1
|
Zhu J, Zhou T, Menggen M, Aimulajiang K, Wen H. Ghrelin regulating liver activity and its potential effects on liver fibrosis and Echinococcosis. Front Cell Infect Microbiol 2024; 13:1324134. [PMID: 38259969 PMCID: PMC10800934 DOI: 10.3389/fcimb.2023.1324134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 12/15/2023] [Indexed: 01/24/2024] Open
Abstract
Ghrelin widely exists in the central nervous system and peripheral organs, and has biological activities such as maintaining energy homeostasis, regulating lipid metabolism, cell proliferation, immune response, gastrointestinal physiological activities, cognition, memory, circadian rhythm and reward effects. In many benign liver diseases, it may play a hepatoprotective role against steatosis, chronic inflammation, oxidative stress, mitochondrial dysfunction, endoplasmic reticulum stress and apoptosis, and improve liver cell autophagy and immune response to improve disease progression. However, the role of Ghrelin in liver Echinococcosis is currently unclear. This review systematically summarizes the molecular mechanisms by which Ghrelin regulates liver growth metabolism, immune-inflammation, fibrogenesis, proliferation and apoptosis, as well as its protective effects in liver fibrosis diseases, and further proposes the role of Ghrelin in liver Echinococcosis infection. During the infectious process, it may promote the parasitism and survival of parasites on the host by improving the immune-inflammatory microenvironment and fibrosis state, thereby accelerating disease progression. However, there is currently a lack of targeted in vitro and in vivo experimental evidence for this viewpoint.
Collapse
Affiliation(s)
- Jiang Zhu
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
- Department of Hepatobiliary and Hydatid Disease, Digestive and Vascular Surgery Center Therapy Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Tanfang Zhou
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
- Department of Hepatobiliary and Hydatid Disease, Digestive and Vascular Surgery Center Therapy Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Meng Menggen
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Kalibixiati Aimulajiang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Hao Wen
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
- Department of Hepatobiliary and Hydatid Disease, Digestive and Vascular Surgery Center Therapy Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| |
Collapse
|
2
|
Kim DM, Lee JH, Pan Q, Han HW, Shen Z, Eshghjoo S, Wu CS, Yang W, Noh JY, Threadgill DW, Guo S, Wright G, Alaniz R, Sun Y. Nutrient-sensing growth hormone secretagogue receptor in macrophage programming and meta-inflammation. Mol Metab 2024; 79:101852. [PMID: 38092245 PMCID: PMC10772824 DOI: 10.1016/j.molmet.2023.101852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 12/03/2023] [Accepted: 12/08/2023] [Indexed: 12/20/2023] Open
Abstract
OBJECTIVE Obesity-associated chronic inflammation, aka meta-inflammation, is a key pathogenic driver for obesity-associated comorbidity. Growth hormone secretagogue receptor (GHSR) is known to mediate the effects of nutrient-sensing hormone ghrelin in food intake and fat deposition. We previously reported that global Ghsr ablation protects against diet-induced inflammation and insulin resistance, but the site(s) of action and mechanism are unknown. Macrophages are key drivers of meta-inflammation. To unravel the role of GHSR in macrophages, we generated myeloid-specific Ghsr knockout mice (LysM-Cre;Ghsrf/f). METHODS LysM-Cre;Ghsrf/f and control Ghsrf/f mice were subjected to 5 months of high-fat diet (HFD) feeding to induce obesity. In vivo, metabolic profiling of food intake, physical activity, and energy expenditure, as well as glucose and insulin tolerance tests (GTT and ITT) were performed. At termination, peritoneal macrophages (PMs), epididymal white adipose tissue (eWAT), and liver were analyzed by flow cytometry and histology. For ex vivo studies, bone marrow-derived macrophages (BMDMs) were generated from the mice and treated with palmitic acid (PA) or lipopolysaccharide (LPS). For in vitro studies, macrophage RAW264.7 cells with Ghsr overexpression or Insulin receptor substrate 2 (Irs2) knockdown were studied. RESULTS We found that Ghsr expression in PMs was increased under HFD feeding. In vivo, HFD-fed LysM-Cre;Ghsrf/f mice exhibited significantly attenuated systemic inflammation and insulin resistance without affecting food intake or body weight. Tissue analysis showed that HFD-fed LysM-Cre;Ghsrf/f mice have significantly decreased monocyte/macrophage infiltration, pro-inflammatory activation, and lipid accumulation, showing elevated lipid-associated macrophages (LAMs) in eWAT and liver. Ex vivo, Ghsr-deficient macrophages protected against PA- or LPS-induced pro-inflammatory polarization, showing reduced glycolysis, increased fatty acid oxidation, and decreased NF-κB nuclear translocation. At molecular level, GHSR metabolically programs macrophage polarization through PKA-CREB-IRS2-AKT2 signaling pathway. CONCLUSIONS These novel results demonstrate that macrophage GHSR plays a key role in the pathogenesis of meta-inflammation, and macrophage GHSR promotes macrophage infiltration and induces pro-inflammatory polarization. These exciting findings suggest that GHSR may serve as a novel immunotherapeutic target for the treatment of obesity and its associated comorbidity.
Collapse
Affiliation(s)
- Da Mi Kim
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA
| | - Jong Han Lee
- Department of Marine Bioindustry, Hanseo University, Seosan 31962, South Korea; USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College Medicine, Houston, TX 77030, USA
| | - Quan Pan
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA
| | - Hye Won Han
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA
| | - Zheng Shen
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA
| | - Sahar Eshghjoo
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX 77807, USA; Agilent technologies, Aanta Clara, CA 95051, USA
| | - Chia-Shan Wu
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA; USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College Medicine, Houston, TX 77030, USA
| | - Wanbao Yang
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA
| | - Ji Yeon Noh
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA
| | - David W Threadgill
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA; Texas A&M Institute for Genome Sciences and Society, Department of Cell Biology and Genetics, Texas A&M University, College Station, TX 77843, USA
| | - Shaodong Guo
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA
| | - Gus Wright
- Department of Veterinary Pathobiology, Texas A&M University, College Station, TX 77843, USA
| | - Robert Alaniz
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX 77807, USA; Tlaloc Therapeutics Inc., College Station, TX 77845, USA
| | - Yuxiang Sun
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA; USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College Medicine, Houston, TX 77030, USA.
| |
Collapse
|
3
|
Peris-Sampedro F, Le May MV, Stoltenborg I, Schéle E, Dickson SL. A skeleton in the cupboard in ghrelin research: Where are the skinny dwarfs? J Neuroendocrinol 2021; 33:e13025. [PMID: 34427011 DOI: 10.1111/jne.13025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 07/09/2021] [Accepted: 08/05/2021] [Indexed: 12/15/2022]
Abstract
Based on studies delivering ghrelin or ghrelin receptor agonists, we have learned a great deal about the importance of the brain ghrelin signalling system for a wide range of physiological processes that include feeding behaviours, growth hormone secretion and glucose homeostasis. Because these processes can be considered as essential to life, the question arises as to why mouse models of depleted ghrelin signalling are not all skinny dwarfs with a host of behavioural and metabolic problems. Here, we provide a systematic detailed review of the phenotype of mice with deficient ghrelin signalling to help better understand the relevance and importance of the brain ghrelin signalling system, with a particular emphasis on those questions that remain unanswered.
Collapse
Affiliation(s)
- Fiona Peris-Sampedro
- Department of Physiology/Endocrine, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Marie V Le May
- Department of Physiology/Endocrine, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Iris Stoltenborg
- Department of Physiology/Endocrine, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Erik Schéle
- Department of Physiology/Endocrine, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Suzanne L Dickson
- Department of Physiology/Endocrine, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
4
|
Xi S, Zhai X, Wang Y, Gong Y, Fu B, Gao C, Guo X, Li Y, Wang Z, Huang S, Lu D, Zhao Y, Qian L, Wang Y. The Ciji-Hua'ai-Baosheng II Formula Attenuates Chemotherapy-Induced Anorexia in Mice With H 22 Hepatocellular Carcinoma. Front Pharmacol 2021; 12:715824. [PMID: 34489705 PMCID: PMC8416666 DOI: 10.3389/fphar.2021.715824] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 07/21/2021] [Indexed: 01/04/2023] Open
Abstract
Background: Ciji-Hua’ai-Baosheng II Formula (CHB-II-F) is a traditional Chinese medicine formula, which specifically targets different aspects of chemotherapy-induced adverse effects in patients with cancer. In our clinical application, CHB-II-F significantly alleviated chemotherapy-induced anorexia (loss of appetite) and improved the quality of life for patients with tumor during and after chemotherapy. However, the mechanism of CHB-II-F in alleviation of chemotherapy-induced anorexia remains to be further investigated. Aim of Study: To explore the therapeutic effect and mechanism of CHB-II-F on chemotherapy-induced anorexia in the mice model of H22 hepatoma. Materials and Methods: A total of 72 Kunming mice of SPF grade were inoculated subcutaneously with H22 hepatoma cells into the right anterior armpit of the mice. After 1 week of seeding, mice were injected intraperitoneally with a high dose of 5-fluorouracil (200 mg/kg 5-FU) to establish the model of chemotherapy. The mice were randomly divided into six groups: untreated group, 5-FU group, 5-FU plus Yangzheng Xiaoji capsule (YZXJC) group, and three groups of 5-FU plus different concentrations of CHB-II-F. All the mice in each group were treated for 14 days. The body weight, food intake, tumor volume, and tumor weight of mice were measured, and pathological examinations of tumor tissue, stomach, and duodenum were carried out. Expressions of serum Leptin, Neuropeptide Y (NPY), epidermal cell growth factor (EGF), Motilin (MTL), Orexin A (OXA), Gastrin (GAS), Ghrelin, Prostaglandin E2 (PGE2), and jejunum superoxide dismutase (SOD) activity and malondialdehyde (MDA) content were examined. The protein and mRNA levels of proopiomelanocortin (POMC), Orexin receptor 1 (OX1R), neuropeptide Y (NPY), cocaine and amphetamine regulated transcript peptide (CART), Agouti gene-related protein (AgRP), Leptin receptor (Ob-R), and Ghrelin receptor (GHSR) were examined in hypothalamus, and the protein levels of substance P (SP) and 5-hydroxytryptamine (5-HT) in duodenum were measured. Results: The combination of CHB-II-F and 5-FU could enhance the inhibitory effect of 5-FU on tumor. The tumor inhibition rates of 5-FU group, YZXJC group, CHB-II-F(H) group, CHB-II-F(M) group, and CHB-II-F(L) group were 58.88, 28.08, 54.96, 37.69, and 28.61%, respectively. Compared with untreated group and 5-FU group, CHB-II-F significantly increased the body weight and food intake of tumor-bearing mice; increased the content of NPY, Orexin A, Ghrelin, GAS, MTL, EGF, and PGE2 in serum and the activity of SOD in jejunum; and decreased the content of Leptin in serum and the content of MDA in jejunum. Compared with untreated group and 5-FU group, CHB-II-F also enhanced the expression of OX1R, GHSR, NPY, and AgRP protein and gene and decreased the expression of Ob-R, POMC, and CART protein and gene in hypothalamus of mice, and the gene expression was consistent with the protein expression. In addition, CHB-II-F decreased the expression of 5-HT and SP protein in duodenum. Conclusion: In the murine model of H22 hepatocellular carcinoma (HCC) receiving chemotherapy, CHB-II-F enhances the inhibitory effect of 5-FU on tumor, significantly improves the pathological injury of gastrointestinal tract caused by chemotherapy, and regulates the secretion of gastrointestinal hormones. It may alleviate chemotherapy-induced anorexia by affecting appetite regulatory factors in the feeding area of hypothalamus central nervous system and peripheral appetite regulatory factors.
Collapse
Affiliation(s)
- Shengyan Xi
- Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen, China.,Department of Traditional Chinese Medicine, Xiang'an Hospital of Xiamen University, Xiamen, China
| | - Xiangyang Zhai
- Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen, China
| | - Yanan Wang
- Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen, China
| | - Yuewen Gong
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Biqian Fu
- The First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chunling Gao
- Department of Radiotherapy, Chenggong Hospital of Xiamen University, Xiamen, China
| | - Xuehui Guo
- Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen, China
| | - Yunhong Li
- Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen, China
| | - Zheng Wang
- Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen, China
| | - Shuqiong Huang
- Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen, China
| | - Dawei Lu
- Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen, China
| | - Yufang Zhao
- Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen, China
| | - Linchao Qian
- Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen, China.,Department of Traditional Chinese Medicine, Xiang'an Hospital of Xiamen University, Xiamen, China
| | - Yanhui Wang
- Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen, China.,Department of Traditional Chinese Medicine, Xiang'an Hospital of Xiamen University, Xiamen, China
| |
Collapse
|
5
|
Özcan B, Delhanty PJD, Huisman M, Visser JA, Neggers SJ, van der Lely AJ. Overweight and obesity in type 1 diabetes is not associated with higher ghrelin concentrations. Diabetol Metab Syndr 2021; 13:79. [PMID: 34294136 PMCID: PMC8296697 DOI: 10.1186/s13098-021-00699-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 07/09/2021] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Several studies have demonstrated suppressed levels of acylated (AG) and unacylated ghrelin (UAG) in patients with type 2 diabetes. However, the role of these hormones in type 1 diabetes has not been extensively studied. This study assessed the relationship between AG and UAG levels and body composition in patients with type 1 diabetes. METHODS We selected eighteen patients with type 1 diabetes and divided them into two groups: non-obese (BMI < 25 kg/m2) and overweight (BMI ≥ 25 kg/m2). Demographics, parameters of body composition and serum parameters including AG and UAG, were assessed. RESULTS The patients with a BMI ≥ 25 kg/m2 were older and had a longer duration of diabetes. AG and UAG levels were not significantly different between non-obese and overweight groups (mean AG non-obese ± SD: 44.5 ± 29.4 pg/ml and mean UAG non-obese 42.4 ± 20.7 pg/ml vs mean AG overweight ± SD: 46.1 ± 29.6 pg/ml and mean UAG overweight 47.2 ± 18.2 pg/ml). AG/UAG ratios did not discriminate between these groups. There was a positive association of insuline dose/kg bodyweight with BMI (r2 = 0.45, p = 0.002). CONCLUSIONS Surprisingly, unlike non-diabetics and in T2D, we did not observe a difference in plasma levels of AG and UAG between normal weight and overweight adult type 1 diabetics. However, we did observe a positive correlation between BMI and insuline dose/kg bodyweight, suggesting that exogenous insulin is more important than the ghrelin system in the development of obesity in type 1 diabetes.
Collapse
Affiliation(s)
- Behiye Özcan
- Departments of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, PO Box 2040, 3000 CA, Rotterdam, The Netherlands.
| | - Patric J D Delhanty
- Departments of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Martin Huisman
- Departments of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Jenny A Visser
- Departments of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Sebastian J Neggers
- Departments of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Aart Jan van der Lely
- Departments of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
| |
Collapse
|
6
|
Schalla MA, Taché Y, Stengel A. Neuroendocrine Peptides of the Gut and Their Role in the Regulation of Food Intake. Compr Physiol 2021; 11:1679-1730. [PMID: 33792904 DOI: 10.1002/cphy.c200007] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The regulation of food intake encompasses complex interplays between the gut and the brain. Among them, the gastrointestinal tract releases different peptides that communicate the metabolic state to specific nuclei in the hindbrain and the hypothalamus. The present overview gives emphasis on seven peptides that are produced by and secreted from specialized enteroendocrine cells along the gastrointestinal tract in relation with the nutritional status. These established modulators of feeding are ghrelin and nesfatin-1 secreted from gastric X/A-like cells, cholecystokinin (CCK) secreted from duodenal I-cells, glucagon-like peptide 1 (GLP-1), oxyntomodulin, and peptide YY (PYY) secreted from intestinal L-cells and uroguanylin (UGN) released from enterochromaffin (EC) cells. © 2021 American Physiological Society. Compr Physiol 11:1679-1730, 2021.
Collapse
Affiliation(s)
- Martha A Schalla
- Charité Center for Internal Medicine and Dermatology, Department for Psychosomatic Medicine, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Yvette Taché
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, CURE: Digestive Diseases Research Center, David Geffen School of Medicine, UCLA, Los Angeles, California, USA.,VA Greater Los Angeles Healthcare System, Los Angeles, California, USA
| | - Andreas Stengel
- Charité Center for Internal Medicine and Dermatology, Department for Psychosomatic Medicine, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany.,Department of Psychosomatic Medicine and Psychotherapy, Medical University Hospital Tübingen, Tübingen, Germany
| |
Collapse
|
7
|
"A LEAP 2 conclusions? Targeting the ghrelin system to treat obesity and diabetes". Mol Metab 2020; 46:101128. [PMID: 33246141 PMCID: PMC8085568 DOI: 10.1016/j.molmet.2020.101128] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 11/15/2020] [Accepted: 11/20/2020] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND The hormone ghrelin stimulates food intake, promotes adiposity, increases body weight, and elevates blood glucose. Consequently, alterations in plasma ghrelin levels and the functioning of other components of the broader ghrelin system have been proposed as potential contributors to obesity and diabetes. Furthermore, targeting the ghrelin system has been proposed as a novel therapeutic strategy for obesity and diabetes. SCOPE OF REVIEW The current review focuses on the potential for targeting ghrelin and other proteins comprising the ghrelin system as a treatment for obesity and diabetes. The main components of the ghrelin system are introduced. Data supporting a role for the endogenous ghrelin system in the development of obesity and diabetes along with data that seemingly refute such a role are outlined. An argument for further research into the development of ghrelin system-targeted therapeutic agents is delineated. Also, an evidence-based discussion of potential factors and contexts that might influence the efficacy of this class of therapeutics is provided. MAJOR CONCLUSIONS It would not be a "leap to" conclusions to suggest that agents which target the ghrelin system - including those that lower acyl-ghrelin levels, raise LEAP2 levels, block GHSR activity, and/or raise desacyl-ghrelin signaling - could represent efficacious novel treatments for obesity and diabetes.
Collapse
|
8
|
Sun S, Corbeels K, Desmet L, Segers A, Wang Q, Van Der Schueren B, Depoortere I. Involvement of the GHSR in the developmental programming and metabolic disturbances induced by maternal undernutrition. J Nutr Biochem 2020; 85:108468. [PMID: 32750410 DOI: 10.1016/j.jnutbio.2020.108468] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 07/03/2020] [Accepted: 07/13/2020] [Indexed: 12/18/2022]
Abstract
The mismatch between maternal undernutrition and adequate nutrition after birth increases the risk of developing metabolic diseases. We aimed to investigate whether the hyperghrelinemia during maternal undernourishment rewires the hypothalamic development of the offspring and contributes to the conversion to an obese phenotype when fed a high-fat diet (HFD). Pregnant C57BL/6 J, wild type (WT) and ghrelin receptor (GHSR)-/- mice were assigned to either a normal nourished (NN) group, or an undernutrition (UN) (30% food restricted) group. All pups were fostered by NN Swiss mice. After weaning, pups were fed a normal diet, followed by a HFD from week 9. Plasma ghrelin levels peaked at postnatal day 15 (P15) in both C57BL/6 J UN and NN pups. Hypothalamic Ghsr mRNA expression was upregulated at P15 in UN pups compared to NN pups and inhibited agouti-related peptide (AgRP) projections. Adequate lactation increased body weight of UN WT but not of GHSR-/- pups compared to NN littermates. After weaning with a HFD, body weight and food intake was higher in WT UN pups but lower in GHSR-/- UN pups than in NN controls. The GHSR prevented a decrease in ambulatory activity and oxygen consumption in UN offspring during ad libitum feeding. Maternal undernutrition triggers developmental changes in the hypothalamus in utero which were further affected by adequate feeding after birth during the postnatal period by affecting GHSR signaling. The GHSR contributes to the hyperphagia and the increase in body weight when maternal undernutrition is followed by an obesity prone life environment.
Collapse
Affiliation(s)
- Shu Sun
- Gut Peptide Research Lab, Targid, KU Leuven,3000 Leuven, Belgium
| | - Katrien Corbeels
- Clinical and Experimental Endocrinology, Department of Chronic Diseases, Metabolism and Ageing (CHROMETA), KU Leuven,3000 Leuven, Belgium
| | - Louis Desmet
- Gut Peptide Research Lab, Targid, KU Leuven,3000 Leuven, Belgium
| | - Anneleen Segers
- Gut Peptide Research Lab, Targid, KU Leuven,3000 Leuven, Belgium
| | - Qiaoling Wang
- Gut Peptide Research Lab, Targid, KU Leuven,3000 Leuven, Belgium
| | - Bart Van Der Schueren
- Clinical and Experimental Endocrinology, Department of Chronic Diseases, Metabolism and Ageing (CHROMETA), KU Leuven,3000 Leuven, Belgium
| | - Inge Depoortere
- Gut Peptide Research Lab, Targid, KU Leuven,3000 Leuven, Belgium.
| |
Collapse
|
9
|
Mani BK, Puzziferri N, He Z, Rodriguez JA, Osborne-Lawrence S, Metzger NP, Chhina N, Gaylinn B, Thorner MO, Thomas EL, Bell JD, Williams KW, Goldstone AP, Zigman JM. LEAP2 changes with body mass and food intake in humans and mice. J Clin Invest 2020; 129:3909-3923. [PMID: 31424424 DOI: 10.1172/jci125332] [Citation(s) in RCA: 136] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 06/11/2019] [Indexed: 12/11/2022] Open
Abstract
Acyl-ghrelin administration increases food intake, body weight, and blood glucose. In contrast, mice lacking ghrelin or ghrelin receptors (GHSRs) exhibit life-threatening hypoglycemia during starvation-like conditions, but do not consistently exhibit overt metabolic phenotypes when given ad libitum food access. These results, and findings of ghrelin resistance in obese states, imply nutritional state dependence of ghrelin's metabolic actions. Here, we hypothesized that liver-enriched antimicrobial peptide-2 (LEAP2), a recently characterized endogenous GHSR antagonist, blunts ghrelin action during obese states and postprandially. To test this hypothesis, we determined changes in plasma LEAP2 and acyl-ghrelin due to fasting, eating, obesity, Roux-en-Y gastric bypass (RYGB), vertical sleeve gastrectomy (VSG), oral glucose administration, and type 1 diabetes mellitus (T1DM) using humans and/or mice. Our results suggest that plasma LEAP2 is regulated by metabolic status: its levels increased with body mass and blood glucose and decreased with fasting, RYGB, and in postprandial states following VSG. These changes were mostly opposite of those of acyl-ghrelin. Furthermore, using electrophysiology, we showed that LEAP2 both hyperpolarizes and prevents acyl-ghrelin from activating arcuate NPY neurons. We predict that the plasma LEAP2/acyl-ghrelin molar ratio may be a key determinant modulating acyl-ghrelin activity in response to body mass, feeding status, and blood glucose.
Collapse
Affiliation(s)
- Bharath K Mani
- Division of Hypothalamic Research.,Division of Endocrinology & Metabolism, Department of Internal Medicine.,Department of Psychiatry, and
| | - Nancy Puzziferri
- Department of Surgery, UT Southwestern Medical Center, Dallas, Texas, USA.,Department of Surgery, Veterans Administration North Texas Heath Care System, Dallas, Texas, USA
| | | | - Juan A Rodriguez
- Division of Hypothalamic Research.,Division of Endocrinology & Metabolism, Department of Internal Medicine.,Department of Psychiatry, and
| | - Sherri Osborne-Lawrence
- Division of Hypothalamic Research.,Division of Endocrinology & Metabolism, Department of Internal Medicine.,Department of Psychiatry, and
| | - Nathan P Metzger
- Division of Hypothalamic Research.,Division of Endocrinology & Metabolism, Department of Internal Medicine.,Department of Psychiatry, and
| | - Navpreet Chhina
- PsychoNeuroEndocrinology Research Group, Neuropsychopharmacology Unit, Centre for Psychiatry, and.,Computational, Cognitive and Clinical Neuroimaging Laboratory, Division of Brain Sciences, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | - Bruce Gaylinn
- Department of Endocrinology, University of Virginia, Charlottesville, Virginia, USA
| | - Michael O Thorner
- Department of Endocrinology, University of Virginia, Charlottesville, Virginia, USA
| | - E Louise Thomas
- Research Centre for Optimal Health, University of Westminster, London, United Kingdom
| | - Jimmy D Bell
- Research Centre for Optimal Health, University of Westminster, London, United Kingdom
| | | | - Anthony P Goldstone
- PsychoNeuroEndocrinology Research Group, Neuropsychopharmacology Unit, Centre for Psychiatry, and.,Computational, Cognitive and Clinical Neuroimaging Laboratory, Division of Brain Sciences, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | - Jeffrey M Zigman
- Division of Hypothalamic Research.,Division of Endocrinology & Metabolism, Department of Internal Medicine.,Department of Psychiatry, and
| |
Collapse
|
10
|
Pierre A, Van Schuerbeek A, Allaoui W, Van Laere S, Singewald N, Van Eeckhaut A, Smolders I, De Bundel D. Effects of ghrelin receptor activation on forebrain dopamine release, conditioned fear and fear extinction in C57BL/6J mice. J Neurochem 2020; 154:389-403. [DOI: 10.1111/jnc.14996] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 02/25/2020] [Accepted: 02/26/2020] [Indexed: 12/14/2022]
Affiliation(s)
- Anouk Pierre
- Department of Pharmaceutical Sciences Research Group Experimental Pharmacology Center for Neurosciences (C4N) Vrije Universiteit Brussel Brussels Belgium
| | - Andries Van Schuerbeek
- Department of Pharmaceutical Sciences Research Group Experimental Pharmacology Center for Neurosciences (C4N) Vrije Universiteit Brussel Brussels Belgium
| | - Wissal Allaoui
- Department of Pharmaceutical Sciences Research Group Experimental Pharmacology Center for Neurosciences (C4N) Vrije Universiteit Brussel Brussels Belgium
| | - Sven Van Laere
- Interfaculty Center Data Processing & Statistics Vrije Universiteit Brussel Brussels Belgium
| | - Nicolas Singewald
- Department of Pharmacology and Toxicology Institute of Pharmacy and CMBI University of Innsbruck Innsbruck Austria
| | - Ann Van Eeckhaut
- Department of Pharmaceutical Sciences Research Group Experimental Pharmacology Center for Neurosciences (C4N) Vrije Universiteit Brussel Brussels Belgium
| | - Ilse Smolders
- Department of Pharmaceutical Sciences Research Group Experimental Pharmacology Center for Neurosciences (C4N) Vrije Universiteit Brussel Brussels Belgium
| | - Dimitri De Bundel
- Department of Pharmaceutical Sciences Research Group Experimental Pharmacology Center for Neurosciences (C4N) Vrije Universiteit Brussel Brussels Belgium
| |
Collapse
|
11
|
James J, Mair S, Doll W, Sandefer E, Wurtman D, Maurer A, Deane AM, Harris MS. The effects of ulimorelin, a ghrelin agonist, on liquid gastric emptying and colonic transit in humans. Neurogastroenterol Motil 2020; 32:e13784. [PMID: 32017341 DOI: 10.1111/nmo.13784] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Revised: 10/26/2019] [Accepted: 12/02/2019] [Indexed: 02/08/2023]
Abstract
BACKGROUND Ulimorelin, a small molecule ghrelin agonist and prokinetic agent, was effective in animal models of gastroparesis and delayed transit. However, employing once daily administration, it failed in clinical trials of postoperative ileus (POI), a condition in which colonic motility recovers last. The aim of this study was to evaluate drug dosing and regional differences in drug activity between stomach and colon. METHODS Gastric emptying was assessed by scintigraphy in healthy adults at single doses of 600-1200 µg kg-1 and multiple doses of 80-600 µg kg-1 Q8H for 7 days. Colonic motility was assessed by 7-region scintigraphic analysis at a dose of 600 µg kg-1 for 2 days. The primary endpoints were percent change in time to 50% (∆t50 ) liquid gastric emptying on Days 1, 4, and 6 and the geometric mean center of colonic transit at 24 hours (GC24 ). Plasma concentrations of free and total ulimorelin were measured for pharmacokinetic and exposure-response modeling. KEY RESULTS Ulimorelin 150-600 µg kg-1 every 8 hours resulted in statistically significant improvements (∆t50 = 23% to 46% (P < .05)) in gastric emptying from baseline that were sustained through Day 6. However, no effects on GC24 were observed. Pharmacokinetic analyses suggested that the free concentrations of ulimorelin achieved in POI trials and dosing frequency may have been inadequate. CONCLUSIONS AND INFERENCES Ulimorelin is a potent gastric prokinetic but lacks evidence of activity in the human colon, pointing to the stomach as the predominant site of action of ghrelin in humans; ClinicalTrials.gov NCT02993055.
Collapse
Affiliation(s)
- Joyce James
- Lyric Pharmaceuticals, Inc., South San Francisco, CA, USA
| | | | | | | | - David Wurtman
- Lyric Pharmaceuticals, Inc., South San Francisco, CA, USA
| | - Alan Maurer
- Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Adam M Deane
- Intensive Care Unit, Royal Melbourne Hospital, University of Melbourne, Melbourne, Vic., Australia
| | - M Scott Harris
- Lyric Pharmaceuticals, Inc., South San Francisco, CA, USA
| |
Collapse
|
12
|
Shankar K, Gupta D, Mani BK, Findley BG, Lord CC, Osborne-Lawrence S, Metzger NP, Pietra C, Liu C, Berglund ED, Zigman JM. Acyl-ghrelin Is Permissive for the Normal Counterregulatory Response to Insulin-Induced Hypoglycemia. Diabetes 2020; 69:228-237. [PMID: 31685528 PMCID: PMC6971486 DOI: 10.2337/db19-0438] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 11/01/2019] [Indexed: 02/06/2023]
Abstract
Insulin-induced hypoglycemia leads to far-ranging negative consequences in patients with diabetes. Components of the counterregulatory response (CRR) system that help minimize and reverse hypoglycemia and coordination between those components are well studied but not yet fully characterized. Here, we tested the hypothesis that acyl-ghrelin, a hormone that defends against hypoglycemia in a preclinical starvation model, is permissive for the normal CRR to insulin-induced hypoglycemia. Ghrelin knockout (KO) mice and wild-type (WT) littermates underwent an insulin bolus-induced hypoglycemia test and a low-dose hyperinsulinemic-hypoglycemic clamp procedure. Clamps also were performed in ghrelin-KO mice and C57BL/6N mice administered the growth hormone secretagogue receptor agonist HM01 or vehicle. Results show that hypoglycemia, as induced by an insulin bolus, was more pronounced and prolonged in ghrelin-KO mice, supporting previous studies suggesting increased insulin sensitivity upon ghrelin deletion. Furthermore, during hyperinsulinemic-hypoglycemic clamps, ghrelin-KO mice required a 10-fold higher glucose infusion rate (GIR) and exhibited less robust corticosterone and growth hormone responses. Conversely, HM01 administration, which reduced the GIR required by ghrelin-KO mice during the clamps, increased plasma corticosterone and growth hormone. Thus, our data suggest that endogenously produced acyl-ghrelin not only influences insulin sensitivity but also is permissive for the normal CRR to insulin-induced hypoglycemia.
Collapse
Affiliation(s)
- Kripa Shankar
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX
| | - Deepali Gupta
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX
| | - Bharath K Mani
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX
| | - Brianna G Findley
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX
| | - Caleb C Lord
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX
| | - Sherri Osborne-Lawrence
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX
| | - Nathan P Metzger
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX
| | | | - Chen Liu
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX
| | - Eric D Berglund
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX
| | - Jeffrey M Zigman
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX
- Division of Endocrinology, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX
- Department of Psychiatry, UT Southwestern Medical Center, Dallas, TX
| |
Collapse
|
13
|
Shankar K, Gupta D, Mani BK, Findley BG, Osborne-Lawrence S, Metzger NP, Liu C, Berglund ED, Zigman JM. Ghrelin Protects Against Insulin-Induced Hypoglycemia in a Mouse Model of Type 1 Diabetes Mellitus. Front Endocrinol (Lausanne) 2020; 11:606. [PMID: 33042003 PMCID: PMC7518392 DOI: 10.3389/fendo.2020.00606] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 07/27/2020] [Indexed: 01/28/2023] Open
Abstract
Insulin-induced hypoglycemia is a major limiting factor in maintaining optimal blood glucose in patients with type 1 diabetes and advanced type 2 diabetes. Luckily, a counterregulatory response (1) system exists to help minimize and reverse hypoglycemia, although more studies are needed to better characterize its components. Recently, we showed that the hormone ghrelin is permissive for the normal CRR to insulin-induced hypoglycemia when assessed in mice without diabetes. Here, we tested the hypothesis that ghrelin also is protective against insulin-induced hypoglycemia in the streptozotocin (2) mouse model of type 1 diabetes. STZ-treated ghrelin-knockout (KO) (3) mice as well as STZ-treated wild-type (WT) littermates were subjected to a low-dose hyperinsulinemic-hypoglycemic clamp procedure. The STZ-treated ghrelin-KO mice required a much higher glucose infusion rate than the STZ-treated WT mice. Also, the STZ-treated ghrelin-KO mice exhibited attenuated plasma epinephrine and norepinephrine responses to the insulin-induced hypoglycemia. Taken together, our data suggest that without ghrelin, STZ-treated mice modeling type 1 diabetes are unable to mount the usual CRR to insulin-induced hypoglycemia.
Collapse
Affiliation(s)
- Kripa Shankar
- Department of Internal Medicine, Center for Hypothalamic Research, UT Southwestern Medical Center, Dallas, TX, United States
| | - Deepali Gupta
- Department of Internal Medicine, Center for Hypothalamic Research, UT Southwestern Medical Center, Dallas, TX, United States
| | - Bharath K. Mani
- Department of Internal Medicine, Center for Hypothalamic Research, UT Southwestern Medical Center, Dallas, TX, United States
| | - Brianna G. Findley
- Department of Internal Medicine, Center for Hypothalamic Research, UT Southwestern Medical Center, Dallas, TX, United States
| | - Sherri Osborne-Lawrence
- Department of Internal Medicine, Center for Hypothalamic Research, UT Southwestern Medical Center, Dallas, TX, United States
| | - Nathan P. Metzger
- Department of Internal Medicine, Center for Hypothalamic Research, UT Southwestern Medical Center, Dallas, TX, United States
| | - Chen Liu
- Department of Internal Medicine, Center for Hypothalamic Research, UT Southwestern Medical Center, Dallas, TX, United States
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX, United States
| | - Eric D. Berglund
- Department of Internal Medicine, Center for Hypothalamic Research, UT Southwestern Medical Center, Dallas, TX, United States
| | - Jeffrey M. Zigman
- Department of Internal Medicine, Center for Hypothalamic Research, UT Southwestern Medical Center, Dallas, TX, United States
- Division of Endocrinology, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, United States
- Department of Psychiatry, UT Southwestern Medical Center, Dallas, TX, United States
- *Correspondence: Jeffrey M. Zigman
| |
Collapse
|
14
|
Pierre A, Regin Y, Van Schuerbeek A, Fritz EM, Muylle K, Beckers T, Smolders IJ, Singewald N, De Bundel D. Effects of disrupted ghrelin receptor function on fear processing, anxiety and saccharin preference in mice. Psychoneuroendocrinology 2019; 110:104430. [PMID: 31542636 DOI: 10.1016/j.psyneuen.2019.104430] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 08/26/2019] [Accepted: 09/03/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Obesity is a risk factor for stress-related mental disorders such as post-traumatic stress disorder. The underlying mechanism through which obesity affects mental health remains poorly understood but dysregulation of the ghrelin system may be involved. Stress increases plasma ghrelin levels, which stimulates food intake as a potential stress-coping mechanism. However, diet-induced obesity induces ghrelin resistance which in turn may have deleterious effects on stress-coping. In our study, we explored whether disruption of ghrelin receptor function though high-fat diet or genetic ablation affects fear processing, anxiety-like behavior and saccharin preference in mice. METHODS Adult male C57BL6/J mice were placed on a standard diet or high-fat diet for a total period of 8 weeks. We first established that high-fat diet exposure for 4 weeks elicits ghrelin resistance, evidenced by a blunted hyperphagic response following administration of a ghrelin receptor agonist. We then carried out an experiment in which we subjected mice to auditory fear conditioning after 4 weeks of diet exposure and evaluated effects on fear extinction, anxiety-like behavior and saccharin preference. To explore whether fear conditioning as such may influence the effect of diet exposure, we also subjected mice to auditory fear conditioning prior to diet onset and 4 weeks later we investigated auditory fear extinction, anxiety-like behavior and saccharin preference. In a final experiment, we further assessed lack of ghrelin receptor function by investigating auditory fear processing, anxiety-like behavior and saccharin preference in ghrelin receptor knockout mice and their wild-type littermates. RESULTS High-fat diet exposure had no significant effect on auditory fear conditioning and its subsequent extinction or on anxiety-like behavior but significantly lowered saccharin preference. Similarly, ghrelin receptor knockout mice did not differ significantly from their wild-type littermates for auditory fear processing or anxiety-like behavior but showed significantly lower saccharin preference compared to wild-type littermates. CONCLUSION Taken together, our data suggest that disruption of ghrelin receptor function per se does not affect fear or anxiety-like behavior but may decrease saccharin preference in mice.
Collapse
Affiliation(s)
- A Pierre
- Department of Pharmaceutical Sciences, Research Group Experimental Pharmacology, Center for Neurosciences (C4N), Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium
| | - Y Regin
- Department of Pharmaceutical Sciences, Research Group Experimental Pharmacology, Center for Neurosciences (C4N), Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium
| | - A Van Schuerbeek
- Department of Pharmaceutical Sciences, Research Group Experimental Pharmacology, Center for Neurosciences (C4N), Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium
| | - E M Fritz
- Department of Pharmacology and Toxicology, Institute of Pharmacy and CMBI, University of Innsbruck, Innrain 80/82, Innsbruck, Austria
| | - K Muylle
- Department of Pharmaceutical Sciences, Research Group Experimental Pharmacology, Center for Neurosciences (C4N), Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium
| | - T Beckers
- Departement of Psychology and Leuven Brain Institute, KU Leuven, Tiensestraat 102 box 3712, 3000, Leuven, Belgium
| | - I J Smolders
- Department of Pharmaceutical Sciences, Research Group Experimental Pharmacology, Center for Neurosciences (C4N), Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium
| | - N Singewald
- Department of Pharmacology and Toxicology, Institute of Pharmacy and CMBI, University of Innsbruck, Innrain 80/82, Innsbruck, Austria
| | - D De Bundel
- Department of Pharmaceutical Sciences, Research Group Experimental Pharmacology, Center for Neurosciences (C4N), Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium.
| |
Collapse
|
15
|
Farokhnia M, Faulkner ML, Piacentino D, Lee MR, Leggio L. Ghrelin: From a gut hormone to a potential therapeutic target for alcohol use disorder. Physiol Behav 2019; 204:49-57. [DOI: 10.1016/j.physbeh.2019.02.008] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 01/24/2019] [Accepted: 02/06/2019] [Indexed: 12/22/2022]
|
16
|
Abstract
This review evaluates published studies regarding alpha-melanocyte stimulating hormone (α-MSH) in ghrelin-elicited feeding and gut motility. We have sought to integrate all available evidences to provide a complete review on the properties of melanocortin receptors (MCR) and the potential clinical treatment of α-MSH after ghrelin-elicited feeding and gut motility. The available studies were grouped into four categories: food intake, gastric emptying, small intestinal transit, and colonic transit. As we describe, the literature provides evidence of the ability of ghrelin to increase food intake, gastric emptying, small intestinal transit, and colonic transit. α-MSH, which displays high affinity for the MC3 and MC4 receptors, can competitively activate MCRs with agouti-related protein stimulated by ghrelin, and partly attenuates the effect of acyl ghrelin on food intake. Central ghrelin-induced acceleration of gastric emptying is not mediated by MCRs, but the acceleration of the small intestinal transit is at least partly mediated via MCRs in the brain. Similar to fecal pellets and total fecal weight, distal colonic motility and secretion are partly mediated by MCRs in the brain. The interplay between acyl ghrelin and MCRs may provide a new therapeutic avenue to ameliorate anorexia and constipation.
Collapse
Affiliation(s)
- Hsien-Hao Huang
- Department of Emergency Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Institute of Emergency and Critical Medicine, National Yang-Ming University School of Medicine, Taipei, Taiwan, ROC
| | - Chih-Yen Chen
- Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Faculty of Medicine, National Yang-Ming University School of Medicine, Taipei, Taiwan, ROC
- Taiwan Association for the Study of Small Intestinal Diseases, Guishan, Taiwan, ROC
- Chinese Taipei Society for the Study of Obesity, Taipei, Taiwan, ROC
| |
Collapse
|
17
|
aP2-Cre Mediated Ablation of GHS-R Attenuates Adiposity and Improves Insulin Sensitivity during Aging. Int J Mol Sci 2018; 19:ijms19103002. [PMID: 30275401 PMCID: PMC6213105 DOI: 10.3390/ijms19103002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 09/28/2018] [Accepted: 09/29/2018] [Indexed: 01/08/2023] Open
Abstract
Ghrelin via its receptor, the growth hormone secretagogue receptor (GHS-R), increases food intake and adiposity. The tissue-specific functions of GHS-R in peripheral tissues are mostly unknown. We previously reported that while GHS-R expression is very low in white and brown fat of young mice, expression increases during aging. To investigate whether GHS-R has cell-autonomous effects in adipose tissues, we generated aP2-Cre-mediated GHS-R knockdown mice (aP2-Cre/Ghsrf/f). We studied young (5–6 months) and old (15–17 months) aP2-Cre/Ghsrf/f mice and their age-matched controls. Interestingly, young aP2-Cre/Ghsrf/f mice had normal body weight but reduced fat; old mice showed pronounced reductions of both body weight and body fat. Calorimetry analysis revealed that aP2-Cre/Ghsrf/f mice had normal food intake and locomotor activity at both young and old age; but intriguingly, while energy expenditure was normal at young age, it was significantly increased at old age. Both young and old aP2-Cre/Ghsrf/f mice exhibited improved insulin sensitivity and glucose tolerance. Importantly, old aP2-Cre/Ghsrf/f mice maintained higher core body temperature at 4 °C, and showed higher expression of the thermogenic uncoupling protein 1 (UCP1) gene. The ex vivo studies further demonstrated that GHS-R deficient white adipocytes from old mice exhibit increased glucose uptake and lipolysis, promoting lipid mobilization. Despite the fact that the in vivo phenotypes of aP2-Cre/Ghsrf/f mice may not be exclusively determined by GHS-R knockdown in adipose tissues, our data support that GHS-R has cell-autonomous effects in adipocytes. The anabolic effect of GHS-R in adipocytes is more pronounced in aging, which likely contributes to age-associated obesity and insulin resistance.
Collapse
|
18
|
Vancleef L, Thijs T, Baert F, Ceulemans LJ, Canovai E, Wang Q, Steensels S, Segers A, Farré R, Pirenne J, Lannoo M, Tack J, Depoortere I. Obesity Impairs Oligopeptide/Amino Acid-Induced Ghrelin Release and Smooth Muscle Contractions in the Human Proximal Stomach. Mol Nutr Food Res 2018; 62. [DOI: 10.1002/mnfr.201700804] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 11/29/2017] [Indexed: 01/11/2023]
Affiliation(s)
- Laurien Vancleef
- Translational Research Center for Gastrointestinal Disorders; Department of Clinical & Experimental Medicine; University of Leuven; Leuven Belgium
| | - Theo Thijs
- Translational Research Center for Gastrointestinal Disorders; Department of Clinical & Experimental Medicine; University of Leuven; Leuven Belgium
| | - Florence Baert
- Translational Research Center for Gastrointestinal Disorders; Department of Clinical & Experimental Medicine; University of Leuven; Leuven Belgium
| | - Laurens J. Ceulemans
- Translational Research Center for Gastrointestinal Disorders; Department of Clinical & Experimental Medicine; University of Leuven; Leuven Belgium
| | - Emilio Canovai
- Abdominal Transplant Surgery; University Hospital Gasthuisberg; Leuven Belgium
| | - Qiaoling Wang
- Translational Research Center for Gastrointestinal Disorders; Department of Clinical & Experimental Medicine; University of Leuven; Leuven Belgium
| | - Sandra Steensels
- Translational Research Center for Gastrointestinal Disorders; Department of Clinical & Experimental Medicine; University of Leuven; Leuven Belgium
| | - Anneleen Segers
- Translational Research Center for Gastrointestinal Disorders; Department of Clinical & Experimental Medicine; University of Leuven; Leuven Belgium
| | - Ricard Farré
- Translational Research Center for Gastrointestinal Disorders; Department of Clinical & Experimental Medicine; University of Leuven; Leuven Belgium
| | - Jacques Pirenne
- Translational Research Center for Gastrointestinal Disorders; Department of Clinical & Experimental Medicine; University of Leuven; Leuven Belgium
| | - Matthias Lannoo
- Abdominal Surgery; University Hospital Gasthuisberg; Belgium
| | - Jan Tack
- Translational Research Center for Gastrointestinal Disorders; Department of Clinical & Experimental Medicine; University of Leuven; Leuven Belgium
| | - Inge Depoortere
- Translational Research Center for Gastrointestinal Disorders; Department of Clinical & Experimental Medicine; University of Leuven; Leuven Belgium
| |
Collapse
|
19
|
Mani BK, Zigman JM. Ghrelin as a Survival Hormone. Trends Endocrinol Metab 2017; 28:843-854. [PMID: 29097101 PMCID: PMC5777178 DOI: 10.1016/j.tem.2017.10.001] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 10/03/2017] [Accepted: 10/05/2017] [Indexed: 12/22/2022]
Abstract
Ghrelin administration induces food intake and body weight gain. Based on these actions, the ghrelin system was initially proposed as an antiobesity target. Subsequent studies using genetic mouse models have raised doubts about the role of the endogenous ghrelin system in mediating body weight homeostasis or obesity. However, this is not to say that the endogenous ghrelin system is not important metabolically or otherwise. Here we review an emerging concept in which the endogenous ghrelin system serves an essential function during extreme nutritional and psychological challenges to defend blood glucose, protect body weight, avoid exaggerated depression, and ultimately allow survival.
Collapse
Affiliation(s)
- Bharath K Mani
- Divisions of Hypothalamic Research and Endocrinology, Department of Internal Medicine, and Department of Psychiatry, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390-9077, USA
| | - Jeffrey M Zigman
- Divisions of Hypothalamic Research and Endocrinology, Department of Internal Medicine, and Department of Psychiatry, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390-9077, USA.
| |
Collapse
|
20
|
Mani BK, Uchida A, Lee Y, Osborne-Lawrence S, Charron MJ, Unger RH, Berglund ED, Zigman JM. Hypoglycemic Effect of Combined Ghrelin and Glucagon Receptor Blockade. Diabetes 2017; 66:1847-1857. [PMID: 28487437 PMCID: PMC5482080 DOI: 10.2337/db16-1303] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 04/19/2017] [Indexed: 12/18/2022]
Abstract
Glucagon receptor (GcgR) blockade has been proposed as an alternative to insulin monotherapy for treating type 1 diabetes since deletion or inhibition of GcgRs corrects hyperglycemia in models of diabetes. The factors regulating glycemia in a setting devoid of insulin and glucagon function remain unclear but may include the hormone ghrelin. Not only is ghrelin release controlled by glucose but also ghrelin has many actions that can raise or reduce falls in blood glucose level. Here, we tested the hypothesis that ghrelin rises to prevent hypoglycemia in the absence of glucagon function. Both GcgR knockout (Gcgr-/-) mice and db/db mice that were administered GcgR monoclonal antibody displayed lower blood glucose levels accompanied by elevated plasma ghrelin levels. Although treatment with the pancreatic β-cell toxin streptozotocin induced hyperglycemia and raised plasma ghrelin levels in wild-type mice, hyperglycemia was averted in similarly treated Gcgr-/- mice and the plasma ghrelin level was further increased. Notably, administration of a ghrelin receptor antagonist further reduced blood glucose levels into the markedly hypoglycemic range in overnight-fasted, streptozotocin-treated Gcgr-/- mice. A lowered blood glucose level also was observed in overnight-fasted, streptozotocin-treated ghrelin receptor-null mice that were administered GcgR monoclonal antibody. These data suggest that when glucagon activity is blocked in the setting of type 1 diabetes, the plasma ghrelin level rises, preventing hypoglycemia.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/pharmacology
- Atenolol/pharmacology
- Blood Glucose/drug effects
- Blood Glucose/metabolism
- Cells, Cultured
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/metabolism
- Gastric Mucosa/metabolism
- Ghrelin/metabolism
- Immunohistochemistry
- Insulin/metabolism
- Mice
- Mice, Knockout
- Oligopeptides/pharmacology
- Real-Time Polymerase Chain Reaction
- Receptors, Ghrelin/antagonists & inhibitors
- Receptors, Glucagon/antagonists & inhibitors
- Receptors, Glucagon/genetics
- Receptors, Leptin/genetics
- Sympatholytics/pharmacology
Collapse
Affiliation(s)
- Bharath K Mani
- Divisions of Hypothalamic Research and Endocrinology, Department of Internal Medicine and Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX
| | - Aki Uchida
- Advanced Imaging Center and Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Young Lee
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX
| | - Sherri Osborne-Lawrence
- Divisions of Hypothalamic Research and Endocrinology, Department of Internal Medicine and Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX
| | - Maureen J Charron
- Departments of Biochemistry, Obstetrics and Gynecology and Woman's Health, and Medicine, Albert Einstein College of Medicine, Bronx, NY
| | - Roger H Unger
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX
| | - Eric D Berglund
- Advanced Imaging Center and Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Jeffrey M Zigman
- Divisions of Hypothalamic Research and Endocrinology, Department of Internal Medicine and Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX
| |
Collapse
|
21
|
Coppens J, Bentea E, Bayliss JA, Demuyser T, Walrave L, Albertini G, Van Liefferinge J, Deneyer L, Aourz N, Van Eeckhaut A, Portelli J, Andrews ZB, Massie A, De Bundel D, Smolders I. Caloric Restriction Protects against Lactacystin-Induced Degeneration of Dopamine Neurons Independent of the Ghrelin Receptor. Int J Mol Sci 2017; 18:ijms18030558. [PMID: 28273852 PMCID: PMC5372574 DOI: 10.3390/ijms18030558] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 02/14/2017] [Accepted: 02/20/2017] [Indexed: 02/06/2023] Open
Abstract
Parkinson’s disease (PD) is a neurodegenerative disorder, characterized by a loss of dopamine (DA) neurons in the substantia nigra pars compacta (SNc). Caloric restriction (CR) has been shown to exert ghrelin-dependent neuroprotective effects in the 1-methyl-4-phenyl-1,2,3,6-tetrathydropyridine (MPTP)-based animal model for PD. We here investigated whether CR is neuroprotective in the lactacystin (LAC) mouse model for PD, in which proteasome disruption leads to the destruction of the DA neurons of the SNc, and whether this effect is mediated via the ghrelin receptor. Adult male ghrelin receptor wildtype (WT) and knockout (KO) mice were maintained on an ad libitum (AL) diet or on a 30% CR regimen. After 3 weeks, LAC was injected unilaterally into the SNc, and the degree of DA neuron degeneration was evaluated 1 week later. In AL mice, LAC injection significanty reduced the number of DA neurons and striatal DA concentrations. CR protected against DA neuron degeneration following LAC injection. However, no differences were observed between ghrelin receptor WT and KO mice. These results indicate that CR can protect the nigral DA neurons from toxicity related to proteasome disruption; however, the ghrelin receptor is not involved in this effect.
Collapse
Affiliation(s)
- Jessica Coppens
- Research Group Experimental Pharmacology (EFAR/FASC), Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussel, Belgium.
| | - Eduard Bentea
- Research Group Pharmaceutical Biotechnology and Molecular Biology (MICH), Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussel, Belgium.
| | - Jacqueline A Bayliss
- Department of Physiology, School of Biomedical and Psychological Sciences, Monash University, Clayton, Melbourne 3800, Australia.
| | - Thomas Demuyser
- Research Group Experimental Pharmacology (EFAR/FASC), Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussel, Belgium.
| | - Laura Walrave
- Research Group Experimental Pharmacology (EFAR/FASC), Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussel, Belgium.
| | - Giulia Albertini
- Research Group Experimental Pharmacology (EFAR/FASC), Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussel, Belgium.
| | - Joeri Van Liefferinge
- Research Group Experimental Pharmacology (EFAR/FASC), Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussel, Belgium.
| | - Lauren Deneyer
- Research Group Pharmaceutical Biotechnology and Molecular Biology (MICH), Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussel, Belgium.
| | - Najat Aourz
- Research Group Experimental Pharmacology (EFAR/FASC), Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussel, Belgium.
| | - Ann Van Eeckhaut
- Research Group Experimental Pharmacology (EFAR/FASC), Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussel, Belgium.
| | - Jeanelle Portelli
- Research Group Experimental Pharmacology (EFAR/FASC), Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussel, Belgium.
| | - Zane B Andrews
- Department of Physiology, School of Biomedical and Psychological Sciences, Monash University, Clayton, Melbourne 3800, Australia.
| | - Ann Massie
- Research Group Pharmaceutical Biotechnology and Molecular Biology (MICH), Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussel, Belgium.
| | - Dimitri De Bundel
- Research Group Experimental Pharmacology (EFAR/FASC), Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussel, Belgium.
| | - Ilse Smolders
- Research Group Experimental Pharmacology (EFAR/FASC), Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussel, Belgium.
| |
Collapse
|
22
|
Steensels S, Vancleef L, Depoortere I. The Sweetener-Sensing Mechanisms of the Ghrelin Cell. Nutrients 2016; 8:E795. [PMID: 27941594 PMCID: PMC5188450 DOI: 10.3390/nu8120795] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 11/21/2016] [Accepted: 11/28/2016] [Indexed: 12/12/2022] Open
Abstract
Carbohydrate administration decreases plasma levels of the 'hunger hormone' ghrelin. The ghrelin cell is co-localized with the sweet taste receptor subunit, TAS1R3, and the gustatory G-protein, gustducin, both involved in the sensing of sweeteners by entero-endocrine cells. This study investigated the role of gustducin-mediated sweet taste receptor signaling on ghrelin secretion in a gastric ghrelinoma cell line, tissue segments and mice. The monosaccharide d-glucose and low-intensity sweetener oligofructose (OFS) decreased (p < 0.001) ghrelin secretion while the high-intensity sweetener sucralose increased (p < 0.001) ghrelin secretion in vitro. These effects were not mediated via the sweet taste receptor or glucose transporters (the sodium-dependent glucose cotransporter SGLT-1 and GLUT2). The effect of these compounds was mimicked ex vivo in gastric and jejunal segments from both wild type (WT) and α-gustducin knockout (α-gust-/-) mice. In vivo, the sensing of d-glucose was polarized since intragastric but not intravenous administration of d-glucose decreased (p < 0.05) ghrelin levels in an α-gustducin independent manner which involved inhibition of duodenal ghrelin release. In contrast, neither OFS nor sucralose affected ghrelin secretion in vivo. In conclusion, α-gustducin-mediated sweet taste receptor signaling does not play a functional role in the sensing of carbohydrates, or low- or high-intensity sweeteners by the ghrelin cell.
Collapse
Affiliation(s)
- Sandra Steensels
- Gut Peptide Lab, Translational Research Center for Gastrointestinal Disorders (TARGID), University of Leuven-KU Leuven, 3000 Leuven, Belgium.
| | - Laurien Vancleef
- Gut Peptide Lab, Translational Research Center for Gastrointestinal Disorders (TARGID), University of Leuven-KU Leuven, 3000 Leuven, Belgium.
| | - Inge Depoortere
- Gut Peptide Lab, Translational Research Center for Gastrointestinal Disorders (TARGID), University of Leuven-KU Leuven, 3000 Leuven, Belgium.
| |
Collapse
|
23
|
Lee JH, Lin L, Xu P, Saito K, Wei Q, Meadows AG, Bongmba OYN, Pradhan G, Zheng H, Xu Y, Sun Y. Neuronal Deletion of Ghrelin Receptor Almost Completely Prevents Diet-Induced Obesity. Diabetes 2016; 65:2169-78. [PMID: 27207529 PMCID: PMC4955988 DOI: 10.2337/db15-1587] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 04/28/2016] [Indexed: 12/17/2022]
Abstract
Ghrelin signaling has major effects on energy and glucose homeostasis, but it is unknown whether ghrelin's functions are centrally and/or peripherally mediated. The ghrelin receptor, growth hormone secretagogue receptor (GHS-R), is highly expressed in the brain and detectable in some peripheral tissues. To understand the roles of neuronal GHS-R, we generated a mouse line where Ghsr gene is deleted in all neurons using synapsin 1 (Syn1)-Cre driver. Our data showed that neuronal Ghsr deletion abolishes ghrelin-induced spontaneous food intake but has no effect on total energy intake. Remarkably, neuronal Ghsr deletion almost completely prevented diet-induced obesity (DIO) and significantly improved insulin sensitivity. The neuronal Ghsr-deleted mice also showed improved metabolic flexibility, indicative of better adaption to different fuels. In addition, gene expression analysis suggested that hypothalamus and/or midbrain might be the sites that mediate the effects of GHS-R in thermogenesis and physical activity, respectively. Collectively, our results indicate that neuronal GHS-R is a crucial regulator of energy metabolism and a key mediator of DIO. Neuronal Ghsr deletion protects against DIO by regulating energy expenditure, not by energy intake. These novel findings suggest that suppressing central ghrelin signaling may serve as a unique antiobesity strategy.
Collapse
Affiliation(s)
- Jong Han Lee
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX
| | - Ligen Lin
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Pingwen Xu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX
| | - Kenji Saito
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX
| | - Qiong Wei
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX Division of Endocrinology, Zhongda Hospital, Southeast University, Nanjing, Jiangsu, China
| | - Adelina G Meadows
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX
| | - Odelia Y N Bongmba
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX
| | - Geetali Pradhan
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX
| | - Hui Zheng
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX
| | - Yong Xu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX
| | - Yuxiang Sun
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX Huffington Center on Aging, Baylor College of Medicine, Houston, TX Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX Department of Nutrition and Food Science, Texas A&M University, College Station, TX
| |
Collapse
|
24
|
Coppens J, Aourz N, Walrave L, Fehrentz JA, Martinez J, De Bundel D, Portelli J, Smolders I. Anticonvulsant effect of a ghrelin receptor agonist in 6Hz corneally kindled mice. Epilepsia 2016; 57:e195-9. [DOI: 10.1111/epi.13463] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/13/2016] [Indexed: 11/28/2022]
Affiliation(s)
- Jessica Coppens
- Center for Neurosciences; VUB Free University Brussels; Brussels Belgium
| | - Najat Aourz
- Center for Neurosciences; VUB Free University Brussels; Brussels Belgium
| | - Laura Walrave
- Center for Neurosciences; VUB Free University Brussels; Brussels Belgium
| | - Jean-Alain Fehrentz
- IBMM Institute of Biomolecules Max Mousseron; University of Montpellier; Montpellier France
| | - Jean Martinez
- IBMM Institute of Biomolecules Max Mousseron; University of Montpellier; Montpellier France
| | - Dimitri De Bundel
- Center for Neurosciences; VUB Free University Brussels; Brussels Belgium
| | - Jeanelle Portelli
- Center for Neurosciences; VUB Free University Brussels; Brussels Belgium
- Department of Neurology; UZ Gent; Ghent Belgium
| | - Ilse Smolders
- Center for Neurosciences; VUB Free University Brussels; Brussels Belgium
| |
Collapse
|
25
|
The Gustatory Signaling Pathway and Bitter Taste Receptors Affect the Development of Obesity and Adipocyte Metabolism in Mice. PLoS One 2015; 10:e0145538. [PMID: 26692363 PMCID: PMC4686985 DOI: 10.1371/journal.pone.0145538] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 12/04/2015] [Indexed: 11/20/2022] Open
Abstract
Intestinal chemosensory signaling pathways involving the gustatory G-protein, gustducin, and bitter taste receptors (TAS2R) have been implicated in gut hormone release. Alterations in gut hormone profiles may contribute to the success of bariatric surgery. This study investigated the involvement of the gustatory signaling pathway in the development of diet-induced obesity and the therapeutic potential of targeting TAS2Rs to induce body weight loss. α-gustducin-deficient (α-gust-/-) mice became less obese than wild type (WT) mice when fed a high-fat diet (HFD). White adipose tissue (WAT) mass was lower in α-gust-/- mice due to increased heat production as a result of increases in brown adipose tissue (BAT) thermogenic activity, involving increased protein expression of uncoupling protein 1. Intra-gastric treatment of obese WT and α-gust-/- mice with the bitter agonists denatonium benzoate (DB) or quinine (Q) during 4 weeks resulted in an α-gustducin-dependent decrease in body weight gain associated with a decrease in food intake (DB), but not involving major changes in gut peptide release. Both WAT and 3T3-F442A pre-adipocytes express TAS2Rs. Treatment of pre-adipocytes with DB or Q decreased differentiation into mature adipocytes. In conclusion, interfering with the gustatory signaling pathway protects against the development of HFD-induced obesity presumably through promoting BAT activity. Intra-gastric bitter treatment inhibits weight gain, possibly by directly affecting adipocyte metabolism.
Collapse
|
26
|
Laermans J, Vancleef L, Tack J, Depoortere I. Role of the clock gene Bmal1 and the gastric ghrelin-secreting cell in the circadian regulation of the ghrelin-GOAT system. Sci Rep 2015; 5:16748. [PMID: 26576661 PMCID: PMC4649743 DOI: 10.1038/srep16748] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 10/19/2015] [Indexed: 11/09/2022] Open
Abstract
As adequate food intake is crucial to survival, organisms have evolved endogenous circadian clocks to generate optimal temporal patterns of food-related behavior and physiology. The gastric ghrelin-secreting cell is thought to be part of this network of peripheral food-entrainable oscillators (FEOs), regulating the circadian release of this orexigenic peptide. This study aimed to determine the role of the core clock gene Bmal1 and the gastric ghrelin-secreting cell as an FEO in the circadian rhythmicity of ghrelin expression and secretion in vivo and in vitro. Bmal1-deficient mice not only lacked circadian rhythmicity in plasma ghrelin levels and food intake, but also showed decreased gastric mRNA expression of ghrelin and ghrelin O-acyltransferase (GOAT), the ghrelin activating enzyme. Furthermore, in the absence of the hypothalamic master clock, food-related stimuli entrained the molecular clock of gastric ghrelinoma cells to regulate the rhythmic release of ghrelin. Divergent responses in octanoyl and total ghrelin release towards different food cues were observed, suggesting that the FEO also regulates the circadian rhythmicity of GOAT. Collectively, these findings indicate that circadian rhythmicity of ghrelin signaling requires Bmal1 and is driven by a food-responsive clock in the gastric ghrelin-secreting cell that not only regulates ghrelin, but also GOAT activity.
Collapse
Affiliation(s)
- J Laermans
- Gut Peptide Research Lab, Translational Research Center for Gastrointestinal Disorders (TARGID), University of Leuven, Leuven, Belgium
| | - L Vancleef
- Gut Peptide Research Lab, Translational Research Center for Gastrointestinal Disorders (TARGID), University of Leuven, Leuven, Belgium
| | - J Tack
- Gut Peptide Research Lab, Translational Research Center for Gastrointestinal Disorders (TARGID), University of Leuven, Leuven, Belgium
| | - I Depoortere
- Gut Peptide Research Lab, Translational Research Center for Gastrointestinal Disorders (TARGID), University of Leuven, Leuven, Belgium
| |
Collapse
|
27
|
Avau B, Rotondo A, Thijs T, Andrews CN, Janssen P, Tack J, Depoortere I. Targeting extra-oral bitter taste receptors modulates gastrointestinal motility with effects on satiation. Sci Rep 2015; 5:15985. [PMID: 26541810 PMCID: PMC4635351 DOI: 10.1038/srep15985] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2015] [Accepted: 10/06/2015] [Indexed: 01/01/2023] Open
Abstract
Bitter taste receptors (TAS2Rs) are present in extra-oral tissues, including gut endocrine cells. This study explored the presence and mechanism of action of TAS2R agonists on gut smooth muscle in vitro and investigated functional effects of intra-gastric administration of TAS2R agonists on gastric motility and satiation. TAS2Rs and taste signalling elements were expressed in smooth muscle tissue along the mouse gut and in human gastric smooth muscle cells (hGSMC). Bitter tastants induced concentration and region-dependent contractility changes in mouse intestinal muscle strips. Contractions induced by denatonium benzoate (DB) in gastric fundus were mediated via increases in intracellular Ca2+ release and extracellular Ca2+-influx, partially masked by a hyperpolarizing K+-efflux. Intra-gastric administration of DB in mice induced a TAS2R-dependent delay in gastric emptying. In hGSMC, bitter compounds evoked Ca2+-rises and increased ERK-phosphorylation. Healthy volunteers showed an impaired fundic relaxation in response to nutrient infusion and a decreased nutrient volume tolerance and increased satiation during an oral nutrient challenge test after intra-gastric DB administration. These findings suggest a potential role for intestinal TAS2Rs as therapeutic targets to alter gastrointestinal motility and hence to interfere with hunger signalling.
Collapse
Affiliation(s)
- Bert Avau
- Translational Research Center for Gastrointestinal Disorders, University of Leuven, Leuven, Belgium
| | - Alessandra Rotondo
- Translational Research Center for Gastrointestinal Disorders, University of Leuven, Leuven, Belgium
| | - Theo Thijs
- Translational Research Center for Gastrointestinal Disorders, University of Leuven, Leuven, Belgium
| | - Christopher N Andrews
- Translational Research Center for Gastrointestinal Disorders, University of Leuven, Leuven, Belgium
| | - Pieter Janssen
- Translational Research Center for Gastrointestinal Disorders, University of Leuven, Leuven, Belgium
| | - Jan Tack
- Translational Research Center for Gastrointestinal Disorders, University of Leuven, Leuven, Belgium
| | - Inge Depoortere
- Translational Research Center for Gastrointestinal Disorders, University of Leuven, Leuven, Belgium
| |
Collapse
|
28
|
Di Giovangiulio M, Stakenborg N, Bosmans G, Meroni E, Farro G, Gomez-Pinilla PJ, Depoortere I, Boeckxstaens GE, Matteoli G. Ghrelin receptor modulates T helper cells during intestinal inflammation. Neurogastroenterol Motil 2015; 27:1542-52. [PMID: 26227790 DOI: 10.1111/nmo.12640] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 06/23/2015] [Indexed: 02/08/2023]
Abstract
BACKGROUND The orexigenic peptide ghrelin has anti-inflammatory properties in colitis, however, the mechanism of action and the immune cells targeted remain still to be elucidated. Here, we assessed the possible effect of ghrelin on T helper (Th) cells in a T cell transfer model of chronic colitis. METHODS Disease was induced in the recombination activating gene 1 knockout mice (Rag1(-/-) ) by adoptive transfer of naïve Th cells from ghrelin receptor knockout mice (GRLN-R(-/-) ) or littermate wild-type (WT) mice. The course and severity of colitis was assessed by monitoring body weight, diarrhea score, histological analysis, gene expression, and flow cytometry analysis. The possible effects of ghrelin on Th cell proliferation, polarization, and apoptosis was examined in vitro. KEY RESULTS Our data showed that Rag1(-/-) mice injected with GRLN-R(-/-) Th cells displayed increased severity of colitis compared to mice injected with WT Th cells. In addition, Rag1(-/-) mice injected with GRLN-R(-/-) Th cells had significantly higher intestinal inflammation and increased accumulation of Th1 and Th17 cells in the colon. In vitro, ghrelin directly affected proliferation of Th cells and induced apoptosis whereas it did not influence Th cell polarization. CONCLUSION & INFERENCES Our observations suggest that ghrelin modulates Th effector cells in the gut controlling proliferation and inducing apoptosis. Our findings further support the use of ghrelin as a novel therapeutic option to treat intestinal inflammatory diseases.
Collapse
Affiliation(s)
- M Di Giovangiulio
- Department of Clinical and Experimental Medicine, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - N Stakenborg
- Department of Clinical and Experimental Medicine, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - G Bosmans
- Department of Clinical and Experimental Medicine, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - E Meroni
- Department of Clinical and Experimental Medicine, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - G Farro
- Department of Clinical and Experimental Medicine, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - P J Gomez-Pinilla
- Department of Clinical and Experimental Medicine, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - I Depoortere
- Department of Clinical and Experimental Medicine, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - G E Boeckxstaens
- Department of Clinical and Experimental Medicine, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - G Matteoli
- Department of Clinical and Experimental Medicine, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| |
Collapse
|
29
|
Chemosensory signalling pathways involved in sensing of amino acids by the ghrelin cell. Sci Rep 2015; 5:15725. [PMID: 26510380 PMCID: PMC4625164 DOI: 10.1038/srep15725] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 10/01/2015] [Indexed: 01/06/2023] Open
Abstract
Taste receptors on enteroendocrine cells sense nutrients and transmit signals that control gut hormone release. This study aimed to investigate the amino acid (AA) sensing mechanisms of the ghrelin cell in a gastric ghrelinoma cell line, tissue segments and mice. Peptone and specific classes of amino acids stimulate ghrelin secretion in the ghrelinoma cell line. Sensing of L-Phe occurs via the CaSR, monosodium glutamate via the TAS1R1-TAS1R3 while L-Ala and peptone act via 2 different amino acid taste receptors: CaSR &TAS1R1-TAS1R3 and CaSR &GPRC6A, respectively. The stimulatory effect of peptone on ghrelin release was mimicked ex vivo in gastric but not in jejunal tissue segments, where peptone inhibited ghrelin release. The latter effect could not be blocked by receptor antagonists for CCK, GLP-1 or somatostatin. In vivo, plasma ghrelin levels were reduced both upon intragastric (peptone or L-Phe) or intravenous (L-Phe) administration, indicating that AA- sensing is not polarized and is due to inhibition of ghrelin release from the stomach or duodenum respectively. In conclusion, functional AA taste receptors regulate AA-induced ghrelin release in vitro. The effects differ between stomach and jejunum but these local nutrient sensing mechanisms are overruled in vivo by indirect mechanisms inhibiting ghrelin release.
Collapse
|
30
|
Müller TD, Nogueiras R, Andermann ML, Andrews ZB, Anker SD, Argente J, Batterham RL, Benoit SC, Bowers CY, Broglio F, Casanueva FF, D'Alessio D, Depoortere I, Geliebter A, Ghigo E, Cole PA, Cowley M, Cummings DE, Dagher A, Diano S, Dickson SL, Diéguez C, Granata R, Grill HJ, Grove K, Habegger KM, Heppner K, Heiman ML, Holsen L, Holst B, Inui A, Jansson JO, Kirchner H, Korbonits M, Laferrère B, LeRoux CW, Lopez M, Morin S, Nakazato M, Nass R, Perez-Tilve D, Pfluger PT, Schwartz TW, Seeley RJ, Sleeman M, Sun Y, Sussel L, Tong J, Thorner MO, van der Lely AJ, van der Ploeg LHT, Zigman JM, Kojima M, Kangawa K, Smith RG, Horvath T, Tschöp MH. Ghrelin. Mol Metab 2015; 4:437-60. [PMID: 26042199 PMCID: PMC4443295 DOI: 10.1016/j.molmet.2015.03.005] [Citation(s) in RCA: 760] [Impact Index Per Article: 76.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Revised: 03/11/2015] [Accepted: 03/11/2015] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The gastrointestinal peptide hormone ghrelin was discovered in 1999 as the endogenous ligand of the growth hormone secretagogue receptor. Increasing evidence supports more complicated and nuanced roles for the hormone, which go beyond the regulation of systemic energy metabolism. SCOPE OF REVIEW In this review, we discuss the diverse biological functions of ghrelin, the regulation of its secretion, and address questions that still remain 15 years after its discovery. MAJOR CONCLUSIONS In recent years, ghrelin has been found to have a plethora of central and peripheral actions in distinct areas including learning and memory, gut motility and gastric acid secretion, sleep/wake rhythm, reward seeking behavior, taste sensation and glucose metabolism.
Collapse
Affiliation(s)
- T D Müller
- Institute for Diabetes and Obesity, Helmholtz Zentrum München, München, Germany
| | - R Nogueiras
- Department of Physiology, Centro de Investigación en Medicina Molecular y Enfermedades Crónicas, University of Santiago de Compostela (CIMUS)-Instituto de Investigación Sanitaria (IDIS)-CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, Spain
| | - M L Andermann
- Division of Endocrinology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Z B Andrews
- Department of Physiology, Faculty of Medicine, Monash University, Melbourne, Victoria, Australia
| | - S D Anker
- Applied Cachexia Research, Department of Cardiology, Charité Universitätsmedizin Berlin, Germany
| | - J Argente
- Department of Pediatrics and Pediatric Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain ; Department of Pediatrics, Universidad Autónoma de Madrid and CIBER Fisiopatología de la obesidad y nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - R L Batterham
- Centre for Obesity Research, University College London, London, United Kingdom
| | - S C Benoit
- Metabolic Disease Institute, Division of Endocrinology, Department of Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - C Y Bowers
- Tulane University Health Sciences Center, Endocrinology and Metabolism Section, Peptide Research Section, New Orleans, LA, USA
| | - F Broglio
- Division of Endocrinology, Diabetes and Metabolism, Dept. of Medical Sciences, University of Torino, Torino, Italy
| | - F F Casanueva
- Department of Medicine, Santiago de Compostela University, Complejo Hospitalario Universitario de Santiago (CHUS), CIBER de Fisiopatologia Obesidad y Nutricion (CB06/03), Instituto Salud Carlos III, Santiago de Compostela, Spain
| | - D D'Alessio
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA
| | - I Depoortere
- Translational Research Center for Gastrointestinal Disorders, University of Leuven, Leuven, Belgium
| | - A Geliebter
- New York Obesity Nutrition Research Center, Department of Medicine, St Luke's-Roosevelt Hospital Center, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - E Ghigo
- Department of Pharmacology & Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - P A Cole
- Monash Obesity & Diabetes Institute, Monash University, Clayton, Victoria, Australia
| | - M Cowley
- Department of Physiology, Faculty of Medicine, Monash University, Melbourne, Victoria, Australia ; Monash Obesity & Diabetes Institute, Monash University, Clayton, Victoria, Australia
| | - D E Cummings
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - A Dagher
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - S Diano
- Dept of Neurobiology, Yale University School of Medicine, New Haven, CT, USA
| | - S L Dickson
- Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - C Diéguez
- Department of Physiology, School of Medicine, Instituto de Investigacion Sanitaria (IDIS), University of Santiago de Compostela, Spain
| | - R Granata
- Division of Endocrinology, Diabetes and Metabolism, Dept. of Medical Sciences, University of Torino, Torino, Italy
| | - H J Grill
- Department of Psychology, Institute of Diabetes, Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, USA
| | - K Grove
- Department of Diabetes, Obesity and Metabolism, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA
| | - K M Habegger
- Comprehensive Diabetes Center, University of Alabama School of Medicine, Birmingham, AL, USA
| | - K Heppner
- Division of Diabetes, Obesity, and Metabolism, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - M L Heiman
- NuMe Health, 1441 Canal Street, New Orleans, LA 70112, USA
| | - L Holsen
- Departments of Psychiatry and Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - B Holst
- Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen N, Denmark
| | - A Inui
- Department of Psychosomatic Internal Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - J O Jansson
- Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - H Kirchner
- Medizinische Klinik I, Universitätsklinikum Schleswig-Holstein Campus Lübeck, Lübeck, Germany
| | - M Korbonits
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London, Queen Mary University of London, London, UK
| | - B Laferrère
- New York Obesity Research Center, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - C W LeRoux
- Diabetes Complications Research Centre, Conway Institute, University College Dublin, Ireland
| | - M Lopez
- Department of Physiology, Centro de Investigación en Medicina Molecular y Enfermedades Crónicas, University of Santiago de Compostela (CIMUS)-Instituto de Investigación Sanitaria (IDIS)-CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, Spain
| | - S Morin
- Institute for Diabetes and Obesity, Helmholtz Zentrum München, München, Germany
| | - M Nakazato
- Division of Neurology, Respirology, Endocrinology and Metabolism, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Kiyotake, Miyazaki, Japan
| | - R Nass
- Division of Endocrinology and Metabolism, University of Virginia, Charlottesville, VA, USA
| | - D Perez-Tilve
- Department of Internal Medicine, Department of Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - P T Pfluger
- Institute for Diabetes and Obesity, Helmholtz Zentrum München, München, Germany
| | - T W Schwartz
- Department of Neuroscience and Pharmacology, Laboratory for Molecular Pharmacology, The Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | - R J Seeley
- Department of Surgery, University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - M Sleeman
- Department of Physiology, Faculty of Medicine, Monash University, Melbourne, Victoria, Australia
| | - Y Sun
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - L Sussel
- Department of Genetics and Development, Columbia University, New York, NY, USA
| | - J Tong
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA
| | - M O Thorner
- Division of Endocrinology and Metabolism, University of Virginia, Charlottesville, VA, USA
| | - A J van der Lely
- Department of Medicine, Erasmus University MC, Rotterdam, The Netherlands
| | | | - J M Zigman
- Departments of Internal Medicine and Psychiatry, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - M Kojima
- Molecular Genetics, Institute of Life Science, Kurume University, Kurume, Japan
| | - K Kangawa
- National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
| | - R G Smith
- The Scripps Research Institute, Florida Department of Metabolism & Aging, Jupiter, FL, USA
| | - T Horvath
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Section of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - M H Tschöp
- Institute for Diabetes and Obesity, Helmholtz Zentrum München, München, Germany ; Division of Metabolic Diseases, Department of Medicine, Technical University Munich, Munich, Germany
| |
Collapse
|
31
|
Mayerl S, Liebsch C, Visser TJ, Heuer H. Absence of TRH receptor 1 in male mice affects gastric ghrelin production. Endocrinology 2015; 156:755-67. [PMID: 25490146 DOI: 10.1210/en.2014-1395] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
TRH not only functions as a thyrotropin releasing hormone but also acts as a neuropeptide in central circuits regulating food intake and energy expenditure. As one suggested mode of action, TRH expressed in the caudal brainstem influences vagal activity by activating TRH receptor 1 (TRH-R1). In order to evaluate the impact of a diminished medullary TRH signaling on ghrelin metabolism, we analyzed metabolic changes of TRH-R1 knockout (R1ko) mice in response to 24 hours of food deprivation. Because R1ko mice are hypothyroid, we also studied eu- and hypothyroid wild-type (wt) animals and R1ko mice rendered euthyroid by thyroid hormone treatment. Independent of their thyroidal state, R1ko mice displayed a higher body weight loss than wt animals and a delayed reduction in locomotor activity upon fasting. Ghrelin transcript levels in the stomach as well as total ghrelin levels in the circulation were equally high in fasted wt and R1ko mice. In contrast, only wt mice responded to fasting with a rise in ghrelin-O-acyltransferase mRNA expression and consequently an increase in serum levels of acylated ghrelin. Together, our data suggest that an up-regulation of medullary TRH expression and subsequently enhanced activation of TRH-R1 in the vagal system represents a critical step in the stimulation of ghrelin-O-acyltransferase expression upon starvation that in turn is important for adjusting the circulating levels of acylated ghrelin to the fasting condition.
Collapse
Affiliation(s)
- Steffen Mayerl
- Leibniz Institute for Age Research/Fritz Lipmann Institute (S.M., C.L., H.H.), D-07745 Jena, Germany; Brandenburg University of Technology Cottbus-Senftenberg (C.L.), D-01968 Senftenberg, Germany; Department of Internal Medicine (T.J.V.), Erasmus Medical Center, Rotterdam, The Netherlands; and Leibniz Research Institute for Environmental Medicine (H.H.), Düsseldorf, Germany
| | | | | | | |
Collapse
|
32
|
Laermans J, Broers C, Beckers K, Vancleef L, Steensels S, Thijs T, Tack J, Depoortere I. Shifting the circadian rhythm of feeding in mice induces gastrointestinal, metabolic and immune alterations which are influenced by ghrelin and the core clock gene Bmal1. PLoS One 2014; 9:e110176. [PMID: 25329803 PMCID: PMC4199674 DOI: 10.1371/journal.pone.0110176] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Accepted: 09/12/2014] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND In our 24-hour society, an increasing number of people are required to be awake and active at night. As a result, the circadian rhythm of feeding is seriously compromised. To mimic this, we subjected mice to restricted feeding (RF), a paradigm in which food availability is limited to short and unusual times of day. RF induces a food-anticipatory increase in the levels of the hunger hormone ghrelin. We aimed to investigate whether ghrelin triggers the changes in body weight and gastric emptying that occur during RF. Moreover, the effect of genetic deletion of the core clock gene Bmal1 on these physiological adaptations was studied. METHODS Wild-type, ghrelin receptor knockout and Bmal1 knockout mice were fed ad libitum or put on RF with a normal or high-fat diet (HFD). Plasma ghrelin levels were measured by radioimmunoassay. Gastric contractility was studied in vitro in muscle strips and in vivo (13C breath test). Cytokine mRNA expression was quantified and infiltration of immune cells was assessed histologically. RESULTS The food-anticipatory increase in plasma ghrelin levels induced by RF with normal chow was abolished in HFD-fed mice. During RF, body weight restoration was facilitated by ghrelin and Bmal1. RF altered cytokine mRNA expression levels and triggered contractility changes resulting in an accelerated gastric emptying, independent from ghrelin signaling. During RF with a HFD, Bmal1 enhanced neutrophil recruitment to the stomach, increased gastric IL-1α expression and promoted gastric contractility changes. CONCLUSIONS This is the first study demonstrating that ghrelin and Bmal1 regulate the extent of body weight restoration during RF, whereas Bmal1 controls the type of inflammatory infiltrate and contractility changes in the stomach. Disrupting the circadian rhythm of feeding induces a variety of diet-dependent metabolic, immune and gastrointestinal alterations, which may explain the higher prevalence of obesity and immune-related gastrointestinal disorders among shift workers.
Collapse
Affiliation(s)
- Jorien Laermans
- Gut Peptide Research Lab, Translational Research Center for Gastrointestinal Disorders, KU Leuven - University of Leuven, Leuven, Belgium
| | - Charlotte Broers
- Gut Peptide Research Lab, Translational Research Center for Gastrointestinal Disorders, KU Leuven - University of Leuven, Leuven, Belgium
| | - Kelly Beckers
- Gut Peptide Research Lab, Translational Research Center for Gastrointestinal Disorders, KU Leuven - University of Leuven, Leuven, Belgium
| | - Laurien Vancleef
- Gut Peptide Research Lab, Translational Research Center for Gastrointestinal Disorders, KU Leuven - University of Leuven, Leuven, Belgium
| | - Sandra Steensels
- Gut Peptide Research Lab, Translational Research Center for Gastrointestinal Disorders, KU Leuven - University of Leuven, Leuven, Belgium
| | - Theo Thijs
- Gut Peptide Research Lab, Translational Research Center for Gastrointestinal Disorders, KU Leuven - University of Leuven, Leuven, Belgium
| | - Jan Tack
- Gut Peptide Research Lab, Translational Research Center for Gastrointestinal Disorders, KU Leuven - University of Leuven, Leuven, Belgium
| | - Inge Depoortere
- Gut Peptide Research Lab, Translational Research Center for Gastrointestinal Disorders, KU Leuven - University of Leuven, Leuven, Belgium
- * E-mail:
| |
Collapse
|
33
|
Seki M, Masaki H, Arauchi T, Nakauchi H, Sugano S, Suzuki Y. A comparison of the rest complex binding patterns in embryonic stem cells and epiblast stem cells. PLoS One 2014; 9:e95374. [PMID: 24752154 PMCID: PMC3994037 DOI: 10.1371/journal.pone.0095374] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Accepted: 03/26/2014] [Indexed: 10/29/2022] Open
Abstract
We detected and characterized the binding sites of the representative Rest complex components Rest, Sin3A, and Lsd1. We compared their binding patterns in mouse embryonic stem (ES) cells and epiblast stem (EpiS) cells. We found few Rest sites unique to the EpiS cells. The ES-unique site features were distinct from those of the common sites, namely, the signal intensities were weaker, and the characteristic gene function categories differed. Our analyses showed that the Rest binding sites do not always overlap with the Sin3A and Lsd1 binding sites. The Sin3A binding pattern differed remarkably between the ES and EpiS cells and was accompanied by significant changes in acetylated-histone patterns in the surrounding regions. A series of transcriptome analyses in the same cell types unexpectedly showed that the putative target gene transcript levels were not dramatically different despite dynamic changes in the Rest complex binding patterns and chromatin statuses, which suggests that Rest is not the sole determinant of repression at its targets. Nevertheless, we identified putative Rest targets with explicitly enhanced transcription upon Rest knock-down in 143 and 60 common and ES-unique Rest target genes, respectively. Among such sites, several genes are involved in ES cell proliferation. In addition, we also found that long, intergenic non-coding RNAs were apparent Rest targets and shared similar features with the protein-coding target genes. Interestingly, such non-coding target genes showed less conservation through evolution than protein-coding targets. As a result of differences in the components and targets of the Rest complex, its functional roles may differ in ES and EpiS cells.
Collapse
Affiliation(s)
- Masahide Seki
- Department of Medical Genome Sciences, Graduate School of Frontier Sciences, the University of Tokyo, Kashiwa, Chiba, Japan
| | - Hideki Masaki
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, the University of Tokyo, Minato-ku, Tokyo, Japan
| | - Takako Arauchi
- Department of Medical Genome Sciences, Graduate School of Frontier Sciences, the University of Tokyo, Kashiwa, Chiba, Japan
| | - Hiromitsu Nakauchi
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, the University of Tokyo, Minato-ku, Tokyo, Japan
| | - Sumio Sugano
- Department of Medical Genome Sciences, Graduate School of Frontier Sciences, the University of Tokyo, Kashiwa, Chiba, Japan
| | - Yutaka Suzuki
- Department of Medical Genome Sciences, Graduate School of Frontier Sciences, the University of Tokyo, Kashiwa, Chiba, Japan
- * E-mail:
| |
Collapse
|
34
|
Aourz N, Portelli J, Coppens J, De Bundel D, Di Giovanni G, Van Eeckhaut A, Michotte Y, Smolders I. Cortistatin-14 mediates its anticonvulsant effects via sst2 and sst3 but not ghrelin receptors. CNS Neurosci Ther 2014; 20:662-70. [PMID: 24685142 DOI: 10.1111/cns.12259] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Revised: 02/28/2014] [Accepted: 03/04/2014] [Indexed: 12/12/2022] Open
Abstract
Cortistatin (CST)-14, a neuropeptide that is structurally and functionally related to somatostatin-14 (SRIF) binds all five somatostatin receptor subtypes (sst1-sst5). Using in vivo microdialysis and telemetry-based electroencephalographic recordings, we provide the first experimental evidence for anticonvulsive effects of CST-14 in a pilocarpine-induced seizure model in rats and mice and for the involvement of sst2 and sst3 receptors in these anticonvulsant actions of CST-14. Both receptor subtypes are required for the anticonvulsant effects of CST-14 given that co-perfusion of a selective sst2 antagonist (cyanamid15486) or a selective sst3 antagonist (SST3-ODN-8) reversed anticonvulsant effect of CST-14, and this, independently of each other. Next, as the ghrelin receptor has been proposed as a target for the biological effects of CST-14, we used ghrelin receptor knockout mice and their wild type littermates to study the involvement of this receptor in the anticonvulsive actions of CST-14. Our results show a significant decrease in seizure duration in both genotypes when CST-14 treated mice were compared with corresponding control animals receiving only pilocarpine. In addition, this CST-14-induced decrease was comparable in both genotypes. We here thus provide the first evidence that ghrelin receptors are not involved in mediating anticonvulsant actions of CST-14 in vivo.
Collapse
Affiliation(s)
- Najat Aourz
- Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information, Research Group Experimental Pharmacology, Center for Neurosciences (C4N), Vrije Universiteit Brussel, Brussels, Belgium
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Abstract
Obesity is one of the largest and fastest growing public health problems in the world. Last century social changes have set an obesogenic milieu that calls for micro and macro environment interventions for disease prevention, while treatment is mandatory for individuals already obese. The cornerstone of overweight and obesity treatment is diet and physical exercise. However, many patients find lifestyle modifications difficult to comply and prone to failure in the long-term; therefore many patients consider anti-obesity drugs an important adjuvant if not a better alternative to behavioral approach or obesity surgery. Since the pharmacological options for obesity treatment remain quite limited, this is an exciting research area, with new treatment targets and strategies on the horizon. This review discusses the development of innovative therapeutic agents, focusing in energy homeostasis regulation and the use of molecular vaccines, targeting hormones such as somatostatin, GIP and ghrelin, to reduce body weight.
Collapse
Affiliation(s)
- Mariana P Monteiro
- Department of Anatomy; Unit for Multidisciplinary Biomedical Research (UMIB); Institute for Biomedical Sciences Abel Salazar (ICBAS); University of Porto; Porto, Portugal
| |
Collapse
|
36
|
Delporte C. Structure and physiological actions of ghrelin. SCIENTIFICA 2013; 2013:518909. [PMID: 24381790 PMCID: PMC3863518 DOI: 10.1155/2013/518909] [Citation(s) in RCA: 102] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 11/10/2013] [Indexed: 05/30/2023]
Abstract
Ghrelin is a gastric peptide hormone, discovered as being the endogenous ligand of growth hormone secretagogue receptor. Ghrelin is a 28 amino acid peptide presenting a unique n-octanoylation modification on its serine in position 3, catalyzed by ghrelin O-acyl transferase. Ghrelin is mainly produced by a subset of stomach cells and also by the hypothalamus, the pituitary, and other tissues. Transcriptional, translational, and posttranslational processes generate ghrelin and ghrelin-related peptides. Homo- and heterodimers of growth hormone secretagogue receptor, and as yet unidentified receptors, are assumed to mediate the biological effects of acyl ghrelin and desacyl ghrelin, respectively. Ghrelin exerts wide physiological actions throughout the body, including growth hormone secretion, appetite and food intake, gastric secretion and gastrointestinal motility, glucose homeostasis, cardiovascular functions, anti-inflammatory functions, reproductive functions, and bone formation. This review focuses on presenting the current understanding of ghrelin and growth hormone secretagogue receptor biology, as well as the main physiological effects of ghrelin.
Collapse
Affiliation(s)
- Christine Delporte
- Laboratory of Pathophysiological and Nutritional Biochemistry, Université Libre de Bruxelles, 808 Route de Lennik, Bat G/E-CP611, 1070 Brussels, Belgium
| |
Collapse
|
37
|
Avau B, Carbone F, Tack J, Depoortere I. Ghrelin signaling in the gut, its physiological properties, and therapeutic potential. Neurogastroenterol Motil 2013; 25:720-32. [PMID: 23910374 DOI: 10.1111/nmo.12193] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Accepted: 06/26/2013] [Indexed: 02/08/2023]
Abstract
BACKGROUND Ghrelin, an orexigenic hormone secreted from the stomach, was soon after its discovery hypothesized to be a prokinetic agent, due to its homology to motilin. Studies in animals and humans, using ghrelin and ghrelin receptor agonists, confirmed this hypothesis, suggesting a therapeutic potential for the ghrelin receptor in the treatment of gastrointestinal motility disorders. Precilinical studies demonstrated that ghrelin can act directly on ghrelin receptors on the enteric nervous system, but the predominant route of action under physiological circumstances is signaling via the vagus nerve in the upper gastrointestinal tract and the pelvic nerves in the colon. Different pharmaceutical companies have designed stable ghrelin mimetics that revealed promising results in trials for the treatment of diabetic gastroparesis and post-operative ileus. Nevertheless, no drug was able to reach the market so far, facing problems proving superiority over placebo treatment in larger trials. PURPOSE This review aims to summarize the road that led to the current knowledge concerning the prokinetic properties of ghrelin with a focus on the therapeutic potential of ghrelin receptor agonists in the treatment of hypomotility disorders. In addition, we outline some of the problems that could be at the basis of the negative outcome of the trials with ghrelin agonists and question whether the right target groups were selected. It is clear that a new approach is needed to develop marketable drugs with this class of gastroprokinetic agents.
Collapse
Affiliation(s)
- B Avau
- Translational Research Center for Gastrointestinal Disorders, University of Leuven, Leuven, Belgium
| | | | | | | |
Collapse
|
38
|
Creedon CT, Verhulst PJ, Choi KM, Mason JE, Linden DR, Szurszewski JH, Gibbons SJ, Farrugia G. Assessment of gastric emptying in non-obese diabetic mice using a [13C]-octanoic acid breath test. J Vis Exp 2013:e50301. [PMID: 23542813 DOI: 10.3791/50301] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Gastric emptying studies in mice have been limited by the inability to follow gastric emptying changes in the same animal since the most commonly used techniques require killing of the animals and postmortem recovery of the meal(1,2). This approach prevents longitudinal studies to determine changes in gastric emptying with age and progression of disease. The commonly used [(13)C]-octanoic acid breath test for humans(3) has been modified for use in mice(4-6) and rats(7) and we previously showed that this test is reliable and responsive to changes in gastric emptying in response to drugs and during diabetic disease progression(8). In this video presentation the principle and practical implementation of this modified test is explained. As in the previous study, NOD LtJ mice are used, a model of type 1 diabetes(9). A proportion of these mice develop the symptoms of gastroparesis, a complication of diabetes characterized by delayed gastric emptying without mechanical obstruction of the stomach(10). This paper demonstrates how to train the mice for testing, how to prepare the test meal and obtain 4 hr gastric emptying data and how to analyze the obtained data. The carbon isotope analyzer used in the present study is suitable for the automatic sampling of the air samples from up to 12 mice at the same time. This technique allows the longitudinal follow-up of gastric emptying from larger groups of mice with diabetes or other long-standing diseases.
Collapse
Affiliation(s)
- Christopher T Creedon
- Enteric Neuroscience Program, Department of Physiology and Biomedical Engineering, Mayo Clinic
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Verhulst PJ, Depoortere I. Ghrelin's second life: From appetite stimulator to glucose regulator. World J Gastroenterol 2012; 18:3183-95. [PMID: 22783041 PMCID: PMC3391754 DOI: 10.3748/wjg.v18.i25.3183] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2011] [Revised: 12/01/2011] [Accepted: 01/18/2012] [Indexed: 02/06/2023] Open
Abstract
Ghrelin, a 28 amino acid peptide hormone produced by the stomach, was the first orexigenic hormone to be discovered from the periphery. The octanoyl modification at Ser3, mediated by ghrelin O-acyltransferase (GOAT), is essential for ghrelin’s biological activity. Ghrelin stimulates food intake through binding to its receptor (GRLN-R) on neurons in the arcuate nucleus of the hypothalamus. Ghrelin is widely expressed throughout the body; accordingly, it is implicated in several other physiological functions, which include growth hormone release, gastric emptying, and body weight regulation. Ghrelin and GRLN-R expression are also found in the pancreas, suggesting a local physiological role. Accordingly, several recent studies now point towards an important role for ghrelin and its receptor in the regulation of blood glucose homeostasis, which is the main focus of this review. Several mechanisms of this regulation by ghrelin have been proposed, and one possibility is through the regulation of insulin secretion. Despite some controversy, most studies suggest that ghrelin exerts an inhibitory effect on insulin secretion, resulting in increased circulating glucose levels. Ghrelin may thus be a diabetogenic factor. Obesity-related type 2 diabetes has become an increasingly important health problem, almost reaching epidemic proportions in the world; therefore, antagonists of the ghrelin-GOAT signaling pathway, which will tackle both energy- and glucose homeostasis, may be considered as promising new therapies for this disease.
Collapse
|
40
|
Portelli J, Thielemans L, Ver Donck L, Loyens E, Coppens J, Aourz N, Aerssens J, Vermoesen K, Clinckers R, Schallier A, Michotte Y, Moechars D, Collingridge GL, Bortolotto ZA, Smolders I. Inactivation of the constitutively active ghrelin receptor attenuates limbic seizure activity in rodents. Neurotherapeutics 2012; 9:658-72. [PMID: 22669710 PMCID: PMC3441926 DOI: 10.1007/s13311-012-0125-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Ghrelin is a pleiotropic neuropeptide that has been recently implicated in epilepsy. Animal studies performed to date indicate that ghrelin has anticonvulsant properties; however, its mechanism of anticonvulsant action is unknown. Here we show that the anticonvulsant effects of ghrelin are mediated via the growth hormone secretagogue receptor (GHSR). To our surprise, however, we found that the GHSR knockout mice had a higher seizure threshold than their wild-type littermates when treated with pilocarpine. Using both in vivo and in vitro models, we further discovered that inverse agonism and desensitization/internalization of the GHSR attenuate limbic seizures in rats and epileptiform activity in hippocampal slices. This constitutes a novel mechanism of anticonvulsant action, whereby an endogenous agonist reduces the activity of a constitutively active receptor.
Collapse
Affiliation(s)
- Jeanelle Portelli
- Center for Neurosciences, Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium
| | - Leen Thielemans
- Janssen Research and Development, a Division of Janssen Pharmaceutica NV, 2340 Beerse, Belgium
| | - Luc Ver Donck
- Janssen Research and Development, a Division of Janssen Pharmaceutica NV, 2340 Beerse, Belgium
| | - Ellen Loyens
- Center for Neurosciences, Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium
| | - Jessica Coppens
- Center for Neurosciences, Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium
| | - Najat Aourz
- Center for Neurosciences, Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium
| | - Jeroen Aerssens
- Janssen Research and Development, a Division of Janssen Pharmaceutica NV, 2340 Beerse, Belgium
| | - Katia Vermoesen
- Center for Neurosciences, Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium
| | - Ralph Clinckers
- Center for Neurosciences, Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium
| | - Anneleen Schallier
- Center for Neurosciences, Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium
| | - Yvette Michotte
- Center for Neurosciences, Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium
| | - Dieder Moechars
- Janssen Research and Development, a Division of Janssen Pharmaceutica NV, 2340 Beerse, Belgium
| | - Graham L. Collingridge
- MRC Centre for Synaptic Plasticity, School of Physiology and Pharmacology, University of Bristol, BS8 1TD Bristol, United Kingdom
- Department of Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, 151-746 Seoul, Gwanakgu Korea
| | - Zuner A. Bortolotto
- MRC Centre for Synaptic Plasticity, School of Physiology and Pharmacology, University of Bristol, BS8 1TD Bristol, United Kingdom
| | - Ilse Smolders
- Center for Neurosciences, Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium
| |
Collapse
|
41
|
Janssen S, Laermans J, Iwakura H, Tack J, Depoortere I. Sensing of fatty acids for octanoylation of ghrelin involves a gustatory G-protein. PLoS One 2012; 7:e40168. [PMID: 22768248 PMCID: PMC3387020 DOI: 10.1371/journal.pone.0040168] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2012] [Accepted: 06/05/2012] [Indexed: 11/30/2022] Open
Abstract
Background Ghrelin is an important regulator of energy – and glucose homeostasis. The octanoylation at Ser3 is essential for ghrelin’s biological effects but the mechanisms involved in the octanoylation are unknown. We investigated whether the gustatory G-protein, α-gustducin, and the free fatty acid receptors GPR40 and GPR120 are involved in the fatty acid sensing mechanisms of the ghrelin cell. Methods Wild-type (WT) and α-gustducin knockout (gust−/−) mice were fed a glyceryl trioctanoate-enriched diet (OD) during 2 weeks. Ghrelin levels and gastric emptying were determined. Co-localization between GPR40, GPR120 and ghrelin or α-gustducin/α-transducin was investigated by immunofluorescence staining. The role of GPR120 in the effect of medium and long chain fatty acids on the release of ghrelin was studied in the ghrelinoma cell line, MGN3-1. The effect of the GPR40 agonist, MEDICA16, and the GPR120 agonist, grifolic acid, on ghrelin release was studied both in vitro and in vivo. Results Feeding an OD specifically increased octanoyl ghrelin levels in the stomach of WT mice but not of gust−/− mice. Gastric emptying was accelerated in WT but not in gust−/− mice. GPR40 was colocalized with desoctanoyl but not with octanoyl ghrelin, α-gustducin or α-transducin positive cells in the stomach. GPR120 only colocalized with ghrelin in the duodenum. Addition of octanoic acid or α-linolenic acid to MGN3-1 cells increased and decreased octanoyl ghrelin levels, respectively. Both effects could not be blocked by GPR120 siRNA. MEDICA16 and grifolic acid did not affect ghrelin secretion in vitro but oral administration of grifolic acid increased plasma ghrelin levels. Conclusion This study provides the first evidence that α-gustducin is involved in the octanoylation of ghrelin and shows that the ghrelin cell can sense long- and medium-chain fatty acids directly. GPR120 but not GPR40 may play a role in the lipid sensing cascade of the ghrelin cell.
Collapse
Affiliation(s)
- Sara Janssen
- Translational Research Center for Gastrointestinal Disorders, Catholic University of Leuven, Leuven, Belgium
| | - Jorien Laermans
- Translational Research Center for Gastrointestinal Disorders, Catholic University of Leuven, Leuven, Belgium
| | - Hiroshi Iwakura
- Departments of Medicine, Clinical Science, Endocrinology, and Metabolism, Translational Research Center, Kyoto University Graduate School of Medicine, Kyoto University Hospital, Kyoto, Japan
| | - Jan Tack
- Translational Research Center for Gastrointestinal Disorders, Catholic University of Leuven, Leuven, Belgium
| | - Inge Depoortere
- Translational Research Center for Gastrointestinal Disorders, Catholic University of Leuven, Leuven, Belgium
- * E-mail:
| |
Collapse
|
42
|
Lee J, Cummings BP, Martin E, Sharp JW, Graham JL, Stanhope KL, Havel PJ, Raybould HE. Glucose sensing by gut endocrine cells and activation of the vagal afferent pathway is impaired in a rodent model of type 2 diabetes mellitus. Am J Physiol Regul Integr Comp Physiol 2011; 302:R657-66. [PMID: 22160540 DOI: 10.1152/ajpregu.00345.2011] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Glucose in the gut lumen activates gut endocrine cells to release 5-HT, glucagon-like peptide 1/2 (GLP-1/2), and glucose-dependent insulinotropic polypeptide (GIP), which act to change gastrointestinal function and regulate postprandial plasma glucose. There is evidence that both release and action of incretin hormones is reduced in type 2 diabetes (T2D). We measured cellular activation of enteroendocrine and enterochromaffin cells, enteric neurons, and vagal afferent neurons in response to intestinal glucose in a model of type 2 diabetes mellitus, the UCD-T2DM rat. Prediabetic (PD), recent-diabetic (RD, 2 wk postonset), and 3-mo diabetic (3MD) fasted UCD-T2DM rats were given an orogastric gavage of vehicle (water, 0.5 ml /100 g body wt) or glucose (330 μmol/100 g body wt); after 6 min tissue was removed and cellular activation was determined by immunohistochemistry for phosphorylated calcium calmodulin-dependent kinase II (pCaMKII). In PD rats, pCaMKII immunoreactivity was increased in duodenal 5-HT (P < 0.001), K (P < 0.01) and L (P < 0.01) cells in response to glucose; glucose-induced activation of all three cell types was significantly reduced in RD and 3MD compared with PD rats. Immunoreactivity for GLP-1, but not GIP, was significantly reduced in RD and 3MD compared with PD rats (P < 0.01). Administration of glucose significantly increased pCaMKII in enteric and vagal afferent neurons in PD rats; glucose-induced pCaMKII immunoreactivity was attenuated in enteric and vagal afferent neurons (P < 0.01, P < 0.001, respectively) in RD and 3MD. These data suggest that glucose sensing in enteroendocrine and enterochromaffin cells and activation of neural pathways is markedly impaired in UCD-T2DM rats.
Collapse
Affiliation(s)
- Jennifer Lee
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California, Davis, California 95616, USA
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Role of the AMP-activated protein kinase (AMPK) signaling pathway in the orexigenic effects of endogenous ghrelin. ACTA ACUST UNITED AC 2011; 173:27-35. [PMID: 21963822 DOI: 10.1016/j.regpep.2011.09.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2011] [Revised: 08/08/2011] [Accepted: 09/16/2011] [Indexed: 12/14/2022]
Abstract
Ghrelin, released from the stomach, stimulates food intake through activation of the ghrelin receptor (GHS-R) located on neuropeptide Y (NPY)/agouti-related peptide (AgRP) neurons in the hypothalamus. A role for the energy sensor AMP-activated protein kinase (AMPK) and its downstream effector uncoupling protein 2 (UCP2) in the stimulatory effect of exogenous ghrelin on NPY/AgRP expression and food intake has been suggested. This study aimed to investigate whether a rise in endogenous ghrelin levels is able to influence hypothalamic AMPK activity, pACC, UCP2 and NPY/AgRP expression through activation of GHS-R. An increase in endogenous ghrelin levels was established by fasting (24h) or by induction of streptozotocin(STZ)-diabetes (15 days) in GHS-R(+/+) and GHS-R(-/-) mice. GHS-R(+/+) mice showed a significant increase in AgRP and NPY mRNA expression after fasting, which was not observed in GHS-R(-/-) mice. Fasting did not affect AMPK activity nor ACC phosphorylation in both genotypes and increased UCP2 mRNA expression. The hyperghrelinemia associated with STZ-induced diabetes was accompanied by an increased NPY and AgRP expression in GHS-R(+/+) but not in GHS-R(-/-) mice. AMPK activity and UCP2 expression in GHS-R(+/+) mice after induction of diabetes were decreased to a similar extent in both genotypes. Exogenous ghrelin administration tended to decrease hypothalamic AMPK activity. In conclusion, an increase in endogenous ghrelin levels triggered by fasting or STZ-induced diabetes stimulates the expression of AgRP and NPY via interaction with the GHS-R. The changes in AMPK activity, pACC and UCP2 occur independently from GHS-R suggesting that they do not play a major role in the orexigenic effect of endogenous ghrelin.
Collapse
|
44
|
Bitter taste receptors and α-gustducin regulate the secretion of ghrelin with functional effects on food intake and gastric emptying. Proc Natl Acad Sci U S A 2011; 108:2094-9. [PMID: 21245306 DOI: 10.1073/pnas.1011508108] [Citation(s) in RCA: 256] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Ghrelin is a hunger hormone with gastroprokinetic properties but the factors controlling ghrelin secretion from the stomach are unknown. Bitter taste receptors (T2R) and the gustatory G proteins, α-gustducin (gust) and α-transducin, are expressed in the gut and are involved in the chemosensation of nutrients. This study aimed to investigate whether T2R-agonists affect (i) ghrelin release via α-gustducin and (ii) food intake and gastric emptying via the release of ghrelin. The mouse stomach contains two ghrelin cell populations: cells containing octanoyl and desoctanoyl ghrelin, which were colocalized with α-gustducin and α-transducin, and cells staining for desoctanoyl ghrelin. Gavage of T2R-agonists increased plasma octanoyl ghrelin levels in WT mice but the effect was partially blunted in gust(-/-) mice. Intragastric administration of T2R-agonists increased food intake during the first 30 min in WT but not in gust(-/-) and ghrelin receptor knockout mice. This increase was accompanied by an increase in the mRNA expression of agouti-related peptide in the hypothalamus of WT but not of gust(-/-) mice. The temporary increase in food intake was followed by a prolonged decrease (next 4 h), which correlated with an inhibition of gastric emptying. The delay in emptying, which was partially counteracted by ghrelin, was not mediated by cholecystokinin and GLP-1 but involved a direct inhibitory effect of T2R-agonists on gastric contractility. This study is unique in providing functional evidence that activation of bitter taste receptors stimulates ghrelin secretion. Modulation of endogenous ghrelin levels by tastants may provide novel therapeutic applications for the treatment of weight -and gastrointestinal motility disorders.
Collapse
|
45
|
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 2011; 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] [MESH Headings] [Grants] [Track Full Text] [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
|
46
|
Gallas S, Sinno MH, Boukhettala N, Coëffier M, Dourmap N, Gourcerol G, Ducrotté P, Déchelotte P, Leroi AM, Fetissov SO. Gastric electrical stimulation increases ghrelin production and inhibits catecholaminergic brainstem neurons in rats. Eur J Neurosci 2010; 33:276-84. [PMID: 21059113 DOI: 10.1111/j.1460-9568.2010.07474.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Gastric electrical stimulation (GES) is a new therapeutic option for functional dyspepsia and gastroparesis. In addition to ameliorating nausea and vomiting, GES results in improved appetite which is not always associated with accelerated gastric emptying. To explore the central and peripheral factors underlying GES-associated improvement of appetite we developed a GES model in anaesthetized Wistar rats. During laparotomy, two electrodes were implanted into the stomach and high-frequency low-energy GES (14 Hz, 5 mA) was applied. The effects of 1 h GES were compared with sham stimulation. After GES, c-Fos expression was increased in the mucosal and submucosal layers of the stimulated area (174%). In the stomach, GES increased ghrelin mRNA (178%) and doubled the number of ghrelin-positive cells, resulting in elevated plasma levels of ghrelin (2.3 ± 0.2 vs. 1.6 ± 0.2 ng/mL). In the arcuate nucleus of the hypothalamus, GES increased c-Fos (277%) and agouti-related protein (AgRP) mRNA expression (135%). GES reduced the number of c-Fos-positive cells throughout the nucleus of the solitary tract (between 93 and 75% from rostral to caudal levels) including catecholaminergic neurons (81% at caudal level). Gastric emptying, plasma glucose and heart rate variability were not affected by GES. This study shows that GES may improve appetite via stimulation of main orexigenic pathways, including ghrelin production in the stomach and AgRP in the hypothalamus, as well as by reducing the activity of catecholaminergic brainstem neurons.
Collapse
Affiliation(s)
- Syrine Gallas
- Digestive System & Nutrition Laboratory (ADEN EA4311), Institute of Medical Research and Innovation, Rouen University & Hospital, IFR23, Rouen 76183, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Yakabi K, Kurosawa S, Tamai M, Yuzurihara M, Nahata M, Ohno S, Ro S, Kato S, Aoyama T, Sakurada T, Takabayashi H, Hattori T. Rikkunshito and 5-HT2C receptor antagonist improve cisplatin-induced anorexia via hypothalamic ghrelin interaction. ACTA ACUST UNITED AC 2010; 161:97-105. [PMID: 20171995 DOI: 10.1016/j.regpep.2010.02.003] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2009] [Revised: 02/10/2010] [Accepted: 02/11/2010] [Indexed: 12/24/2022]
Abstract
Circulating ghrelin concentration regulates appetite behavior, but no study thus far has focused on the role of central ghrelin in anorexia after chemotherapy. To clarify the action mechanisms of rikkunshito (RKT), a traditional Japanese medicine, on cisplatin-induced anorexia, we attempted to elucidate its effect on hypothalamic ghrelin receptor expression in cisplatin-induced anorexia. We first examined the effects of an intracerebroventricular (ICV) injection of exogenous ghrelin on food intake with or without cisplatin treatment, and the effects of cisplatin or m-chlorophenylpiperazine (mCPP), a 5-HT2C receptor agonist, on hypothalamic growth hormone secretagogue receptor 1a (GHS-R1a) mRNA expression. To identify the mechanism of cisplatin-induced decrease in hypothalamic GHS-R1a mRNA expression, we evaluated the effects of SB242084HCl, a 5-HT2C receptor antagonist, and RKT on hypothalamic GHS-R1a gene expression, along with the effect of coadministration of a GHS-R1a antagonist on decreased food intake. Compared to vehicle controls, an ICV-injected rat ghrelin failed to inhibit the decrease in food intake in cisplatin-treated rats. Hypothalamic GHS-R1a gene expression was significantly reduced after cisplatin or mCPP treatment, and the induced decrease was reversed by SB242084HCl or RKT, but not granisetron or ondansetron, both of which are 5-HT3 receptor antagonists. Their suppressive effect on the decrease in food intake was abolished by coadministration of the GHS-R1a antagonist. Administration of RKT or SB242084HCl reversed the decrease in food intake induced by mCPP injection. The improvement by RKT on decreased food intake after cisplatin treatment was partly mediated by hesperidin and isoliquiritigenin, components of RKT. Cisplatin-induced anorexia may worsen because of decreased hypothalamic GHS-R1a gene expression. A 5-HT2C receptor antagonist and RKT suppressed cisplatin-induced anorexia by inhibiting reduction of GHS-R1a signal transduction in the hypothalamus.
Collapse
Affiliation(s)
- Koji Yakabi
- Department of Gastroenterology and Hepatology, Saitama Medical Center, Saitama Medical University, 1981 Tsujido-machi, Kamoda, Kawagoe-city, Saitama 350-8550, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
De Smet B, Mitselos A, Depoortere I. Motilin and ghrelin as prokinetic drug targets. Pharmacol Ther 2009; 123:207-23. [DOI: 10.1016/j.pharmthera.2009.04.004] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2009] [Accepted: 04/09/2009] [Indexed: 12/13/2022]
|
49
|
Depoortere I. Targeting the ghrelin receptor to regulate food intake. ACTA ACUST UNITED AC 2009; 156:13-23. [PMID: 19362579 DOI: 10.1016/j.regpep.2009.04.002] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2009] [Accepted: 04/05/2009] [Indexed: 01/28/2023]
Affiliation(s)
- Inge Depoortere
- Centre for Gastroenterological Research, Gasthuisberg, O&N, box 701, B-3000 Leuven, Belgium.
| |
Collapse
|