1
|
Wang Z, Xu Q, Hou L, He Z, Christian M, Dai X. Food-derived polysaccharides and anti-obesity effects through enhancing adipose thermogenesis: structure-activity relationships, mechanisms, and regulation of gut microecology. Crit Rev Food Sci Nutr 2025:1-22. [PMID: 40351151 DOI: 10.1080/10408398.2025.2500675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/14/2025]
Abstract
Polysaccharides represent a crucial and extensively utilized bioactive fraction in natural products, which are employed in the treatment of metabolic disorders due to their significant therapeutic potential. Recently, food-derived polysaccharides (FPs) have emerged as significant substances in obesity management, valued for their ability to activate thermogenic fat. This review discusses the correlation between the structural features of FPs and their efficacy in combating obesity. Moreover, the molecular mechanism by which FPs regulate thermogenic fat and how the intestinal microecology induces thermogenic fat activity is elucidated. The anti-obesity effects of FPs depend on their structure, including molecular weight, composition, linkages, conformation, and branching. Furthermore, FPs regulate fat thermogenesis via multiple mechanisms, including AMPK, p38, AKT, PGC-1α-FNDC5/irisin, and miRNA signaling pathways. Importantly, gut microbiota, together with its associated metabolites and gut-derived hormones, are pivotal in the regulatory control of brown fat by FPs. This work provides an in-depth examination of how adipose tissue thermogenesis contributes to the anti-obesity effects of FPs, shedding light on their potential in preventing obesity and informing the formulation of natural weight-loss remedies.
Collapse
Affiliation(s)
- Zhenyu Wang
- College of Life Sciences, China Jiliang University, Hangzhou, China
| | - Qiyu Xu
- College of Life Sciences, China Jiliang University, Hangzhou, China
| | - Lijuan Hou
- College of Life Sciences, China Jiliang University, Hangzhou, China
| | - Zhiyong He
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
| | - Mark Christian
- School of Science and Technology, Nottingham Trent University, Clifton, Nottingham, United Kingdom
| | - Xianjun Dai
- College of Life Sciences, China Jiliang University, Hangzhou, China
| |
Collapse
|
2
|
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
|
3
|
Gong B, Yao Z, Zhou C, Wang W, Sun L, Han J. Glucagon-like peptide-1 analogs: Miracle drugs are blooming? Eur J Med Chem 2024; 269:116342. [PMID: 38531211 DOI: 10.1016/j.ejmech.2024.116342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 03/11/2024] [Accepted: 03/15/2024] [Indexed: 03/28/2024]
Abstract
Glucagon-like peptide-1 (GLP-1), secreted by L cells in the small intestine, assumes a central role in managing type 2 diabetes mellitus (T2DM) and obesity. Its influence on insulin secretion and gastric emptying positions it as a therapeutic linchpin. However, the limited applicability of native GLP-1 stems from its short half-life, primarily due to glomerular filtration and the inactivating effect of dipeptidyl peptidase-IV (DPP-IV). To address this, various structural modification strategies have been developed to extend GLP-1's half-life. Despite the commendable efficacy displayed by current GLP-1 receptor agonists, inherent limitations persist. A paradigm shift emerges with the advent of unimolecular multi-agonists, such as the recently introduced tirzepatide, wherein GLP-1 is ingeniously combined with other gastrointestinal hormones. This novel approach has captured the spotlight within the diabetes and obesity research community. This review summarizes the physiological functions of GLP-1, systematically explores diverse structural modifications, delves into the realm of unimolecular multi-agonists, and provides a nuanced portrayal of the developmental prospects that lie ahead for GLP-1 analogs.
Collapse
Affiliation(s)
- Binbin Gong
- College of Medicine, Jiaxing University, Jiaxing, 314001, China; College of Pharmacy, Zhejiang University of Technology, Hangzhou, 310000, China
| | - Zhihong Yao
- College of Medicine, Jiaxing University, Jiaxing, 314001, China; College of Pharmacy, Zhejiang University of Technology, Hangzhou, 310000, China
| | - Chenxu Zhou
- College of Medicine, Jiaxing University, Jiaxing, 314001, China
| | - Wenxi Wang
- College of Pharmacy, Zhejiang University of Technology, Hangzhou, 310000, China
| | - Lidan Sun
- College of Medicine, Jiaxing University, Jiaxing, 314001, China.
| | - Jing Han
- School of Chemistry & Materials Science, Jiangsu Normal University, Xuzhou, 221116, China.
| |
Collapse
|
4
|
Brzozowska MM, Isaacs M, Bliuc D, Baldock PA, Eisman JA, White CP, Greenfield JR, Center JR. Effects of bariatric surgery and dietary intervention on insulin resistance and appetite hormones over a 3 year period. Sci Rep 2023; 13:6032. [PMID: 37055514 PMCID: PMC10102182 DOI: 10.1038/s41598-023-33317-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 04/11/2023] [Indexed: 04/15/2023] Open
Abstract
To examine an impact of three types of bariatric surgery compared with dietary intervention (DIET), on concurrent changes in Homeostatic Model Assessment for Insulin Resistance (HOMA-IR) and appetite hormones over 3 years. Fifty-five adults were studied during phase of weight loss (0-12 months) and during weight stability (12-36 months) post intervention. Measurements of HOMA-IR, fasting and postprandial PYY and GLP1, adiponectin, CRP, RBP4, FGF21 hormones and dual-Xray absorptiometry were performed throughout the study. All surgical groups achieved significant reductions in HOMA-IR with greatest difference between Roux-en-Y gastric bypass and DIET (- 3.7; 95% CI - 5.4, - 2.1; p = 0.001) at 12-36 months. Initial (0-12 months) HOMA-IR values were no different to DIET after adjustment for the lost weight. During 12-36 months, after controlling for treatment procedure and weight, for every twofold increase in postprandial PYY and adiponectin, HOMA-IR decreased by 0.91 (95% CI - 1.71, - 0.11; p = 0.030) and by 0.59 (95% CI - 1.10, - 0.10; p = 0.023) respectively. Initial, non-sustained changes in RBP4 and FGF21 were not associated with HOMA-IR values. While initial rapid weight loss reduces insulin resistance, the enhanced secretions of PYY and adiponectin may contribute to weight-independent improvements in HOMA-IR during weight stability.Clinical trial registration: Australian New Zealand Clinical Trials Registry (ANZCTR): ACTRN12613000188730.
Collapse
Affiliation(s)
- Malgorzata M Brzozowska
- Endocrinology, The Sutherland Hospital, Caringbah, Australia.
- Faculty of Medicine, UNSW Sydney, Sydney, Australia.
- Garvan Institute of Medical Research, Healthy Ageing Theme, Darlinghurst, Australia.
| | - Michelle Isaacs
- Faculty of Medicine, UNSW Sydney, Sydney, Australia
- Endocrinology, St Vincent's Hospital Sydney, Darlinghurst, Australia
| | - Dana Bliuc
- Faculty of Medicine, UNSW Sydney, Sydney, Australia
- Garvan Institute of Medical Research, Healthy Ageing Theme, Darlinghurst, Australia
| | - Paul A Baldock
- Faculty of Medicine, UNSW Sydney, Sydney, Australia
- Garvan Institute of Medical Research, Healthy Ageing Theme, Darlinghurst, Australia
- School of Medicine, The University of Notre Dame Australia, Darlinghurst, Australia
| | - John A Eisman
- Faculty of Medicine, UNSW Sydney, Sydney, Australia
- Garvan Institute of Medical Research, Healthy Ageing Theme, Darlinghurst, Australia
- Endocrinology, St Vincent's Hospital Sydney, Darlinghurst, Australia
- School of Medicine, The University of Notre Dame Australia, Darlinghurst, Australia
| | - Chris P White
- Faculty of Medicine, UNSW Sydney, Sydney, Australia
- Prince of Wales Hospital, NSW Health Pathology, Randwick, Australia
- Endocrinology, Prince of Wales Hospital, Randwick, Australia
| | - Jerry R Greenfield
- Faculty of Medicine, UNSW Sydney, Sydney, Australia
- Garvan Institute of Medical Research, Healthy Ageing Theme, Darlinghurst, Australia
- Endocrinology, St Vincent's Hospital Sydney, Darlinghurst, Australia
| | - Jacqueline R Center
- Faculty of Medicine, UNSW Sydney, Sydney, Australia
- Garvan Institute of Medical Research, Healthy Ageing Theme, Darlinghurst, Australia
- Endocrinology, St Vincent's Hospital Sydney, Darlinghurst, Australia
- School of Medicine, The University of Notre Dame Australia, Darlinghurst, Australia
| |
Collapse
|
5
|
Sridhar A, Khan D, Flatt PR, Irwin N, Moffett RC. PYY (3-36) protects against high fat feeding induced changes of pancreatic islet and intestinal hormone content and morphometry. Biochim Biophys Acta Gen Subj 2023; 1867:130359. [PMID: 37001706 DOI: 10.1016/j.bbagen.2023.130359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 03/16/2023] [Accepted: 03/23/2023] [Indexed: 03/31/2023]
Abstract
BACKGROUND Prolonged high fat feeding negatively impacts pancreatic and intestinal morphology. In this regard, direct effects of PYY(3-36) on intestinal cell and pancreatic islet morphometry are yet to be fully explored in the setting of obesity. METHODS We examined the influence of 21-days twice daily treatment with PYY(3-36) on these parameters in mice fed a high fat diet (HFD). RESULTS PYY(3-36) treatment decreased food intake, body weight and circulating glucose in HFD mice. In terms of intestinal morphology, crypt depth was restored to control levels by PYY(3-36), with an additional enlargement of villi length. PYY(3-36) also reversed HFD-induced decreases of ileal PYY, and especially GLP-1, content. HFD increased numbers of PYY and GIP positive ileal cells, with PYY(3-36) fully reversing the effect on PYY cell detection. There were no obvious differences in the overall number of GLP-1 positive ileal cells in all mice, barring PYY(3-36) marginally decreasing GLP-1 villi cell immunoreactivity. Within pancreatic islets, PYY(3-36) significantly decreased alpha-cell area, whilst islet, beta-, PYY- and delta-cell areas remained unchanged. However, PYY(3-36) increased the percentage of beta-cells while also reducing percentage alpha-cell area. This was related to PYY(3-36)-induced reductions of beta-cell proliferation and apoptosis frequencies. Co-localisation of islet PYY with glucagon or somatostatin was elevated by PYY(3-36), with GLP-1/glucagon co-visualisation increased when compared to lean controls. CONCLUSION PYY(3-36) exerts protective effects on pancreatic and intestinal morphology in HFD mice linked to elevated ileal GLP-1 content. GENERAL SIGNIFICANCE These observations highlight mechanisms linked to the metabolic and weight reducing benefits of PYY(3-36).
Collapse
Affiliation(s)
- A Sridhar
- Biomedical Sciences Research Institute, School of Biomedical Sciences, Ulster University, Coleraine, N. Ireland, UK
| | - D Khan
- Biomedical Sciences Research Institute, School of Biomedical Sciences, Ulster University, Coleraine, N. Ireland, UK
| | - P R Flatt
- Biomedical Sciences Research Institute, School of Biomedical Sciences, Ulster University, Coleraine, N. Ireland, UK
| | - N Irwin
- Biomedical Sciences Research Institute, School of Biomedical Sciences, Ulster University, Coleraine, N. Ireland, UK.
| | - R C Moffett
- Biomedical Sciences Research Institute, School of Biomedical Sciences, Ulster University, Coleraine, N. Ireland, UK
| |
Collapse
|
6
|
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: 3.3] [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
|
7
|
Yang CH, Onda DA, Oakhill JS, Scott JW, Galic S, Loh K. Regulation of Pancreatic β-Cell Function by the NPY System. Endocrinology 2021; 162:6213414. [PMID: 33824978 DOI: 10.1210/endocr/bqab070] [Citation(s) in RCA: 5] [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: 12/22/2020] [Indexed: 01/24/2023]
Abstract
The neuropeptide Y (NPY) system has been recognized as one of the most critical molecules in the regulation of energy homeostasis and glucose metabolism. Abnormal levels of NPY have been shown to contribute to the development of metabolic disorders including obesity, cardiovascular diseases, and diabetes. NPY centrally promotes feeding and reduces energy expenditure, while the other family members, peptide YY (PYY) and pancreatic polypeptide (PP), mediate satiety. New evidence has uncovered additional functions for these peptides that go beyond energy expenditure and appetite regulation, indicating a more extensive function in controlling other physiological functions. In this review, we will discuss the role of the NPY system in the regulation of pancreatic β-cell function and its therapeutic implications for diabetes.
Collapse
Affiliation(s)
- Chieh-Hsin Yang
- St. Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia
| | - Danise-Ann Onda
- St. Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia
| | - Jonathan S Oakhill
- St. Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia
- Department of Medicine, University of Melbourne, Parkville, VIC 3010, Australia
- Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, VIC 3000, Australia
| | - John W Scott
- St. Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia
- Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, VIC 3000, Australia
- The Florey Institute of Neuroscience and Mental Health, Parkville, VIC 3010, Australia
| | - Sandra Galic
- St. Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia
- Department of Medicine, University of Melbourne, Parkville, VIC 3010, Australia
| | - Kim Loh
- St. Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia
- Department of Medicine, University of Melbourne, Parkville, VIC 3010, Australia
| |
Collapse
|
8
|
Schalla MA, Taché Y, Stengel A. Neuroendocrine Peptides of the Gut and Their Role in the Regulation of Food Intake. Compr Physiol 2021; 11:1679-1730. [PMID: 33792904 DOI: 10.1002/cphy.c200007] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The regulation of food intake encompasses complex interplays between the gut and the brain. Among them, the gastrointestinal tract releases different peptides that communicate the metabolic state to specific nuclei in the hindbrain and the hypothalamus. The present overview gives emphasis on seven peptides that are produced by and secreted from specialized enteroendocrine cells along the gastrointestinal tract in relation with the nutritional status. These established modulators of feeding are ghrelin and nesfatin-1 secreted from gastric X/A-like cells, cholecystokinin (CCK) secreted from duodenal I-cells, glucagon-like peptide 1 (GLP-1), oxyntomodulin, and peptide YY (PYY) secreted from intestinal L-cells and uroguanylin (UGN) released from enterochromaffin (EC) cells. © 2021 American Physiological Society. Compr Physiol 11:1679-1730, 2021.
Collapse
Affiliation(s)
- Martha A Schalla
- Charité Center for Internal Medicine and Dermatology, Department for Psychosomatic Medicine, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Yvette Taché
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, CURE: Digestive Diseases Research Center, David Geffen School of Medicine, UCLA, Los Angeles, California, USA.,VA Greater Los Angeles Healthcare System, Los Angeles, California, USA
| | - Andreas Stengel
- Charité Center for Internal Medicine and Dermatology, Department for Psychosomatic Medicine, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany.,Department of Psychosomatic Medicine and Psychotherapy, Medical University Hospital Tübingen, Tübingen, Germany
| |
Collapse
|
9
|
Alhabeeb H, AlFaiz A, Kutbi E, AlShahrani D, Alsuhail A, AlRajhi S, Alotaibi N, Alotaibi K, AlAmri S, Alghamdi S, AlJohani N. Gut Hormones in Health and Obesity: The Upcoming Role of Short Chain Fatty Acids. Nutrients 2021; 13:nu13020481. [PMID: 33572661 PMCID: PMC7911102 DOI: 10.3390/nu13020481] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 12/21/2020] [Accepted: 12/30/2020] [Indexed: 12/13/2022] Open
Abstract
We are currently facing an obesity pandemic, with worldwide obesity rates having tripled since 1975. Obesity is one of the main risk factors for the development of non-communicable diseases, which are now the leading cause of death worldwide. This calls for urgent action towards understanding the underlying mechanisms behind the development of obesity as well as developing more effective treatments and interventions. Appetite is carefully regulated in humans via the interaction between the central nervous system and peripheral hormones. This involves a delicate balance in external stimuli, circulating satiating and appetite stimulating hormones, and correct functioning of neuronal signals. Any changes in this equilibrium can lead to an imbalance in energy intake versus expenditure, which often leads to overeating, and potentially weight gain resulting in overweight or obesity. Several lines of research have shown imbalances in gut hormones are found in those who are overweight or obese, which may be contributing to their condition. Therefore, this review examines the evidence for targeting gut hormones in the treatment of obesity by discussing how their dysregulation influences food intake, the potential possibility of altering the circulating levels of these hormones for treating obesity, as well as the role of short chain fatty acids and protein as novel treatments.
Collapse
Affiliation(s)
- Habeeb Alhabeeb
- Research Center, King Fahad Medical City—KFMC, Riyadh 11525, Saudi Arabia; (A.A.); (E.K.); (D.A.); (A.A.); (S.A.); (S.A.)
- Correspondence:
| | - Ali AlFaiz
- Research Center, King Fahad Medical City—KFMC, Riyadh 11525, Saudi Arabia; (A.A.); (E.K.); (D.A.); (A.A.); (S.A.); (S.A.)
| | - Emad Kutbi
- Research Center, King Fahad Medical City—KFMC, Riyadh 11525, Saudi Arabia; (A.A.); (E.K.); (D.A.); (A.A.); (S.A.); (S.A.)
| | - Dayel AlShahrani
- Research Center, King Fahad Medical City—KFMC, Riyadh 11525, Saudi Arabia; (A.A.); (E.K.); (D.A.); (A.A.); (S.A.); (S.A.)
| | - Abdullah Alsuhail
- Research Center, King Fahad Medical City—KFMC, Riyadh 11525, Saudi Arabia; (A.A.); (E.K.); (D.A.); (A.A.); (S.A.); (S.A.)
| | - Saleh AlRajhi
- Family Medicine, King Fahad Medical City—KFMC, Riyadh 11525, Saudi Arabia;
| | - Nemer Alotaibi
- College of Medicine, Shaqra University, Shaqra 11961, Saudi Arabia; (N.A.); (K.A.)
| | - Khalid Alotaibi
- College of Medicine, Shaqra University, Shaqra 11961, Saudi Arabia; (N.A.); (K.A.)
| | - Saad AlAmri
- Research Center, King Fahad Medical City—KFMC, Riyadh 11525, Saudi Arabia; (A.A.); (E.K.); (D.A.); (A.A.); (S.A.); (S.A.)
| | - Saleh Alghamdi
- Research Center, King Fahad Medical City—KFMC, Riyadh 11525, Saudi Arabia; (A.A.); (E.K.); (D.A.); (A.A.); (S.A.); (S.A.)
| | - Naji AlJohani
- Obesity, Endocrine, and Metabolism Center, King Fahad Medical City—KFMC, Riyadh 11525, Saudi Arabia;
| |
Collapse
|
10
|
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: 0.2] [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
|
11
|
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: 0.4] [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
|
12
|
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: 47] [Impact Index Per Article: 9.4] [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
|
13
|
Gimeno RE, Briere DA, Seeley RJ. Leveraging the Gut to Treat Metabolic Disease. Cell Metab 2020; 31:679-698. [PMID: 32187525 PMCID: PMC7184629 DOI: 10.1016/j.cmet.2020.02.014] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 11/23/2019] [Accepted: 02/20/2020] [Indexed: 02/07/2023]
Abstract
25 years ago, the future of treating obesity and diabetes focused on end organs known to be involved in energy balance and glucose regulation, including the brain, muscle, adipose tissue, and pancreas. Today, the most effective therapies are focused around the gut. This includes surgical options, such as vertical sleeve gastrectomy and Roux-en-Y gastric bypass, that can produce sustained weight loss and diabetes remission but also extends to pharmacological treatments that simulate or amplify various signals that come from the gut. The purpose of this Review is to discuss the wealth of approaches currently under development that seek to further leverage the gut as a source of novel therapeutic opportunities with the hope that we can achieve the effects of surgical interventions with less invasive and more scalable solutions.
Collapse
Affiliation(s)
- Ruth E Gimeno
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46225, USA
| | - Daniel A Briere
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46225, USA
| | - Randy J Seeley
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
14
|
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: 20] [Impact Index Per Article: 4.0] [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
|
15
|
Jones ES, Nunn N, Chambers AP, Østergaard S, Wulff BS, Luckman SM. Modified Peptide YY Molecule Attenuates the Activity of NPY/AgRP Neurons and Reduces Food Intake in Male Mice. Endocrinology 2019; 160:2737-2747. [PMID: 31074796 PMCID: PMC6806261 DOI: 10.1210/en.2019-00100] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 05/06/2019] [Indexed: 01/15/2023]
Abstract
To study the effects of an analog of the gut-produced hormone peptide YY (PYY3-36), which has increased selectivity for the Y2 receptor; specifically, to record its effects on food intake and on hypothalamic neuropeptide Y/agouti-related peptide (NPY/AgRP) neuron activity. NNC0165-1273, a modified form of the peptide hormone PYY3-36 with potent selectivity at Y2 receptor (>5000-fold over Y1, 1250-fold over Y4, and 650-fold over Y5 receptor), was tested in vivo and in vitro in mouse models. NNC0165-1273 has fivefold lower relative affinity for Y2 compared with PYY3-36, but >250-, 192-, and 400-fold higher selectivity, respectively, for the Y1, Y4, and Y5 receptors. NNC0165-1273 produced a reduction in nighttime feeding at a dose at which PYY3-36 loses efficacy. The normal behavioral satiety sequence observed suggests that NNC0165-1273 is not nauseating and, instead, reduces food intake by producing early satiety. Additionally, NNC0165-1273 blocked ghrelin-induced cFos expression in NPY/AgRP neurons. In vitro electrophysiological recordings showed that, opposite to ghrelin, NNC0165-1273 hyperpolarized NPY/AgRP neurons and reduced action potential frequency. Administration of NNC0165-1273 via subcutaneous osmotic minipump caused a dose-dependent decrease in body weight and fat mass in an obese mouse model. Finally, NNC0165-1273 attenuated the feeding response when NPY/AgRP neurons were activated using ghrelin or more selectively with designer receptors. NNC0165-1273 is nonnauseating and stimulates a satiety response through, at least in part, a direct action on hypothalamic NPY/AgRP neurons. Modification of PYY3-36 to produce compounds with increased affinity to Y2 receptors may be useful as antiobesity therapies in humans.
Collapse
Affiliation(s)
- Edward S Jones
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Nicolas Nunn
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Adam P Chambers
- GLP-1 & T2D Pharmacology, Novo Nordisk A/S, Novo Nordisk Park, Måløv, Denmark
| | - Søren Østergaard
- Research Chemistry 2, Novo Nordisk A/S, Novo Nordisk Park, Måløv, Denmark
| | - Birgitte S Wulff
- Obesity Research, Novo Nordisk A/S, Novo Nordisk Park, Måløv, Denmark
| | - Simon M Luckman
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
16
|
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: 2.3] [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
|
17
|
Elmansi AM, Awad ME, Eisa NH, Kondrikov D, Hussein KA, Aguilar-Pérez A, Herberg S, Periyasamy-Thandavan S, Fulzele S, Hamrick MW, McGee-Lawrence ME, Isales CM, Volkman BF, Hill WD. What doesn't kill you makes you stranger: Dipeptidyl peptidase-4 (CD26) proteolysis differentially modulates the activity of many peptide hormones and cytokines generating novel cryptic bioactive ligands. Pharmacol Ther 2019; 198:90-108. [PMID: 30759373 PMCID: PMC7883480 DOI: 10.1016/j.pharmthera.2019.02.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Dipeptidyl peptidase 4 (DPP4) is an exopeptidase found either on cell surfaces where it is highly regulated in terms of its expression and surface availability (CD26) or in a free/circulating soluble constitutively available and intrinsically active form. It is responsible for proteolytic cleavage of many peptide substrates. In this review we discuss the idea that DPP4-cleaved peptides are not necessarily inactivated, but rather can possess either a modified receptor selectivity, modified bioactivity, new antagonistic activity, or even a novel activity relative to the intact parent ligand. We examine in detail five different major DPP4 substrates: glucagon-like peptide 1 (GLP-1), glucose-dependent insulinotropic polypeptide (GIP), peptide tyrosine-tyrosine (PYY), and neuropeptide Y (NPY), and stromal derived factor 1 (SDF-1 aka CXCL12). We note that discussion of the cleaved forms of these five peptides are underrepresented in the research literature, and are both poorly investigated and poorly understood, representing a serious research literature gap. We believe they are understudied and misinterpreted as inactive due to several factors. This includes lack of accurate and specific quantification methods, sample collection techniques that are inherently inaccurate and inappropriate, and a general perception that DPP4 cleavage inactivates its ligand substrates. Increasing evidence points towards many DPP4-cleaved ligands having their own bioactivity. For example, GLP-1 can work through a different receptor than GLP-1R, DPP4-cleaved GIP can function as a GIP receptor antagonist at high doses, and DPP4-cleaved PYY, NPY, and CXCL12 can have different receptor selectivity, or can bind novel, previously unrecognized receptors to their intact ligands, resulting in altered signaling and functionality. We believe that more rigorous research in this area could lead to a better understanding of DPP4's role and the biological importance of the generation of novel cryptic ligands. This will also significantly impact our understanding of the clinical effects and side effects of DPP4-inhibitors as a class of anti-diabetic drugs that potentially have an expanding clinical relevance. This will be specifically relevant in targeting DPP4 substrate ligands involved in a variety of other major clinical acute and chronic injury/disease areas including inflammation, immunology, cardiology, stroke, musculoskeletal disease and injury, as well as cancer biology and tissue maintenance in aging.
Collapse
Affiliation(s)
- Ahmed M Elmansi
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29403, United States; Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC 29403, United States
| | - Mohamed E Awad
- Department of Oral Biology, School of Dentistry, Augusta University, Augusta, GA 30912, United States
| | - Nada H Eisa
- Georgia Cancer Center, Augusta University, Augusta, GA 30912, United States; Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| | - Dmitry Kondrikov
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29403, United States; Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC 29403, United States
| | - Khaled A Hussein
- Department of Surgery and Medicine, National Research Centre, Cairo, Egypt
| | - Alexandra Aguilar-Pérez
- Department of Anatomy and Cell Biology, Indiana University School of Medicine in Indianapolis, IN, United States; Department of Cellular and Molecular Biology, School of Medicine, Universidad Central del Caribe, Bayamon, 00956, Puerto Rico; Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States
| | - Samuel Herberg
- Departments of Ophthalmology & Cell and Dev. Bio., SUNY Upstate Medical University, Syracuse, NY 13210, United States
| | | | - Sadanand Fulzele
- Department of Orthopaedic Surgery, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States; Center for Healthy Aging, Medical College of Georgia, Augusta University, Augusta, GA, 30912, United States
| | - Mark W Hamrick
- Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States; Department of Orthopaedic Surgery, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States; Center for Healthy Aging, Medical College of Georgia, Augusta University, Augusta, GA, 30912, United States
| | - Meghan E McGee-Lawrence
- Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States; Department of Orthopaedic Surgery, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States; Center for Healthy Aging, Medical College of Georgia, Augusta University, Augusta, GA, 30912, United States
| | - Carlos M Isales
- Department of Orthopaedic Surgery, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States; Center for Healthy Aging, Medical College of Georgia, Augusta University, Augusta, GA, 30912, United States; Division of Endocrinology, Diabetes and Metabolism, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States
| | - Brian F Volkman
- Biochemistry Department, Medical College of Wisconsin, Milwaukee, WI 53226, United States
| | - William D Hill
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29403, United States; Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC 29403, United States; Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States; Department of Orthopaedic Surgery, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States; Center for Healthy Aging, Medical College of Georgia, Augusta University, Augusta, GA, 30912, United States.
| |
Collapse
|
18
|
|
19
|
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: 53] [Impact Index Per Article: 8.8] [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
|
20
|
Nutritive, Post-ingestive Signals Are the Primary Regulators of AgRP Neuron Activity. Cell Rep 2018; 21:2724-2736. [PMID: 29212021 DOI: 10.1016/j.celrep.2017.11.036] [Citation(s) in RCA: 148] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 11/08/2017] [Accepted: 11/10/2017] [Indexed: 11/21/2022] Open
Abstract
The brain regulates food intake by processing sensory cues and peripheral physiological signals, but the neural basis of this integration remains unclear. Hypothalamic, agouti-related protein (AgRP)-expressing neurons are critical regulators of food intake. AgRP neuron activity is high during hunger and is rapidly reduced by the sight and smell of food. Here, we reveal two distinct components of AgRP neuron activity regulation: a rapid but transient sensory-driven signal and a slower, sustained calorie-dependent signal. We discovered that nutrients are necessary and sufficient for sustained reductions in AgRP neuron activity and that activity reductions are proportional to the calories obtained. This change in activity is recapitulated by exogenous administration of gut-derived satiation signals. Furthermore, we showed that the nutritive value of food trains sensory systems-in a single trial-to drive rapid, anticipatory AgRP neuron activity inhibition. Together, these data demonstrate that nutrients are the primary regulators of AgRP neuron activity.
Collapse
|
21
|
Li R, Li Y, Su Y, Shen D, Dai P, Li C. Short-term ingestion of deoxynivalenol in naturally contaminated feed alters piglet performance and gut hormone secretion. Anim Sci J 2018; 89:1134-1143. [PMID: 29808618 DOI: 10.1111/asj.13034] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 03/06/2018] [Indexed: 12/19/2022]
Abstract
The mycotoxin deoxynivalenol (DON) generally exists in cereals and affects human and animal health. The aim of this study is to analyze the impacts of DON in naturally contaminated feed on piglet growth performance and intestinal hormone secretion in the short term. We randomly divided 5-week-old piglets into four groups: Control, DON 1,000, DON 2,000 and DON 3,000 groups. Piglets received a feed naturally contaminated with DON (approximately 400, 1,000, 2,000 or 3,000 μg/kg) for 21 days. Body weight showed no significant difference following exposure to DON. The balance of anti-oxidation and oxidation was disrupted by DON after 21 days. The concentration of tumor necrosis factor-alpha (TNF-α) and cyclooxgenase-2 (COX-2) significantly increased (p < .001) in all DON-treated groups. Gut anorexigenic hormone secretion of peptide YY (PYY) and cholecystokinin (CCK) had a time- and dose-dependent relationship with DON exposure; however, there was no effect on orexigenic hormone ghrelin secretion. Changes of histomorphology in the jejunum were observed in DON-treated groups, including villi flattening and fusion, and apical necrosis of villi. These results indicated that DON could suppress piglet growth performance and alter gut hormone secretion in the short term.
Collapse
Affiliation(s)
- Ruonan Li
- Jiangsu Provincial Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Yansen Li
- Jiangsu Provincial Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Yongteng Su
- Jiangsu Aomai Bio-Tech Company, Nanjing White Horse National Modern Agricultural High-Tech Industrial Park, Nanjing, China
| | - Dan Shen
- Jiangsu Provincial Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Pengyuan Dai
- Jiangsu Provincial Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Chunmei Li
- Jiangsu Provincial Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
22
|
Fernandez-Cachon ML, Pedersen SL, Rigbolt KT, Zhang C, Fabricius K, Hansen HH, Elster L, Fink LN, Schäfer M, Rhee NA, Langholz E, Wandall E, Friis SU, Vilmann P, Kristiansen VB, Schmidt C, Schreiter K, Breitschopf K, Hübschle T, Jorsal T, Vilsbøll T, Schmidt T, Theis S, Knop FK, Larsen PJ, Jelsing J. Guanylin and uroguanylin mRNA expression is increased following Roux-en-Y gastric bypass, but guanylins do not play a significant role in body weight regulation and glycemic control. Peptides 2018; 101:32-43. [PMID: 29289697 DOI: 10.1016/j.peptides.2017.12.024] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 12/18/2017] [Accepted: 12/24/2017] [Indexed: 02/07/2023]
Abstract
AIM To determine whether intestinal expression of guanylate cyclase activator 2A (GUCA2A) and guanylate cyclase activator 2B (GUCA2B) genes is regulated in obese humans following Roux-en-Y gastric bypass (RYGB), and to evaluate the corresponding guanylin (GN) and uroguanylin (UGN) peptides for potentially contributing to the beneficial metabolic effects of RYGB. METHODS Enteroendocrine cells were harvested peri- and post-RYGB, and GUCA2A/GUCA2B mRNA expression was compared. GN, UGN and their prohormones (proGN, proUGN) were administered subcutaneously in normal-weight mice to evaluate effects on food intake and glucose regulation. The effect of pro-UGN or UGN overexpression, using adeno-associated virus (AAV) vectors, was assessed in diet-induced obese (DIO) mice. Intracerebroventricular administration of GN and UGN was performed in rats for assessment of putative centrally mediated effects on food intake. GN and UGN, as well as their prohormones, were evaluated for effects on glucose-stimulated insulin secretion (GSIS) in rat pancreatic islets and perfused rat pancreas. RESULTS GUCA2A and GUCA2B mRNA expression was significantly upregulated in enteroendocrine cells after RYGB. Peripheral administration of guanylins or prohormones did not influence food intake, oral glucose tolerance, and GSIS. Central administration of GN and UGN did not affect food intake in rats. Chronic AVV-mediated overexpression of UGN and proUGN had no effect on body weight or glucose homeostasis in DIO mice. CONCLUSION GN and UGN, as well as their prohormones, do not seem to play a significant role in body weight regulation and glycemic control, suggesting that guanylin-family peptides do not show promise as targets for the treatment of obesity or diabetes.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Nicolai A Rhee
- Center for Diabetes Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
| | - Ebbe Langholz
- Department of Medicine, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
| | - Erik Wandall
- Department of Medicine, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
| | - Steffen U Friis
- Department of Medicine, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
| | - Peter Vilmann
- Gastro Unit, Herlev Hospital, University of Copenhagen, Herlev, Denmark
| | | | | | | | | | | | - Tina Jorsal
- Center for Diabetes Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Tina Vilsbøll
- Center for Diabetes Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
| | | | | | - Filip K Knop
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Center for Diabetes Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
| | | | | |
Collapse
|
23
|
Guida C, McCulloch LJ, Godazgar M, Stephen SD, Baker C, Basco D, Dong J, Chen D, Clark A, Ramracheya RD. Sitagliptin and Roux-en-Y gastric bypass modulate insulin secretion via regulation of intra-islet PYY. Diabetes Obes Metab 2018; 20:571-581. [PMID: 28892258 PMCID: PMC5836881 DOI: 10.1111/dom.13113] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 08/28/2017] [Accepted: 09/05/2017] [Indexed: 12/14/2022]
Abstract
AIMS The gut hormone peptide tyrosine tyrosine (PYY) is critical for maintaining islet integrity and restoring islet function following Roux-en-Y gastric bypass (RYGB). The expression of PYY and its receptors (NPYRs) in islets has been documented but not fully characterized. Modulation of islet PYY by the proteolytic enzyme dipeptidyl peptidase IV (DPP-IV) has not been investigated and the impact of DPP-IV inhibition on islet PYY function remains unexplored. Here we have addressed these gaps and their effects on glucose-stimulated insulin secretion (GSIS). We have also investigated changes in pancreatic PYY in diabetes and following RYGB. METHODS Immunohistochemistry and gene expression analysis were used to assess PYY, NPYRs and DPP-IV expression in rodent and human islets. DPP-IV activity inhibition was achieved by sitagliptin. Secretion studies were used to test PYY and the effects of sitagliptin on insulin release, and the involvement of GLP-1. Radioimmunoassays were used to measure hormone content in islets. RESULTS PYY and DPP-IV localized in different cell types in islets while NPYR expression was confined to the beta-cells. Chronic PYY application enhanced GSIS in rodent and diabetic human islets. DPP-IV inhibition by sitagliptin potentiated GSIS; this was mediated by locally-produced PYY, and not GLP-1. Pancreatic PYY was markedly reduced in diabetes. RYGB strongly increased islet PYY content, but did not lead to full restoration of pancreatic GLP-1 levels. CONCLUSION Local regulation of pancreatic PYY, rather than GLP-1, by DPP-IV inhibition or RYGB can directly modulate the insulin secretory response to glucose, indicating a novel role of pancreatic PYY in diabetes and weight-loss surgery.
Collapse
Affiliation(s)
- Claudia Guida
- Oxford Centre for Diabetes, Endocrinology and MetabolismChurchill Hospital, Oxford UniversityOxfordUK
| | - Laura J. McCulloch
- Oxford Centre for Diabetes, Endocrinology and MetabolismChurchill Hospital, Oxford UniversityOxfordUK
| | - Mahdieh Godazgar
- Oxford Centre for Diabetes, Endocrinology and MetabolismChurchill Hospital, Oxford UniversityOxfordUK
| | - Sam D. Stephen
- Oxford Centre for Diabetes, Endocrinology and MetabolismChurchill Hospital, Oxford UniversityOxfordUK
| | - Charlotte Baker
- Oxford Centre for Diabetes, Endocrinology and MetabolismChurchill Hospital, Oxford UniversityOxfordUK
| | - Davide Basco
- Center for Integrative GenomicsUniversity of LausanneLausanneSwitzerland
| | | | - Duan Chen
- Department of Clinical and Molecular MedicineNorwegian University of Science and TechnologyTrondheimNorway
| | - Anne Clark
- Oxford Centre for Diabetes, Endocrinology and MetabolismChurchill Hospital, Oxford UniversityOxfordUK
| | - Reshma D Ramracheya
- Oxford Centre for Diabetes, Endocrinology and MetabolismChurchill Hospital, Oxford UniversityOxfordUK
| |
Collapse
|
24
|
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: 55] [Impact Index Per Article: 7.9] [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
|
25
|
Hopkins M, Beaulieu K, Myers A, Gibbons C, Blundell JE. Mechanisms responsible for homeostatic appetite control: theoretical advances and practical implications. Expert Rev Endocrinol Metab 2017; 12:401-415. [PMID: 30063436 DOI: 10.1080/17446651.2017.1395693] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Homeostatic appetite control is part of a psychobiological system that has evolved to maintain an adequate supply of nutrients for growth and maintenance. The system links the physiological needs for energy with the behaviour that satisfies these needs (feeding), and is shaped by excitatory and inhibitory signals. Owing to rapid shifts in the food environment, homeostatic appetite control is not well adapted for modern-day human functioning. Areas covered: Homeostatic appetite control has two divisions. Tonic processes exert stable and enduring influences, with signals arising from bodily tissues and metabolism. Episodic processes fluctuate rapidly and are related to nutrient ingestion and the composition of foods consumed. Research in these areas incorporates potent endocrine signals that can influence behaviour. Expert commentary: The regulation of adipose tissue, and its impact on appetite (energy) homeostasis, has been heavily researched. More recently however, it has been demonstrated that fat-free mass has the potential to act as a tonic driver of food intake. A challenging issue is to determine how the post-prandial action of episodic satiety hormones and gastrointestinal mechanisms can effectively brake the metabolic drive to eat, in order to keep food intake under control and prevent a positive energy balance and fat accumulation.
Collapse
Affiliation(s)
- Mark Hopkins
- a School of Food Science and Nutrition, Faculty of Mathematics and Physical Sciences , University of Leeds , Leeds , UK
| | - Kristine Beaulieu
- b School of Psychology, Faculty of Medicine and Health , University of Leeds , Leeds , UK
| | - Anna Myers
- b School of Psychology, Faculty of Medicine and Health , University of Leeds , Leeds , UK
| | - Catherine Gibbons
- b School of Psychology, Faculty of Medicine and Health , University of Leeds , Leeds , UK
| | - John E Blundell
- b School of Psychology, Faculty of Medicine and Health , University of Leeds , Leeds , UK
| |
Collapse
|
26
|
Shi YC, Ip CK, Reed F, Sarruf DA, Wulff BS, Herzog H. Y5 receptor signalling counteracts the anorectic effects of PYY3-36 in diet-induced obese mice. J Neuroendocrinol 2017; 29. [PMID: 28485050 DOI: 10.1111/jne.12483] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 05/03/2017] [Accepted: 05/03/2017] [Indexed: 12/14/2022]
Abstract
Peptide YY 3-36 (PYY3-36) is known as a critical satiety factor that reduces food intake both in rodents and humans. Although the anorexic effect of PYY3-36 is assumed to be mediated mainly by the Y2 receptor, the involvement of other Y-receptors in this process has never been conclusively resolved. Amongst them, the Y5 receptor (Y5R) is the most likely candidate to also be a target for PYY3-36, which is considered to counteract the anorectic effects of Y2R activation. In the present study, we show that short-term treatment of diet-induced obese wild-type (WT) and Y5R knockout mice (Y5KO) with PYY3-36 leads to a significantly reduced food intake in both genotypes, which is more pronounced in Y5R KO mice. Interestingly, chronic PYY3-36 infusion via minipumps to WT mice causes an increased cumulative food intake, which is associated with increased body weight gain. By contrast, lack of Y5R reversed this effect. Consistent with the observed increased body weight and fat mass in WT-treated mice, glucose tolerance was also impaired by chronic PYY3-36 treatment. Again, this was less affected in Y5KO mice, suggestive of a role of Y5R in the regulation of glucose homeostasis. Taken together, our data suggest that PYY3-36 mediated signalling via Y5 receptors may counteract the anorectic effects that it mediates via the Y2 receptor (Y2R), consequently lowering bodyweight in the absence of Y5 signalling. These findings open the potential of combination therapy using PYY3-36 and Y5R antagonists to enhance the food intake reducing effects of PYY3-36.
Collapse
Affiliation(s)
- Y-C Shi
- Neuroscience Division, Garvan Institute of Medical Research, St Vincent's Hospital, Sydney, NSW, Australia
- Faculty of Medicine, UNSW Australia, Sydney, NSW, Australia
| | - C K Ip
- Neuroscience Division, Garvan Institute of Medical Research, St Vincent's Hospital, Sydney, NSW, Australia
| | - F Reed
- Neuroscience Division, Garvan Institute of Medical Research, St Vincent's Hospital, Sydney, NSW, Australia
| | - D A Sarruf
- Incretin and Obesity Research, Novo Nordisk, Maaloev, Denmark
| | - B S Wulff
- Incretin and Obesity Research, Novo Nordisk, Maaloev, Denmark
| | - H Herzog
- Neuroscience Division, Garvan Institute of Medical Research, St Vincent's Hospital, Sydney, NSW, Australia
- Faculty of Medicine, UNSW Australia, Sydney, NSW, Australia
| |
Collapse
|
27
|
Guida C, Stephen S, Guitton R, Ramracheya RD. The Role of PYY in Pancreatic Islet Physiology and Surgical Control of Diabetes. Trends Endocrinol Metab 2017; 28:626-636. [PMID: 28533020 DOI: 10.1016/j.tem.2017.04.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 04/27/2017] [Indexed: 12/30/2022]
Abstract
Bariatric surgery in obese individuals leads to rapid and lasting remission of type 2 diabetes (T2D). This phenomenon occurs independently of weight loss possibly via a combination of factors. The incretin hormone GLP-1 has so far been recognised as a critical factor. However, recent data have indicated that elevation in another gut hormone, peptide tyrosine tyrosine (PYY), may drive the beneficial effects of surgery. Here we discuss recent findings on PYY-mediated control of glucose homeostasis and its role in diabetes, in the context of what is known for GLP-1. Identification of factors that increase the expression of PYY following bariatric surgery and elucidation of its role in diabetes reversal may have clinical relevance as a nonsurgical therapy for T2D.
Collapse
Affiliation(s)
- Claudia Guida
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, OX37LJ Oxford, UK
| | - Sam Stephen
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, OX37LJ Oxford, UK
| | - Romain Guitton
- Angers University Hospital, 18 Avenue du Général Patton, 49000 Angers, France
| | - Reshma D Ramracheya
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, OX37LJ Oxford, UK.
| |
Collapse
|
28
|
Statistical Methods for Latent Class Quantitative Trait Loci Mapping. Genetics 2017; 206:1309-1317. [PMID: 28550015 PMCID: PMC5500132 DOI: 10.1534/genetics.117.203885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 05/18/2017] [Indexed: 11/18/2022] Open
Abstract
Identifying the genetic basis of complex traits is an important problem with the potential to impact a broad range of biological endeavors. A number of effective statistical methods are available for quantitative trait loci (QTL) mapping that allow for the efficient identification of multiple, potentially interacting, loci under a variety of experimental conditions. Although proven useful in hundreds of studies, the majority of these methods assumes a single model common to each subject, which may reduce power and accuracy when genetically distinct subclasses exist. To address this, we have developed an approach to enable latent class QTL mapping. The approach combines latent class regression with stepwise variable selection and traditional QTL mapping to estimate the number of subclasses in a population, and to identify the genetic model that best describes each subclass. Simulations demonstrate good performance of the method when latent classes are present as well as when they are not, with accurate estimation of QTL. Application of the method to case studies of obesity and diabetes in mouse gives insight into the genetic basis of related complex traits.
Collapse
|
29
|
Schiess S, Platz S, Kemper M, Schreiner M, Mewis I, Rohn S, Bumke-Vogt C, Pivovarova O, Pfeiffer AF. Oral administration of nasturtium affects peptide YY secretion in male subjects. Mol Nutr Food Res 2017; 61. [DOI: 10.1002/mnfr.201600886] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2016] [Revised: 01/22/2017] [Accepted: 02/15/2017] [Indexed: 12/21/2022]
Affiliation(s)
- Sonja Schiess
- German Institute of Human Nutrition Potsdam-Rehbrücke; Department of Clinical Nutrition; Arthur-Scheunert-Allee 114-116 D-14558 Nuthetal Germany
| | - Stefanie Platz
- University of Hamburg; HAMBURG SCHOOL OF FOOD SCIENCE; Institute of Food Chemistry; Grindelallee 117 D-20146 Hamburg Germany
| | - Margrit Kemper
- German Institute of Human Nutrition Potsdam-Rehbrücke; Department of Clinical Nutrition; Arthur-Scheunert-Allee 114-116 D-14558 Nuthetal Germany
- Charité University Medicine; Department of Endocrinology; Diabetes and Nutrition; Campus Benjamin Franklin; Hindenburgdamm 30 D-12203 Berlin Berlin Germany
- German Center for Diabetes Research (DZD); 85764 München-Neuherberg Germany
| | - Monika Schreiner
- Leibniz Institute of Vegetable and Ornamental Crops; Großbeeren/Erfurt e.V.; Department Plant Quality; Theodor-Echtermeyer-Weg 1 D-14979 Großbeeren Germany
| | - Inga Mewis
- Leibniz Institute of Vegetable and Ornamental Crops; Großbeeren/Erfurt e.V.; Department Plant Quality; Theodor-Echtermeyer-Weg 1 D-14979 Großbeeren Germany
| | - Sascha Rohn
- University of Hamburg; HAMBURG SCHOOL OF FOOD SCIENCE; Institute of Food Chemistry; Grindelallee 117 D-20146 Hamburg Germany
| | - Christiane Bumke-Vogt
- Leibniz Institute of Vegetable and Ornamental Crops; Großbeeren/Erfurt e.V.; Department Plant Quality; Theodor-Echtermeyer-Weg 1 D-14979 Großbeeren Germany
| | - Olga Pivovarova
- German Institute of Human Nutrition Potsdam-Rehbrücke; Department of Clinical Nutrition; Arthur-Scheunert-Allee 114-116 D-14558 Nuthetal Germany
- Charité University Medicine; Department of Endocrinology; Diabetes and Nutrition; Campus Benjamin Franklin; Hindenburgdamm 30 D-12203 Berlin Berlin Germany
- German Center for Diabetes Research (DZD); 85764 München-Neuherberg Germany
| | - Andreas F.H. Pfeiffer
- German Institute of Human Nutrition Potsdam-Rehbrücke; Department of Clinical Nutrition; Arthur-Scheunert-Allee 114-116 D-14558 Nuthetal Germany
- Charité University Medicine; Department of Endocrinology; Diabetes and Nutrition; Campus Benjamin Franklin; Hindenburgdamm 30 D-12203 Berlin Berlin Germany
- German Center for Diabetes Research (DZD); 85764 München-Neuherberg Germany
| |
Collapse
|
30
|
Khan D, Vasu S, Moffett RC, Irwin N, Flatt PR. Islet distribution of Peptide YY and its regulatory role in primary mouse islets and immortalised rodent and human beta-cell function and survival. Mol Cell Endocrinol 2016; 436:102-13. [PMID: 27465830 DOI: 10.1016/j.mce.2016.07.020] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 07/18/2016] [Accepted: 07/19/2016] [Indexed: 12/25/2022]
Abstract
Recent evidence suggests that the classic gut peptide, Peptide YY (PYY), could play a fundamental role in endocrine pancreatic function. In the present study expression of PYY and its NPY receptors on mouse islets and immortalised rodent and human beta-cells was examined together with the effects of both major circulating forms of PYY, namely PYY(1-36) and PYY(3-36), on beta-cell function, murine islet adaptions to insulin deficiency/resistance, as well as direct effects on cultured beta-cell proliferation and apoptosis. In vivo administration of PYY(3-36), but not PYY(1-36), markedly (p < 0.05) decreased food intake in overnight fasted mice. Neither form of PYY affected glucose disposal or insulin secretion following an i.p. glucose challenge. However, in vitro, PYY(1-36) and PYY(3-36) inhibited (p < 0.05 to p < 0.001) glucose, alanine and GLP-1 stimulated insulin secretion from immortalised rodent and human beta-cells, as well as isolated mouse islets, by impeding alterations in membrane potential, [Ca(2+)]i and elevations of cAMP. Mice treated with multiple low dose streptozotocin presented with severe (p < 0.01) loss of beta-cell mass accompanied by notable increases (p < 0.001) in alpha and PP cell numbers. In contrast, hydrocortisone-induced insulin resistance increased islet number (p < 0.01) and beta-cell mass (p < 0.001). PYY expression was consistently observed in alpha-, PP- and delta-, but not beta-cells. Streptozotocin decreased islet PYY co-localisation with PP (p < 0.05) and somatostatin (p < 0.001), whilst hydrocortisone increased PYY co-localisation with glucagon (p < 0.05) in mice. More detailed in vitro investigations revealed that both forms of PYY augmented (p < 0.05 to p < 0.01) immortalised human and rodent beta-cell proliferation and protected against streptozotocin-induced cytotoxicity, to a similar or superior extent as the well characterised beta-cell proliferative and anti-apoptotic agent GLP-1. Taken together, these data highlight the significance and potential offered by modulation of pancreatic islet NPY receptor signalling pathways for preservation of beta-cell mass in diabetes.
Collapse
Affiliation(s)
- Dawood Khan
- SAAD Centre for Pharmacy and Diabetes, University of Ulster, Coleraine, Northern Ireland, UK
| | - Srividya Vasu
- SAAD Centre for Pharmacy and Diabetes, University of Ulster, Coleraine, Northern Ireland, UK
| | - R Charlotte Moffett
- SAAD Centre for Pharmacy and Diabetes, University of Ulster, Coleraine, Northern Ireland, UK
| | - Nigel Irwin
- SAAD Centre for Pharmacy and Diabetes, University of Ulster, Coleraine, Northern Ireland, UK.
| | - Peter R Flatt
- SAAD Centre for Pharmacy and Diabetes, University of Ulster, Coleraine, Northern Ireland, UK
| |
Collapse
|
31
|
Ramracheya RD, McCulloch LJ, Clark A, Wiggins D, Johannessen H, Olsen MK, Cai X, Zhao CM, Chen D, Rorsman P. PYY-Dependent Restoration of Impaired Insulin and Glucagon Secretion in Type 2 Diabetes following Roux-En-Y Gastric Bypass Surgery. Cell Rep 2016; 15:944-950. [PMID: 27117413 PMCID: PMC5063952 DOI: 10.1016/j.celrep.2016.03.091] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Revised: 02/18/2016] [Accepted: 03/25/2016] [Indexed: 02/07/2023] Open
Abstract
Roux-en-Y gastric bypass (RYGB) is a weight-reduction procedure resulting in rapid resolution of type 2 diabetes (T2D). The role of pancreatic islet function in this restoration of normoglycemia has not been fully elucidated. Using the diabetic Goto-Kakizaki (GK) rat model, we demonstrate that RYGB restores normal glucose regulation of glucagon and insulin secretion and normalizes islet morphology. Culture of isolated islets with serum from RYGB animals mimicked these effects, implicating a humoral factor. These latter effects were reversed following neutralization of the gut hormone peptide tyrosine tyrosine (PYY) but persisted in the presence of a glucagon-like peptide-1 (GLP-1) receptor antagonist. The effects of RYGB on secretion were replicated by chronic exposure of diabetic rat islets to PYY in vitro. These findings indicate that the mechanism underlying T2D remission may be mediated by PYY and suggest that drugs promoting PYY release or action may restore pancreatic islet function in T2D. Roux-en-Y gastric bypass rapidly restores islet function and morphology in diabetic GK rats The effects of RYGB on islet function are mediated by the gut hormone PYY and not GLP-1 In vitro PYY application to diabetic islets restores insulin and glucagon secretion
Collapse
Affiliation(s)
- Reshma D Ramracheya
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, Oxon OX3 7LE, UK.
| | - Laura J McCulloch
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, Oxon OX3 7LE, UK
| | - Anne Clark
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, Oxon OX3 7LE, UK
| | - David Wiggins
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, Oxon OX3 7LE, UK
| | - Helene Johannessen
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, 7491 Trondheim, Norway
| | - Magnus Kringstad Olsen
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, 7491 Trondheim, Norway
| | - Xing Cai
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, 7491 Trondheim, Norway
| | - Chun-Mei Zhao
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, 7491 Trondheim, Norway
| | - Duan Chen
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, 7491 Trondheim, Norway
| | - Patrik Rorsman
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, Oxon OX3 7LE, UK; Metabolic Research, Department of Physiology, Institute of Neuroscience and Physiology, University of Goteborg, Box 432, 40530 Göteborg, Sweden
| |
Collapse
|
32
|
Olsen J, Kofoed J, Østergaard S, Wulff BS, Nielsen FS, Jorgensen R. Metabolism of peptide YY 3-36 in Göttingen mini-pig and rhesus monkey. Peptides 2016; 78:59-67. [PMID: 26774588 DOI: 10.1016/j.peptides.2016.01.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 01/04/2016] [Accepted: 01/11/2016] [Indexed: 11/16/2022]
Abstract
Peptide YY 3-36-amide (PYY3-36) is a peptide hormone, which is known to decrease appetite and food-intake by activation of the Y2 receptor. The current studies were designed to identify the metabolites of PYY3-36 in mini-pig and rhesus monkey. Plasma samples were analyzed by high resolution LC-MS (and MS/MS) in order to unambiguously identify the metabolites of PYY3-36. In summary, the metabolism of PYY3-36 was similar in mini-pig and rhesus monkey. Several metabolites were identified and PYY3-34 was identified at the highest levels in plasma. In addition, mini-pigs were also dosed with PYY1-36-amide, PYY3-35, PYY3-34 and [N-methyl 34Q]-PYY3-36-amide in order to investigate the mechanisms by which PYY was metabolized. PYY3-35 was rapidly converted to PYY3-34 whereas dosing of PYY3-34 to mini-pigs only showed circulating degradation products at low levels, i.e., PYY3-34 was metabolically more stable than PYY3-36 and PYY3-35. [N-methyl 34Q]-PYY3-36-amide was hypothesized to be stable toward cleavage between 34Q and 35R and after i.v. administration to mini-pigs, one major cleavage product was identified as [N-methyl 34Q]-PYY3-35. Overall, this showed that cleavage between 35R and 36Y was possible as well as between 34Q and 35R (as shown for PYY3-35), which indicated that metabolism of PYY3-36 to PYY3-34 may be a two-step process. PYY1-36 was also dosed to mini-pigs, which showed that PYY1-36 was metabolized in the C-terminal as PYY3-36. The overall degradation pattern of PYY1-36 was more complex due to the simultaneous enzymatic degradation in the N-terminal to form PYY2-34/36 and PYY3-34/36. In vitro incubations with heparin stabilized plasma showed that PYY3-36 was degraded with a half-life of 175 min, whereas incubations with PYY3-35 (half-life of 6 min) showed a rapid formation of PYY3-34. In conclusion, the present studies showed that PYY3-36 underwent enzymatic degradation in the C-terminal part and that the major circulating metabolite was PYY3-34. Furthermore, it may be a sequential two-step process leading to the formation of PYY3-35 and subsequently the metabolically more stable PYY3-34.
Collapse
Affiliation(s)
- Jørgen Olsen
- Discovery ADME, Novo Nordisk A/S, Måløv, Denmark.
| | - Jacob Kofoed
- Protein & Peptide Chemistry 3, Novo Nordisk A/S, Måløv, Denmark
| | | | | | | | - Rasmus Jorgensen
- Histology & Diabetes Pharmacology Novo Nordisk A/S, Måløv, Denmark
| |
Collapse
|
33
|
Richards L, Li M, van Esch B, Garssen J, Folkerts G. The effects of short-chain fatty acids on the cardiovascular system. PHARMANUTRITION 2016. [DOI: 10.1016/j.phanu.2016.02.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
34
|
Abstract
The gastrointestinal hormone peptide tyrosine tyrosine 3-36 (PYY(3-36)) has attained broad recognition with respect to its involvement in energy homeostasis and the control of food intake. It is mainly secreted by distal intestinal enteroendocrine L-cells in response to eating and exerts neurally mediated, paracrine and endocrine effects on various target organs. In addition to its gastrointestinal effects, PYY(3-36) has long been known to inhibit food intake. Recent closer examination of the effects of PYY(3-36) revealed that this gut-derived peptide also influences a wide spectrum of behavioral and cognitive functions that are pivotal for basic processes of perception and judgment, including central information processing, salience learning, working memory, and behavioral responding to novelty. Here, we review the effects of PYY(3-36) that go beyond food intake and provide a conceptual framework suggesting that several apparently unrelated behavioral actions of PYY(3-36) may actually reflect different manifestations of modulating the central dopamine system.
Collapse
|
35
|
Kuehl PJ, Boyden T, Dobry DE, Doyle-Eisele M, Friesen DT, McDonald JD, Murri BG, Vodak DT, Lyon DK. Inhaled PYY(3-36) dry-powder formulation for appetite suppression. Drug Dev Ind Pharm 2015; 42:150-156. [PMID: 26006332 DOI: 10.3109/03639045.2015.1036067] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
OBJECTIVE Peptide YY3-36 [PYY(3-36)] has shown efficacy in appetite suppression when dosed by injection modalities (intraperitoneal (IP)/subcutaneous). Transitioning to needle-free delivery, towards inhalation, often utilizes systemic pharmacokinetics as a key endpoint to compare different delivery methods and doses. Systemic pharmacokinetics were evaluated for PYY3-36 when delivered by IP, subcutaneous, and inhalation, the systemic pharmacokinetics were then used to select doses in an appetite suppression pharmacodynamic study. METHODS Dry-powder formulations were manufactured by spray drying and delivered to mice via nose only inhalation. The systemic plasma, lung tissue, and bronchoalveolar lavage fluid pharmacokinetics of different inhalation doses of PYY(3-36) were compared to IP and subcutaneous efficacious doses. Based on these pharmacokinetic data, inhalation doses of 70:30 PYY(3-36):Dextran T10 were evaluated in a mouse model of appetite suppression and compared to IP and subcutaneous data. RESULTS Inhalation pharmacokinetic studies showed that plasma exposure was similar for a 2 × higher inhalation dose when compared to subcutaneous and IP delivery. Inhalation doses of 0.22 and 0.65 mg/kg were for efficacy studies. The results showed a dose-dependent (not dose proportional) decrease in food consumption over 4 h, which is similar to IP and subcutaneous delivery routes. CONCLUSIONS The pharmacokinetic and pharmacodynamics results substantiate the ability of pharmacokinetic data to inform pharmacodynamics dose selection for inhalation delivery of the peptide PYY(3-36). Additionally, engineered PYY(3-36):Dextran T10 particles delivered to the respiratory tract show promise as a non-invasive therapeutic for appetite suppression.
Collapse
Affiliation(s)
- Philip J Kuehl
- a Lovelace Respiratory Research Institute , Albuquerque , NM , USA
| | | | | | | | | | - Jacob D McDonald
- a Lovelace Respiratory Research Institute , Albuquerque , NM , USA
| | | | | | | |
Collapse
|
36
|
The role of short chain fatty acids in appetite regulation and energy homeostasis. Int J Obes (Lond) 2015; 39:1331-8. [PMID: 25971927 PMCID: PMC4564526 DOI: 10.1038/ijo.2015.84] [Citation(s) in RCA: 439] [Impact Index Per Article: 43.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Revised: 12/23/2014] [Accepted: 04/19/2015] [Indexed: 12/24/2022]
Abstract
Over the last 20 years there has been an increasing interest in the influence of the gastrointestinal tract on appetite regulation. Much of the focus has been on the neuronal and hormonal relationship between the gastrointestinal tract and the brain. There is now mounting evidence that the colonic microbiota and their metabolic activity have a significant role in energy homeostasis. The supply of substrate to the colonic microbiota has a major impact on the microbial population and the metabolites they produce, particularly short chain fatty acids (SCFAs). SCFAs are produced when non-digestible carbohydrates, namely dietary fibres and resistant starch, undergo fermentation by the colonic microbiota. Both the consumption of fermentable carbohydrates and the administration of SCFAs have been reported to result in a wide range of health benefits including improvements in body composition, glucose homeostasis, blood lipid profiles and reduced body weight and colon cancer risk. However, published studies tend to report the effects that fermentable carbohydrates and SCFAs have on specific tissues and metabolic processes, and fail to explain how these local effects translate into systemic effects and the mitigation of disease risk. Moreover, studies tend to investigate SCFAs collectively and neglect to report the effects associated with individual SCFAs. Here, we bring together the recent evidence and suggest an overarching model for the effects of SCFAs on one of their beneficial aspects: appetite regulation and energy homeostasis.
Collapse
|
37
|
Henry KE, Elfers CT, Burke RM, Chepurny OG, Holz GG, Blevins JE, Roth CL, Doyle RP. Vitamin B12 conjugation of peptide-YY(3-36) decreases food intake compared to native peptide-YY(3-36) upon subcutaneous administration in male rats. Endocrinology 2015; 156:1739-49. [PMID: 25658456 PMCID: PMC4398759 DOI: 10.1210/en.2014-1825] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Challenges to peptide-based therapies include rapid clearance, ready degradation by hydrolysis/proteolysis, and poor intestinal uptake and/or a need for blood brain barrier transport. This work evaluates the efficacy of conjugation of vitamin B12 (B12) on sc administered peptide tyrosine tyrosine (PYY)(3-36) function. In the current experiments, a B12-PYY(3-36) conjugate was tested against native PYY(3-36), and an inactive conjugate B12-PYYC36 (null control) in vitro and in vivo. In vitro experiments demonstrated similar agonism for the neuropeptide Y2 receptor by the B12-PYY(3-36) conjugate (EC50 26.5 nM) compared with native PYY(3-36) (EC50 16.0 nM), with the null control having an EC50 of 1.8 μM. In vivo experiments were performed in young adult male Sprague Dawley rats (9 wk). Daily treatments were delivered sc in five 1-hour pulses, each pulse delivering 5-10 nmol/kg, by implanted microinfusion pumps. Increases in hindbrain Fos expression were comparable 90 minutes after B12-PYY(3-36) or PYY3-36 injection relative to saline or B12-PYYC36. Food intake was reduced during a 5-day treatment for both B12-PYY(3-36)- (24%, P = .001) and PYY(3-36)-(13%, P = .008) treated groups relative to baseline. In addition, reduction of food intake after the three dark cycle treatment pulses was more consistent with B12-PYY(3-36) treatment (-26%, -29%, -27%) compared with the PYY(3-36) treatment (-3%, -21%, -16%), and B12-PYY(3-36) generated a significantly longer inhibition of food intake vs. PYY(3-36) treatment after the first two pulses (P = .041 and P = .036, respectively). These findings demonstrate a stronger, more consistent, and longer inhibition of food intake after the pulses of B12-PYY(3-36) conjugate compared with the native PYY(3-36).
Collapse
Affiliation(s)
- Kelly E Henry
- Department of Chemistry (K.E.H., R.M.B., R.P.D.), Center for Science and Technology, Syracuse University, Syracuse, New York 13244; Center for Integrative Brain Research (C.T.E., C.L.R.), Division of Endocrinology, Seattle Children's Research Institute, Seattle, Washington 98101; Departments of Medicine (O.G.C., G.G.H., R.P.D.) and Pharmacology (G.G.H.), State University of New York, Upstate Medical University, Syracuse, New York 13210; Research and Development Service (J.E.B.), Veterans Affairs Puget Sound Health Care System, Seattle, Washington 98108; Department of Medicine (J.E.B.), Division of Metabolism, Endocrinology, and Nutrition, University of Washington, Seattle, Washington 98195; and Division of Endocrinology (C.L.R.), Department of Pediatrics, University of Washington, Seattle, Washington 98105
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Abstract
As obesity continues to be a global epidemic, research into the mechanisms of hunger and satiety and how those signals act to regulate energy homeostasis persists. Peptide YY (PYY) is an acute satiety signal released upon nutrient ingestion and has been shown to decrease food intake when administered exogenously. More recently, investigators have studied how different factors influence PYY release and circulating levels in humans. Some of these factors include exercise, macronutrient composition of the diet, body-weight status, adiposity levels, sex, race and ageing. The present article provides a succinct and comprehensive review of the recent literature published on the different factors that influence PYY release and circulating levels in humans. Where human data are insufficient, evidence in animal or cell models is summarised. Additionally, the present review explores the recent findings on PYY responses to different dietary fatty acids and how this new line of research will make an impact on future studies on PYY. Human demographics, such as sex and age, do not appear to influence PYY levels. Conversely, adiposity or BMI, race and acute exercise all influence circulating PYY levels. Both dietary fat and protein strongly stimulate PYY release. Furthermore, MUFA appear to result in a smaller PYY response compared with SFA and PUFA. PYY levels appear to be affected by acute exercise, macronutrient composition, adiposity, race and the composition of fatty acids from dietary fat.
Collapse
|
39
|
Saeed S, Bech PR, Hafeez T, Alam R, Falchi M, Ghatei MA, Bloom SR, Arslan M, Froguel P. Changes in levels of peripheral hormones controlling appetite are inconsistent with hyperphagia in leptin-deficient subjects. Endocrine 2014; 45:401-8. [PMID: 23824601 DOI: 10.1007/s12020-013-0009-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2013] [Accepted: 06/22/2013] [Indexed: 01/05/2023]
Abstract
Congenital leptin deficiency, a rare genetic disorder due to a homozygous mutation in the leptin gene (LEP), is accompanied by extreme obesity and hyperphagia. A number of gastrointestinal hormones have been shown to critically regulate food intake but their physiological role in hyperphagic response in congenital leptin deficiency has not been elucidated. This study is the first to evaluate the fasting and postprandial profiles of gut-derived hormones in homozygous and heterozygous carriers of LEP mutation. The study subjects from two consanguineous families consisted of five homozygous and eight heterozygous carriers of LEP mutation, c.398delG. Ten wild-type normal-weight subjects served as controls. Fasting and 1-h postprandial plasma ghrelin, glucagon-like peptide (GLP) 1, peptide YY (PYY), leptin and insulin levels were measured by immunoassays. Fasting plasma ghrelin levels in homozygotes remained remarkably unchanged following food consumption (P = 0.33) in contrast to a significant decline in heterozygous (P < 0.03) and normal (P < 0.02) subjects. A significant postprandial increase in PYY was observed in heterozygous (P < 0.02) and control subjects (P < 0.01), but not in the homozygous group (P = 0.22). A postprandial rise in GLP-1 levels was significant (P < 0.02) in all groups. Interestingly, fasting leptin levels in heterozygotes were not significantly different from controls and did not change significantly following meal. Our results demonstrate that gut hormones play little or no physiological role in driving the hyperphagic response of leptin-deficient subjects. In contrast, fasting and postprandial levels of gut hormones in heterozygous mutation carriers were comparable to those of normal-weight controls.
Collapse
Affiliation(s)
- Sadia Saeed
- Department of Genomics of Common Disease, Hammersmith Hospital, Imperial College London, Burlington-Danes Building, Du Cane Road, London, W12 0NN, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Abstract
Over the past 30 years, it has been established that hormones produced by the gut, pancreas, and adipose tissue are key players in the control of body weight. These hormones act through a complex neuroendocrine system, including the hypothalamus, to regulate metabolism and energy homeostasis. In obesity, this homeostatic balance is disrupted, either through alterations in the levels of these hormones or through resistance to their actions. Alterations in gut hormone secretion following gastric bypass surgery are likely to underlie the dramatic and persistent loss of weight following this procedure, as well as the observed amelioration in type 2 diabetes mellitus. Medications based on the gut hormone GLP-1 are currently in clinical use to treat type 2 diabetes mellitus and have been shown to produce weight loss. Further therapies for obesity based on other gut hormones are currently in development.
Collapse
Affiliation(s)
- Rebecca Scott
- Division of Diabetes, Endocrinology, Metabolism, Hammersmith Hospital, Imperial College London, London, United Kingdom.
| | | | | |
Collapse
|
41
|
Loktev AV, Jackson PK. Neuropeptide Y family receptors traffic via the Bardet-Biedl syndrome pathway to signal in neuronal primary cilia. Cell Rep 2013; 5:1316-29. [PMID: 24316073 DOI: 10.1016/j.celrep.2013.11.011] [Citation(s) in RCA: 153] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Revised: 10/21/2013] [Accepted: 11/06/2013] [Indexed: 10/25/2022] Open
Abstract
Human monogenic obesity syndromes, including Bardet-Biedl syndrome (BBS), implicate neuronal primary cilia in regulation of energy homeostasis. Cilia in hypothalamic neurons have been hypothesized to sense and regulate systemic energy status, but the molecular mechanism of this signaling remains unknown. Here, we report a comprehensive localization screen of 42 G-protein-coupled receptors (GPCR) revealing seven ciliary GPCRs, including the neuropeptide Y (NPY) receptors NPY2R and NPY5R. We show that mice modeling BBS disease or obese tubby mice fail to localize NPY2R to cilia in the hypothalamus and that BBS mutant mice fail to activate c-fos or decrease food intake in response to the NPY2R ligand PYY3-36. We find that cells with ciliary NPY2R show augmented PYY3-36-dependent cAMP signaling. Our data demonstrate that ciliary targeting of NPY receptors is important for controlling energy balance in mammals, revealing a physiologically defined ligand-receptor pathway signaling within neuronal cilia.
Collapse
Affiliation(s)
- Alexander V Loktev
- Research Oncology, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Peter K Jackson
- Research Oncology, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA.
| |
Collapse
|
42
|
Administration of the Y2 receptor agonist PYY3-36 in mice induces multiple behavioral changes relevant to schizophrenia. Neuropsychopharmacology 2013; 38:2446-55. [PMID: 23748226 PMCID: PMC3799064 DOI: 10.1038/npp.2013.146] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Revised: 05/24/2013] [Accepted: 06/05/2013] [Indexed: 12/22/2022]
Abstract
Functional changes in neuropeptide Y (NPY) signaling at the Y2 receptor subtype have been widely implicated in stress-related neuropsychiatric illnesses such as depression and anxiety disorders. Altered Y2 receptor signaling may also play a role in the precipitation of behavioral and cognitive symptoms associated with schizophrenia. To seek preclinical evidence for this possibility, we explored the functional consequences of treatment with the selective Y2 receptor agonist PYY(3-36) using translational tests for the assessment of schizophrenia-relevant behavioral and cognitive deficits in mice. We found that acute systemic administration of PYY(3-36) at a low dose (1 μg/100 g body weight) or high dose (20 μg/100 g body weight) profoundly impaired social interaction without affecting innate anxiety. PYY(3-36) treatment at the high dose further led to a disruption of sensorimotor gating in the form of prepulse inhibition deficiency. This effect was fully antagonized by acute treatment with the preferential dopamine D2 receptor antagonist haloperidol, but not with clozapine. In addition, both doses of PYY(3-36) impaired selective associative learning in the latent inhibition paradigm and spatial working memory in a matching-to-position water maze test. The wide range of abnormalities induced by PYY(3-36) suggests that signaling at the Y2 subtype of NPY receptors is critical for a number of behavioral and cognitive functions, some of which are highly relevant to schizophrenia and related psychotic disorders. At least some of the behavioral deficits induced by augmentation of Y2 receptor signaling may involve increased dopaminergic activity.
Collapse
|
43
|
Gut hormones as therapeutic agents in treatment of diabetes and obesity. Curr Opin Pharmacol 2013; 13:996-1001. [PMID: 24060699 DOI: 10.1016/j.coph.2013.09.005] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Revised: 08/19/2013] [Accepted: 09/04/2013] [Indexed: 12/24/2022]
Abstract
Obesity and Type 2 Diabetes Mellitus (T2DM) present an ever-increasing threat to global health. Although bariatric surgery is an effective treatment, it cannot be applied to the vast majority of patients. The beneficial effects of bariatric surgery are related to complex alterations in the secretion of gut hormones. By recapitulation of the changes of gut hormone secretion after bariatric surgery, drugs based on gut hormones represent an exciting possibility for the treatment of T2DM and obesity. We review the rapidly emerging role of GLP-1 based treatments as well as the future for new drugs based on other gut hormones such as GIP, ghrelin, oxyntomodulin and peptide YY.
Collapse
|
44
|
Tatarkiewicz K, Belanger P, Gu G, Parkes D, Roy D. No evidence of drug-induced pancreatitis in rats treated with exenatide for 13 weeks. Diabetes Obes Metab 2013; 15:417-26. [PMID: 23163898 PMCID: PMC3654567 DOI: 10.1111/dom.12040] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Revised: 09/29/2012] [Accepted: 11/12/2012] [Indexed: 12/18/2022]
Abstract
AIMS The potential association of glucagon-like peptide receptor agonists (GLP-1RAs) with the development of pancreatitis or pancreatic malignancies in patients with diabetes has been suggested. This study evaluated the long-term effects of the GLP-1RA exenatide on pancreatic exocrine structure and function in the Zucker diabetic fatty (ZDF) rat model of type 2 diabetes. METHODS Rats received subcutaneous twice-daily injections of 0 (control), 6, 40 and 250 µg/kg/day exenatide for 3 months. Clinical signs, body and pancreas weight, food consumption, HbA1c, fasting serum amylase, lipase, glucose and insulin concentrations were evaluated during treatment and after a 28-day off-drug period to assess the reversibility of any observed effects. Morphometric analysis of pancreatic ductal cell proliferation and apoptosis were performed. RESULTS Plasma exenatide concentrations were several-fold higher than therapeutic levels observed in humans. No exenatide-related effects were observed on clinical signs, lipase concentration, pancreatic weight, pancreatic histology, ductal cell proliferation or apoptosis. Exenatide improved animal survival, physical condition, glucose concentrations and HbA1c, decreased food intake, and increased serum insulin concentration. Total amylase concentrations, although within normal ranges, were slightly higher in exenatide-treated rats; following the off-drug period, total amylase concentrations were comparable in treated and untreated rats. Exenatide-related minimal-to-moderate islet hypertrophy was observed at doses ≥6 µg/kg/day, with dose-related increases in incidence and degree. These changes were still present after the off-drug period. CONCLUSIONS Chronic administration of exenatide in ZDF rats resulted in the expected metabolic benefits and improved animal survival, with no adverse effects noted on pancreatic exocrine structure and function.
Collapse
|
45
|
Li B, Zhou X, Wu J, Zhou H. From gut changes to type 2 diabetes remission after gastric bypass surgeries. Front Med 2013; 7:191-200. [PMID: 23553469 DOI: 10.1007/s11684-013-0258-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Accepted: 01/14/2013] [Indexed: 12/18/2022]
Abstract
Increasing evidence suggests that the gut may influence the host's metabolism and ultimately change the outcomes of type 2 diabetes mellitus (T2DM). We review the evidence on the relationship between the gut and T2DM remission after gastric bypass surgery, and discuss the potential mechanisms underlying the above relationship: gut anatomical rearrangement, microbial composition changes, altered gut cells, and gut hormone modulation. However, the exact changes and their relative importance in the metabolic improvements after gastric bypass surgery remain to be further clarified. Elucidating the precise metabolic mechanisms of T2DM resolution after bypass surgery will help to reveal the molecular mechanisms of pathogenesis, and facilitate the development of novel diagnoses and preventative interventions for this common disease.
Collapse
Affiliation(s)
- Bing Li
- Key Laboratory of Systems Biology, SIBS-Novo Nordisk Translational Research Centre for PreDiabetes, Shanghai Institutes for Biological Sciences, CAS, Shanghai, China
| | | | | | | |
Collapse
|
46
|
Chandarana K, Gelegen C, Irvine EE, Choudhury AI, Amouyal C, Andreelli F, Withers DJ, Batterham RL. Peripheral activation of the Y2-receptor promotes secretion of GLP-1 and improves glucose tolerance. Mol Metab 2013; 2:142-52. [PMID: 24049729 DOI: 10.1016/j.molmet.2013.03.001] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Revised: 03/04/2013] [Accepted: 03/05/2013] [Indexed: 12/15/2022] Open
Abstract
The effect of peptide tyrosine-tyrosine (PYY) on feeding is well established but currently its role in glucose homeostasis is poorly defined. Here we show in mice, that intraperitoneal (ip) injection of PYY3-36 or Y2R agonist improves nutrient-stimulated glucose tolerance and enhances insulin secretion; an effect blocked by peripheral, but not central, Y2R antagonist administration. Studies on isolated mouse islets revealed no direct effect of PYY3-36 on insulin secretion. Bariatric surgery in mice, enterogastric anastomosis (EGA), improved glucose tolerance in wild-type mice and increased circulating PYY and active GLP-1. In contrast, in Pyy-null mice, post-operative glucose tolerance and active GLP-1 levels were similar in EGA and sham-operated groups. PYY3-36 ip increased hepato-portal active GLP-1 plasma levels, an effect blocked by ip Y2R antagonist. Collectively, these data suggest that PYY3-36 therefore acting via peripheral Y2R increases hepato-portal active GLP-1 plasma levels and improves nutrient-stimulated glucose tolerance.
Collapse
Key Words
- AUC, area under the curve
- CNS, central nervous system
- DPP-4, di-peptidyl peptidase-4
- EGA, entero-gastric anastomosis
- GLP-1
- Glucose homeostasis
- HFD, high-fat diet
- ICV, intracerebroventricular
- IPGTT, intraperitoneal glucose tolerance test
- PYY
- PYY, peptide tyrosine–tyrosine
- T2DM, type 2 diabetes mellitus
- WT, wild-type
- Y2-receptor
- Y2R, Y2-receptor
- aCSF, artificial cerebrospinal fluid
- active GLP-1, glucagon-like peptide-1(7-36)amide
- ip, intraperitoneal
Collapse
Affiliation(s)
- Keval Chandarana
- Centre for Obesity Research, Department of Medicine, University College London, Rayne Institute, 5 University Street, WC1E 6JJ, London, UK
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Jones KW, Eller LK, Parnell JA, Doyle-Baker PK, Edwards AL, Reimer RA. Effect of a dairy- and calcium-rich diet on weight loss and appetite during energy restriction in overweight and obese adults: a randomized trial. Eur J Clin Nutr 2013; 67:371-6. [PMID: 23462943 PMCID: PMC3948984 DOI: 10.1038/ejcn.2013.52] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND/OBJECTIVES A diet rich in dairy and calcium (Ca) has been variably associated with improvements in body composition and decreased risk of type 2 diabetes. Our objective was to determine if a dietary pattern high in dairy and Ca improves weight loss and subjective appetite to a greater extent than a low dairy/Ca diet during energy restriction in overweight and obese adults with metabolic syndrome. SUBJECTS/METHODS A total of 49 participants were randomized to one of two treatment groups: Control (low dairy, ≈ 700 mg/day Ca, -500 kcal/day) or Dairy/Ca (high dairy, ≈ 1400 mg/day Ca, -500 kcal/day) for 12 weeks. Body composition, subjective ratings of appetite, food intake, plasma satiety hormones, glycemic response and inflammatory cytokines were measured. RESULTS Control (-2.2 ± 0.5 kg) and Dairy/Ca (-3.3 ± 0.6 kg) had similar weight loss. Based on self-reported energy intake, the percentage of expected weight loss achieved was higher with Dairy/Ca (82.1 ± 19.4%) than Control (32.2 ± 7.7%; P=0.03). Subjects in the Dairy/Ca group reported feeling more satisfied (P=0.01) and had lower dietary fat intake (P=0.02) over 12 weeks compared with Control. Compared with Control, Dairy/Ca had higher plasma levels of peptide tyrosine tyrosine (PYY, P=0.01) during the meal tolerance test at week 12. Monocyte chemoattractant protein-1 was reduced at 30 min with Dairy/Ca compared with Control (P=0.04). CONCLUSIONS In conclusion, a dairy- and Ca-rich diet was not associated with greater weight loss than control. Modest increases in plasma PYY concentrations with increased dairy/Ca intake, however, may contribute to enhanced sensations of satisfaction and reduced dietary fat intake during energy restriction.
Collapse
Affiliation(s)
- K W Jones
- Faculty of Kinesiology, University of Calgary, Calgary, Alberta, Canada
| | | | | | | | | | | |
Collapse
|
48
|
Wu W, Bates MA, Bursian SJ, Flannery B, Zhou HR, Link JE, Zhang H, Pestka JJ. Peptide YY3-36 and 5-hydroxytryptamine mediate emesis induction by trichothecene deoxynivalenol (vomitoxin). Toxicol Sci 2013; 133:186-95. [PMID: 23457120 DOI: 10.1093/toxsci/kft033] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Deoxynivalenol (DON, vomitoxin), a trichothecene mycotoxin produced by Fusarium sp. that frequently occurs in cereal grains, has been associated with human and animal food poisoning. Although a common hallmark of DON-induced toxicity is the rapid onset of emesis, the mechanisms for this adverse effect are not fully understood. Recently, our laboratory has demonstrated that the mink (Neovison vison) is a suitable small animal model for investigating trichothecene-induced emesis. The goal of this study was to use this model to determine the roles of two gut satiety hormones, peptide YY3-36 (PYY3-36) and cholecystokinin (CCK), and the neurotransmitter 5-hydroxytryptamine (5-HT) in DON-induced emesis. Following ip exposure to DON at 0.1 and 0.25mg/kg bw, emesis induction ensued within 15-30min and then persisted up to 120min. Plasma DON measurement revealed that this emesis period correlated with the rapid distribution and clearance of the toxin. Significant elevations in both plasma PYY3-36 (30-60min) and 5-HT (60min) but not CCK were observed during emesis. Pretreatment with the neuropeptide Y2 receptor antagonist JNJ-31020028 attenuated DON- and PYY-induced emesis, whereas the CCK1 receptor antagonist devezapide did not alter DON's emetic effects. The 5-HT3 receptor antagonist granisetron completely suppressed induction of vomiting by DON and the 5-HT inducer cisplatin. Granisetron pretreatment also partially blocked PYY3-36-induced emesis, suggesting a potential upstream role for this gut satiety hormone in 5-HT release. Taken together, the results suggest that both PYY3-36 and 5-HT play contributory roles in DON-induced emesis.
Collapse
Affiliation(s)
- Wenda Wu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P.R. China
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Pedragosa-Badia X, Stichel J, Beck-Sickinger AG. Neuropeptide Y receptors: how to get subtype selectivity. Front Endocrinol (Lausanne) 2013; 4:5. [PMID: 23382728 PMCID: PMC3563083 DOI: 10.3389/fendo.2013.00005] [Citation(s) in RCA: 122] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2012] [Accepted: 01/09/2013] [Indexed: 11/13/2022] Open
Abstract
The neuropeptide Y (NPY) system is a multireceptor/multiligand system consisting of four receptors in humans (hY(1), hY(2), hY(4), hY(5)) and three agonists (NPY, PYY, PP) that activate these receptors with different potency. The relevance of this system in diseases like obesity or cancer, and the different role that each receptor plays influencing different biological processes makes this system suitable for the design of subtype selectivity studies. In this review we focus on the latest findings within the NPY system, we summarize recent mutagenesis studies, structure activity relationship studies, receptor chimera, and selective ligands focusing also on the binding mode of the native agonists.
Collapse
Affiliation(s)
| | | | - Annette G. Beck-Sickinger
- Institute of Biochemistry, Faculty of Biosciences, Pharmacy and Psychology, Universität LeipzigLeipzig, Germany
| |
Collapse
|
50
|
Falor AE, Zobel M, Kaji A, Neville A, De Virgilio C. Admission variables predictive of gangrenous cholecystitis. Am Surg 2012; 22:1648-57. [PMID: 23025944 DOI: 10.1007/s11695-012-0698-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The objective of the present study was to identify admission clinical factors associated with gangrenous cholecystitis (GC) and factors associated with conversion to open cholecystectomy. We retrospectively evaluated 391 patients over a 17-month period who underwent urgent laparoscopic cholecystectomy for a diagnosis of acute cholecystitis. Eighty-nine patients with pathologically proven GC were compared with 302 patients without GC. On multivariable logistic regression, predictors of GC included male gender, white blood cell count greater than 14,000/mm3, heart rate greater than 90 beats per minute, and sodium 135 mg/dL or less. Conversion rate to open cholecystectomy was 7.9 per cent overall, 4 per cent for non-GC, and 19 per cent for GC (odds ratio, 0.2; 95% confidence interval, 0.1 to 0.4; P<0.00001). Conversion was predicted by increasing number of days to surgery, total bilirubin, and white blood cell count. Complication rate was higher in the GC group (10.1 vs 3.6% in the acute cholecystitis group, P=0.01). The increased rate of conversion observed with surgery delay suggests that early laparoscopic cholecystectomy may be preferable in most patients.
Collapse
Affiliation(s)
- Ann E Falor
- Department of Surgery, Harbor-UCLA Medical Center, Torrance, California 90509, USA
| | | | | | | | | |
Collapse
|