesis for GEP44 in contrast to Ex-4. Collectively,
these data demonstrate that targeting GLP-1R and Y2-R with chimeric
single peptides offers a route to new glucoregulatory treatments that
are well-tolerated and have improved weight loss when compared directly
to Ex-4.
Collapse
Affiliation(s)
- Brandon T Milliken
- Department of Chemistry, Syracuse University, 111 College Place, Syracuse, New York 13244, United States
| | - Clinton Elfers
- Department of Pediatrics, Seattle Children's Hospital, University of Washington, Seattle, Washington 98105, United States
| | - Oleg G Chepurny
- Department of Medicine, State University of New York, Upstate Medical University, Syracuse, New York 13210, United States
| | - Kylie S Chichura
- Department of Chemistry, Syracuse University, 111 College Place, Syracuse, New York 13244, United States
| | - Ian R Sweet
- Diabetes Research Institute, University of Washington, Seattle, Washington 98105, United States
| | - Tito Borner
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Matthew R Hayes
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Bart C De Jonghe
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - George G Holz
- Department of Medicine, State University of New York, Upstate Medical University, Syracuse, New York 13210, United States
| | - Christian L Roth
- Department of Pediatrics, Seattle Children's Hospital, University of Washington, Seattle, Washington 98105, United States
| | - Robert P Doyle
- Department of Chemistry, Syracuse University, 111 College Place, Syracuse, New York 13244, United States.,Department of Medicine, State University of New York, Upstate Medical University, Syracuse, New York 13210, United States
| |
Collapse
|
23
|
Müller M, Ryoo MCK, Roura E. Gut sensing of dietary amino acids, peptides and proteins, and feed-intake regulation in pigs. ANIMAL PRODUCTION SCIENCE 2021. [DOI: 10.1071/an21185] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
24
|
Boer GA, Holst JJ. Incretin Hormones and Type 2 Diabetes-Mechanistic Insights and Therapeutic Approaches. BIOLOGY 2020; 9:biology9120473. [PMID: 33339298 PMCID: PMC7766765 DOI: 10.3390/biology9120473] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 12/09/2020] [Accepted: 12/10/2020] [Indexed: 02/06/2023]
Abstract
Simple Summary When we ingest a meal, our intestine secretes hormones that are released into the bloodstream. Amongst these hormones are the incretins hormones which stimulate the release of insulin from the pancreas which is essential for the regulation of in particular postprandial glucose concentrations. In patients with type 2 diabetes, the effect of the incretins is diminished. This is thought to contribute importantly to the pathophysiology of the disease. However, in pharmacological amounts, the incretins may still influence insulin secretion and metabolism. Much research has therefore been devoted to the development of incretin-based therapies for type 2 diabetes. These therapies include compounds that strongly resemble the incretins, hereby stimulating their effects as well as inhibitors of the enzymatic degradation of the hormones, thereby increasing the concentration of incretins in the blood. Both therapeutic approaches have been implemented successfully, but research is still ongoing aimed at the development of further optimized therapies. Abstract Glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) are secreted from the gut upon nutrient stimulation and regulate postprandial metabolism. These hormones are known as classical incretin hormones and are responsible for a major part of postprandial insulin release. The incretin effect is severely reduced in patients with type 2 diabetes, but it was discovered that administration of GLP-1 agonists was capable of normalizing glucose control in these patients. Over the last decades, much research has been focused on the development of incretin-based therapies for type 2 diabetes. These therapies include incretin receptor agonists and inhibitors of the incretin-degrading enzyme dipeptidyl peptidase-4. Especially the development of diverse GLP-1 receptor agonists has shown immense success, whereas studies of GIP monotherapy in patients with type 2 diabetes have consistently been disappointing. Interestingly, both GIP-GLP-1 co-agonists and GIP receptor antagonists administered in combination with GLP-1R agonists appear to be efficient with respect to both weight loss and control of diabetes, although the molecular mechanisms behind these effects remain unknown. This review describes our current knowledge of the two incretin hormones and the development of incretin-based therapies for treatment of type 2 diabetes.
Collapse
Affiliation(s)
- Geke Aline Boer
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark;
- NNF Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Jens Juul Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark;
- NNF Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark
- Correspondence: ; Tel.: +45-2875-7518
| |
Collapse
|
25
|
Maroni MJ, Capri KM, Cushman AV, Deane HV, Concepcion H, DeCourcey H, Seggio JA. The timing of fasting leads to different levels of food consumption and PYY 3-36 in nocturnal mice. Hormones (Athens) 2020; 19:549-558. [PMID: 32572709 DOI: 10.1007/s42000-020-00221-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 06/09/2020] [Indexed: 12/21/2022]
Abstract
PURPOSE The daily circadian cycle is known to modulate both feeding behavior and metabolism. As such, the timing of food consumption can play a role in regulating overall health. The purpose of this study is to determine whether fasting at different times of the day alters subsequent food consumption and levels of PYY3-36, a hormone secreted after a meal which inhibits appetite. METHODS Separate groups of mice were fasted at different times of the day: (1) start of the day, (2) middle of the day, (3) start of the night, and (4) middle of the night, and either injected with vehicle or PYY3-36 to assess their subsequent food consumption patterns, PYY3-36 levels, and glucose and insulin levels. We also investigated whether light exposure during the night would alter food consumption and PYY3-36 levels after fasting. RESULTS Mice fasted during the start of the daytime exhibited increased food consumption post-fast compared to mice fasted during the night. Injections of PYY3-36 during the night were more effective in reducing food consumption compared to PYY3-36 administration during the day. Constant light exposure suppressed food consumption after fasting and increased fasting PYY3-36 levels. CONCLUSIONS These results indicate that mice exhibit distinct food consumption patterns after being presented with a fast at different times of the day. Light exposure also modulates both food consumption after a fast and levels of PYY3-36.
Collapse
Affiliation(s)
- Marissa J Maroni
- Department of Biological Sciences, Bridgewater State University, 24 Park Ave., Bridgewater State University, Bridgewater, MA, 02325, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Kimberly M Capri
- Department of Biological Sciences, Bridgewater State University, 24 Park Ave., Bridgewater State University, Bridgewater, MA, 02325, USA
- Boston University, Boston, MA, 02215, USA
| | - Alexis V Cushman
- Department of Biological Sciences, Bridgewater State University, 24 Park Ave., Bridgewater State University, Bridgewater, MA, 02325, USA
| | - Hannah V Deane
- Department of Biological Sciences, Bridgewater State University, 24 Park Ave., Bridgewater State University, Bridgewater, MA, 02325, USA
| | - Holly Concepcion
- Department of Biological Sciences, Bridgewater State University, 24 Park Ave., Bridgewater State University, Bridgewater, MA, 02325, USA
| | - Holly DeCourcey
- Department of Biological Sciences, Bridgewater State University, 24 Park Ave., Bridgewater State University, Bridgewater, MA, 02325, USA
| | - Joseph A Seggio
- Department of Biological Sciences, Bridgewater State University, 24 Park Ave., Bridgewater State University, Bridgewater, MA, 02325, USA.
| |
Collapse
|
26
|
Lafferty RA, Tanday N, Flatt PR, Irwin N. Generation and characterisation of C-terminally stabilised PYY molecules with potential in vivo NPYR2 activity. Metabolism 2020; 111:154339. [PMID: 32777442 DOI: 10.1016/j.metabol.2020.154339] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 07/09/2020] [Accepted: 07/30/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND Activation of neuropeptide Y2 receptors (NPYR2) by the N-terminally truncated, dipeptidyl peptidase-4 (DPP-4) generated, Peptide YY (PYY) metabolite, namely PYY(3-36), results in satiating actions. However, PYY(3-36) is also subject to C-terminal enzymatic cleavage, which annuls anorectic effects. METHODS Substitution of l-Arg35 with d-Arg35 in the DPP-4 stable sea lamprey PYY(1-36) peptide imparts full C-terminal stability. In the current study, we have taken this molecule and introduced DPP-4 susceptibility by Iso3 substitution. RESULTS As expected, [Iso3]sea lamprey PYY(1-36) and [Iso3](d-Arg35)sea lamprey PYY(1-36) were N-terminally degraded to respective PYY(3-36) metabolites in plasma. Only [Iso3](d-Arg35)sea lamprey PYY(1-36) was C-terminally stable. Both peptides possessed similar insulinostatic and anti-apoptotic biological actions to native PYY(1-36) in beta-cells. Unlike native PYY(1-36) and [Iso3](d-Arg35)sea lamprey PYY(1-36), [Iso3]sea lamprey PYY(1-36) displayed some proliferative actions in Npyr1 knockout beta-cells. In addition, [Iso3]sea lamprey PYY(1-36) induced more rapid NPYR2-dependent appetite suppressive effects in mice than its C-terminally stable counterpart. Twice daily administration of either peptide to high fat fed (HFF) mice resulted in significant body weight reduction and improvements in circulating triglyceride levels. [Iso3]sea lamprey PYY(1-36) treatment also prevented elevations in glucagon. Both peptides, and especially [Iso3]sea lamprey PYY(1-36), improved glucose tolerance. The treatment interventions also partially reversed the deleterious effects of sustained high fat feeding on pancreatic islet morphology. CONCLUSION The present study confirms that sustained NPYR2 receptor activation by [Iso3](d-Arg35)sea lamprey induced significant weight lowering actions. However, identifiable benefits of this peptide over [Iso3]sea lamprey PYY(1-36), which was not protected against C-terminal degradation, were not pronounced.
Collapse
Affiliation(s)
- Ryan A Lafferty
- SAAD Centre for Pharmacy and Diabetes, University of Ulster, Coleraine, Northern Ireland BT52 1SA, UK
| | - Neil Tanday
- SAAD Centre for Pharmacy and Diabetes, University of Ulster, Coleraine, Northern Ireland BT52 1SA, UK
| | - Peter R Flatt
- SAAD Centre for Pharmacy and Diabetes, University of Ulster, Coleraine, Northern Ireland BT52 1SA, UK
| | - Nigel Irwin
- SAAD Centre for Pharmacy and Diabetes, University of Ulster, Coleraine, Northern Ireland BT52 1SA, UK.
| |
Collapse
|
27
|
Koliaki C, Liatis S, Dalamaga M, Kokkinos A. The Implication of Gut Hormones in the Regulation of Energy Homeostasis and Their Role in the Pathophysiology of Obesity. Curr Obes Rep 2020; 9:255-271. [PMID: 32647952 DOI: 10.1007/s13679-020-00396-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW This review provides an update on the role of gut hormones and their interactions in the regulation of energy homeostasis, describes gut hormone adaptations in obesity and in response to weight loss, and summarizes the current evidence on the role of gut hormone-based therapies for obesity treatment. RECENT FINDINGS Gut hormones play a key role in regulating eating behaviour, energy and glucose homeostasis. Dysregulated gut hormone responses have been proposed to be pathogenetically involved in the development and perpetuation of obesity. Summarizing the major gut hormone changes in obesity, obese individuals are characterized by blunted postprandial ghrelin suppression, loss of premeal ghrelin peaks, impaired diurnal ghrelin variability and reduced fasting and postprandial levels of anorexigenic peptides. Adaptive alterations of gut hormone levels are implicated in weight regain, thus complicating hypocaloric dietary interventions, and can further explain the profound weight loss and metabolic improvement following bariatric surgery. A plethora of compounds mimicking gut hormone changes after bariatric surgery are currently under investigation, introducing a new era in the pharmacotherapy of obesity. The current trend is to combine different gut hormone receptor agonists and target multiple systems simultaneously, in order to replicate as closely as possible the gut hormone milieu after bariatric surgery and circumvent the counter-regulatory adaptive changes associated with dietary energy restriction. An increasing number of preclinical and early-phase clinical trials reveal the additive benefits obtained with dual or triple gut peptide receptor agonists in reducing body weight and improving glycaemia. Gut hormones act as potent regulators of energy and glucose homeostasis. Therapeutic strategies targeting their levels or receptors emerge as a promising approach to treat patients with obesity and hyperglycaemia.
Collapse
Affiliation(s)
- Chrysi Koliaki
- First Department of Propaedeutic Internal Medicine, Medical School, Laiko General Hospital, National Kapodistrian University of Athens, 17 Agiou Thoma Street, 11527, Athens, Greece.
| | - Stavros Liatis
- First Department of Propaedeutic Internal Medicine, Medical School, Laiko General Hospital, National Kapodistrian University of Athens, 17 Agiou Thoma Street, 11527, Athens, Greece
| | - Maria Dalamaga
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Alexander Kokkinos
- First Department of Propaedeutic Internal Medicine, Medical School, Laiko General Hospital, National Kapodistrian University of Athens, 17 Agiou Thoma Street, 11527, Athens, Greece
| |
Collapse
|
28
|
Lafferty RA, Tanday N, McCloskey A, Bompada P, De Marinis Y, Flatt PR, Irwin N. Peptide YY (1-36) peptides from phylogenetically ancient fish targeting mammalian neuropeptide Y1 receptors demonstrate potent effects on pancreatic β-cell function, growth and survival. Diabetes Obes Metab 2020; 22:404-416. [PMID: 31692207 DOI: 10.1111/dom.13908] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 10/31/2019] [Accepted: 10/31/2019] [Indexed: 02/06/2023]
Abstract
AIM To investigate the antidiabetic efficacy of enzymatically stable Peptide YY (PYY) peptides from phylogenetically ancient fish. MATERIALS AND METHODS N-terminally stabilized, PYY (1-36) sequences from Amia calva (bowfin), Oncorhynchus mykiss (trout), Petromyzon marinus (sea lamprey) and Scaphirhynchus albus (sturgeon), were synthesized, and both biological actions and antidiabetic therapeutic efficacy were assessed. RESULTS All fish PYY (1-36) peptides were resistant to dipeptidyl peptidase-4 (DPP-4) degradation and inhibited glucose- and alanine-induced (P < 0.05 to P < 0.001) insulin secretion. In addition, PYY (1-36) peptides imparted significant (P < 0.05 to P < 0.001) β-cell proliferative and anti-apoptotic benefits. Proliferative effects were almost entirely absent in β cells with CRISPR-Cas9-induced knockout of Npyr1. In contrast to human PYY (1-36), the piscine-derived peptides lacked appetite-suppressive actions. Twice-daily administration of sea lamprey PYY (1-36), the superior bioactive peptide, for 21 days significantly (P < 0.05 to P < 0.001) decreased fluid intake, non-fasting glucose and glucagon in streptozotocin (STZ)-induced diabetic mice. In addition, glucose tolerance, insulin sensitivity, pancreatic insulin and glucagon content were significantly improved. Metabolic benefits were linked to positive changes in pancreatic islet morphology as a result of augmented (P < 0.001) proliferation and decreased apoptosis of β cells. Sturgeon PYY (1-36) exerted similar but less impressive effects in STZ mice. CONCLUSION These observations reveal, for the first time, that PYY (1-36) peptide sequences from phylogenetically ancient fish replicate the pancreatic β-cell benefits of human PYY (1-36) and have clear potential for the treatment of type 2 diabetes.
Collapse
Affiliation(s)
- Ryan A Lafferty
- SAAD Centre for Pharmacy and Diabetes, Biomedical Sciences Research Institute, University of Ulster, Coleraine, UK
| | - Neil Tanday
- SAAD Centre for Pharmacy and Diabetes, Biomedical Sciences Research Institute, University of Ulster, Coleraine, UK
| | - Andrew McCloskey
- SAAD Centre for Pharmacy and Diabetes, Biomedical Sciences Research Institute, University of Ulster, Coleraine, UK
| | - Pradeep Bompada
- Genomics, Diabetes and Endocrinology, Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - Yang De Marinis
- Genomics, Diabetes and Endocrinology, Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - Peter R Flatt
- SAAD Centre for Pharmacy and Diabetes, Biomedical Sciences Research Institute, University of Ulster, Coleraine, UK
| | - Nigel Irwin
- SAAD Centre for Pharmacy and Diabetes, Biomedical Sciences Research Institute, University of Ulster, Coleraine, UK
| |
Collapse
|
29
|
Kim TY, Shoback DM, Black DM, Rogers SJ, Stewart L, Carter JT, Posselt AM, King NJ, Schafer AL. Increases in PYY and uncoupling of bone turnover are associated with loss of bone mass after gastric bypass surgery. Bone 2020; 131:115115. [PMID: 31689523 PMCID: PMC6930344 DOI: 10.1016/j.bone.2019.115115] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 10/07/2019] [Accepted: 10/17/2019] [Indexed: 02/08/2023]
Abstract
CONTEXT The gut hormones peptide YY (PYY) and ghrelin mediate in part the metabolic benefits of Roux-en-Y gastric bypass (RYGB) surgery. However, preclinical data suggest these hormones also affect the skeleton and could contribute to postoperative bone loss. OBJECTIVE We investigated whether changes in fasting serum total PYY and ghrelin were associated with bone turnover marker levels and loss of bone mineral density (BMD) after RYGB. DESIGN, SETTING, PARTICIPANTS Prospective cohort of adults undergoing RYGB (n=44) at San Francisco academic hospitals. MAIN OUTCOME MEASURES We analyzed 6-month changes in PYY, ghrelin, bone turnover markers, and BMD by dual-energy x-ray absorptiometry (DXA) and quantitative computed tomography (QCT). We calculated the uncoupling index (UI), reflecting the relative balance of bone resorption and formation. RESULTS Postoperatively, there was a trend for an increase in PYY (+25pg/mL, p=0.07) and a significant increase in ghrelin (+192pg/mL, p<0.01). PYY changes negatively correlated with changes in spine BMD by QCT (r=-0.36, p=0.02) and bone formation marker P1NP (r=-0.30, p=0.05). Relationships were significant after adjustments for age, sex, and weight loss. No consistent relationships were found between ghrelin and skeletal outcomes. Mean 6-month UI was -3.3; UI correlated with spine BMD loss by QCT (r=0.40, p=0.01). CONCLUSIONS Postoperative PYY increases were associated with attenuated increases in P1NP and greater declines in spine BMD by QCT. Uncoupling of bone turnover correlated with BMD loss. These findings suggest a role for PYY in loss of bone mass after RYGB and highlight the relationship between intestinal and skeletal metabolism.
Collapse
Affiliation(s)
- Tiffany Y Kim
- Departments of Medicine, University of California, San Francisco, San Francisco, USA; Endocrine Research Unit, San Francisco VA Health Care System, San Francisco, USA.
| | - Dolores M Shoback
- Departments of Medicine, University of California, San Francisco, San Francisco, USA; Endocrine Research Unit, San Francisco VA Health Care System, San Francisco, USA
| | - Dennis M Black
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, USA
| | - Stanley J Rogers
- Department of Surgery, University of California, San Francisco, San Francisco, USA
| | - Lygia Stewart
- Department of Surgery, University of California, San Francisco, San Francisco, USA; Surgical Services, San Francisco VA Health Care System, San Francisco, USA
| | - Jonathan T Carter
- Department of Surgery, University of California, San Francisco, San Francisco, USA
| | - Andrew M Posselt
- Department of Surgery, University of California, San Francisco, San Francisco, USA
| | - Nicole J King
- Departments of Medicine, University of California, San Francisco, San Francisco, USA; Endocrine Research Unit, San Francisco VA Health Care System, San Francisco, USA
| | - Anne L Schafer
- Departments of Medicine, University of California, San Francisco, San Francisco, USA; Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, USA; Endocrine Research Unit, San Francisco VA Health Care System, San Francisco, USA
| |
Collapse
|
30
|
Jain AK, le Roux CW, Puri P, Tavakkoli A, Gletsu-Miller N, Laferrère B, Kellermayer R, DiBaise JK, Martindale RG, Wolfe BM. Proceedings of the 2017 ASPEN Research Workshop-Gastric Bypass: Role of the Gut. JPEN J Parenter Enteral Nutr 2019; 42:279-295. [PMID: 29443403 DOI: 10.1002/jpen.1121] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 11/16/2017] [Indexed: 12/11/2022]
Abstract
The goal of the National Institutes of Health-funded American Society for Parenteral and Enteral Nutrition 2017 research workshop (RW) "Gastric Bypass: Role of the Gut" was to focus on the exciting research evaluating gut-derived signals in modulating outcomes after bariatric surgery. Although gastric bypass surgery has undoubted positive effects, the mechanistic basis of improved outcomes cannot be solely explained by caloric restriction. Emerging data suggest that bile acid metabolic pathways, luminal contents, energy balance, gut mucosal integrity, as well as the gut microbiota are significantly modulated after bariatric surgery and may be responsible for the variable outcomes, each of which was rigorously evaluated. The RW served as a timely and novel academic meeting that brought together clinicians and researchers across the scientific spectrum, fostering a unique venue for interdisciplinary collaboration among investigators. It promoted engaging discussion and evolution of new research hypotheses and ideas, driving the development of novel ameliorative, therapeutic, and nonsurgical interventions targeting obesity and its comorbidities. Importantly, a critical evaluation of the current knowledge regarding gut-modulated signaling after bariatric surgery, potential pitfalls, and lacunae were thoroughly addressed.
Collapse
Affiliation(s)
- Ajay Kumar Jain
- Department of Pediatrics, SSM Cardinal Glennon Children's Medical Center, Saint Louis University School of Medicine, Saint Louis, Missouri, USA
| | - Carel W le Roux
- Diabetes Complications Research Center, University College Dublin, School of Medicine, Dublin, Ireland
| | - Puneet Puri
- Division of Gastroenterology, Hepatology and Nutrition, Virginia Commonwealth University, Richmond, Vieginia, USA
| | - Ali Tavakkoli
- Brigham and Women's Hospital, Center for Weight Management and Metabolic Surgery, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Blandine Laferrère
- Department of Medicine, Division of Endocrinology, Columbia University, New York, New York, USA
| | | | - John K DiBaise
- Division of Gastroenterology and Hepatology, Mayo Clinic, Phoenix, Arizona, USA
| | | | - Bruce M Wolfe
- Oregon Health and Science University, Portland, Oregon, USA
| |
Collapse
|
31
|
Sanches E, Timmermans M, Topal B, Celik A, Sundbom M, Ribeiro R, Parmar C, Ugale S, Proczko M, Stepaniak PS, Pujol Rafols J, Mahawar K, Buise MP, Neimark A, Severin R, Pouwels S. Cardiac remodeling in obesity and after bariatric and metabolic surgery; is there a role for gastro-intestinal hormones? Expert Rev Cardiovasc Ther 2019; 17:771-790. [PMID: 31746657 DOI: 10.1080/14779072.2019.1690991] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction: Obesity is associated with various diseases such as type 2 diabetes, hypertension, obstructive sleep apnea syndrome (OSAS), metabolic syndrome, and cardiovascular diseases. It affects several organ systems, including the pulmonary and cardiac systems. Furthermore, it induces pulmonary and cardiac changes that can result in right and/or left heart failure.Areas covered: In this review, authors provide an overview of obesity and cardiovascular remodeling, the individual actions of the gut hormones (like GLP-1 and PYY), the effects after bariatric/metabolic surgery and its influence on cardiac remodeling. In this review, we focussed and searched for literature in Pubmed and The Cochrane library (from the earliest date until April 2019), regarding cardiac function changes before and after bariatric surgery and literature regarding changes in gastrointestinal hormones.Expert opinion: Regarding the surgical treatment of obesity and metabolic diseases there is recognition of the importance of both weight loss (bariatric surgery) and improvement in metabolic milieu (metabolic surgery). A growing body of evidence further suggests that bariatric surgical procedures [like the Sleeve Gastrectomy (SG), Roux-en Y Gastric Bypass (RYGB), or One Anastomosis Gastric Bypass (OAGB)] have can improve outcomes of patients suffering from a number of cardiovascular diseases, including heart failure.
Collapse
Affiliation(s)
- Elijah Sanches
- Department of Surgery, Haaglanden Medical Center, The Hague, The Netherlands
| | - Marieke Timmermans
- Department of Surgery, Haaglanden Medical Center, The Hague, The Netherlands
| | - Besir Topal
- Department of Cardiothoracic Surgery, Onze Lieve Vrouwe Gasthuis, Amsterdam, The Netherlands
| | - Alper Celik
- Department of Bariatric and Metabolic Surgery, Metabolic Surgery Clinic, Sisli, Turkey
| | - Magnus Sundbom
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Rui Ribeiro
- Centro Multidisciplinar da Doença Metabólica, Clínica de Santo António, Lisbon, Portugal
| | - Chetan Parmar
- Department of Surgery, Whittington Hospital, London, UK
| | - Surendra Ugale
- Bariatric & Metabolic Surgery Clinic, Kirloskar Hospital, Hyderabad, India
| | - Monika Proczko
- Department of General, Endocrine and Transplant Surgery, University Medical Center, Gdansk University, Gdansk, Poland
| | - Pieter S Stepaniak
- Department of Operating Rooms, Catharina Hospital, Eindhoven, The Netherlands
| | | | - Kamal Mahawar
- Bariatric Unit, Sunderland Royal Hospital, Sunderland, UK
| | - Marc P Buise
- Department of Anesthesiology, Intensive Care and Pain Medicine, Catharina Hospital, Eindhoven, The Netherlands
| | - Aleksandr Neimark
- Department of Surgery, Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - Rich Severin
- Department of Physical Therapy, College of Applied Health Sciences, University of Illinois at Chicago, Chicago, IL, USA.,Doctor of Physical Therapy Program, Robbins College of Health and Human Sciences, Baylor University, Waco, TX, USA
| | - Sjaak Pouwels
- Department of Surgery, Haaglanden Medical Center, The Hague, The Netherlands
| |
Collapse
|
32
|
Kokkinos A, Tsilingiris D, le Roux CW, Rubino F, Mantzoros CS. Will medications that mimic gut hormones or target their receptors eventually replace bariatric surgery? Metabolism 2019; 100:153960. [PMID: 31412266 DOI: 10.1016/j.metabol.2019.153960] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 07/31/2019] [Accepted: 08/05/2019] [Indexed: 02/07/2023]
Abstract
Bariatric surgery is currently the most effective therapeutic modality through which sustained beneficial effects on weight loss and metabolic improvement are achieved. During recent years, indications for bariatric surgery have been expanded to include cases of poorly controlled type 2 (T2DM) diabetes mellitus in lesser extremes of body weight. A spectrum of the beneficial effects of surgery is attributed to robust changes of postprandial gut peptide responses that are observed post operatively. Consolidated knowledge regarding gut peptide physiology as well as emerging new evidence shedding light on the mode of action of previously overlooked gut hormones provide appealing potential obesity and T2DM therapeutic perspectives. The accumulation of evidence from the effect of exogenous administration of native gut peptides alone or in combinations to humans as well as the development of mimetic agents exerting agonistic effects on combinations of gut hormone receptors pave the way for future integrated gut peptide-based treatments, which may mimic the effects of bariatric surgery.
Collapse
Affiliation(s)
- Alexander Kokkinos
- First Department of Propaedeutic Internal Medicine, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece.
| | - Dimitrios Tsilingiris
- First Department of Propaedeutic Internal Medicine, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
| | - Carel W le Roux
- Diabetes Complications Research Centre, University College Dublin, Dublin, Ireland
| | - Francesco Rubino
- Department of Metabolic and Bariatric Surgery, Diabetes and Nutritional Science Division, King's College Hospital, London, United Kingdom
| | - Christos S Mantzoros
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, USA
| |
Collapse
|
33
|
Holst JJ, Albrechtsen NJW, Rosenkilde MM, Deacon CF. Physiology of the Incretin Hormones,
GIP
and
GLP
‐1—Regulation of Release and Posttranslational Modifications. Compr Physiol 2019; 9:1339-1381. [DOI: 10.1002/cphy.c180013] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
34
|
Boutari C, Polyzos SA, Mantzoros CS. Of mice and men: Why progress in the pharmacological management of obesity is slower than anticipated and what could be done about it? Metabolism 2019; 96:vi-xi. [PMID: 30910448 DOI: 10.1016/j.metabol.2019.03.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 03/16/2019] [Indexed: 01/02/2023]
Affiliation(s)
- Chrysoula Boutari
- Second Propedeutic Department of Internal Medicine, Faculty of Medicine, Aristotle University, Hippokration Hospital, Thessaloniki, Greece; Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Stergios A Polyzos
- First Department of Pharmacology, Faculty of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Christos S Mantzoros
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA; Section of Endocrinology, Boston VA Healthcare System, Boston, MA, USA.
| |
Collapse
|
35
|
Lafferty RA, Gault VA, Flatt PR, Irwin N. Effects of 2 Novel PYY(1-36) Analogues, (P 3L 31P 34)PYY(1-36) and PYY(1-36)(Lys 12PAL), on Pancreatic Beta-Cell Function, Growth, and Survival. CLINICAL MEDICINE INSIGHTS-ENDOCRINOLOGY AND DIABETES 2019; 12:1179551419855626. [PMID: 31244528 PMCID: PMC6580715 DOI: 10.1177/1179551419855626] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 05/15/2019] [Indexed: 12/14/2022]
Abstract
Recent studies have identified a beneficial role for peptide tyrosine tyrosine
(PYY) on pancreatic beta-cell function and survival. These effects are linked to
the activation of neuropeptide Y1 receptors (NPYR1s) by PYY(1-36). However,
PYY(1-36) is subject to rapid degradation by dipeptidyl peptidase-4 (DPP-4),
resulting is the loss of NPYR1 activity. Therefore, the aim of this study was to
develop 2 enzymatically stable PYY(1-36) analogues, namely,
(P3L31P34)PYY(1-36) and
PYY(1-36)(Lys12PAL), with further structural modifications to
enhance NPYR1 specificity. As expected,
(P3L31P34)PYY(1-36) was fully resistant to
DPP-4-mediated degradation in vitro, whereas PYY(1-36) and
PYY(1-36)(Lys12PAL) were both liable to DPP-4 breakdown.
PYY(1-36) and (P3L31P34)PYY(1-36) induced
significant reductions in glucose-stimulated insulin secretion (GSIS) from BRIN
BD11 cells, but only PYY(1-36) diminished alanine-stimulated insulin secretion.
In contrast, PYY(1-36)(Lys12PAL) had no impact on GSIS or
alanine-induced insulin release. All 3 PYY peptides significantly enhanced
proliferation in BRIN BD11 and 1.1B4 beta-cell lines, albeit only at the highest
concentration examined, 10-6 M, for
(P3L31P34)PYY(1-36) and
PYY(1-36)(Lys12PAL) in BRIN BD11 cells. Regarding the protection
of beta-cells against cytokine-induced apoptosis, PYY(1-36) induced clear
protective effects. Both (P3L31P34)PYY(1-36)
and PYY(1-36)(Lys12PAL) offered some protection against apoptosis in
BRIN BD11 cells, but were significantly less efficacious than PYY(1-36).
Similarly, in 1.1B4 cells, both PYY analogues (10-6 M) protected
against cytokine-induced apoptosis, but
(P3L31P34)PYY(1-36) was significantly less
effective than PYY(1-36). All 3 PYY peptides had no impact on refeeding in
overnight fasted mice. These data underline the beta-cell benefits of PYY(1-36)
and highlight the challenges of synthesising stable, bioactive, NPYR1-specific,
PYY(1-36) analogues.
Collapse
Affiliation(s)
- Ryan A Lafferty
- SAAD Centre for Pharmacy and Diabetes, University of Ulster, Coleraine, UK.,Diabetes Research Group, University of Ulster, Coleraine, UK
| | - Victor A Gault
- SAAD Centre for Pharmacy and Diabetes, University of Ulster, Coleraine, UK
| | - Peter R Flatt
- SAAD Centre for Pharmacy and Diabetes, University of Ulster, Coleraine, UK
| | - Nigel Irwin
- SAAD Centre for Pharmacy and Diabetes, University of Ulster, Coleraine, UK
| |
Collapse
|
36
|
Shao J, Chen MJ, Kuehl PJ, Hochhaus G. Pharmacokinetic and pharmacodynamic modeling of gut hormone peptide YY (3-36) after pulmonary delivery. Drug Dev Ind Pharm 2019; 45:1101-1110. [PMID: 31039626 DOI: 10.1080/03639045.2019.1593443] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Peptide YY(3-36) (PYY(3-36)) is an endogenous appetite suppressing peptide. The present research was to perform pharmacokinetic/pharmacodynamic (PK/PD) analysis for predicting the concentration- and response-time profiles of PYY(3-36) after systemic and pulmonary delivery in mice, with the goal of suggesting a potential pulmonary dosing regimen in humans. A PK/PD model was developed to describe PYY(3-36) plasma concentration - and relative food intake rate ratio (as % of control) - time profiles after intraperitoneal and subcutaneous administration, and inhalation in mice. The absorption of inhaled PYY(3-36) from the lungs of mice could only be described with a combined slow (absorption rate of 0.147 L/h) and fast (absorption rate of 104.4 L/h) absorption process, presumably related to absorption from the central and peripheral regions of the lungs. The estimates for IC50 and Imax were 6.8 ng/mL and 63.5%, respectively, based on inhibitory Emax model. The PK parameters, such as clearance (CL), volume of distribution at steady state (Vdss), and the absorption rates (ka), were then scaled to human's. The scaled human CL and Vdss for obese subjects were 24.8 L/h and 9.0 L, respectively. The model predicted human plasma PYY(3-36) concentrations agreed reasonably well with placebo-normalized plasma PYY(3-36) concentrations after short-term infusion and SC injection in literature. An inhalation dose of PYY(3-36) of about 100 µg was proposed for obese subjects based on simulations. This PK/PD analysis satisfactorily described PYY(3-36) concentration-time and relative food intake rate ratio- time profiles at all doses and routes. The developed model might facilitate the inhalation dose selection of PYY(3-36).
Collapse
Affiliation(s)
- Jie Shao
- a Department of Pharmaceutics, College of Pharmacy , University of Florida , Gainesville , Florida , USA
| | - Mong-Jen Chen
- a Department of Pharmaceutics, College of Pharmacy , University of Florida , Gainesville , Florida , USA
| | - Philip J Kuehl
- b Lovelace Respiratory Research Institute , Albuquerque , New Mexico , USA
| | - Guenther Hochhaus
- a Department of Pharmaceutics, College of Pharmacy , University of Florida , Gainesville , Florida , USA
| |
Collapse
|
37
|
Changes in Energy Expenditure of Patients with Obesity Following Bariatric Surgery: a Systematic Review of Prospective Studies and Meta-analysis. Obes Surg 2019; 29:2318-2337. [PMID: 31016456 DOI: 10.1007/s11695-019-03851-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
We herein summarize the available literature on the effects of bariatric surgery (BS) on energy expenditure in individuals with obesity. We conducted a systematic literature review, and 35 prospective studies met our inclusion criteria. The findings indicate that BS contributes to increased diet-induced thermogenesis (DIT) and decreased total energy expenditure (TEE) and resting energy expenditure (REE) in patients with obesity. The meta-analysis demonstrated a significant decrease in TEE and REE within 6 months following BS. With the sustained decrease in REE, there was no further decrease in TEE between the 6- and 12-month follow-up. Increased DIT might explain the variance between the patterns of REE and TEE change. The postoperative decrease in REE/FFM and increase in REE/BW were observed. The changes in substrate utilization might be consistent with the change in the respiration quotient postoperatively.
Collapse
|
38
|
Schiellerup SP, Skov-Jeppesen K, Windeløv JA, Svane MS, Holst JJ, Hartmann B, Rosenkilde MM. Gut Hormones and Their Effect on Bone Metabolism. Potential Drug Therapies in Future Osteoporosis Treatment. Front Endocrinol (Lausanne) 2019; 10:75. [PMID: 30863364 PMCID: PMC6399108 DOI: 10.3389/fendo.2019.00075] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 01/28/2019] [Indexed: 12/19/2022] Open
Abstract
Bone homeostasis displays a circadian rhythm with increased resorption during the night time as compared to day time, a difference that seems-at least partly-to be caused by food intake during the day. Thus, ingestion of a meal results in a decrease in bone resorption, but people suffering from short bowel syndrome lack this response. Gut hormones, released in response to a meal, contribute to this link between the gut and bone metabolism. The responsible hormones appear to include glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-1 (GLP-1), known as incretin hormones due to their role in regulating glucose homeostasis by enhancing insulin release in response to food intake. They interact with their cognate receptors (GIPR and GLP-1R), which are both members of the class B G protein-coupled receptors (GPCRs), and already recognized as targets for treatment of metabolic diseases, such as type 2 diabetes mellitus (T2DM) and obesity. Glucagon-like peptide-2 (GLP-2), secreted concomitantly with GLP-1, acting via another class B receptor (GLP-2R), is also part of this gut-bone axis. Several studies, including human studies, have indicated that these three hormones inhibit bone resorption and, moreover, that GIP increases bone formation. Another hormone, peptide YY (PYY), is also secreted from the enteroendocrine L-cells (together with GLP-1 and GLP-2), and acts mainly via interaction with the class A GPCR NPY-R2. PYY is best known for its effect on appetite regulation, but recent studies have also shown an effect of PYY on bone metabolism. The aim of this review is to summarize the current knowledge of the actions of GIP, GLP-1, GLP-2, and PYY on bone metabolism, and to discuss future therapies targeting these receptors for the treatment of osteoporosis.
Collapse
Affiliation(s)
- Sine Paasch Schiellerup
- Laboratory of Molecular Pharmacology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kirsa Skov-Jeppesen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Faculty of Health and Medical Sciences, Novo Nordisk Foundation (NNF) Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Johanne Agerlin Windeløv
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Faculty of Health and Medical Sciences, Novo Nordisk Foundation (NNF) Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Maria Saur Svane
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens Juul Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Faculty of Health and Medical Sciences, Novo Nordisk Foundation (NNF) Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Bolette Hartmann
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Faculty of Health and Medical Sciences, Novo Nordisk Foundation (NNF) Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Mette Marie Rosenkilde
- Laboratory of Molecular Pharmacology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
39
|
Di Domenico M, Pinto F, Quagliuolo L, Contaldo M, Settembre G, Romano A, Coppola M, Ferati K, Bexheti-Ferati A, Sciarra A, Nicoletti GF, Ferraro GA, Boccellino M. The Role of Oxidative Stress and Hormones in Controlling Obesity. Front Endocrinol (Lausanne) 2019; 10:540. [PMID: 31456748 PMCID: PMC6701166 DOI: 10.3389/fendo.2019.00540] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 07/19/2019] [Indexed: 12/15/2022] Open
Abstract
The accumulation of adipose tissue in the body occurs because the energy introduced with food and drink exceeds that expense, but to understand why this imbalance is established and why it is maintained over time, it is important to consider the main causes and risk factors of excess weight. In this review, we will refer to the main factors linked to obesity, starting from oxidative stress to hormonal factors including the role of obesity in breast cancer. Among the many hypotheses formulated on the etiopathology of obesity, a key role can be attributed to the relationship between stress oxidative and intestinal microbiota. Multiple evidences tend to show that genetic, epigenetic, and lifestyle factors contribute to determine in the obese an imbalance of the redox balance correlated with the alteration of the intestinal microbial flora. Obesity acts negatively on the wound healing, in fact several studies indicate morbid obesity significantly increased the risk of a post-operative wound complication and infection. Currently, in the treatment of obesity, medical interventions are aimed not only at modifying caloric intake, but also to modulate and improve the composition of diet with the aim of rebalancing the microbiota-redox state axis.
Collapse
Affiliation(s)
- Marina Di Domenico
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
- Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA, United States
| | - Federica Pinto
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Lucio Quagliuolo
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Maria Contaldo
- Multidisciplinary Department of Medical-Surgical and Dental Specialties, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Giuliana Settembre
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Antonio Romano
- Multidisciplinary Department of Medical-Surgical and Dental Specialties, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Mario Coppola
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Kenan Ferati
- Faculty of Medicine, University of Tetovo, Tetovo, Macedonia
| | | | - Antonella Sciarra
- Department of Translational Medicad Sciences, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Giovanni Francesco Nicoletti
- Plastic Surgery Unit, Multidisciplinary Department of Medical-Surgical and Dental Specialties, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Giuseppe Andrea Ferraro
- Plastic Surgery Unit, Multidisciplinary Department of Medical-Surgical and Dental Specialties, University of Campania Luigi Vanvitelli, Naples, Italy
- *Correspondence: Giuseppe Andrea Ferraro
| | | |
Collapse
|
40
|
Veedfald S, Wu T, Bound M, Grivell J, Hartmann B, Rehfeld JF, Deacon CF, Horowitz M, Holst JJ, Rayner CK. Hyperosmolar Duodenal Saline Infusion Lowers Circulating Ghrelin and Stimulates Intestinal Hormone Release in Young Men. J Clin Endocrinol Metab 2018; 103:4409-4418. [PMID: 30053031 DOI: 10.1210/jc.2018-00699] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 07/17/2018] [Indexed: 02/07/2023]
Abstract
CONTEXT The mechanisms regulating the postprandial suppression of ghrelin secretion remain unclear, but recent observations in rats indicate that an increase in duodenal osmolarity is associated with a reduction in ghrelin levels. Several hormones have been implicated in the regulation of ghrelin. OBJECTIVE We hypothesized that intraduodenal infusion of a hyperosmolar solution would lower plasma ghrelin concentrations. DESIGN, SETTING, PARTICIPANTS, AND INTERVENTIONS Eighteen healthy young men were studied after an overnight fast on two occasions in a randomized double-blinded fashion. A nasoduodenal catheter was positioned and isoosmolar (300 mOsm/L) or hyperosmolar (1500 mOsm/L) saline was infused intraduodenally (4 mL/min, t = 0 to 45 minutes). Venous blood was sampled at t = -45, -30, -15, 0, 15, 30, 45, 60, 75, 90, 120, and 180 minutes. MAIN OUTCOME MEASURES Plasma concentrations of ghrelin, glucagonlike peptide-1 (GLP-1), glucose-dependent insulinotropic polypeptide (GIP), cholecystokinin (CCK), glucagon, pancreatic polypeptide (PP), neurotensin (NT), peptide YY (PYY), motilin, and glucose. RESULTS Ghrelin concentrations were suppressed with hyperosmolar when compared with isoosmolar saline, and remained lower until t = 180 minutes. CCK, NT, GLP-1, PYY, and glucagon all increased during hyperosmolar, but not isoosmolar, saline infusion (P < 0.01 for all), whereas GIP, PP, and motilin levels were not affected by either infusion. CONCLUSIONS Plasma ghrelin concentrations are lowered, whereas CCK, GLP-1, PYY, NT, and glucagon concentrations are augmented, by hyperosmolar duodenal content in healthy individuals. These observations have implications for the evaluation of studies comparing the effects of different types and loads of nutrients and chemicals on gut hormone secretion.
Collapse
Affiliation(s)
- Simon Veedfald
- Discipline of Medicine and National Health and Medical Research Council Centre of Research Excellence in Nutritional Physiology, Interventions and Outcomes, University of Adelaide, Adelaide, South Australia, Australia
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Tongzhi Wu
- Discipline of Medicine and National Health and Medical Research Council Centre of Research Excellence in Nutritional Physiology, Interventions and Outcomes, University of Adelaide, Adelaide, South Australia, Australia
| | - Michelle Bound
- Discipline of Medicine and National Health and Medical Research Council Centre of Research Excellence in Nutritional Physiology, Interventions and Outcomes, University of Adelaide, Adelaide, South Australia, Australia
| | - Jacqueline Grivell
- Discipline of Medicine and National Health and Medical Research Council Centre of Research Excellence in Nutritional Physiology, Interventions and Outcomes, University of Adelaide, Adelaide, South Australia, Australia
| | - Bolette Hartmann
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Jens F Rehfeld
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen, Denmark
| | - Carolyn F Deacon
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Michael Horowitz
- Discipline of Medicine and National Health and Medical Research Council Centre of Research Excellence in Nutritional Physiology, Interventions and Outcomes, University of Adelaide, Adelaide, South Australia, Australia
| | - Jens J Holst
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Christopher K Rayner
- Discipline of Medicine and National Health and Medical Research Council Centre of Research Excellence in Nutritional Physiology, Interventions and Outcomes, University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
41
|
Gut adaptation after metabolic surgery and its influences on the brain, liver and cancer. Nat Rev Gastroenterol Hepatol 2018; 15:606-624. [PMID: 30181611 DOI: 10.1038/s41575-018-0057-y] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Metabolic surgery is the best treatment for long-term weight loss maintenance and comorbidity control. Metabolic operations were originally intended to change anatomy to alter behaviour, but we now understand that the anatomical changes can modulate physiology to change behaviour. They are no longer considered only mechanically restrictive and/or malabsorptive procedures; rather, they are considered metabolic procedures involving complex physiological changes, whereby gut adaptation influences signalling pathways in several other organs, including the liver and the brain, regulating hunger, satiation, satiety, body weight, glucose metabolism and immune functions. The integrative physiology of gut adaptation after these operations consists of a complex mechanistic web of communication between gut hormones, bile acids, gut microbiota, the brain and both enteric and central nervous systems. The understanding of nutrient sensing via enteroendocrine cells, the enteric nervous system, hypothalamic peptides and adipose tissue and of the role of inflammation has advanced our knowledge of this integrative physiology. In this Review, we focus on the adaptation of gut physiology to the anatomical alterations from Roux-en-Y gastric bypass and vertical sleeve gastrectomy and the influence of these procedures on food intake, weight loss, nonalcoholic fatty liver disease (NAFLD) and cancer. We also aim to demonstrate the underlying mechanisms that could explain how metabolic surgery could be used as a therapeutic option in NAFLD and certain obesity-related cancers.
Collapse
|
42
|
Holst JJ, Madsbad S, Bojsen-Møller KN, Svane MS, Jørgensen NB, Dirksen C, Martinussen C. Mechanisms in bariatric surgery: Gut hormones, diabetes resolution, and weight loss. Surg Obes Relat Dis 2018; 14:708-714. [PMID: 29776493 PMCID: PMC5974695 DOI: 10.1016/j.soard.2018.03.003] [Citation(s) in RCA: 125] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 03/04/2018] [Indexed: 12/11/2022]
Abstract
Gastric bypass surgery leads to profound changes in the secretion of gut hormones with effects on metabolism, appetite, and food intake. Here, we discuss their contributions to the improvement in glucose tolerance and the weight loss that results from the operations. We find that the improved glucose tolerance is due the following events: a negative energy balance and resulting weight loss, which improve first hepatic and later peripheral insulin sensitivity, in combination with increased postprandial insulin secretion elicited particularly by exaggerated glucagon-like peptide-1 responses. The weight loss is due to loss of appetite resulting in reduced energy intake, and we find it probable that this process is driven by exaggerated secretion of appetite-regulating gut hormones including, but probably not limited to, glucagon-like peptide-1 and peptide-YY. The increased secretion is due to an accelerated exposure to and absorption of nutrients in the small intestine. This places the weight loss and the gut hormones in key positions with respect to the metabolic improvements after bypass surgery.
Collapse
Affiliation(s)
- Jens Juul Holst
- NNF Center for Basic Metabolic Research and Dept. Biomedical Sciences, the Panum Institute, University of Copenhagen, Copenhagen, Denmark.
| | - Sten Madsbad
- NNF Center for Basic Metabolic Research and Dept. Biomedical Sciences, the Panum Institute, University of Copenhagen, Copenhagen, Denmark; Department of Endocrinology, Hvidovre Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Kirstine N Bojsen-Møller
- NNF Center for Basic Metabolic Research and Dept. Biomedical Sciences, the Panum Institute, University of Copenhagen, Copenhagen, Denmark; Department of Endocrinology, Hvidovre Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Maria Saur Svane
- NNF Center for Basic Metabolic Research and Dept. Biomedical Sciences, the Panum Institute, University of Copenhagen, Copenhagen, Denmark; Department of Endocrinology, Hvidovre Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Nils Bruun Jørgensen
- NNF Center for Basic Metabolic Research and Dept. Biomedical Sciences, the Panum Institute, University of Copenhagen, Copenhagen, Denmark; Department of Endocrinology, Hvidovre Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Carsten Dirksen
- NNF Center for Basic Metabolic Research and Dept. Biomedical Sciences, the Panum Institute, University of Copenhagen, Copenhagen, Denmark; Department of Endocrinology, Hvidovre Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Christoffer Martinussen
- NNF Center for Basic Metabolic Research and Dept. Biomedical Sciences, the Panum Institute, University of Copenhagen, Copenhagen, Denmark; Department of Endocrinology, Hvidovre Hospital, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
43
|
Davis DB, Khoraki J, Ziemelis M, Sirinvaravong S, Han JY, Campos GM. Roux en Y gastric bypass hypoglycemia resolves with gastric feeding or reversal: Confirming a non-pancreatic etiology. Mol Metab 2018; 9:15-27. [PMID: 29449181 PMCID: PMC5869737 DOI: 10.1016/j.molmet.2017.12.011] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 12/14/2017] [Accepted: 12/18/2017] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVE Postprandial hypoglycemia is an infrequent but disabling complication of Roux-en-Y gastric bypass (RYGB) surgery. Controversy still exists as to whether the postprandial hyperinsulinemia observed is due to inherent changes in pancreatic β-cell mass or function or to reversible alterations caused by RYGB anatomy. We aimed to determine if gastric feeding or reversal of RYGB would normalize postprandial glucose and hormone excursions in patients with symptomatic hypoglycemia. METHODS We completed a prospective study of six patients with severe symptomatic RYGB hypoglycemia who underwent RYGB reversal. An additional subject without hypoglycemia who underwent RYGB reversal was also studied prospectively. Mixed meal tolerance testing (MTT) was done orally (RYGB anatomy), via gastrostomy tube in the excluded stomach in the setting of RYGB, and several months after RYGB reversal. RESULTS All subjects reported symptomatic improvement of hypoglycemia after reversal of RYGB. Weight gain after reversal was moderate and variable. Postprandial glucose, insulin, and GLP-1 excursions were significantly diminished with gastric feeding and after reversal. Insulin secretion changed proportional to glucose levels and insulin clearance increased after reversal. Glucagon/insulin ratios were similar throughout study. We further compared the impact of modified sleeve gastrectomy reversal surgery to those with restoration of complete stomach and found no significant differences in weight regain or in postprandial glucose or hormone levels. CONCLUSIONS Reversal of RYGB is an effective treatment option for severe postprandial hypoglycemia. The pathophysiology of this disorder is primarily due to RYGB anatomy resulting in altered glucose, gut, and pancreatic hormone levels and decreased insulin clearance, rather than inherent β-cell hyperplasia or hyperfunction.
Collapse
Affiliation(s)
- Dawn Belt Davis
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Wisconsin, 1685 Highland Avenue, Madison, WI, 53705, USA; William S. Middleton Memorial Veterans Hospital, 2500 Overlook Terrace, Madison, WI 53705, USA.
| | - Jad Khoraki
- Division of General Surgery, Department of Surgery, 600 Highland Avenue, University of Wisconsin, Madison, WI, 53705, USA; Division of Bariatric and Gastrointestinal Surgery, Department of Surgery, Virginia Commonwealth University, 1200 East Broad Street, Richmond, VA, 23298, USA
| | - Martynas Ziemelis
- Division of General Surgery, Department of Surgery, 600 Highland Avenue, University of Wisconsin, Madison, WI, 53705, USA
| | - Sirinart Sirinvaravong
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Wisconsin, 1685 Highland Avenue, Madison, WI, 53705, USA
| | - Jee Young Han
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Wisconsin, 1685 Highland Avenue, Madison, WI, 53705, USA
| | - Guilherme M Campos
- Division of General Surgery, Department of Surgery, 600 Highland Avenue, University of Wisconsin, Madison, WI, 53705, USA; Division of Bariatric and Gastrointestinal Surgery, Department of Surgery, Virginia Commonwealth University, 1200 East Broad Street, Richmond, VA, 23298, USA.
| |
Collapse
|
44
|
Lafferty RA, Flatt PR, Irwin N. Emerging therapeutic potential for peptide YY for obesity-diabetes. Peptides 2018; 100:269-274. [PMID: 29412828 DOI: 10.1016/j.peptides.2017.11.005] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Revised: 11/06/2017] [Accepted: 11/06/2017] [Indexed: 12/25/2022]
Abstract
The vast majority of research to date on the gut hormone Peptide YY (PYY) has focused on appetite suppression and body weight regulation effects. These biological actions are believed to occur through interaction of PYY with hypothalamic Y2 receptors. However, more recent studies have added additional knowledge to understanding of the physiological, and potential therapeutic, roles of PYY beyond obesity alone. Thus, PYY has now been shown to impart improvements in pancreatic beta-cell survival and function, with obvious benefits for diabetes. This effect has been linked mainly to binding and activation of Y1 receptors by PYY, but more evidence is still required in this regard. Given the potential therapeutic promise of PYY-derived compounds, and complexity of receptor interactions, it is important to fully understand the complete biological action profile of PYY. Therefore, the current review aims to compile, evaluate and summarise current knowledge on PYY, with particular emphasis on obesity and diabetes treatment, and the importance of specific Y receptor interactions for this.
Collapse
Affiliation(s)
- Ryan A Lafferty
- SAAD Centre for Pharmacy and Diabetes, University of Ulster, Coleraine, Northern Ireland, BT52 1SA, UK
| | - Peter R Flatt
- SAAD Centre for Pharmacy and Diabetes, University of Ulster, Coleraine, Northern Ireland, BT52 1SA, UK
| | - Nigel Irwin
- SAAD Centre for Pharmacy and Diabetes, University of Ulster, Coleraine, Northern Ireland, BT52 1SA, UK.
| |
Collapse
|
45
|
Sun EWL, Martin AM, Young RL, Keating DJ. The Regulation of Peripheral Metabolism by Gut-Derived Hormones. Front Endocrinol (Lausanne) 2018; 9:754. [PMID: 30662430 PMCID: PMC6328484 DOI: 10.3389/fendo.2018.00754] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 11/27/2018] [Indexed: 12/13/2022] Open
Abstract
Enteroendocrine cells lining the gut epithelium constitute the largest endocrine organ in the body and secrete over 20 different hormones in response to cues from ingested foods and changes in nutritional status. Not only do these hormones convey signals from the gut to the brain via the gut-brain axis, they also act directly on metabolically important peripheral targets in a highly concerted fashion to maintain energy balance and glucose homeostasis. Gut-derived hormones released during fasting tend to be orexigenic and have hyperglycaemic potential. Conversely, gut hormones secreted postprandially generally promote satiety and facilitate glucose clearance. Although some of the metabolic benefits conferred by bariatric surgeries have been ascribed to changes in the secretory profiles of various gut hormones, the therapeutic potential of the enteroendocrine system as a viable target against metabolic diseases remain largely underexploited, except for incretin-mimetics. This review provides a brief overview of the physiological importance and highlights the therapeutic potential of the following gut hormones: serotonin, glucose-dependent insulinotropic peptide, glucagon-like peptide 1, oxyntomodulin, peptide YY, insulin-like peptide 5, and ghrelin.
Collapse
Affiliation(s)
- Emily W. L. Sun
- College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Alyce M. Martin
- College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Richard L. Young
- Nutrition and Metabolism, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
| | - Damien J. Keating
- College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
- Nutrition and Metabolism, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
- *Correspondence: Damien J. Keating
| |
Collapse
|
46
|
Gut check on diabesity: leveraging gut mechanisms for the treatment of type 2 diabetes and obesity. Curr Opin Pharmacol 2017; 37:10-15. [PMID: 28802873 DOI: 10.1016/j.coph.2017.07.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 07/20/2017] [Indexed: 01/20/2023]
Abstract
Gut hormones have long been understood to regulate food intake and metabolism. Bariatric surgery significantly elevates circulating gut hormone levels and is proven to affect acute remission of type 2 diabetes before any weight loss is observed. Subsequent weight loss is accrued over weeks to months but is sustained into the long term. Hence, there exists great enthusiasm to recapitulate these changes in gut hormones in the form of novel combination drugs for type 2 diabetes and obesity.
Collapse
|
47
|
Abstract
AbstractA better understanding of the factors that influence eating behaviour is of importance as our food choices are associated with the risk of developing chronic diseases such as obesity, CVD, type 2 diabetes or some forms of cancer. In addition, accumulating evidence suggests that the industrial food production system is a major contributor to greenhouse gas emission and may be unsustainable. Therefore, our food choices may also contribute to climate change. By identifying the factors that influence eating behaviour new interventions may be developed, at the individual or population level, to modify eating behaviour and contribute to society’s health and environmental goals. Research indicates that eating behaviour is dictated by a complex interaction between physiology, environment, psychology, culture, socio-economics and genetics that is not fully understood. While a growing body of research has identified how several single factors influence eating behaviour, a better understanding of how these factors interact is required to facilitate the developing new models of eating behaviour. Due to the diversity of influences on eating behaviour this would probably necessitate a greater focus on multi-disciplinary research. In the present review, the influence of several salient physiological and environmental factors (largely related to food characteristics) on meal initiation, satiation (meal size) and satiety (inter-meal interval) are briefly discussed. Due to the large literature this review is not exhaustive but illustrates the complexity of eating behaviour. The present review will also highlight several limitations that apply to eating behaviour research.
Collapse
|
48
|
Colonic infusions of short-chain fatty acid mixtures promote energy metabolism in overweight/obese men: a randomized crossover trial. Sci Rep 2017; 7:2360. [PMID: 28539646 PMCID: PMC5443817 DOI: 10.1038/s41598-017-02546-x] [Citation(s) in RCA: 203] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 04/12/2017] [Indexed: 12/15/2022] Open
Abstract
Short-chain fatty acids (SCFA), formed by microbial fermentation, are believed to be involved in the aetiology of obesity and diabetes. This study investigated the effects of colonic administration of physiologically relevant SCFA mixtures on human substrate and energy metabolism. In this randomized, double-blind, crossover study, twelve normoglycaemic men (BMI 25–35 kg/m2) underwent four investigational days, during which SCFA mixtures (200 mmol/L) high in either acetate (HA), propionate (HP), butyrate (HB) or placebo (PLA) were rectally administered during fasting and postprandial conditions (oral glucose load). Before and for two hours after colonic infusions, indirect calorimetry was performed and blood samples were collected. All three SCFA mixtures increased fasting fat oxidation (P < 0.01), whilst resting energy expenditure increased after HA and HP compared with PLA (P < 0.05). In addition, all three SCFA mixtures increased fasting and postprandial plasma peptide YY (PYY) concentrations, and attenuated fasting free glycerol concentrations versus PLA (P < 0.05). Colonic infusions of SCFA mixtures, in concentrations and ratios reached after fibre intake, increased fat oxidation, energy expenditure and PYY, and decreased lipolysis in overweight/obese men. Human intervention studies are warranted to investigate whether these effects translate into long-term benefits for body weight control and insulin sensitivity in the obese insulin resistant state.
Collapse
|
49
|
Wu W, Zhou HR, Bursian SJ, Link JE, Pestka JJ. Calcium-Sensing Receptor and Transient Receptor Ankyrin-1 Mediate Emesis Induction by Deoxynivalenol (Vomitoxin). Toxicol Sci 2017; 155:32-42. [PMID: 27667315 PMCID: PMC6366674 DOI: 10.1093/toxsci/kfw191] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The common foodborne mycotoxin deoxynivalenol (DON, vomitoxin) can negatively impact animal and human health by causing food refusal and vomiting. Gut enteroendocrine cells (EECs) secrete hormones that mediate DON's anorectic and emetic effects. In prior work utilizing a cloned EEC model, our laboratory discovered that DON-induced activation of calcium-sensing receptor (CaSR), a G-coupled protein receptor (GPCR), and transient receptor ankyrin-1 (TRPA1), a transient receptor potential (TRP) channel, drives Ca2+-mediated hormone secretion. Consistent with these in vitro findings, CaSR and TRPA1 mediate DON-induced satiety hormone release and food refusal in the mouse, an animal model incapable of vomiting. However, the roles of this GPCR and TRP in DON's emetic effects remain to be determined. To address this, we tested the hypothesis that DON triggers emesis in mink by activating CaSR and TRPA1. Oral gavage with selective agonists for CaSR (R-568) or TRPA1 (allyl isothiocyanate; AITC) rapidly elicited emesis in the mink in dose-dependent fashion. Oral pretreatment of the animals with the CaSR antagonist NPS-2143 or the TRP antagonist ruthenium red (RR), respectively, inhibited these responses. Importantly, DON-induced emesis in mink was similarly inhibited by oral pretreatment with NPS-2143 or RR. In addition, these antagonists suppressed concurrent DON-induced elevations in plasma peptide YY3-36 and 5-hydroxytryptamine-hormones previously demonstrated to mediate the toxin's emetic effects in mink. Furthermore, antagonist co-treatment additively suppressed DON-induced emesis and peptide YY 3-36 release. To summarize, the observations here strongly suggest that activation of CaSR and TRPA1 might have critical roles in DON-induced emesis.
Collapse
Affiliation(s)
- Wenda Wu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, Michigan 48824
| | - Hui-Ren Zhou
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, Michigan 48824
| | - Steven J Bursian
- Center for Integrative Toxicology, Michigan State University, East Lansing, Michigan 48824
- Department of Animal Science, Michigan State University, East Lansing, Michigan 48824
| | - Jane E Link
- Department of Animal Science, Michigan State University, East Lansing, Michigan 48824
| | - James J Pestka
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, Michigan 48824;
- Center for Integrative Toxicology, Michigan State University, East Lansing, Michigan 48824
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan 48824
| |
Collapse
|
50
|
Burcelin R, Gourdy P. Harnessing glucagon-like peptide-1 receptor agonists for the pharmacological treatment of overweight and obesity. Obes Rev 2017; 18:86-98. [PMID: 27636208 DOI: 10.1111/obr.12465] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 08/01/2016] [Accepted: 08/01/2016] [Indexed: 01/07/2023]
Abstract
Over the past 30 years, there has been a dramatic rise in global obesity prevalence, resulting in significant economic and social consequences. Attempts to develop pharmacological agents to treat obesity have met with many obstacles including the lack of long-term effectiveness and the potential for adverse effects. Historically, there have been limited treatment options for overweight and obesity; however, since 2012, a number of new drugs have become available. A number of peptides produced in the gut act as key mediators of the gut-brain axis, which is involved in appetite regulation. This review discusses the role of the gut-brain axis in appetite regulation with special focus on glucagon-like peptide-1. Liraglutide 3.0 mg, a glucagon-like peptide-1 receptor agonist that targets this pathway, is now approved for the treatment of obesity and overweight (body mass index ≥27 kg/m2 ) with comorbidities such as type 2 diabetes, high blood pressure, high cholesterol or obstructive sleep apnoea. In addition, other glucagon-like peptide-1 receptor agonists offer promise for obesity management in the future. This review examines how glucagon-like peptide-1 receptor agonists promote weight loss and summarizes the clinical data on weight loss with glucagon-like peptide-1 receptor agonists.
Collapse
Affiliation(s)
- R Burcelin
- Inserm U1048, Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France
| | - P Gourdy
- Inserm U1048, Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France.,Diabetology Department, Toulouse University Hospital, Toulouse, France
| |
Collapse
|