1
|
Bhargava R, Luur S, Rodriguez Flores M, Emini M, Prechtl CG, Goldstone AP. Postprandial Increases in Liver-Gut Hormone LEAP2 Correlate with Attenuated Eating Behavior in Adults Without Obesity. J Endocr Soc 2023; 7:bvad061. [PMID: 37287649 PMCID: PMC10243873 DOI: 10.1210/jendso/bvad061] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Indexed: 06/09/2023] Open
Abstract
Background The novel liver-gut hormone liver-expressed antimicrobial peptide-2 (LEAP2) is a centrally acting inverse agonist, and competitive antagonist of orexigenic acyl ghrelin (AG), at the GH secretagogue receptor, reducing food intake in rodents. In humans, the effects of LEAP2 on eating behavior and mechanisms behind the postprandial increase in LEAP2 are unclear, though this is reciprocal to the postprandial decrease in plasma AG. Methods Plasma LEAP2 was measured in a secondary analysis of a previous study. Twenty-two adults without obesity attended after an overnight fast, consuming a 730-kcal meal without or with subcutaneous AG administration. Postprandial changes in plasma LEAP2 were correlated with postprandial changes in appetite, high-energy (HE) or low-energy (LE) food cue reactivity using functional magnetic resonance imaging, ad libitum food intake, and plasma/serum AG, glucose, insulin, and triglycerides. Results Postprandial plasma LEAP2 increased by 24.5% to 52.2% at 70 to 150 minutes, but was unchanged by exogenous AG administration. Postprandial increases in LEAP2 correlated positively with postprandial decreases in appetite, and cue reactivity to HE/LE and HE food in anteroposterior cingulate cortex, paracingulate cortex, frontal pole, and middle frontal gyrus, with similar trend for food intake. Postprandial increases in LEAP2 correlated negatively with body mass index, but did not correlate positively with increases in glucose, insulin, or triglycerides, nor decreases in AG. Conclusions These correlational findings are consistent with a role for postprandial increases in plasma LEAP2 in suppressing human eating behavior in adults without obesity. Postprandial increases in plasma LEAP2 are unrelated to changes in plasma AG and the mediator(s) remain uncertain.
Collapse
Affiliation(s)
- Raghav Bhargava
- PsychoNeuroEndocrinology Research Group, Division of Psychiatry, Department of Brain Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, W12 0NN, UK
| | - Sandra Luur
- PsychoNeuroEndocrinology Research Group, Division of Psychiatry, Department of Brain Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, W12 0NN, UK
| | - Marcela Rodriguez Flores
- PsychoNeuroEndocrinology Research Group, Division of Psychiatry, Department of Brain Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, W12 0NN, UK
| | - Mimoza Emini
- PsychoNeuroEndocrinology Research Group, Division of Psychiatry, Department of Brain Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, W12 0NN, UK
| | - Christina G Prechtl
- School of Public Health, Faculty of Medicine, Imperial College London, St. Mary's Hospital, London, W2 1PG, UK
| | - Anthony P Goldstone
- PsychoNeuroEndocrinology Research Group, Division of Psychiatry, Department of Brain Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, W12 0NN, UK
| |
Collapse
|
2
|
Kehagias D, Georgopoulos N, Habeos I, Lampropoulos C, Mulita F, Kehagias I. The role of the gastric fundus in glycemic control. Hormones (Athens) 2023; 22:151-163. [PMID: 36705877 DOI: 10.1007/s42000-023-00429-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 01/10/2023] [Indexed: 01/28/2023]
Abstract
PURPOSE Ghrelin, one of the most studied gut hormones, is mainly produced by the gastric fundus. Abundant evidence exists from preclinical and clinical studies underlining its contribution to glucose regulation. In the following narrative review, the role of the gastric fundus in glucose regulation is summarized and we investigate whether its resection enhances glycemic control. METHODS An electronic search was conducted in the PubMed® database and in Google Scholar® using a combination of medical subject headings (MeSH). We examined types of metabolic surgery, including, in particular, gastric fundus resection, either as part of laparoscopic sleeve gastrectomy (LSG) or modified laparoscopic gastric bypass with fundus resection (LRYGBP + FR), and the contribution of ghrelin reduction to glucose regulation. RESULTS Fourteen human studies were judged to be eligible and included in this narrative review. Reduction of ghrelin levels after fundus resection might be related to early glycemic improvement before significant weight loss is achieved. Long-term data regarding the role of ghrelin reduction in glucose homeostasis are sparse. CONCLUSION The exact role of ghrelin in achieving glycemic control is still ambiguous. Data from human studies reveal a potential contribution of ghrelin reduction to early glycemic improvement, although further well-designed studies are needed.
Collapse
Affiliation(s)
- Dimitrios Kehagias
- Department of General Surgery, General University Hospital of Patras, 26504, Rio, Greece.
| | - Neoklis Georgopoulos
- Division of Reproductive Endocrinology, Department of Obstetrics and Gynecology, University of Patras Medical School, 26504, Rio, Greece
| | - Ioannis Habeos
- Division of Endocrinology and Diabetes, Department of Internal Medicine, University Hospital of Patras, 26504, Rio, Greece
| | | | - Francesk Mulita
- Department of General Surgery, General University Hospital of Patras, 26504, Rio, Greece
| | - Ioannis Kehagias
- Division of Bariatric and Metabolic Surgery, Department of Surgery, General University Hospital of Patras, 26504, Rio, Greece
| |
Collapse
|
3
|
Zhao Y, Shu Y, Zhao N, Zhou Z, Jia X, Jian C, Jin S. Insulin resistance induced by long-term sleep deprivation in rhesus macaques can be attenuated by Bifidobacterium. Am J Physiol Endocrinol Metab 2022; 322:E165-E172. [PMID: 34843659 DOI: 10.1152/ajpendo.00329.2021] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Long-term sleep deprivation (SD) is a bad lifestyle habit, especially among specific occupational practitioners, characterized by circadian rhythm misalignment and abnormal sleep/wake cycles. SD is closely associated with an increased risk of metabolic disturbance, particularly obesity and insulin resistance. The incretin hormone, glucagon-like peptide-1 (GLP-1), is a critical insulin release determinant secreted by the intestinal L-cell upon food intake. Besides, the gut microbiota participates in metabolic homeostasis and regulates GLP-1 release in a circadian rhythm manner. As a commonly recognized intestinal probiotic, Bifidobacterium has various clinical indications regarding its curative effect. However, few studies have investigated the effect of Bifidobacterium supplementation on sleep disorders. In the present study, we explored the impact of long-term SD on the endocrine metabolism of rhesus monkeys and determined the effect of Bifidobacterium supplementation on the SD-induced metabolic status. Lipid concentrations, body weight, fast blood glucose, and insulin levels increased after SD. Furthermore, after 2 mo of long-term SD, the intravenous glucose tolerance test showed that the glucose metabolism was impaired and the insulin sensitivity decreased. Moreover, 1 mo of Bifidobacterium oral administration significantly reduced blood glucose and attenuated insulin resistance in rhesus macaques. Overall, our results suggested that Bifidobacterium might be used to alleviate SD-induced aberrant glucose metabolism and improve insulin resistance. Also, it might help in better understanding the mechanisms governing the beneficial effects of Bifidobacterium.NEW & NOTEWORTHY Our findings demonstrated that long-term sleep deprivation is closely associated with metabolic syndromes. Bifidobacterium administration showed a superior effect on insulin resistance caused by sleep deprivation. Overall, we provide prevention and treatment methods for long-term sleep deprivation, a bad lifestyle habit among specific occupational practitioners, such as irregular shift workers.
Collapse
Affiliation(s)
- Ying Zhao
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Yan Shu
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Ning Zhao
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Zili Zhou
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Xiong Jia
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Chenxing Jian
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Si Jin
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| |
Collapse
|
4
|
Nunez‐Salces M, Li H, Feinle‐Bisset C, Young RL, Page AJ. The regulation of gastric ghrelin secretion. Acta Physiol (Oxf) 2021; 231:e13588. [PMID: 33249751 DOI: 10.1111/apha.13588] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 11/19/2020] [Accepted: 11/23/2020] [Indexed: 12/13/2022]
Abstract
Ghrelin is a gastric hormone with multiple physiological functions, including the stimulation of food intake and adiposity. It is well established that circulating ghrelin levels are closely associated with feeding patterns, rising strongly before a meal and lowering upon food intake. However, the mechanisms underlying the modulation of ghrelin secretion are not fully understood. The purpose of this review is to discuss current knowledge on the circadian oscillation of circulating ghrelin levels, the neural mechanisms stimulating fasting ghrelin levels and peripheral mechanisms modulating postprandial ghrelin levels. Furthermore, the therapeutic potential of targeting the ghrelin pathway is discussed in the context of the treatment of various metabolic disorders, including obesity, type 2 diabetes, diabetic gastroparesis and Prader-Willi syndrome. Moreover, eating disorders including anorexia nervosa, bulimia nervosa and binge-eating disorder are also discussed.
Collapse
Affiliation(s)
- Maria Nunez‐Salces
- Vagal Afferent Research Group Adelaide Medical School The University of Adelaide Adelaide SA Australia
- Centre of Research Excellence in Translating Nutritional Science to Good Health Adelaide Medical School The University of Adelaide Adelaide SA Australia
- Nutrition, Diabetes & Gut Health, Lifelong Health Theme South Australian Health & Medical Research Institute (SAHMRI) Adelaide SA Australia
| | - Hui Li
- Vagal Afferent Research Group Adelaide Medical School The University of Adelaide Adelaide SA Australia
- Centre of Research Excellence in Translating Nutritional Science to Good Health Adelaide Medical School The University of Adelaide Adelaide SA Australia
- Nutrition, Diabetes & Gut Health, Lifelong Health Theme South Australian Health & Medical Research Institute (SAHMRI) Adelaide SA Australia
| | - Christine Feinle‐Bisset
- Centre of Research Excellence in Translating Nutritional Science to Good Health Adelaide Medical School The University of Adelaide Adelaide SA Australia
| | - Richard L. Young
- Centre of Research Excellence in Translating Nutritional Science to Good Health Adelaide Medical School The University of Adelaide Adelaide SA Australia
- Nutrition, Diabetes & Gut Health, Lifelong Health Theme South Australian Health & Medical Research Institute (SAHMRI) Adelaide SA Australia
- Intestinal Nutrient Sensing Group Adelaide Medical School The University of Adelaide Adelaide SA Australia
| | - Amanda J. Page
- Vagal Afferent Research Group Adelaide Medical School The University of Adelaide Adelaide SA Australia
- Centre of Research Excellence in Translating Nutritional Science to Good Health Adelaide Medical School The University of Adelaide Adelaide SA Australia
- Nutrition, Diabetes & Gut Health, Lifelong Health Theme South Australian Health & Medical Research Institute (SAHMRI) Adelaide SA Australia
| |
Collapse
|
5
|
Cornejo MP, Mustafá ER, Cassano D, Banères JL, Raingo J, Perello M. The ups and downs of growth hormone secretagogue receptor signaling. FEBS J 2021; 288:7213-7229. [PMID: 33460513 DOI: 10.1111/febs.15718] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 01/05/2021] [Accepted: 01/14/2021] [Indexed: 12/14/2022]
Abstract
The growth hormone secretagogue receptor (GHSR) has emerged as one of the most fascinating molecules from the perspective of neuroendocrine control. GHSR is mainly expressed in the pituitary and the brain, and plays key roles regulating not only growth hormone secretion but also food intake, adiposity, body weight, glucose homeostasis and other complex functions. Quite atypically, GHSR signaling displays a basal constitutive activity that can be up- or downregulated by two digestive system-derived hormones: the octanoylated-peptide ghrelin and the liver-expressed antimicrobial peptide 2 (LEAP2), which was recently recognized as an endogenous GHSR ligand. The existence of two ligands with contrary actions indicates that GHSR activity can be tightly regulated and that the receptor displays the capability to integrate such opposing inputs in order to provide a balanced intracellular signal. This article provides a summary of the current understanding of the biology of ghrelin, LEAP2 and GHSR and discusses the reconceptualization of the cellular and physiological implications of the ligand-regulated GHSR signaling, based on the latest findings.
Collapse
Affiliation(s)
- María P Cornejo
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET), Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], Buenos Aires, Argentina
| | - Emilio R Mustafá
- Laboratory of Electrophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET), Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], Buenos Aires, Argentina
| | - Daniela Cassano
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET), Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], Buenos Aires, Argentina
| | - Jean-Louis Banères
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, CNRS, Université de Montpellier, Ecole Nationale Supérieure de Chimie de Montpellier, Faculté de Pharmacie, Montpellier cedex 5, France
| | - Jesica Raingo
- Laboratory of Electrophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET), Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], Buenos Aires, Argentina
| | - Mario Perello
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET), Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], Buenos Aires, Argentina
| |
Collapse
|
6
|
Duszka K, Gregor A, Reichel MW, Baierl A, Fahrngruber C, König J. Visual stimulation with food pictures in the regulation of hunger hormones and nutrient deposition, a potential contributor to the obesity crisis. PLoS One 2020; 15:e0232099. [PMID: 32330183 PMCID: PMC7182185 DOI: 10.1371/journal.pone.0232099] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 04/07/2020] [Indexed: 12/11/2022] Open
Abstract
Food cues affect hunger and nutritional choices. Omnipresent stimulation with palatable food contributes to the epidemics of obesity. The objective of the study was to investigate the impact of food cues on appetite-related hormones and to assess the functionality of the secreted hormones on macronutrient uptake in healthy subjects. Additionally, we aimed at verifying differences in the response of total and active ghrelin to stimulation with food pictures and to a meal followed by the stimulation. We were also interested in the identification of factors contributing to response to food cues. We recruited healthy, non-obese participants for two independent cross-over studies. During the first study, the subjects were presented random non-food pictures on the first day and pictures of foods on the second day of the study. Throughout the second study, following the picture session, the participants were additionally asked to drink a milkshake. Concentrations of blood glucose, triglycerides and hunger-related hormones were measured. The results showed that concentrations of several hormones measured in the blood are interdependent. In the case of ghrelin and gastric inhibitory peptide (GIP) as well as ghrelin and glucagon-like peptide-1 (GLP-1), this co-occurrence relies on the visual cues. Regulation of total ghrelin concentration following food stimulation is highly individual and responders showed upregulated total ghrelin, while the concentration of active ghrelin decreases following a meal. Protein content and colour intensity of food pictures reversely correlated with participants’ rating of the pictures. We conclude that observation of food pictures influences the concentration of several appetite-related hormones. The close link of visual clues to physiological responses is likely of clinical relevance. Additionally, the protein content of displayed foods and green colour intensity in pictures may serve as a predictor of subjective attractiveness of the presented meal.
Collapse
Affiliation(s)
- Kalina Duszka
- Department of Nutritional Sciences, University of Vienna, Vienna, Austria
- * E-mail:
| | - András Gregor
- Department of Nutritional Sciences, University of Vienna, Vienna, Austria
| | | | - Andreas Baierl
- Department of Statistics and Operations Research, University of Vienna, Vienna, Austria
| | | | - Jürgen König
- Department of Nutritional Sciences, University of Vienna, Vienna, Austria
| |
Collapse
|
7
|
Fakhry J, Stebbing MJ, Hunne B, Bayguinov Y, Ward SM, Sasse KC, Callaghan B, McQuade RM, Furness JB. Relationships of endocrine cells to each other and to other cell types in the human gastric fundus and corpus. Cell Tissue Res 2018; 376:37-49. [PMID: 30467709 DOI: 10.1007/s00441-018-2957-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 11/01/2018] [Indexed: 02/07/2023]
Abstract
Gastric endocrine cell hormones contribute to the control of the stomach and to signalling to the brain. In other gut regions, enteroendocrine cells (EECs) exhibit extensive patterns of colocalisation of hormones. In the current study, we characterise EECs in the human gastric fundus and corpus. We utilise immunohistochemistry to investigate EECs with antibodies to ghrelin, serotonin (5-HT), somatostatin, peptide YY (PYY), glucagon-like peptide 1, calbindin, gastrin and pancreastatin, the latter as a marker of enterochromaffin-like (ECL) cells. EECs were mainly located in regions of the gastric glands populated by parietal cells. Gastrin cells were absent and PYY cells were very rare. Except for about 25% of 5-HT cells being a subpopulation of ECL cells marked by pancreastatin, colocalisation of hormones in gastric EECs was infrequent. Ghrelin cells were distributed throughout the fundus and corpus; most were basally located in the glands, often very close to parietal cells and were closed cells i.e., not in contact with the lumen. A small proportion had long processes located close to the base of the mucosal epithelium. The 5-HT cells were of at least three types: small, round, closed cells; cells with multiple, often very long, processes; and a subgroup of ECL cells. Processes were in contact with their surrounding cells, including parietal cells. Mast cells had very weak or no 5-HT immunoreactivity. Somatostatin cells were a closed type with long processes. In conclusion, four major chemically defined EEC types occurred in the human oxyntic mucosa. Within each group were cells with distinct morphologies and relationships to other mucosal cells.
Collapse
Affiliation(s)
- Josiane Fakhry
- Department of Anatomy and Neuroscience, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Martin J Stebbing
- Department of Anatomy and Neuroscience, University of Melbourne, Parkville, VIC, 3010, Australia.,Florey Institute of Neuroscience and Mental Health, Parkville, VIC, 3010, Australia
| | - Billie Hunne
- Department of Anatomy and Neuroscience, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Yulia Bayguinov
- Department of Physiology and Cell Biology, Reno School of Medicine, University of Nevada, Reno, NV, 89557, USA
| | - Sean M Ward
- Department of Physiology and Cell Biology, Reno School of Medicine, University of Nevada, Reno, NV, 89557, USA
| | - Kent C Sasse
- School of Medicine, Universiity of Nevada, Reno, NV, 89557, USA.,Renown Regional Medical Center, Reno, NV, 89502, USA
| | - Brid Callaghan
- Department of Anatomy and Neuroscience, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Rachel M McQuade
- Department of Anatomy and Neuroscience, University of Melbourne, Parkville, VIC, 3010, Australia.,Florey Institute of Neuroscience and Mental Health, Parkville, VIC, 3010, Australia
| | - John B Furness
- Department of Anatomy and Neuroscience, University of Melbourne, Parkville, VIC, 3010, Australia. .,Florey Institute of Neuroscience and Mental Health, Parkville, VIC, 3010, Australia.
| |
Collapse
|
8
|
Blanco AM, Bertucci JI, Valenciano AI, Delgado MJ, Unniappan S. Ghrelin suppresses cholecystokinin (CCK), peptide YY (PYY) and glucagon-like peptide-1 (GLP-1) in the intestine, and attenuates the anorectic effects of CCK, PYY and GLP-1 in goldfish (Carassius auratus). Horm Behav 2017; 93:62-71. [PMID: 28506816 DOI: 10.1016/j.yhbeh.2017.05.004] [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: 10/17/2016] [Revised: 04/27/2017] [Accepted: 05/12/2017] [Indexed: 02/06/2023]
Abstract
Ghrelin is an important gut-derived hormone with an appetite stimulatory role, while most of the intestinal hormones, including cholecystokinin (CCK), peptide YY (PYY) and glucagon-like peptide-1 (GLP-1), are appetite-inhibitors. Whether these important peptides with opposing roles on food intake interact to regulate energy balance in fish is currently unknown. The aim of this study was to characterize the putative crosstalk between ghrelin and CCK, PYY and GLP-1 in goldfish (Carassius auratus). We first determined the localization of CCK, PYY and GLP-1 in relation to ghrelin and its main receptor GHS-R1a (growth hormone secretagogue 1a) in the goldfish intestine by immunohistochemistry. Colocalization of ghrelin/GHS-R1a and CCK/PYY/GLP-1 was found primarily in the luminal border of the intestinal mucosa. In an intestinal explant culture, a significant decrease in prepro-cck, prepro-pyy and proglucagon transcript levels was observed after 60min of incubation with ghrelin, which was abolished by preincubation with the GHS-R1a ghrelin receptor antagonist [D-Lys3]-GHRP-6 (except for proglucagon). The protein expression of PYY and GLP-1 was also downregulated by ghrelin. Finally, intraperitoneal co-administration of CCK, PYY or GLP-1 with ghrelin results in no modification of food intake in goldfish. Overall, results of the present study show for the first time in fish that ghrelin exerts repressive effects on enteric anorexigens. It is likely that these interactions mediate the stimulatory effects of ghrelin on feeding and metabolism in fish.
Collapse
Affiliation(s)
- Ayelén Melisa Blanco
- Departamento de Fisiología (Fisiología Animal II), Facultad de Biología, Universidad Complutense de Madrid, José Antonio Nováis 12, 28040 Madrid, Spain; Laboratory of Integrative Neuroendocrinology, Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, 52 Campus Drive, S7N 5B4 Saskatoon, Saskatchewan, Canada.
| | - Juan Ignacio Bertucci
- Laboratory of Integrative Neuroendocrinology, Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, 52 Campus Drive, S7N 5B4 Saskatoon, Saskatchewan, Canada; Instituto de Investigaciones Biotecnológicas-Instituto Tecnológico Chascomús, Avenida Intendente Marinos Km. 8,2, 7130 Chascomús, Buenos Aires, Argentina.
| | - Ana Isabel Valenciano
- Departamento de Fisiología (Fisiología Animal II), Facultad de Biología, Universidad Complutense de Madrid, José Antonio Nováis 12, 28040 Madrid, Spain.
| | - María Jesús Delgado
- Departamento de Fisiología (Fisiología Animal II), Facultad de Biología, Universidad Complutense de Madrid, José Antonio Nováis 12, 28040 Madrid, Spain.
| | - Suraj Unniappan
- Laboratory of Integrative Neuroendocrinology, Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, 52 Campus Drive, S7N 5B4 Saskatoon, Saskatchewan, Canada.
| |
Collapse
|
9
|
Abstract
Many of the neurocircuits and hormones known to underlie the sensations of hunger and satiety also substantially alter the activity of the dopaminergic reward system. Much interest lies in the ways that hunger, satiety, and reward tie together, as the epidemic of obesity seems tied to the recent development and mass availability of highly palatable foods. In this review, we will first discuss the basic neurocircuitry of the midbrain and basal forebrain reward system. We will elaborate how several important mediators of hunger-the agouti-related protein neurons of the arcuate nucleus, the lateral hypothalamic nucleus, and ghrelin-enhance the sensitivity of the dopaminergic reward system. Then, we will elaborate how mediators of satiety-the nucleus tractus solitarius, pro-opiomelanocortin neurons of the arcuate nucleus, and its peripheral hormonal influences such as leptin-reduce the reward system sensitivity. We hope to provide a template by which future research may identify the ways in which highly rewarding foods bypass this balanced system to produce excessive food consumption.
Collapse
Affiliation(s)
- Ryan Michael Cassidy
- Brown Foundation of the Institute of Molecular Medicine for the Prevention of Human Diseases of McGovern Medical School, Neuroscience Program MD Anderson Cancer Center and UTHealth Graduate School of Biological Sciences, The University of Texas Health Science Center at Houston, Houston, TX, USA
- *Correspondence: Ryan Michael Cassidy,
| | - Qingchun Tong
- Brown Foundation of the Institute of Molecular Medicine for the Prevention of Human Diseases of McGovern Medical School, Neuroscience Program MD Anderson Cancer Center and UTHealth Graduate School of Biological Sciences, The University of Texas Health Science Center at Houston, Houston, TX, USA
| |
Collapse
|
10
|
Hong X, Zhang H, Liang H, Li D, Huang J, Li Z, Jiang S, Zhang W, Xu G. Exendin-4 decreases ghrelin levels through mTOR signaling. Mol Cell Endocrinol 2016; 437:201-212. [PMID: 27569528 DOI: 10.1016/j.mce.2016.08.039] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 08/11/2016] [Accepted: 08/24/2016] [Indexed: 12/13/2022]
Abstract
Exendin-4 (EX-4), a long-acting glucagon-like peptide-1 receptor (GLP-1R) agonist, regulates feeding behavior through its ability to inhibit gastric emptying. Ghrelin, a gastric hormone, provides a hunger signal to the central nervous system to stimulate appetite. Here, we report that EX-4 suppresses ghrelin production through the mTORC1-dependent mechanism. Central administration of EX-4 reduces gastric, hypothalamic and plasma ghrelin in both C57BL/6J mice and diet induced obese mice. These changes were associated with a significant increase in mTORC1 activity. Both GLP-1 and EX-4 suppressed the expression and secretion of ghrelin in cultured mHypoE-42 cells, a hypothalamic cell line. These effects were associated with significant changes in mTOR signaling. Inhibition of mTORC1 activity by mTOR siRNA or rapamycin abolished the suppression of ghrelin production induced by GLP-1 and EX-4 in mHypoE-42 cells. Our results identify mTORC1 as a critical signaling pathway for the downregulation of ghrelin induced by activation of GLP-1R.
Collapse
Affiliation(s)
- Xiaosi Hong
- Department of Physiology, School of Medicine, Jinan University, 601 Huangpu Avenue West, Tianhe District, Guangzhou, Guangdong, 510632, China
| | - Heng Zhang
- Department of Physiology, School of Medicine, Jinan University, 601 Huangpu Avenue West, Tianhe District, Guangzhou, Guangdong, 510632, China
| | - Hongbin Liang
- Department of Physiology, School of Medicine, Jinan University, 601 Huangpu Avenue West, Tianhe District, Guangzhou, Guangdong, 510632, China
| | - Danjie Li
- Department of Physiology, School of Medicine, Jinan University, 601 Huangpu Avenue West, Tianhe District, Guangzhou, Guangdong, 510632, China
| | - Jiana Huang
- Department of Physiology, School of Medicine, Jinan University, 601 Huangpu Avenue West, Tianhe District, Guangzhou, Guangdong, 510632, China
| | - Zhi Li
- Department of Physiology, School of Medicine, Jinan University, 601 Huangpu Avenue West, Tianhe District, Guangzhou, Guangdong, 510632, China
| | - Sibo Jiang
- Departments of Pharmaceutics, University of Florida, 6550 Sanger Rd, Orlando, FL 32827, USA
| | - Weizhen Zhang
- Shenzhen University Diabetes Center, Shenzhen University Health Science Center, Shenzhen, Guangdong, China; Department of Surgery, University of Michigan Medical Center, Ann Arbor, MI 48109-0346, USA.
| | - Geyang Xu
- Department of Physiology, School of Medicine, Jinan University, 601 Huangpu Avenue West, Tianhe District, Guangzhou, Guangdong, 510632, China.
| |
Collapse
|
11
|
Santiago-García PA, López MG. Agavins from Agave angustifolia and Agave potatorum affect food intake, body weight gain and satiety-related hormones (GLP-1 and ghrelin) in mice. Food Funct 2015; 5:3311-9. [PMID: 25367106 DOI: 10.1039/c4fo00561a] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Agavins act as a fermentable dietary fiber and have attracted attention due to their potential for reducing the risk of disease. Therefore, we evaluated the effect of supplementation using 10% agavins with a short-degree of polymerization (SDP) from Agave angustifolia Haw. (AASDP) or Agave potatorum Zucc. (APSDP) along with chicory fructans (RSE) as a reference for 5 weeks, on the energy intake, body weight gain, satiety-related hormones from the gut and blood (GLP-1 and ghrelin), blood glucose and lipids, and short-chain fatty acids (SCFAs) from the gut of ad libitum-fed mice. We evaluated the energy intake daily and weight gain every week. At the end of the experiment, portal vein blood samples as well as intestinal segments and the stomach were collected to measure glucagon-like peptide-1 (GLP-1) and ghrelin using RIA and ELISA kits, respectively. Colon SCFAs were measured using gas chromatography. The energy intake, body weight gain, and triglycerides were lower in the fructan-fed mice than in the STD-fed mice. The AASDP, APSDP, and RSE diets increased the serum levels of GLP-1 (40, 93, and 16%, respectively vs. STD) (P ≤ 0.05), whereas ghrelin was decreased (16, 38, and 42%, respectively) (P ≤ 0.05). Butyric acid increased significantly in the APSDP-fed mice (26.59 mmol g(-1), P ≤ 0.001) compared with that in the AASDP- and RSE-fed mice. We concluded that AASDP and APSDP are able to promote the secretion of the peptides involved in appetite regulation, which might help to control obesity and its associated metabolic disorder.
Collapse
|
12
|
Ali SS, Hamed EA, Ayuob NN, Shaker Ali A, Suliman MI. Effects of different routes of nicotine administration on gastric morphology and hormonal secretion in rats. Exp Physiol 2015; 100:881-95. [DOI: 10.1113/ep085015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 05/06/2015] [Indexed: 12/16/2022]
Affiliation(s)
- Soad Shaker Ali
- Anatomy Department, Faculty of Medicine; King Abdulaziz University; Jeddah Saudi Arabia
| | - Enas Ahmed Hamed
- Physiology Department, Faculty of Medicine; Assuit University; Asyut Egypt
| | - Nasra Naeim Ayuob
- Anatomy Department, Faculty of Medicine; King Abdulaziz University; Jeddah Saudi Arabia
- Histology Department, Faculty of Medicine; Mansoura University; Mansoura Egypt
| | - Ahmed Shaker Ali
- Pharmacology Department, Faculty of Medicine; King Abdulaziz University; Jeddah Saudi Arabia
| | - Mansour Ibrahem Suliman
- Pharmacology Department, Faculty of Medicine; King Abdulaziz University; Jeddah Saudi Arabia
| |
Collapse
|
13
|
Iwakura H, Kangawa K, Nakao K. The regulation of circulating ghrelin - with recent updates from cell-based assays. Endocr J 2015; 62:107-22. [PMID: 25273611 DOI: 10.1507/endocrj.ej14-0419] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Ghrelin is a stomach-derived orexigenic hormone with a wide range of physiological functions. Elucidation of the regulation of the circulating ghrelin level would lead to a better understanding of appetite control in body energy homeostasis. Earlier studies revealed that circulating ghrelin levels are under the control of both acute and chronic energy status: at the acute scale, ghrelin levels are increased by fasting and decreased by feeding, whereas at the chronic scale, they are high in obese subjects and low in lean subjects. Subsequent studies revealed that nutrients, hormones, or neural activities can influence circulating ghrelin levels in vivo. Recently developed in vitro assay systems for ghrelin secretion can assess whether and how individual factors affect ghrelin secretion from cells. In this review, on the basis of numerous human, animal, and cell-based studies, we summarize current knowledge on the regulation of circulating ghrelin levels and enumerate the factors that influence ghrelin levels.
Collapse
Affiliation(s)
- Hiroshi Iwakura
- Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan
| | | | | |
Collapse
|
14
|
Liu R, Ma D, Li Y, Hu R, Peng Y, Wang Q. The anorexic effect of Ex4/Fc through GLP-1 receptor activation in high-fat diet fed mice. Acta Biochim Biophys Sin (Shanghai) 2014; 46:675-81. [PMID: 24951724 DOI: 10.1093/abbs/gmu044] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Exendin-4 (Ex4), a peptide initially found in the saliva of the Gila monster, can activate the signaling pathway of the incretin hormone glucagon-like peptide-1 (GLP-1) through the GLP-1 receptor (GLP-1R). We previously reported that a chimera protein consisting of Ex4 and mouse IgG heavy chain constant regions (Ex4/Fc) can exert biological effects of GLP-1, such as improving glycemic control and ameliorating manifestations in diabetic mice. The aim of this study was to determine whether Ex4/Fc is effective in modulating energy homeostasis in mice. Our results showed that in vivo expression of Ex4/Fc by intramuscular injection of the plasmid encoding Ex4/Fc followed by local electroporation effectively decreased food intake in the mice on high-fat diet (HFD) feeding. In addition, the reduced energy intake was associated with the decreased excrements from the Ex4/Fc-treated HFD mice but not the Fc control mice. Remarkably, the Ex4/Fctreated HFD mice displayed significantly lower triglyceride (TG) levels when compared with the control mice. Interestingly, while the leptin levels were not changed, the circulating ghrelin levels were higher in Ex4/Fc mice than those in the Fc control mice. These results suggested that Ex4/Fc can improve energy metabolism and lipid metabolism through GLP-1R in mice under excessive nutrition conditions.
Collapse
|
15
|
López M, Huazano-García A, García-Pérez M, García-Vieyra M. Agave Fiber Structure Complexity and Its Impact on Health. POLYSACCHARIDES 2014. [DOI: 10.1201/b17121-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
|
16
|
Engelstoft MS, Park WM, Sakata I, Kristensen LV, Husted AS, Osborne-Lawrence S, Piper PK, Walker AK, Pedersen MH, Nøhr MK, Pan J, Sinz CJ, Carrington PE, Akiyama TE, Jones RM, Tang C, Ahmed K, Offermanns S, Egerod KL, Zigman JM, Schwartz TW. Seven transmembrane G protein-coupled receptor repertoire of gastric ghrelin cells. Mol Metab 2013; 2:376-92. [PMID: 24327954 DOI: 10.1016/j.molmet.2013.08.006] [Citation(s) in RCA: 242] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Accepted: 08/26/2013] [Indexed: 12/18/2022] Open
Abstract
The molecular mechanisms regulating secretion of the orexigenic-glucoregulatory hormone ghrelin remain unclear. Based on qPCR analysis of FACS-purified gastric ghrelin cells, highly expressed and enriched 7TM receptors were comprehensively identified and functionally characterized using in vitro, ex vivo and in vivo methods. Five Gαs-coupled receptors efficiently stimulated ghrelin secretion: as expected the β1-adrenergic, the GIP and the secretin receptors but surprisingly also the composite receptor for the sensory neuropeptide CGRP and the melanocortin 4 receptor. A number of Gαi/o-coupled receptors inhibited ghrelin secretion including somatostatin receptors SSTR1, SSTR2 and SSTR3 and unexpectedly the highly enriched lactate receptor, GPR81. Three other metabolite receptors known to be both Gαi/o- and Gαq/11-coupled all inhibited ghrelin secretion through a pertussis toxin-sensitive Gαi/o pathway: FFAR2 (short chain fatty acid receptor; GPR43), FFAR4 (long chain fatty acid receptor; GPR120) and CasR (calcium sensing receptor). In addition to the common Gα subunits three non-common Gαi/o subunits were highly enriched in ghrelin cells: GαoA, GαoB and Gαz. Inhibition of Gαi/o signaling via ghrelin cell-selective pertussis toxin expression markedly enhanced circulating ghrelin. These 7TM receptors and associated Gα subunits constitute a major part of the molecular machinery directly mediating neuronal and endocrine stimulation versus metabolite and somatostatin inhibition of ghrelin secretion including a series of novel receptor targets not previously identified on the ghrelin cell.
Collapse
Key Words
- 7TM, seven transmembrane segment
- BAC, bacterial artificial chromosome
- CCK, cholecystokinin
- CFMB, (S)-2-(4-chlorophenyl)-3,3-dimethyl-N-(5-phenylthiazol-2-yl)butamide
- CGRP, calcitonin gene-related peptide
- CHBA, 3-chloro-5-hydroxybenzoic acid
- Enteroendocrine
- G protein signaling
- GIP, glucose-dependent insulinotropic polypeptide
- GLP-1, glucagon-like peptide 1
- GPCR
- Ghrelin
- Metabolites
- PTx, Bordetella pertussis toxin
- PYY, peptide YY
- Secretion
- hrGFP, humanized Renilla reniformis green fluorescent protein
Collapse
Affiliation(s)
- Maja S Engelstoft
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section for Metabolic Receptology and Enteroendocrinology, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen, Denmark ; Laboratory for Molecular Pharmacology, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen, Denmark
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Du G, Liu M, Parvizi N, Zhao R. Ectopic expression of ghrelin affects gastric H+–K+-ATPase activity and expression of GHR/IGF-1 system in weaned mice. ACTA ACUST UNITED AC 2013; 186:12-7. [DOI: 10.1016/j.regpep.2013.06.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Revised: 03/10/2013] [Accepted: 06/19/2013] [Indexed: 10/26/2022]
|
18
|
Liraglutide suppresses the plasma levels of active and des-acyl ghrelin independently of active glucagon-like Peptide-1 levels in mice. ISRN ENDOCRINOLOGY 2013; 2013:184753. [PMID: 23997963 PMCID: PMC3755422 DOI: 10.1155/2013/184753] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2013] [Accepted: 07/14/2013] [Indexed: 12/01/2022]
Abstract
Glucagon-like peptide-1 (GLP-1), an insulinotropic gastrointestinal peptide that is primarily produced by intestinal endocrine L-cells, stimulates satiety. Ghrelin, a hormone that is produced predominantly by the stomach stimulates hunger. There are two forms of ghrelin: active ghrelin and inactive des-acyl ghrelin. After depriving mice of food for 24 h, we demonstrated that the systemic administration of liraglutide (100 μg/kg), a human GLP-1 analog that binds to the GLP-1 receptor, increased (1.4-fold) the plasma levels of active GLP-1 and suppressed the plasma levels of active and des-acyl ghrelin after 1 h. Despite the elevated plasma levels of active GLP-1 (11-fold), liraglutide had no effect on the plasma levels of active or des-acyl ghrelin after 12 h. These findings demonstrated that liraglutide suppresses the plasma levels of active and des-acyl ghrelin independently of active GLP-1 levels in fasted mice, suggesting a novel in vivo biological effect of liraglutide beyond regulating plasma GLP-1.
Collapse
|
19
|
Belza A, Ritz C, Sørensen MQ, Holst JJ, Rehfeld JF, Astrup A. Contribution of gastroenteropancreatic appetite hormones to protein-induced satiety. Am J Clin Nutr 2013; 97:980-9. [PMID: 23466396 DOI: 10.3945/ajcn.112.047563] [Citation(s) in RCA: 128] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Effects of protein intake on appetite-regulating hormones and their dynamics are unclear. OBJECTIVES We investigated the satiating effects of meals with varying protein contents and whether there was an effect of dose on appetite-regulating hormones and appetite ratings. DESIGN Twenty-five men [mean ± SD age: 30.0 ± 8.7 y; body mass index (BMI; in kg/m(2)): 25.9 ± 4.7] participated in the 3-way, randomized, double-blind crossover study. Test meals were isocaloric with 30% of energy from fat and protein content adjusted at the expense of carbohydrate. Test meals were normal protein (NP; 14% of energy from protein), medium-high protein (MHP; 25% of energy from protein), and high protein (HP, 50% of energy from protein). Appetite ratings and blood samples were assessed every 0.5 h for 4 h. An ad libitum lunch was served 4 h after the meal. RESULTS Protein increased dose-dependently glucagon-like peptide-1 (GLP-1), peptide YY (PYY) 3-36, and glucagon; MHP produced 10%, 7%, and 47% greater responses, respectively; and HP produced 20%, 14%, and 116% greater responses, respectively, than did NP (P < 0.03). Compared with NP, HP increased insulin and cholecystokinin and decreased ghrelin and glucose-dependent insulinotropic polypeptide (P < 0.05). Satiety and fullness dose-dependently increased by 7% and 6% for MHP and 16% and 19% for HP compared with NP (P < 0.001). Hunger and prospective consumption dose-dependently decreased by 15% and 13% for MHP and by 25% and 26% for HP compared with NP (P < 0.0003). There was a combined effect of GLP-1 and PYY 3-36 (P = 0.03) next to the additive effect of GLP-1 (P = 0.006) on the composite appetite score. No difference was shown in ad libitum energy intake. CONCLUSION Protein dose-dependently increased satiety and GLP-1, PYY 3-36, and glucagon, which may, at least in part, be responsible for the satiety-stimulating effect of protein. This trial was registered at clinicaltrials.gov as NCT01561235.
Collapse
Affiliation(s)
- Anita Belza
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | | | | |
Collapse
|
20
|
Lu X, Zhao X, Feng J, Liou AP, Anthony S, Pechhold S, Sun Y, Lu H, Wank S. Postprandial inhibition of gastric ghrelin secretion by long-chain fatty acid through GPR120 in isolated gastric ghrelin cells and mice. Am J Physiol Gastrointest Liver Physiol 2012; 303:G367-76. [PMID: 22678998 PMCID: PMC3774249 DOI: 10.1152/ajpgi.00541.2011] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Ghrelin is a gastric peptide hormone that controls appetite and energy homeostasis. Plasma ghrelin levels rise before a meal and fall quickly thereafter. Elucidation of the regulation of ghrelin secretion has been hampered by the difficulty of directly interrogating ghrelin cells diffusely scattered within the complex gastric mucosa. Therefore, we generated transgenic mice with ghrelin cell expression of green fluorescent protein (GFP) to enable characterization of ghrelin secretion in a pure population of isolated gastric ghrelin-expressing GFP (Ghr-GFP) cells. Using quantitative RT-PCR and immunofluorescence staining, we detected a high level of expression of the long-chain fatty acid (LCFA) receptor GPR120, while the other LCFA receptor, GPR40, was undetectable. In short-term-cultured pure Ghr-GFP cells, the LCFAs docosadienoic acid, linolenic acid, and palmitoleic acid significantly suppressed ghrelin secretion. The physiological mechanism of LCFA inhibition on ghrelin secretion was studied in mice. Serum ghrelin levels were transiently suppressed after gastric gavage of LCFA-rich lipid in mice with pylorus ligation, indicating that the ghrelin cell may directly sense increased gastric LCFA derived from ingested intraluminal lipids. Meal-induced increase in gastric mucosal LCFA was assessed by measuring the transcripts of markers for tissue uptake of LCFA, lipoprotein lipase (LPL), fatty acid translocase (CD36), glycosylphosphatidylinositol-anchored HDL-binding protein 1, and nuclear fatty acid receptor peroxisome proliferator-activated receptor-γ. Quantitative RT-PCR studies indicate significantly increased mRNA levels of lipoprotein lipase, glycosylphosphatidylinositol-anchored HDL-binding protein 1, and peroxisome proliferator-activated receptor-γ in postprandial gastric mucosa. These results suggest that meal-related increases in gastric mucosal LCFA interact with GPR120 on ghrelin cells to inhibit ghrelin secretion.
Collapse
Affiliation(s)
- Xinping Lu
- 1Digestive Diseases Branch, National Institute of Diabetes, Digestive, and Kidney Diseases, National Institutes of Health, Bethesda, Maryland;
| | - Xilin Zhao
- 1Digestive Diseases Branch, National Institute of Diabetes, Digestive, and Kidney Diseases, National Institutes of Health, Bethesda, Maryland;
| | - Jianying Feng
- 1Digestive Diseases Branch, National Institute of Diabetes, Digestive, and Kidney Diseases, National Institutes of Health, Bethesda, Maryland;
| | - Alice P. Liou
- 1Digestive Diseases Branch, National Institute of Diabetes, Digestive, and Kidney Diseases, National Institutes of Health, Bethesda, Maryland;
| | - Shari Anthony
- 1Digestive Diseases Branch, National Institute of Diabetes, Digestive, and Kidney Diseases, National Institutes of Health, Bethesda, Maryland;
| | - Susanne Pechhold
- 2Diabetes Branch, National Institute of Diabetes, Digestive, and Kidney Diseases, National Institutes of Health, Bethesda, Maryland;
| | - Yuxiang Sun
- 3Children's Nutrition Research Center, Huffington Center on Aging, Departments of Pediatrics and Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas; and
| | - Huiyan Lu
- 4Transgenic Facility, National Institute of Diabetes, Digestive, and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Stephen Wank
- 1Digestive Diseases Branch, National Institute of Diabetes, Digestive, and Kidney Diseases, National Institutes of Health, Bethesda, Maryland;
| |
Collapse
|
21
|
Stengel A, Taché Y. Yin and Yang - the Gastric X/A-like Cell as Possible Dual Regulator of Food Intake. J Neurogastroenterol Motil 2012; 18:138-49. [PMID: 22523723 PMCID: PMC3325299 DOI: 10.5056/jnm.2012.18.2.138] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2012] [Accepted: 03/11/2012] [Indexed: 01/14/2023] Open
Abstract
Ingestion of food affects secretion of hormones from enteroendocrine cells located in the gastrointestinal mucosa. These hormones are involved in the regulation of various gastrointestinal functions including the control of food intake. One cell in the stomach, the X/A-like has received much attention over the past years due to the production of ghrelin. Until now, ghrelin is the only known orexigenic hormone that is peripherally produced and centrally acting to stimulate food intake. Subsequently, additional peptide products of this cell have been described including desacyl ghrelin, obestatin and nesfatin-1. Desacyl ghrelin seems to be involved in the regulation of food intake as well and could play a counter-balancing role of ghrelin's orexigenic effect. In contrast, the initially proposed anorexigenic action of obestatin did not hold true and therefore the involvement of this peptide in the regulation of feeding is questionable. Lastly, the identification of nesfatin-1 in the same cell in different vesicles than ghrelin extended the function of this cell type to the inhibition of feeding. Therefore, this X/A-like cell could play a unique role by encompassing yin and yang properties to mediate not only hunger but also satiety.
Collapse
Affiliation(s)
- Andreas Stengel
- Department of Medicine, Division Psychosomatic Medicine and Psychotherapy, Charité, Campus Mitte, Universitätsmedizin Berlin, Berlin, Germany
| | | |
Collapse
|
22
|
Stengel A, Taché Y. Ghrelin - a pleiotropic hormone secreted from endocrine x/a-like cells of the stomach. Front Neurosci 2012; 6:24. [PMID: 22355282 PMCID: PMC3280431 DOI: 10.3389/fnins.2012.00024] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2011] [Accepted: 01/29/2012] [Indexed: 12/13/2022] Open
Abstract
The gastric X/A-like endocrine cell receives growing attention due to its peptide products with ghrelin being the best characterized. This peptide hormone was identified a decade ago as a stimulator of food intake and to date remains the only known peripherally produced and centrally acting orexigenic hormone. In addition, subsequent studies identified numerous other functions of this peptide including the stimulation of gastrointestinal motility, the maintenance of energy homeostasis and an impact on reproduction. Moreover, ghrelin is also involved in the response to stress and assumed to play a role in coping functions and exert a modulatory action on immune pathways. Our knowledge on the regulation of ghrelin has markedly advanced during the past years by the identification of the ghrelin acylating enzyme, ghrelin-O-acyltransferase, and by the description of changes in expression, activation, and release under different metabolic as well as physically and psychically challenging conditions. However, our insight on regulatory processes of ghrelin at the cellular and subcellular levels is still very limited and warrants further investigation.
Collapse
Affiliation(s)
- Andreas Stengel
- Division Psychosomatic Medicine and Psychotherapy, Department of Medicine, Charité - Universitätsmedizin Berlin Berlin, Germany
| | | |
Collapse
|
23
|
Beta glucan: health benefits in obesity and metabolic syndrome. J Nutr Metab 2011; 2012:851362. [PMID: 22187640 PMCID: PMC3236515 DOI: 10.1155/2012/851362] [Citation(s) in RCA: 215] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2011] [Accepted: 10/27/2011] [Indexed: 12/27/2022] Open
Abstract
Despite the lack of international agreement regarding the definition and classification of fiber, there is established evidence on the role of dietary fibers in obesity and metabolic syndrome. Beta glucan (β-glucan) is a soluble fiber readily available from oat and barley grains that has been gaining interest due to its multiple functional and bioactive properties. Its beneficial role in insulin resistance, dyslipidemia, hypertension, and obesity is being continuously documented. The fermentability of β-glucans and their ability to form highly viscous solutions in the human gut may constitute the basis of their health benefits. Consequently, the applicability of β-glucan as a food ingredient is being widely considered with the dual purposes of increasing the fiber content of food products and enhancing their health properties. Therefore, this paper explores the role of β-glucans in the prevention and treatment of characteristics of the metabolic syndrome, their underlying mechanisms of action, and their potential in food applications.
Collapse
|
24
|
Hamed EA, Zakary MM, Ahmed NS, Gamal RM. Circulating leptin and insulin in obese patients with and without type 2 diabetes mellitus: relation to ghrelin and oxidative stress. Diabetes Res Clin Pract 2011; 94:434-41. [PMID: 21924513 DOI: 10.1016/j.diabres.2011.08.023] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2011] [Revised: 08/02/2011] [Accepted: 08/19/2011] [Indexed: 12/22/2022]
Abstract
AIM This case control study aimed to investigate relationship between appetite hormones (ghrelin and leptin) and body mass index (BMI), insulin and oxidative stress in simple obese and type 2 diabetes (T2DM) obese patients. METHODS Thirty healthy controls; 30 simple obese and 30 T2DM obese patients were enrolled. Demographic and clinical data of all participants were reported. Serum levels of fasting blood glucose (FBG), postprandial blood glucose (PBG), lipid peroxide (LPO) and nitric oxide (NO) were measured by chemical methods while, insulin, leptin and ghrelin by ELISA kits. RESULTS Serum levels of insulin, leptin, LPO were significantly higher while, ghrelin was significantly lower in simple obese and obese patients with diabetes versus controls. Insulin resistance was found in 76.67% simple obese and 93.33% obese patients with diabetes. Ghrelin showed a positive correlation with PBG in controls; but negative correlation with BMI in simple obese and with NO in obese patients with diabetes. Positive correlations were found between LPO and FBG, insulin, homeostasis model assessment of insulin resistance (HOMA-IR) and between leptin and FBG in obese patients with diabetes. CONCLUSIONS Our results suggested that hyperinsulinemia and hyperleptinemia may be most important mechanisms in decreasing ghrelin and inducing oxidative stress in simple obese and T2DM obese patients.
Collapse
Affiliation(s)
- Enas A Hamed
- Department of Physiology, Faculty of Medicine, Assiut University, P.O. Box 71526, Assiut, Egypt.
| | | | | | | |
Collapse
|
25
|
Zhao H, Yannaing S, Thanthan S, Kuwayama H. Sulfated gastrin stimulates ghrelin and growth hormone release but inhibits insulin secretion in cattle. Peptides 2011; 32:2218-24. [PMID: 22001226 DOI: 10.1016/j.peptides.2011.09.023] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2011] [Revised: 09/27/2011] [Accepted: 09/29/2011] [Indexed: 11/25/2022]
Abstract
This study was designed to determine the effects of gastrin on the circulating levels of ghrelin, growth hormone (GH), insulin, glucagon and glucose in ruminants. Two experiments were done in eight Holstein steers. Animals were randomly assigned to receive intravenous bolus injections: (1) 0.1% bovine serum albumin in saline as vehicle, 0.8, 4.0 and 20.0 μg/kg body weight (BW) of bovine sulfated gastrin-34; (2) vehicle, 0.53 μg/kg BW of bovine sulfated gastrin-17 alone or combined with 20.0 μg/kg BW of [D-Lys(3)]-GHRP-6, the selective antagonist of GHS-R1a. Blood samples were collected from -10 to 150 min relative to injection time. Concentrations of acyl and total ghrelin in response to gastrin-34 injection were significantly increased in a dose-dependent manner. Concentrations of GH were also markedly elevated by gastrin-34 injection; however, the effect of 20.0 μg/kg was weaker than that of 4.0 μg/kg. The three doses of gastrin-34 equally decreased insulin levels within 15 min and maintained the level until the time of last sampling. Gastrin-34 had no effect (P > 0.05) on the levels of glucagon and glucose. Levels of acyl ghrelin increased after administration of gastrin-17 alone or combined with [D-Lys(3)]-GHRP-6; however, [D-Lys(3)]-GHRP-6 did not block the elevation of GH by gastrin-17. The present results indicate that sulfated gastrin stimulates both ghrelin and GH release, but the GHS-R1a may not contribute to the release of GH by gastrin. Moreover, sulfated gastrin seems to indirectly maintain the homeostasis of blood glucose through the down-regulation of insulin in ruminants.
Collapse
Affiliation(s)
- Hongqiong Zhao
- Department of Life Science and Agriculture, Obihiro University of Agriculture and Veterinary Medicine, Nishi 2-11, Inada, Obihiro 080-8555, Japan.
| | | | | | | |
Collapse
|
26
|
Rudovich NN, Nikiforova VJ, Otto B, Pivovarova O, Gögebakan O, Erban A, Möhlig M, Weickert MO, Spranger J, Tschöp MH, Willmitzer L, Nauck M, Pfeiffer AFH. Metabolomic linkage reveals functional interaction between glucose-dependent insulinotropic polypeptide and ghrelin in humans. Am J Physiol Endocrinol Metab 2011; 301:E608-17. [PMID: 21586700 DOI: 10.1152/ajpendo.00154.2011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The gastric peptide ghrelin promotes energy storage, appetite, and food intake. Nutrient intake strongly suppresses circulating ghrelin via molecular mechanisms possibly involving insulin and gastrointestinal hormones. On the basis of the growing evidence that glucose-dependent insulinotropic polypeptide (GIP) is involved in the control of fuel metabolism, we hypothesized that GIP and/or insulin, directly or via changes in plasma metabolites, might affect circulating ghrelin. Fourteen obese subjects were infused with GIP (2.0 pmol·kg(-1)·min(-1)) or placebo in the fasting state during either euglycemic hyperinsulinemic (EC) or hyperglycemic hyperinsulinemic clamps (HC). Apart from analysis of plasma ghrelin and insulin levels, GC-TOF/MS analysis was applied to create a hormone-metabolite network for each experiment. The GIP and insulin effects on circulating ghrelin were analyzed within the framework of those networks. In the HC, ghrelin levels decreased in the absence (19.2% vs. baseline, P = 0.028) as well as in the presence of GIP (33.8%, P = 0.018). Ghrelin levels were significantly lower during HC with GIP than with placebo, despite insulin levels not differing significantly. In the GIP network combining data on GIP-infusion, EC+GIP and HC+GIP experiments, ghrelin was integrated into hormone-metabolite networks through a connection to a group of long-chain fatty acids. In contrast, ghrelin was excluded from the network of experiments without GIP. GIP decreased circulating ghrelin and might have affected the ghrelin system via modification of long-chain fatty acid pools. These observations were independent of insulin and offer potential mechanistic underpinnings for the involvement of GIP in systemic control of energy metabolism.
Collapse
Affiliation(s)
- Natalia N Rudovich
- Department of Clinical Nutrition, German Institute of Human Nutrition, Potsdam, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Stengel A, Goebel-Stengel M, Wang L, Shaikh A, Lambrecht NWG, Rivier J, Taché Y. Abdominal surgery inhibits circulating acyl ghrelin and ghrelin-O-acyltransferase levels in rats: role of the somatostatin receptor subtype 2. Am J Physiol Gastrointest Liver Physiol 2011; 301:G239-48. [PMID: 21636529 PMCID: PMC3154605 DOI: 10.1152/ajpgi.00018.2011] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Clinical studies are evaluating the efficacy of synthetic ghrelin agonists in postoperative ileus management. However, the control of ghrelin secretion under conditions of postoperative gastric ileus is largely unknown. Peripheral somatostatin inhibits ghrelin secretion in animals and humans. We investigated the time course of ghrelin changes postsurgery in fasted rats and whether somatostatin receptor subtype 2 (sst(2)) signaling is involved. Abdominal surgery (laparotomy and 1-min cecal palpation) induced a rapid and long-lasting decrease in plasma acyl ghrelin levels as shown by the 64, 67, and 59% reduction at 0.5, 2, and 5 h postsurgery, respectively, compared with sham (anesthesia alone for 10 min, P < 0.05). Levels were partly recovered at 7 h and fully restored at 24 h. The percentage of acyl ghrelin reduction was significantly higher than that of desacyl ghrelin at 2 h postsurgery and not at any other time point. This was associated with a 48 and 23% decrease in gastric and plasma ghrelin-O-acyltransferase protein concentrations, respectively (P < 0.001). Ghrelin-positive cells in the oxyntic mucosa expressed sst(2a) receptor and the sst(2) agonist S-346-011 inhibited fasting acyl ghrelin levels by 64 and 77% at 0.5 and 2 h, respectively. The sst(2) antagonist S-406-028 prevented the abdominal surgery-induced decreased circulating acyl ghrelin but not the delayed gastric emptying assessed 0.5 h postinjection. These data show that activation of sst(2) receptor located on gastric X/A-like cells plays a key role in the rapid inhibition of circulating acyl ghrelin induced by abdominal surgery while not being primarily involved in the early phase of postoperative gastric ileus.
Collapse
Affiliation(s)
- Andreas Stengel
- 1Department of Medicine, CURE Digestive Diseases Research Center, Center for Neurobiology of Stress, Digestive Diseases Division at University of California Los Angeles and Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles;
| | - Miriam Goebel-Stengel
- 1Department of Medicine, CURE Digestive Diseases Research Center, Center for Neurobiology of Stress, Digestive Diseases Division at University of California Los Angeles and Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles;
| | - Lixin Wang
- 1Department of Medicine, CURE Digestive Diseases Research Center, Center for Neurobiology of Stress, Digestive Diseases Division at University of California Los Angeles and Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles;
| | - Almaas Shaikh
- 1Department of Medicine, CURE Digestive Diseases Research Center, Center for Neurobiology of Stress, Digestive Diseases Division at University of California Los Angeles and Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles;
| | - Nils W. G. Lambrecht
- 2Gastrointestinal Endocrinology, Veterans Affairs Long Beach Healthcare System, Long Beach; and
| | - Jean Rivier
- 3Peptide Biology Laboratories, Salk Institute, La Jolla, California
| | - Yvette Taché
- 1Department of Medicine, CURE Digestive Diseases Research Center, Center for Neurobiology of Stress, Digestive Diseases Division at University of California Los Angeles and Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles;
| |
Collapse
|
28
|
Stengel A, Taché Y. Interaction between gastric and upper small intestinal hormones in the regulation of hunger and satiety: ghrelin and cholecystokinin take the central stage. Curr Protein Pept Sci 2011; 12:293-304. [PMID: 21428875 PMCID: PMC3670092 DOI: 10.2174/138920311795906673] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2011] [Accepted: 03/23/2011] [Indexed: 12/13/2022]
Abstract
Several peptides are produced and released from endocrine cells scattered within the gastric oxyntic and the small intestinal mucosa. These peptide hormones are crucially involved in the regulation of gastrointestinal functions and food intake by conveying their information to central regulatory sites located in the brainstem as well as in the forebrain, such as hypothalamic nuclei. So far, ghrelin is the only known hormone that is peripherally produced in gastric X/A-like cells and centrally acting to stimulate food intake, whereas the suppression of feeding seems to be much more redundantly controlled by a number of gut peptides. Cholecystokinin produced in the duodenum is a well established anorexigenic hormone that interacts with ghrelin to modulate food intake indicating a regulatory network located at the first site of contact with nutrients in the stomach and upper small intestine. In addition, a number of peptides including leptin, urocortin 2, amylin and glucagon-like peptide 1 interact synergistically with CCK to potentiate its satiety signaling effect. New developments have led to the identification of additional peptides in X/A-like cells either derived from the pro-ghrelin gene by alternative splicing and posttranslational processing (obestatin) or a distinct gene (nucleobindin2/nesfatin-1) which have been investigated for their influence on food intake.
Collapse
Affiliation(s)
- Andreas Stengel
- Department of Medicine, CURE Digestive Diseases Research Center, Center for Neurobiology of Stress, Digestive Diseases Division UCLA, and VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Yvette Taché
- Department of Medicine, CURE Digestive Diseases Research Center, Center for Neurobiology of Stress, Digestive Diseases Division UCLA, and VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| |
Collapse
|
29
|
Affiliation(s)
- Rao N. Jaladanki
- University of Maryland School of Medicine and Baltimore Veterans Affairs Medical Center
| | - Jian-Ying Wang
- University of Maryland School of Medicine and Baltimore Veterans Affairs Medical Center
| |
Collapse
|
30
|
Abstract
PURPOSE OF REVIEW Disturbances in gastrointestinal hormones have been implicated in the pathogenesis of eating disorders such as anorexia nervosa and bulimia nervosa. However, the contribution of these hormonal changes to the onset and maintenance of eating disorder remains unclear. We focus our review on a selective number of gastrointestinal hormones that are known to play a role in the regulation of short-term or long-term energy balance and examine their association with eating disorder in recently published literature. RECENT FINDINGS Several new studies reported differential changes of ghrelin isoforms during fasting and following nutrient ingestion. New findings on other appetite-regulating hormones (peptide YY, cholecystokinin, incretin hormones and pancreatic polypeptide) at different nutritional states and disease stage have also been reported in subtypes of eating disorder. Most of the changes in peripheral hormones disappeared or partially recovered after the restoration of weight with nutritional and behavioral therapy. SUMMARY Dysregulation of gastrointestinal hormones is more likely to contribute to the maintenance of the disordered eating behavior and related metabolic outcomes as well as the clinical course rather than causing them. A better understanding of this relationship also carries implications for developing targeted hormone-base treatment for eating disorder.
Collapse
Affiliation(s)
- Jenny Tong
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | | |
Collapse
|
31
|
Stengel A, Goebel M, Luckey A, Yuan PQ, Wang L, Taché Y. Cold ambient temperature reverses abdominal surgery-induced delayed gastric emptying and decreased plasma ghrelin levels in rats. Peptides 2010; 31:2229-35. [PMID: 20817059 PMCID: PMC2967623 DOI: 10.1016/j.peptides.2010.08.026] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2010] [Revised: 08/26/2010] [Accepted: 08/26/2010] [Indexed: 02/01/2023]
Abstract
We investigated whether acute cold-induced vagal activation through brainstem thyrotropin-releasing hormone (TRH) signaling influences abdominal surgery-induced delayed gastric emptying (GE) in fasted rats. Laparotomy and cecal palpation or sham (short anesthesia alone) was performed 10 min before or 30 min after cold exposure (4-6°C) lasting 90 min. Non-nutrient GE was assessed during 70-90 min of cold exposure. Control groups remained at room temperature (RT). The stable TRH analog, RX-77368 (50 ng/rat) was injected intracisternally immediately before surgery and GE monitored 30-50 min postsurgery in rats maintained at RT. Plasma acyl (AG) and total ghrelin levels were assessed using the new RAPID blood processing method and radioimmunoassays. Desacyl ghrelin (DAG) was derived from total minus AG. In rats maintained at RT, abdominal surgery decreased GE by 60% compared to sham. Cold before or after surgery or RX-77368 normalized the delayed GE. In non-fasted rats, cold exposure increased plasma AG and DAG levels at 2 h (2.4- and 2.7-times, respectively) and 4 h (2.2- and 2.0-times, respectively) compared to values in rats maintained at RT. In fasted rats, abdominal surgery decreased AG and DAG levels by 2.4- and 2.1-times, respectively, at 90 min. Cold for 90 min after surgery normalized AG and DAG levels to those observed in sham-treated animals kept at RT. These data indicate that endogenous (cold exposure) and exogenous (TRH analog) activation of medullary TRH vagal signaling prevent abdominal surgery-induced delayed GE. The restoration of circulating AG levels inhibited by abdominal surgery may contribute to alleviate postoperative gastric ileus.
Collapse
Affiliation(s)
- Andreas Stengel
- CURE/Digestive Diseases Research Center, Center for Neurobiology of Stress, Department of Medicine, Digestive Diseases Division at the University of California Los Angeles, and VA Greater Los Angeles Health Care System, CA 90073, USA
| | | | | | | | | | | |
Collapse
|
32
|
Yang GK, Yip L, Fredholm BB, Kieffer TJ, Kwok YN. Involvement of adenosine signaling in controlling the release of ghrelin from the mouse stomach. J Pharmacol Exp Ther 2010; 336:77-86. [PMID: 20876230 DOI: 10.1124/jpet.110.171280] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Ghrelin, a potent orexigenic hormone released from the stomach, is important in regulating energy metabolism. Abnormal ghrelin levels are associated with eating disorders and metabolic diseases. However, factors involved in the regulation of ghrelin release remain unclear. Here, we examined the involvement of adenosine signaling in the control of ghrelin release from the perfused mouse stomach. Adenosine stimulated ghrelin release concentration-dependently, and the A(2A) receptor-selective antagonists 4-(2-[7-amino-2-(2-furyl)[1,2,4]triazolo[2,3-a][1,3,5]triazin-5-ylamino]ethyl)phenol (ZM 241385) and 2-(2-furanyl)-7-(2-phenylethyl)-7H-pyrazolo[4,3-e][1,2,4]triazolo[1,5-c]pyrimidin-5-amine (SCH 58261) abolished the increased release. The A(2A) receptor-selective agonist 2-p-(2-carboxyethyl)phenethylamino-5-N-ethylcarboxamidoadenosine hydrochloride (CGS 21680) augmented ghrelin release concentration-dependently, whereas the A(1) receptor-selective agonist 2-chloro-N(6)-cyclopentyladenosine inhibited ghrelin release. In A(2A) receptor knockout mice, adenosine inhibited ghrelin release, and the A(1) receptor-selective antagonist 8-cyclopentyl-1,3-dipropylxanthine blocked this inhibition. The adenosine deaminase inhibitor erythro-9-(2-hydroxy-3-nonyl)adenine hydrochloride increased ghrelin release in wild-type and A(1) receptor knockout mice but not in A(2A) receptor knockout mice. Colocalization of ghrelin immunoreactivity with A(1) and A(2A) receptor immunoreactivities in the gastric nerve fibers were observed. Colocalization was also detected for ghrelin and A(1) receptor immunoreactivities in the gastric mucosa. Blockade of neural activities with tetrodotoxin abolished the stimulatory effect of adenosine on ghrelin release. In conclusion, adenosine exerts predominantly a tonic A(2A) receptor-mediated stimulatory action on gastric ghrelin release, whereas an A(1) receptor-mediated inhibitory action is also apparent when the tonic excitatory effect was removed.
Collapse
Affiliation(s)
- Gary K Yang
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada, V6T 1Z3
| | | | | | | | | |
Collapse
|
33
|
Stengel A, Goebel M, Wang L, Reeve JR, Taché Y, Lambrecht NW. Lipopolysaccharide differentially decreases plasma acyl and desacyl ghrelin levels in rats: potential role of the circulating ghrelin-acylating enzyme GOAT. Peptides 2010; 31:1689-96. [PMID: 20599577 PMCID: PMC4067316 DOI: 10.1016/j.peptides.2010.06.015] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2010] [Revised: 06/17/2010] [Accepted: 06/17/2010] [Indexed: 12/11/2022]
Abstract
Bacterial lipopolysaccharide (LPS) in rodents is an established model for studying innate immune responses to gram-negative bacteria and mimicking symptoms of infections including reduced food intake associated with decreased circulating total ghrelin levels. The ghrelin-acylating enzyme, ghrelin-O-acyltransferase (GOAT) involved in the formation of acyl ghrelin (AG) was recently identified. We investigated changes in circulating AG, desacyl ghrelin (DG) and GOAT induced by intraperitoneal LPS (100 microg/kg) and associated changes in food intake. Plasma AG and total ghrelin were assessed by radioimmunoassay, GOAT protein by Western blot and mRNA by RT-qPCR. DG was derived from total minus AG. Plasma AG and DG were decreased at 2, 5 and 7 h (p<0.01) post-injection compared to vehicle and recovered at 24 h. At 2 h there was a significantly greater decrease of AG (-53%) than DG (-28%) resulting in a decreased AG/DG ratio (1:5, p<0.01), which thereafter returned to pre-injection values (1:3). This altered ratio was associated with a 38% decrease in plasma GOAT protein compared to vehicle (p<0.001), whereas gastric GOAT protein was slightly increased by 10% (p<0.05). GOAT mRNA expression was unchanged. Food intake was reduced by 58% measured during the 1.5-2 h period post-LPS injection. Decreased plasma AG and DG preceded the rise in rectal temperature and blood glucose that peaked at 7 h. These data indicate that LPS induces a long-lasting reduction of AG and DG levels that may have a bearing with the decrease in food intake. The faster drop in AG than DG within 2 h is associated with reduced circulating GOAT.
Collapse
Affiliation(s)
- Andreas Stengel
- CURE/Digestive Diseases Research Center, Center for Neurobiology of Stress, Department of Medicine, Digestive Diseases Division at the University of California Los Angeles, and VA Greater Los Angeles Health Care System, CA 90073, USA
| | - Miriam Goebel
- CURE/Digestive Diseases Research Center, Center for Neurobiology of Stress, Department of Medicine, Digestive Diseases Division at the University of California Los Angeles, and VA Greater Los Angeles Health Care System, CA 90073, USA
| | - Lixin Wang
- CURE/Digestive Diseases Research Center, Center for Neurobiology of Stress, Department of Medicine, Digestive Diseases Division at the University of California Los Angeles, and VA Greater Los Angeles Health Care System, CA 90073, USA
| | - Joseph R. Reeve
- CURE/Digestive Diseases Research Center, Center for Neurobiology of Stress, Department of Medicine, Digestive Diseases Division at the University of California Los Angeles, and VA Greater Los Angeles Health Care System, CA 90073, USA
| | - Yvette Taché
- CURE/Digestive Diseases Research Center, Center for Neurobiology of Stress, Department of Medicine, Digestive Diseases Division at the University of California Los Angeles, and VA Greater Los Angeles Health Care System, CA 90073, USA
| | - Nils W.G. Lambrecht
- Gastrointestinal Endocrinology, Veterans Affairs Long Beach Healthcare System, Long Beach, CA 90822, USA
| |
Collapse
|
34
|
Veldhuis JD, Bowers CY. Integrating GHS into the Ghrelin System. INTERNATIONAL JOURNAL OF PEPTIDES 2010; 2010:879503. [PMID: 20798846 PMCID: PMC2925380 DOI: 10.1155/2010/879503] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/24/2009] [Accepted: 12/30/2009] [Indexed: 12/21/2022]
Abstract
Oligopeptide derivatives of metenkephalin were found to stimulate growth-hormone (GH) release directly by pituitary somatotrope cells in vitro in 1977. Members of this class of peptides and nonpeptidyl mimetics are referred to as GH secretagogues (GHSs). A specific guanosine triphosphatate-binding protein-associated heptahelical transmembrane receptor for GHS was cloned in 1996. An endogenous ligand for the GHS receptor, acylghrelin, was identified in 1999. Expression of ghrelin and homonymous receptor occurs in the brain, pituitary gland, stomach, endothelium/vascular smooth muscle, pancreas, placenta, intestine, heart, bone, and other tissues. Principal actions of this peptidergic system include stimulation of GH release via combined hypothalamopituitary mechanisms, orexigenesis (appetitive enhancement), insulinostasis (inhibition of insulin secretion), cardiovascular effects (decreased mean arterial pressure and vasodilation), stimulation of gastric motility and acid secretion, adipogenesis with repression of fat oxidation, and antiapoptosis (antagonism of endothelial, neuronal, and cardiomyocyte death). The array of known and proposed interactions of ghrelin with key metabolic signals makes ghrelin and its receptor prime targets for drug development.
Collapse
Affiliation(s)
- Johannes D. Veldhuis
- Department of Medicine, Endocrine Research Unit, Mayo School of Graduate Medical Education, Clinical Translational Science Center, Mayo Clinic, Rochester, MN 55905, USA
| | - Cyril Y. Bowers
- Division of Endocrinology, Department of Internal Medicine, Tulane University Health Sciences Center, New Orleans, LA 70112, USA
| |
Collapse
|
35
|
Stengel A, Goebel M, Wang L, Taché Y. Ghrelin, des-acyl ghrelin and nesfatin-1 in gastric X/A-like cells: role as regulators of food intake and body weight. Peptides 2010; 31:357-69. [PMID: 19944123 PMCID: PMC3166546 DOI: 10.1016/j.peptides.2009.11.019] [Citation(s) in RCA: 101] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2009] [Revised: 11/12/2009] [Accepted: 11/16/2009] [Indexed: 02/07/2023]
Abstract
Numerous peptides released from endocrine cells in the intestinal mucosa were established early on to be involved in the physiological regulation of food intake with a prominent role in termination of food ingestion when nutrients pass along the intestinal tract. Recently, peptides released from X/A-like endocrine cells of the gastric oxyntic mucosa were recognized as additional key players in the regulation of feeding and energy expenditure. Gastric X/A-like cells release the octanoylated peptide, ghrelin, the only known peripherally produced hormone stimulating food intake through interaction with growth hormone secretagogue 1a receptor (GHS-R1a). Additionally, non-octanoylated (des-acyl) ghrelin present in the circulation at higher levels than ghrelin is currently discussed as potential modulator of food intake by opposing ghrelin's action independent from GHS-R1a although the functional significance remains to be established. Obestatin, a ghrelin-associated peptide was initially reported as anorexigenic modulator of ghrelin's orexigenic action. However, subsequent reports did not support this contention. Interesting is the recent identification of nesfatin-1, a peptide derived from the nucleobindin2 gene prominently expressed in gastric X/A-like cells in different vesicles than ghrelin. Circulating nesfatin-1 levels vary with metabolic state and peripheral or central injection inhibits dark phase feeding in rodents. Overall, these data point to an important role of gastric X/A-like cells in food intake regulation through the expression of the orexigenic peptide ghrelin along with des-acyl ghrelin and nesfatin-1 capable of reducing food intake upon exogenous injection although their mechanisms of action and functional significance remain to be established.
Collapse
Affiliation(s)
- Andreas Stengel
- CURE: Digestive Diseases Research Center and Neurobiology of Stress, Digestive Diseases Division, David Geffen School of Medicine at University of California Los Angeles and Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA.
| | | | | | | |
Collapse
|
36
|
Interactions of gastrointestinal peptides: ghrelin and its anorexigenic antagonists. INTERNATIONAL JOURNAL OF PEPTIDES 2010; 2010. [PMID: 20798884 PMCID: PMC2925274 DOI: 10.1155/2010/817457] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/13/2009] [Revised: 10/13/2009] [Accepted: 10/19/2009] [Indexed: 12/21/2022]
Abstract
Food intake behaviour and energy homeostasis are strongly regulated by a complex system of humoral factors and nerval structures constituting the brain-gut-axis. To date the only known peripherally produced and centrally acting peptide that stimulates food intake is ghrelin, which is mainly synthesized in the stomach. Recent data indicate that the orexigenic effect of ghrelin might be influenced by other gastrointestinal peptides such as cholecystokinin (CCK), bombesin, desacyl ghrelin, peptide YY (PYY), as well as glucagon-like peptide (GLP). Therefore, we will review on the interactions of ghrelin with several gastrointestinal factors known to be involved in appetite regulation in order to elucidate the interdependency of peripheral orexigenic and anorexigenic peptides in the control of appetite.
Collapse
|
37
|
|
38
|
Abstract
The aim of the present review is to synthesise and summarise our recent knowledge on the involvement of cholecystokinin (CCK) and gastrin peptides and their receptors in the control of digestive functions and more generally their role in the field of nutrition in mammals. First, we examined the release of these peptides from the gut, focusing on their molecular forms, the factors regulating their release and the signalling pathways mediating their effects. Second, general physiological effects of CCK and gastrin peptides are described with regard to their specific receptors and the role of CCK on vagal mucosal afferent nerve activities. Local effects of CCK and gastrin in the gut are also reported, including gut development, gastrointestinal motility and control of pancreatic functions through vagal afferent pathways, including NO. Third, some examples of the intervention of the CCK and gastrin peptides are exposed in diseases, taking into account intervention of the classical receptor subtypes (CCK1 and CCK2 receptors) and their heterodimerisation as well as CCK-C receptor subtype. Finally, applications and future challenges are suggested in the nutritional field (performances) and in therapy with regards to the molecular forms or in relation with the type of receptor as well as new techniques to be utilised in detection or in therapy of disease. In conclusion, the present review underlines recent developments in this field: CCK and gastrin peptides and their receptors are the key factor of nutritional aspects; a better understanding of the mechanisms involved may increase the efficiency of the nutritional functions and the treatment of abnormalities under pathological conditions.
Collapse
|
39
|
Bauer JM, Haack A, Winning K, Wirth R, Fischer B, Uter W, Erdmann J, Schusdziarra V, Sieber CC. Impaired postprandial response of active ghrelin and prolonged suppression of hunger sensation in the elderly. J Gerontol A Biol Sci Med Sci 2009; 65:307-11. [PMID: 19906820 DOI: 10.1093/gerona/glp174] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND The role of the orexigenic hormone ghrelin is of major interest in the altered appetite regulation of the elderly. METHODS Basal and postprandial levels of active and total ghrelin were measured in 15 younger (mean age 35.4 years) and 19 older (80.7 years) participants following a carbohydrate-rich test meal. RESULTS Our results showed that older participants felt postprandially less hungry and more full. Although basal levels were not significantly different, active and total ghrelin levels declined postprandially only in the younger study participants. Highly significant differences between the two age groups were shown for the changes of the area under the curve for active ghrelin (p = .024). CONCLUSIONS Our study demonstrates for the first time that differences in hunger and satiety sensations in relation to age are paralleled by a substantially different response of acylated and total ghrelin, that is, the absence of a postprandial decline in ghrelin levels.
Collapse
Affiliation(s)
- J M Bauer
- Department of Geriatric Medicine, University of Erlangen-Nuremberg, Prof. Ernst-Nathan-Strasse 1, 90419 Nuremberg, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Mizutani M, Atsuchi K, Asakawa A, Matsuda N, Fujimura M, Inui A, Kato I, Fujimiya M. Localization of acyl ghrelin- and des-acyl ghrelin-immunoreactive cells in the rat stomach and their responses to intragastric pH. Am J Physiol Gastrointest Liver Physiol 2009; 297:G974-80. [PMID: 20501445 DOI: 10.1152/ajpgi.00147.2009] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Acyl ghrelin has a 28-amino acid sequence with O-n-octanoyl acid modification at the serine 3 position, whereas des-acyl ghrelin has no octanoyl acid modification. Although these peptides exert different physiological functions, no previous studies have shown the different localization of acyl ghrelin and des-acyl ghrelin in the stomach. Here we have developed an antibody specific for des-acyl ghrelin that does not crossreact with acyl ghrelin. Both acyl ghrelin- and des-acyl ghrelin-immunoreactive cells were distributed in the oxyntic and antral mucosa of the rat stomach, with higher density in the antral mucosa than oxyntic mucosa. Immunofluorescence double staining showed that acyl ghrelin- and des-acyl ghrelin-positive reactions overlapped in closed-type round cells, whereas des-acyl ghrelin-positive reaction was found in open-type cells in which acyl ghrelin was negative. Acyl ghrelin-/des-acyl ghrelin-positive closed-type cells contain obestatin; on the other hand, des-acyl ghrelin-positive open-type cells contain somatostatin. We measured the release of acyl ghrelin and des-acyl ghrelin in vascularly perfused rat stomach by ELISA, and the effects of different intragastric pH levels on the release of each peptide were examined. The release of des-acyl ghrelin from the perfused stomach was greater at pH 2 than at pH 4; however, the release of acyl ghrelin was not affected by intragastric pH. The present study demonstrated the differential localization of acyl ghrelin and des-acyl ghrelin in the rat stomach and their different responses to the intragastric pH.
Collapse
Affiliation(s)
- Makoto Mizutani
- Department of Anatomy, Shiga University of Medical Science, Otsu, Shiga, Japan
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Shrestha YB, Wickwire K, Giraudo SQ. Direct effects of nutrients, acetylcholine, CCK, and insulin on ghrelin release from the isolated stomachs of rats. Peptides 2009; 30:1187-91. [PMID: 19463754 PMCID: PMC2687321 DOI: 10.1016/j.peptides.2009.02.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2009] [Revised: 02/04/2009] [Accepted: 02/05/2009] [Indexed: 11/29/2022]
Abstract
Ghrelin is a powerful orexigenic peptide predominantly secreted by the stomach. Blood concentration of ghrelin increases before meals and fall postprandial. Its regulation appears to be influenced by the type of macronutrient ingested, the vagus nerve stimulation and by other post-meal stimulated hormonal factors. However, the direct role of nutrients (amino acids or lipids), neuronal (vagal neurotransmitter acetylcholine) and satiety-inducing factor such as CCK are not known. To study this we applied amino acids, lipids, acetylcholine and CCK via vascular perfusion to the isolated stomachs and found that amino acids significantly reduced ghrelin release from the isolated stomach by approximately approximately 30% vs. the control while lipids (10% intralipid) had no affect. Acetylcholine (1 microM) increased ghrelin release from the stomach by approximately 37% whereas insulin (10nM) decreased it by approximately 30% vs. the control. Interestingly, CCK (100 nM) potently increased ghrelin release by approximately 200% vs. the control. Therefore it appears that ghrelin secretion from the stomach is under direct influence of amino acids, neurotransmitter acetylcholine and hormones such as insulin and CCK.
Collapse
Affiliation(s)
| | | | - Silvia Q. Giraudo
- Corresponding Author and Address: Dr. Silvia Giraudo, Department of Foods and Nutrition, 280 Dawson Hall, University of Georgia, Athens, Georgia 30602, U.S.A, , Phone: 706-542-6977, Fax: 706-542-5059
| |
Collapse
|
42
|
Forbes AB, Warren M, Upjohn M, Jackson B, Jones J, Charlier J, Fox MT. Associations between blood gastrin, ghrelin, leptin, pepsinogen and Ostertagia ostertagi antibody concentrations and voluntary feed intake in calves exposed to a trickle infection with O. ostertagi. Vet Parasitol 2009; 162:295-305. [PMID: 19342178 DOI: 10.1016/j.vetpar.2009.03.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2008] [Revised: 02/17/2009] [Accepted: 03/02/2009] [Indexed: 12/01/2022]
Abstract
Twenty-five, castrated male Holstein-cross calves, between 4 and 5 months of age, weighing 156.5+/-12.2 kg and reared under conditions designed to minimise the risk of parasitic infection, were allocated to one of the five treatment groups on the basis of initial bodyweight. The groups were (1) ad libitum (ad lib) fed controls (ALC); (2) ad lib fed infected (INF) and treated with topical eprinomectin on Day 56; (3) controls pair-fed with the INF group (PFC); (4) ad lib fed controls treated with eprinomectin on Days 0 and 56 (E-ALC) and (5) ad lib fed, infected and treated with eprinomectin on Days 0 and 56 (E-INF). Infection comprised a trickle infection with the equivalent of 10,000 larvae of Ostertagia ostertagi per day from Day 0 to Day 56 and the study concluded on Day 77. Parameters measured throughout the study included: liveweight, feed intake, faecal egg counts; plasma pepsinogen, gastrin, ghrelin and leptin; plasma antibodies to adult O. ostertagi. No significant differences in feed intake or liveweight gain were observed between any of the different groups, a finding thought to result from the high quality of feed offered. Significant differences between the INF and control groups however were observed in faecal egg counts, plasma pepsinogen, gastrin and O. ostertagi antibodies, which were all elevated, and leptin, which was reduced. Values of these parameters for the E-INF group were intermediate between the INF and ALC groups. Plasma ghrelin showed no association with either feed intake or parasitism. Further studies are needed to fully elucidate the roles of various biochemical and neuroendocrine mediators for inappetence in ruminants with parasitic gastroenteritis.
Collapse
|
43
|
Brennan IM, Little TJ, Feltrin KL, Smout AJPM, Wishart JM, Horowitz M, Feinle-Bisset C. Dose-dependent effects of cholecystokinin-8 on antropyloroduodenal motility, gastrointestinal hormones, appetite, and energy intake in healthy men. Am J Physiol Endocrinol Metab 2008; 295:E1487-94. [PMID: 18957613 DOI: 10.1152/ajpendo.90791.2008] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
CCK mediates the effects of nutrients on gastrointestinal motility and appetite. Intravenously administered CCK stimulates pyloric pressures, increases plasma PYY, and suppresses ghrelin, all of which may be important in the regulation of appetite and energy intake. The dose-related effects of exogenous CCK on gastrointestinal motility and gut hormone release, and the relationships between these effects and those on energy intake, are uncertain. We hypothesized that 1) intravenous CCK-8 would have dose-dependent effects on antropyloroduodenal (APD) pressures, plasma PYY and ghrelin concentrations, appetite, and energy intake and 2) the suppression of energy intake by CCK-8 would be related to the stimulation of pyloric motility. Ten healthy men (age 26 +/- 2 yr) were studied on four separate occasions in double-blind, randomized fashion. APD pressures, plasma PYY and ghrelin, and appetite were measured during 120-min intravenous infusions of 1) saline ("control") or 2) CCK-8 at 0.33 ("CCK0.33"), 3) 0.66 ("CCK0.66"), or 4) 2.0 ("CCK2.0") ng.kg(-1).min(-1). After 90 min, energy intake at a buffet meal was quantified. CCK-8 dose-dependently stimulated phasic and tonic pyloric pressures and plasma PYY concentrations (r > 0.70, P < 0.05) and reduced desire to eat and energy intake (r > -0.60, P < 0.05) without inducing nausea. There were relationships between basal pyloric pressure and isolated pyloric pressure waves (IPPW) with plasma CCK (r > 0.50, P < 0.01) and between energy intake with IPPW (r = -0.70, P < 0.05). Therefore, our study demonstrates that exogenous CCK-8 has dose-related effects on APD motility, plasma PYY, desire to eat, and energy intake and suggests that the suppression of energy intake is related to the stimulation of IPPW.
Collapse
Affiliation(s)
- Ixchel M Brennan
- Univ. of Adelaide Discipline of Medicine, Royal Adelaide Hospital, North Terrace, Adelaide, SA 5000 Australia
| | | | | | | | | | | | | |
Collapse
|
44
|
Aleixandre A, Miguel M. Dietary Fiber in the Prevention and Treatment of Metabolic Syndrome: A Review. Crit Rev Food Sci Nutr 2008; 48:905-12. [DOI: 10.1080/10408390701761886] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
45
|
DePaula AL, Macedo ALV, Schraibman V, Mota BR, Vencio S. Hormonal evaluation following laparoscopic treatment of type 2 diabetes mellitus patients with BMI 20-34. Surg Endosc 2008; 23:1724-32. [PMID: 18830747 DOI: 10.1007/s00464-008-0168-6] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2008] [Accepted: 09/02/2008] [Indexed: 12/17/2022]
Abstract
BACKGROUND A group of patients with type 2 diabetes mellitus (T2DM) and body mass index (BMI) 20-34 kg/m(2) were submitted to laparoscopic interposition of a segment of ileum into the proximal jejunum or into the proximal duodenum associated to a sleeve gastrectomy. The objective of this study is to evaluate the hormonal changes in the pre- and postoperative period. MATERIALS AND METHODS Hormonal evaluation was done in 58 patients operated between April 2005 and July 2006. Mean age was 51.4 years (40-66 years). Mean BMI was 28.2 (20-34.8) kg/m(2). All patients had had the diagnosis of T2DM for at least 3 years. Mean duration of T2DM was 9.6 years (3-22 years). Two techniques were performed, consisting of different combinations of ileal interposition (II) associated to a sleeve gastrectomy (SG). The following hormones were assayed in the pre- and postoperative period (mean 16 months) at the baseline and following specific food stimulation (30, 60, 120 min): glucogen-like protein 1 (GLP-1), glucose-dependent insulinotropic peptide (GIP), insulin, glucagon, C-peptide, amylin, cholecystokinin (CCK), pancreatic polypeptide (PPP), somatostatin, peptide YY (PYY), ghrelin, adiponectin, resistin, leptin, and interleukin-6 (IL-6). RESULTS Thirty patients had II associated to sleeve gastrectomy (II-SG) and 28 had II with diverted sleeve gastrectomy (II-DSG). GLP1 exhibited an important rise following the two operations, especially after II-DSG (p < 0.001). GIP also exhibited an important rise, with both II-SG and II-DSG being equally effective (p < 0.001). Insulin and amylin showed a significant rise at 30 min. Glucagon decreased slightly. CCK measurements were very low after II-DSG. PPP was also slightly altered by the II-DSG. PYY showed an important increase with both operations (p < 0.001). Ghrelin showed a significant decrease following the two operations (p < 0.001). Somatostatin and IL-6 were not affected (p = 0.632). Both leptin and resistin blood levels decreased. Adiponectin showed a slight increase. Mean postoperative follow-up was 19.2 months. Both II-SG and II-DSG were effective in achieving adequate glycemic control (91.2%). CONCLUSIONS There was a significant hormonal change following laparoscopic ileal interposition. These alterations may explain the promising good results associated to these operations for the treatment of T2DM in the nonmorbidly obese population.
Collapse
|
46
|
Fukumoto K, Nakahara K, Katayama T, Miyazatao M, Kangawa K, Murakami N. Synergistic action of gastrin and ghrelin on gastric acid secretion in rats. Biochem Biophys Res Commun 2008; 374:60-3. [PMID: 18611393 DOI: 10.1016/j.bbrc.2008.06.114] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2008] [Accepted: 06/24/2008] [Indexed: 10/21/2022]
Abstract
Gastrin and ghrelin are secreted from G cells and X/A-like cells in the stomach, respectively, and respective hormones stimulate gastric acid secretion by acting through histamine and the vagus nerve. In this study, we examined the relationship between gastrin, ghrelin and gastric acid secretion in rats. Intravenous (iv) administration of 3 and 10 nmol of gastrin induced transient increases of ghrelin levels within 10 min in a dose-dependent manner. Double immunostaining for ghrelin and gastrin receptor revealed that a proportion of ghrelin cells possess gastrin receptors. Although (iv) administration of gastrin or ghrelin induced significant gastric acid secretion, simultaneous treatment with both hormones resulted in a synergistic, rather than additive, increase of gastric acid secretion. This synergistic increase was not observed in vagotomized rats. These results suggest that gastrin may directly stimulate ghrelin release from the stomach, and that both hormones may increase gastric acid secretion synergistically.
Collapse
Affiliation(s)
- Kaori Fukumoto
- Department of Veterinary Physiology, Faculty of Agriculture, University of Miyazaki, Miyazaki 889-2192, Japan
| | | | | | | | | | | |
Collapse
|
47
|
Ariga H, Imai K, Chen C, Mantyh C, Pappas TN, Takahashi T. Does ghrelin explain accelerated gastric emptying in the early stages of diabetes mellitus? Am J Physiol Regul Integr Comp Physiol 2008; 294:R1807-12. [PMID: 18385464 DOI: 10.1152/ajpregu.00785.2007] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
During the early stages of diabetes, gastric emptying is often accelerated, rather than delayed. The mechanism of accelerated gastric emptying in diabetes has not been fully studied. A recent study showed that plasma ghrelin levels were elevated in diabetes. As postprandial antropyloric coordination plays an important role in mediating solid gastric emptying, we hypothesize that the elevated plasma ghrelin levels increase postprandial antropyloric coordination to accelerate emptying in the early stages of diabetes. To test this hypothesis, rats were made diabetic by streptozotocin (STZ; 50 mg/kg) injection, and, 2 wk later, pre- and postprandial plasma ghrelin levels, antropyloric coordination, and solid gastric emptying were determined. In control rats, plasma ghrelin levels were immediately reduced after feeding. In contrast, plasma ghrelin levels remained within the fasted levels in STZ rats after feeding. In STZ rats, gastric emptying was significantly accelerated (77.4 +/- 3.2%, n = 6), compared with that of control rats (58.8 +/- 2.5%, n = 6, P < 0.05). Treatments with anti-ghrelin antibodies attenuated accelerated gastric emptying in STZ rats (50.1 +/- 3.5%, n = 6, P < 0.05), while having little effect in vehicle control rats. The incidence of postprandial antropyloric coordination was significantly increased in STZ rats, compared with that of control rats (P < 0.05). Treatments with anti-ghrelin antibodies suppressed this enhanced antropyloric coordination in STZ rats. Our study suggests that elevated endogenous ghrelin enhances antropyloric coordination, which accelerates gastric emptying in the early stages of diabetes.
Collapse
Affiliation(s)
- Hajime Ariga
- Department of Surgery, Duke University Medical Center and Durham Veterans Affairs Medical Center, Durham, NC, USA
| | | | | | | | | | | |
Collapse
|
48
|
Erdmann J, Tahbaz R, Lippl F, Wagenpfeil S, Schusdziarra V. Plasma ghrelin levels during exercise — Effects of intensity and duration. ACTA ACUST UNITED AC 2007; 143:127-35. [PMID: 17570540 DOI: 10.1016/j.regpep.2007.05.002] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2006] [Revised: 04/30/2007] [Accepted: 05/01/2007] [Indexed: 11/26/2022]
Abstract
Ghrelin, a recently discovered hormone of gastric origin has been shown to stimulate appetite and food intake. In man it is considered to play a role in energy homeostasis and regulation of somatropic function. As exercise affects hunger/satiety sensations and food intake, at least under some experimental conditions, we investigated the effect of exercise intensity and duration on ghrelin release and subsequent ad libitum food intake in normal weight subjects. Bicycle exercise on an ergometer for 30 min at 50 W which was below the aerob-anaerobic threshold led to an increase of ghrelin which remained unchanged during the higher intensity at 100 W. Respective hunger/satiety ratings and subsequent food intake and postprandial ghrelin suppression were identical and not different from controls. In a second group 7 subjects cycled at 50 W for 30, 60 and 120 min, respectively. Ghrelin concentrations rose significantly by 50-70 pg/ml above baseline for the respective period of exercise. While postexercise premeal ghrelin levels were not significantly different subsequent food intake after 120 min of cycling was significantly greater compared to control, 30 min and 60 min exercise, respectively. The present data suggest that low rather than high-intensity exercise stimulates ghrelin levels independent of exercise duration. Stimulation of food intake during prolonged exercise is most likely not due to changes of ghrelin.
Collapse
Affiliation(s)
- Johannes Erdmann
- Else-Kröner-Fresenius Center of Nutritional Medicine, Technical University of Munich, Germany.
| | | | | | | | | |
Collapse
|
49
|
Katayama T, Shimamoto S, Oda H, Nakahara K, Kangawa K, Murakami N. Glucagon receptor expression and glucagon stimulation of ghrelin secretion in rat stomach. Biochem Biophys Res Commun 2007; 357:865-70. [PMID: 17462598 DOI: 10.1016/j.bbrc.2007.04.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2007] [Accepted: 04/02/2007] [Indexed: 11/23/2022]
Abstract
The present study was performed to evaluate the role of glucagon in the regulation of ghrelin secretion from the rat stomach. mRNA for ghrelin and glucagon receptor was expressed predominantly in the lower body and pylorus of stomach, but little or not in the upper body and cardia. Ghrelin- and glucagon receptor-immunoreactive cells were detected in lamina propria mucosae of stomach and some cells expressed both. Intravenous administration of glucagon caused transient increases in both acyl- and desacyl-ghrelin levels in the gastric vein within 10 min, which was followed by gradual increases in desacyl-ghrelin levels until 60 min. Steady state levels of ghrelin mRNA in the stomach were increased by 1.9-fold 20 min after glucagon administration, but not at 5 or 120 min. These results suggest that glucagon stimulates acute release of both forms of ghrelin and thereafter upregulates synthesis and release of desacyl-ghrelin in the rat stomach.
Collapse
Affiliation(s)
- Tetsuro Katayama
- Frontier Science Research Center, Kibana Campus, University of Miyazaki, Miyazaki 889-2192, Japan.
| | | | | | | | | | | |
Collapse
|
50
|
Erdmann J, Hebeisen Y, Lippl F, Wagenpfeil S, Schusdziarra V. Food intake and plasma ghrelin response during potato-, rice- and pasta-rich test meals. Eur J Nutr 2007; 46:196-203. [PMID: 17497076 DOI: 10.1007/s00394-007-0649-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2006] [Accepted: 02/26/2007] [Indexed: 10/23/2022]
Abstract
OBJECTIVE Complex carbohydrates such as potato, rice and pasta are frequently consumed accompaniments of meat meals and have different effects on satiety, food intake, glucose, and insulin concentrations. The orexigenic gastric hormone ghrelin contributes to feeding regulation and as yet it is unknown whether there is any differential ghrelin response to these starchy food items corresponding to their effects on food intake. METHODS In 11 subjects the effect of satiating amounts of potatoes, rice or pasta consumed together with 150 g pork steak was examined on hunger/satiety ratings, food intake, plasma insulin, glucose and ghrelin concentrations. RESULTS All meals led to comparable quantities of food intake while energy intake was significantly lower after potatoes. Satiety/hunger ratings were significantly different from basal for the entire 4 h period after rice and pasta meals, while they had returned to basal during the 4th hour after potatoes. After rice and pasta insulin rose significantly for 4 h. Ghrelin decreased during the 2nd and 3rd hour. In contrast potatoes stimulated insulin for the initial 2 h only while ghrelin rose significantly by 120 pg/ml over the 4 h period. A significant correlation was observed between ghrelin and hunger ratings while subsequent second meal food and energy intake did not differ irrespective of the preceding ghrelin concentration. CONCLUSION Compared to rice and pasta satiating amounts of potatoes coingested with meat result in lower energy intake and postprandial insulin concentrations, which is not counterbalanced during subsequent food intake despite higher ghrelin concentrations. The present data support the concept that ghrelin can affect hunger sensations but not necessarily food and energy intake.
Collapse
Affiliation(s)
- Johannes Erdmann
- Else-Kröner-Fresenius Centre of Nutritional Medicine, Technical University of Munich, Ismaninger Strasse 22, 81675 Munich, Germany.
| | | | | | | | | |
Collapse
|