1
|
Dezaki K, Yada T. Status of ghrelin as an islet hormone and paracrine/autocrine regulator of insulin secretion. Peptides 2022; 148:170681. [PMID: 34728253 DOI: 10.1016/j.peptides.2021.170681] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 10/06/2021] [Accepted: 10/28/2021] [Indexed: 12/20/2022]
Abstract
Ghrelin is expressed in the pancreatic islet cells as well as the stomach. In the perfused pancreas and isolated islets, GHS-R antagonism, ghrelin immunoneutralization and ghrelin-knockout (Ghr-KO) all increase glucose-induced insulin release. Thus, pharmacological, immunological and genetic blockades of ghrelin in the pancreatic islets all markedly augment glucose-induced insulin release, showing that islet-derived ghrelin physiologically restricts insulin release in rodents. In this review, we focus on the current understanding of the following key questions: 1) from which islet cells ghrelin is released, 2) on which islet cells ghrelin acts, and 3) mechanisms by which the islet-derived ghrelin inhibits insulin secretion.
Collapse
Affiliation(s)
- Katsuya Dezaki
- Iryo Sosei University, Faculty of Pharmacy, 5-5-1, Chuodai Iino, Iwaki, Fukushima, 970-8551, Japan.
| | - Toshihiko Yada
- Kansai Electric Power Medical Research Institute, Center for Integrative Physiology, Division of Integrative Physiology, 1-5-6 Minatojimaminamimachi, Chuo-ku, Kobe, 650-0047, Japan; Division of Diabetes, Metabolism and Endocrinology, Kobe University Graduate School of Medicine, 7-5-2 Kusunoki-Cho, Chuo-Ku, Kobe, 650-0017, Japan.
| |
Collapse
|
2
|
Song L, Yuan J, Liu Y, Zhang D, Zhang C, Lin Q, Li M, Su K, Li Y, Gao G, Ma R, Dong J. Ghrelin system is involved in improvements in glucose metabolism mediated by hyperbaric oxygen treatment in a streptozotocin‑induced type 1 diabetes mouse model. Mol Med Rep 2020; 22:3767-3776. [PMID: 32901885 PMCID: PMC7533472 DOI: 10.3892/mmr.2020.11481] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 07/28/2020] [Indexed: 12/17/2022] Open
Abstract
Type 1 diabetes mellitus (T1DM) is an autoimmune disorder for which the only effective therapy is insulin replacement. Hyperbaric oxygen (HBO) therapy has demonstrated potential in improving hyperglycemia and as a treatment option for T1DM. Ghrelin and HBO have been previously reported to exert proliferative, anti-apoptotic and anti-inflammatory effects in pancreatic cells. The present study investigated the mechanism underlying HBO- and ghrelin system-mediated regulation of glucose metabolism. Male C57BL/6 mice were intraperitoneally injected with streptozotocin (STZ; 150 mg/kg) to induce T1DM before the diabetic mice were randomly assigned into the T1DM and T1DM + HBO groups. Mice in the T1DM + HBO group received HBO (1 h; 100% oxygen; 2 atmospheres absolute) daily for 2 weeks. Significantly lower blood glucose levels and food intake were observed in mice in the T1DM + HBO group. Following HBO treatment, islet β-cell area were increased whereas those of α-cell were decreased in the pancreas. In addition, greater hepatic glycogen storage in liver was observed, which coincided with higher pancreatic glucose transporter 2 (GLUT2) expression levels and reduced hepatic GLUT2 membrane trafficking. There were also substantially higher total plasma ghrelin concentrations and gastric ghrelin-O-acyl transferase (GOAT) expression levels in mice in the T1DM + HBO group. HBO treatment also abolished reductions in pancreatic GOAT expression levels in T1DM mice. Additionally, hepatic growth hormone secretagogue receptor-1a levels were found to be lower in mice in the T1DM + HBO group compared with those in the T1DM group. These results suggest that HBO administration improved glucose metabolism in a STZ-induced T1DM mouse model. The underlying mechanism involves improved insulin-release, glucose-sensing and regulation of hepatic glycogen storage, an observation that was also likely dependent on the ghrelin signalling system.
Collapse
Affiliation(s)
- Limin Song
- Department of Special Medicine, Medical College, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Junhua Yuan
- Department of Special Medicine, Medical College, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Yuan Liu
- Department of Special Medicine, Medical College, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Di Zhang
- Department of Special Medicine, Medical College, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Caishun Zhang
- Department of Special Medicine, Medical College, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Qian Lin
- Department of Special Medicine, Medical College, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Manwen Li
- Department of Special Medicine, Medical College, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Kaizhen Su
- Department of Clinical Medicine, Medical College, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Yanrun Li
- Department of Clinical Medicine, Medical College, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Guangkai Gao
- Department of Hyperbaric Medicine, Hospital of Chinese People's Liberation Army, Qingdao, Shandong 266072, P.R. China
| | - Ruixia Ma
- Department of Nephrology, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266005, P.R. China
| | - Jing Dong
- Department of Special Medicine, Medical College, Qingdao University, Qingdao, Shandong 266071, P.R. China
| |
Collapse
|
3
|
Camacho-Ramírez A, Mayo-Ossorio MÁ, Pacheco-García JM, Almorza-Gomar D, Ribelles-García A, Belmonte-Núñez A, Prada-Oliveira JA, Pérez-Arana GM. Pancreas is a preeminent source of ghrelin after sleeve gastrectomy in Wistar rats. Histol Histopathol 2020; 35:801-809. [PMID: 31951010 DOI: 10.14670/hh-18-200] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Many surgical techniques are employed in the treatment of severe obesity. A main consequence of these techniques is the improvement of type 2 Diabetes mellitus. Ghrelin is a gut hormone released in the gastric fundus and corpus, which has been related to diabetic improvement as mentioned in these papers. Sleeve gastrectomy and Roux-en Y Gastric Bypass are surgical techniques broadly employed in humans; both severely reduce the gastric surface. Paradoxically, the serum level of ghrelin in patients is preserved. We hypothesized about the role of embryonic pancreatic epsilon cells, which have the capacity to release ghrelin. We studied the changes in the epsilon cells and differentiation markers with immunostaining and ghrelin serum level and after surgery. We employed euglycemic male Wistar rats: two surgical groups (Sleeve gastrectomy and Roux-en Y Gastric Bypass) and two control groups. We reported a significant increase of ghrelin epsilon-cells in the pancreas and basal serum after Sleeve gastrectomy versus the control groups. The epsilon cellular increment was related to neogenesis, as the neurogenin-3 marker revealed. The Roux-en Y Gastric Bypass showed neither epsilon cell increase nor basal serum changes in ghrelin release. As a conclusion, we reported that the severe suppression of the fundus gastric produced the recovery of ghrelin released by the epsilon cells, which was indicative of an ontogenic embryonic pancreatic function.
Collapse
Affiliation(s)
- Alonso Camacho-Ramírez
- Surgery Unit, Puerta del Mar University Hospital, University of Cádiz, Cádiz, Spain.,Biomedical Science Research and Innovation Institute (INIBICA), Puerta del Mar University Hospital, Cádiz, Spain.,Asociación Gaditana de Apoyo al Investigador AGAI, Cádiz, Spain
| | - María Ángeles Mayo-Ossorio
- Surgery Unit, Puerta del Mar University Hospital, University of Cádiz, Cádiz, Spain.,Biomedical Science Research and Innovation Institute (INIBICA), Puerta del Mar University Hospital, Cádiz, Spain
| | - José Manuel Pacheco-García
- Surgery Unit, Puerta del Mar University Hospital, University of Cádiz, Cádiz, Spain.,Biomedical Science Research and Innovation Institute (INIBICA), Puerta del Mar University Hospital, Cádiz, Spain
| | - David Almorza-Gomar
- Biomedical Science Research and Innovation Institute (INIBICA), Puerta del Mar University Hospital, Cádiz, Spain.,Department of Operative Statistic and Research, University of Cádiz, Cádiz, Spain
| | - Antonio Ribelles-García
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Cádiz, Cádiz, Spain.,Sustainable Social Development Research Institute (INDESS), University of Cádiz, Cádiz, Spain
| | - Ana Belmonte-Núñez
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Cádiz, Cádiz, Spain
| | - J Arturo Prada-Oliveira
- Biomedical Science Research and Innovation Institute (INIBICA), Puerta del Mar University Hospital, Cádiz, Spain.,Asociación Gaditana de Apoyo al Investigador AGAI, Cádiz, Spain.,Department of Human Anatomy and Embryology, Faculty of Medicine, University of Cádiz, Cádiz, Spain.
| | - Gonzalo M Pérez-Arana
- Biomedical Science Research and Innovation Institute (INIBICA), Puerta del Mar University Hospital, Cádiz, Spain.,Department of Human Anatomy and Embryology, Faculty of Medicine, University of Cádiz, Cádiz, Spain.,Asociación Gaditana de Apoyo al Investigador AGAI, Cádiz, Spain
| |
Collapse
|
4
|
Gray SM, Page LC, Tong J. Ghrelin regulation of glucose metabolism. J Neuroendocrinol 2019; 31:e12705. [PMID: 30849212 PMCID: PMC6688917 DOI: 10.1111/jne.12705] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 03/04/2019] [Accepted: 03/05/2019] [Indexed: 12/14/2022]
Abstract
Ghrelin and its receptor, the growth hormone secretagogue receptor 1a (GHSR1a), are implicated in the regulation of glucose metabolism via direct actions in the pancreatic islet, as well as peripheral insulin-sensitive tissues and the brain. Although many studies have explored the role of ghrelin in glucose tolerance and insulin secretion, a complete mechanistic understanding remains to be clarified. This review highlights the local expression and function of ghrelin and GHSR1a in pancreatic islets and how this axis may modulate insulin secretion from pancreatic β-cells. Additionally, we discuss the effect of ghrelin on in vivo glucose metabolism in rodents and humans, as well as the metabolic circumstances under which the action of ghrelin may predominate.
Collapse
Affiliation(s)
- Sarah. M. Gray
- Duke Molecular Physiology Institute, Duke University, Durham, NC 27701
| | - Laura C. Page
- Division of Endocrinology, Department of Pediatrics, Duke University, Durham, NC 27701
| | - Jenny Tong
- Duke Molecular Physiology Institute, Duke University, Durham, NC 27701
- Division of Endocrinology, Department of Pediatrics, Duke University, Durham, NC 27701
- Division of Endocrinology, Metabolism, and Nutrition, Department of Medicine, Duke University, Durham, NC 27701
| |
Collapse
|
5
|
Zhang CS, Wang LX, Wang R, Liu Y, Song LM, Yuan JH, Wang B, Dong J. The Correlation Between Circulating Ghrelin and Insulin Resistance in Obesity: A Meta-Analysis. Front Physiol 2018; 9:1308. [PMID: 30298019 PMCID: PMC6160589 DOI: 10.3389/fphys.2018.01308] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 08/29/2018] [Indexed: 11/13/2022] Open
Abstract
Background: Ghrelin, a peptide mainly produced by stomach X-A cells. It plays a pivotal role in the regulation of food intake and energy metabolism, including glucose metabolism and insulin sensitivity. However, the correlation between circulating ghrelin levels and insulin resistance in obesity remained uncertain. This meta-analysis aimed to clarify the association between ghrelin and IR in obesity. Methods: A systematic literature search was performed using PubMed, EMBASE, Cochrane Library and Web of Science until April 18, 2018 with the keywords “ghrelin” and “insulin resistance.” Two independent reviewers selected studies and assessed data. Subgroup analyses were performed to search for sources of heterogeneity. Funnel plots and Egger's test were used to detect publication bias. A random-effects model was used to calculate the pooled effect size. Results: Ten studies with 546 participants were included in this meta-analysis. We found that ghrelin levels were negatively correlated with IR in obese individuals. (r = −0.31; 95% CI: −0.45 to −0.18). Subgroup analysis revealed that circulating ghrelin levels were significantly negatively correlated with IR in people with normal fasting blood glucose (FBG) (<6.9 mmol/dl) (r = −0.28; 95% CI: −0.47 to −0.09, I2 = 39.5%), while there was no relationship between circulating ghrelin levels and IR in the high FBG group (>6.9 mmol/dl) (r = −0.15; 95% CI: −0.33 to 0.03, I2 = 0.0%). Publication bias was insignificant (Egger's test: P = 0.425). Conclusion: In obesity, circulating ghrelin levels were significantly negative correlated with insulin resistance in individuals with normal fasting blood glucose.
Collapse
Affiliation(s)
- Cai-Shun Zhang
- Department of Special Medicine, Medical College, Qingdao University, Qingdao, China
| | - Liu-Xin Wang
- Department of Special Medicine, Medical College, Qingdao University, Qingdao, China
| | - Rui Wang
- Department of Special Medicine, Medical College, Qingdao University, Qingdao, China
| | - Yuan Liu
- Department of Special Medicine, Medical College, Qingdao University, Qingdao, China
| | - Li-Min Song
- Department of Special Medicine, Medical College, Qingdao University, Qingdao, China
| | - Jun-Hua Yuan
- Department of Special Medicine, Medical College, Qingdao University, Qingdao, China
| | - Bin Wang
- Department of Special Medicine, Medical College, Qingdao University, Qingdao, China.,Department of Medical Microbiology, Medical College, Qingdao University, Qingdao, China
| | - Jing Dong
- Department of Special Medicine, Medical College, Qingdao University, Qingdao, China.,Department of Physiology, Medical College, Qingdao University, Qingdao, China
| |
Collapse
|
6
|
Stengel A, Taché Y. Gut-Brain Neuroendocrine Signaling Under Conditions of Stress-Focus on Food Intake-Regulatory Mediators. Front Endocrinol (Lausanne) 2018; 9:498. [PMID: 30210455 PMCID: PMC6122076 DOI: 10.3389/fendo.2018.00498] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 08/08/2018] [Indexed: 12/12/2022] Open
Abstract
The gut-brain axis represents a bidirectional communication route between the gut and the central nervous system comprised of neuronal as well as humoral signaling. This system plays an important role in the regulation of gastrointestinal as well as homeostatic functions such as hunger and satiety. Recent years also witnessed an increased knowledge on the modulation of this axis under conditions of exogenous or endogenous stressors. The present review will discuss the alterations of neuroendocrine gut-brain signaling under conditions of stress and the respective implications for the regulation of food intake.
Collapse
Affiliation(s)
- Andreas Stengel
- Department of Psychosomatic Medicine and Psychotherapy, University Hospital Tübingen, Tübingen, Germany
- Charité Center for Internal Medicine and Dermatology, Department for Psychosomatic Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Yvette Taché
- CURE/Digestive Diseases Research Center, Vatche and Tamar Manoukian Digestive Diseases Division, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- VA Greater Los Angeles Health Care System, Los Angeles, CA, United States
| |
Collapse
|
7
|
Improvement of Adipose Macrophage Polarization in High Fat Diet-Induced Obese GHSR Knockout Mice. BIOMED RESEARCH INTERNATIONAL 2018; 2018:4924325. [PMID: 30112394 PMCID: PMC6077514 DOI: 10.1155/2018/4924325] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 05/06/2018] [Accepted: 05/28/2018] [Indexed: 12/16/2022]
Abstract
Purpose Adipose tissue inflammation is the key linking obesity to insulin resistance. Over 50% of the interstitial cells in adipose tissue are macrophages, which produce inflammatory cytokines and therefore play an important role in the progression of insulin resistance. Within this classification view, macrophage biology is driven by two polarization phenotypes, M1 (proinflammatory) and M2 (anti-inflammatory). The unique functional receptor of ghrelin, growth hormone secretagogue receptor (GHSR), is a classic seven-transmembrane G protein-coupled receptor that is linked to multiple intracellular signaling pathways. Knockout of GHSR improves the obesity and glucose metabolic disorders, suggesting a crucial role of ghrelin activity in insulin resistance. Here, we discussed whether macrophage polarization phenotypes in adipose tissue were changed in GHSR knockout (GHSR-/-) mice. Methods GHSR-/- mice were fed with normal chow diet (NCD) or high fat diet (HFD). Markers of different macrophage polarization phenotypes were detected by real-time RT-PCR. Results The size of adipocytes decreased and interstitial cells, especially infiltrated macrophages, reduced in epididymal adipose tissue of GHSR-/- mice fed with HFD. Compared with wild type mice, the mRNA levels of inflammatory adipokines such as resistin, IL-6, and PAI-1 were significantly lower in epididymal adipose tissue of GHSR-/- mice, whereas anti-inflammatory adipokine, adiponectin, was significantly higher. M1 markers, MCP-1, TNF-α, and iNOS, were significantly lower in epididymal adipose tissue of GHSR-/- mice, whereas M2 markers, Arg-1, Mgl-1, were Mrc1, were significantly higher. Conclusion The GHSR-/- mice fed with HFD showed suppressed adipose inflammation, reduced macrophage infiltration, and enhanced M2 polarization of macrophages in adipose tissue, which improved insulin sensitivity.
Collapse
|
8
|
Okuhara Y, Kaiya H, Teraoka H, Kitazawa T. Structural determination, distribution, and physiological actions of ghrelin in the guinea pig. Peptides 2018; 99:70-81. [PMID: 29183755 DOI: 10.1016/j.peptides.2017.11.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Revised: 11/20/2017] [Accepted: 11/20/2017] [Indexed: 12/13/2022]
Abstract
We identified guinea pig ghrelin (gp-ghrelin), and examined its distribution and physiological actions in the guinea-pig. Gp-ghrelin is a 28-amino acid peptide (GASFR SPEHH SAQQR KESRK LPAKI QPR); seven amino acids are different from that of rat ghrelin at positions 2, 5, 10, 11, 19, 21, and 25, which include the conserved region known in mammals. The third serine residue is mainly modified by n-decanoyl acid. Both gp-ghrelin and rat ghrelin increased intracellular Ca2+ concentration of HEK293 cells expressing guinea pig growth hormone secretagogue receptor 1a (GHS-R1a), and the affinity of gp-ghrelin was slightly higher than that of rat ghrelin. In addition, gp-ghrelin was also effective in CHO cells expressing rat GHS-R1a with similar affinity to that of rat ghrelin. Gp-ghrelin mRNA was predominantly expressed in the stomach, whereas the expression levels in other organs was low. High levels of GHS-R1a mRNA expression were observed in the pituitary, medulla oblongata, and kidney, while medium levels were noted in the thalamus, pons, olfactory bulb, and heart. Immunohistochemistry identified gp-ghrelin-immunopositive cells in the gastric mucosa and pancreas. Intraperitoneal injection of gp-ghrelin increased food intake in the guinea pig. Gp-ghrelin did not cause any mechanical responses in isolated gastrointestinal smooth muscles in vitro, similar to rat ghrelin. In conclusion, the N-terminal structures that are conserved in mammals were different in gp-ghrelin. Moreover, the functional characteristics of gp-ghrelin, other than its distribution, were dissimilar from those in other Rodentia.
Collapse
Affiliation(s)
- Yuji Okuhara
- School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Hokkaido 069-8501, Japan; Pathology Research, Safety Research Laboratory, Kissei Pharmaceutical Co., Ltd., 2320-1, Maki, Hotaka, Azumino, Nagano 399-8305, Japan
| | - Hiroyuki Kaiya
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka 565-8565, Japan
| | - Hiroki Teraoka
- School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Hokkaido 069-8501, Japan
| | - Takio Kitazawa
- School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Hokkaido 069-8501, Japan.
| |
Collapse
|
9
|
Wang Q, Tang W, Rao WS, Song X, Shan CX, Zhang W. Changes of Ghrelin/GOAT axis and mTOR pathway in the hypothalamus after sleeve gastrectomy in obese type-2 diabetes rats. World J Gastroenterol 2017; 23:6231-6241. [PMID: 28974889 PMCID: PMC5603489 DOI: 10.3748/wjg.v23.i34.6231] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 05/02/2017] [Accepted: 06/01/2017] [Indexed: 02/06/2023] Open
Abstract
AIM To examine the changes of the ghrelin/ghrelin O-acyltransferase (GOAT) axis and the mammalian target of rapamycin (mTOR) pathway in the hypothalamus after sleeve gastrectomy. METHODS A total of 30 obese type-2 diabetes Sprague-Dawley (SD) rats, 6 wk of age, fed with high-sugar and high-fat fodder for 2 mo plus intraperitoneal injection of streptozotocin were randomly divided into three groups: non-operation group (S0 group, n = 10), sham operation group (Sh group, n = 10) and sleeve gastrectomy group (SG group, n = 10). Data of body mass, food intake, oral glucose tolerance test (OGTT), acylated ghrelin (AG) and total ghrelin (TG) were collected and measured at the first day (when the rats were 6 wk old), preoperative day 3 and postoperative week 8. The mRNA expression of preproghrelin, GOAT and neuropeptide Y (NPY), and protein expression of ghrelin, GOAT, GHSR and the mTOR pathway (p-Akt, p-mTOR and p-S6) were measured in the hypothalamus. RESULTS SG can significantly improve metabolic symptoms by reducing body mass and food intake. The obese rats showed lower serum TG levels and no change in AG, but the ratio of AG/TG was increased. When compared with the S0 and Sh groups, the SG group showed decreased TG (1482.03 ± 26.55, 1481.49 ± 23.30 and 1206.63 ± 52.02 ng/L, respectively, P < 0.05), but unchanged AG (153.06 ± 13.74, 155.37 ± 19.30 and 144.44 ± 16.689 ng/L, respectively, P > 0.05). As a result, the ratio of AG/TG further increased in the SG group (0.103 ± 0.009, 0.105 ± 0.013 and 0.12 ± 0.016, respectively, P < 0.05). When compared with the S0 group, SG suppressed mRNA and protein levels of preproghrelin (0.63 ± 0.12 vs 0.5 ± 0.11, P < 0.05) and GOAT (0.96 ± 0.09 vs 0.87 ± 0.08, P < 0.05), but did not change NPY mRNA expression (0.61 ± 0.04 vs 0.65 ± 0.07, P > 0.05) in the hypothalamus. The protein levels of p-Akt, p-mTOR and p-S6 were higher in the SG group, which indicated that the hypothalamic mTOR pathway was activated after SG at the postoperative week 8. CONCLUSION The reduction of ghrelin expression and activation of the mTOR pathway might have opposite effects on food intake, as SG improves obesity and T2DM.
Collapse
MESH Headings
- Acylation
- Acyltransferases/metabolism
- Animals
- Diabetes Mellitus, Experimental/blood
- Diabetes Mellitus, Experimental/chemically induced
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/surgery
- Diabetes Mellitus, Type 2/blood
- Diabetes Mellitus, Type 2/chemically induced
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/surgery
- Eating
- Gastrectomy/methods
- Gastroplasty/methods
- Ghrelin/metabolism
- Glucose Tolerance Test
- Humans
- Hypothalamus/metabolism
- Male
- Obesity/blood
- Obesity/complications
- Obesity/metabolism
- Obesity/surgery
- RNA, Messenger/metabolism
- Rats
- Rats, Sprague-Dawley
- Signal Transduction
- TOR Serine-Threonine Kinases/metabolism
- Weight Loss
Collapse
Affiliation(s)
- Qiang Wang
- Department of General Surgery, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Wei Tang
- Department of Surgery, University of Tokyo Hospital, Tokyo 113-8655, Japan
| | - Wen-Sheng Rao
- Department of General Surgery, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Xin Song
- Department of General Surgery, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Cheng-Xiang Shan
- Department of General Surgery, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Wei Zhang
- Department of General Surgery, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| |
Collapse
|
10
|
Characterization of Ghrelin O-Acyltransferase (GOAT) in goldfish (Carassius auratus). PLoS One 2017; 12:e0171874. [PMID: 28178327 PMCID: PMC5298278 DOI: 10.1371/journal.pone.0171874] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 01/26/2017] [Indexed: 12/21/2022] Open
Abstract
Ghrelin is the only known hormone posttranslationally modified with an acylation. This modification is crucial for most of ghrelin’s physiological effects and is catalyzed by the polytopic enzyme ghrelin O-acyltransferase (GOAT). The aim of this study was to characterize GOAT in a teleost model, goldfish (Carassius auratus). First, the full-length cDNA sequence was obtained by RT-PCR and rapid amplification of cDNA ends methods. Two highly homologous cDNAs of 1491 and 1413 bp, respectively, named goat-V1 and goat-V2 were identified. Deduced protein sequences (393 and 367 amino acids, respectively) are predicted to present 11 and 9 transmembrane regions, respectively, and both contain two conserved key residues proposed to be involved in catalysis: asparagine 273 and histidine 304. RT-qPCR revealed that both forms of goat mRNAs show a similar widespread tissue distribution, with the highest expression in the gastrointestinal tract and gonads and less but considerable expression in brain, pituitary, liver and adipose tissue. Immunostaining of intestinal sections showed the presence of GOAT immunoreactive cells in the intestinal mucosa, some of which colocalize with ghrelin. Using an in vitro approach, we observed that acylated ghrelin downregulates GOAT gene and protein levels in cultured intestine in a time-dependent manner. Finally, we found a rhythmic oscillation of goat mRNA expression in the hypothalamus, pituitary and intestinal bulb of goldfish fed at midday, but not at midnight. Together, these findings report novel data characterizing GOAT, and offer new information about the ghrelinergic system in fish.
Collapse
|
11
|
Hormaechea-Agulla D, Gómez-Gómez E, Ibáñez-Costa A, Carrasco-Valiente J, Rivero-Cortés E, L-López F, Pedraza-Arevalo S, Valero-Rosa J, Sánchez-Sánchez R, Ortega-Salas R, Moreno MM, Gahete MD, López-Miranda J, Requena MJ, Castaño JP, Luque RM. Ghrelin O-acyltransferase (GOAT) enzyme is overexpressed in prostate cancer, and its levels are associated with patient's metabolic status: Potential value as a non-invasive biomarker. Cancer Lett 2016; 383:125-134. [PMID: 27693462 DOI: 10.1016/j.canlet.2016.09.022] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2016] [Revised: 09/01/2016] [Accepted: 09/02/2016] [Indexed: 12/31/2022]
Abstract
Ghrelin-O-acyltransferase (GOAT) is the key enzyme regulating ghrelin activity, and has been proposed as a potential therapeutic target for obesity/diabetes and as a biomarker in some endocrine-related cancers. However, GOAT presence and putative role in prostate-cancer (PCa) is largely unknown. Here, we demonstrate, for the first time, that GOAT is overexpressed (mRNA/protein-level) in prostatic tissues (n = 52) and plasma/urine-samples (n = 85) of PCa-patients, compared with matched controls [healthy prostate tissues (n = 12) and plasma/urine-samples from BMI-matched controls (n = 28), respectively]. Interestingly, GOAT levels in PCa-patients correlated with aggressiveness and metabolic conditions (i.e. diabetes). Actually, GOAT expression was regulated by metabolic inputs (i.e. In1-ghrelin, insulin/IGF-I) in cultured normal prostate cells and PCa-cell lines. Importantly, ROC-curve analysis unveiled a valuable diagnostic potential for GOAT to discriminate PCa at the tissue/plasma/urine-level with high sensitivity/specificity, particularly in non-diabetic individuals. Moreover, we discovered that GOAT is secreted by PCa-cells, and that its levels are higher in urine samples from a stimulated post-massage vs. pre-massage prostate-test. In conclusion, plasmatic GOAT levels exhibit high specificity/sensitivity to predict PCa-presence compared with other PCa-biomarkers, especially in non-diabetic individuals, suggesting that GOAT holds potential as a novel non-invasive PCa-biomarker.
Collapse
Affiliation(s)
- Daniel Hormaechea-Agulla
- Maimonides Institute of Biomedical Research of Córdoba (IMIBIC), Córdoba, Spain; Department of Cell Biology, Physiology and Immunology, University of Córdoba, Córdoba, Spain; Hospital Universitario Reina Sofía (HURS), Córdoba, Spain; CIBERobn, Córdoba, Spain; ceiA3, Córdoba, Spain
| | - Enrique Gómez-Gómez
- Maimonides Institute of Biomedical Research of Córdoba (IMIBIC), Córdoba, Spain; Hospital Universitario Reina Sofía (HURS), Córdoba, Spain; Urology Service, HURS, Córdoba, Spain
| | - Alejandro Ibáñez-Costa
- Maimonides Institute of Biomedical Research of Córdoba (IMIBIC), Córdoba, Spain; Department of Cell Biology, Physiology and Immunology, University of Córdoba, Córdoba, Spain; Hospital Universitario Reina Sofía (HURS), Córdoba, Spain; CIBERobn, Córdoba, Spain; ceiA3, Córdoba, Spain
| | - Julia Carrasco-Valiente
- Maimonides Institute of Biomedical Research of Córdoba (IMIBIC), Córdoba, Spain; Hospital Universitario Reina Sofía (HURS), Córdoba, Spain; Urology Service, HURS, Córdoba, Spain
| | - Esther Rivero-Cortés
- Maimonides Institute of Biomedical Research of Córdoba (IMIBIC), Córdoba, Spain; Department of Cell Biology, Physiology and Immunology, University of Córdoba, Córdoba, Spain; Hospital Universitario Reina Sofía (HURS), Córdoba, Spain; CIBERobn, Córdoba, Spain; ceiA3, Córdoba, Spain
| | - Fernando L-López
- Maimonides Institute of Biomedical Research of Córdoba (IMIBIC), Córdoba, Spain; Department of Cell Biology, Physiology and Immunology, University of Córdoba, Córdoba, Spain; Hospital Universitario Reina Sofía (HURS), Córdoba, Spain; CIBERobn, Córdoba, Spain; ceiA3, Córdoba, Spain
| | - Sergio Pedraza-Arevalo
- Maimonides Institute of Biomedical Research of Córdoba (IMIBIC), Córdoba, Spain; Department of Cell Biology, Physiology and Immunology, University of Córdoba, Córdoba, Spain; Hospital Universitario Reina Sofía (HURS), Córdoba, Spain; CIBERobn, Córdoba, Spain; ceiA3, Córdoba, Spain
| | - José Valero-Rosa
- Maimonides Institute of Biomedical Research of Córdoba (IMIBIC), Córdoba, Spain; Hospital Universitario Reina Sofía (HURS), Córdoba, Spain; Urology Service, HURS, Córdoba, Spain
| | - Rafael Sánchez-Sánchez
- Maimonides Institute of Biomedical Research of Córdoba (IMIBIC), Córdoba, Spain; Hospital Universitario Reina Sofía (HURS), Córdoba, Spain; Anatomical Pathology Service, HURS, Córdoba, Spain
| | - Rosa Ortega-Salas
- Maimonides Institute of Biomedical Research of Córdoba (IMIBIC), Córdoba, Spain; Hospital Universitario Reina Sofía (HURS), Córdoba, Spain; Anatomical Pathology Service, HURS, Córdoba, Spain
| | - María M Moreno
- Maimonides Institute of Biomedical Research of Córdoba (IMIBIC), Córdoba, Spain; Hospital Universitario Reina Sofía (HURS), Córdoba, Spain; Anatomical Pathology Service, HURS, Córdoba, Spain
| | - Manuel D Gahete
- Maimonides Institute of Biomedical Research of Córdoba (IMIBIC), Córdoba, Spain; Department of Cell Biology, Physiology and Immunology, University of Córdoba, Córdoba, Spain; Hospital Universitario Reina Sofía (HURS), Córdoba, Spain; CIBERobn, Córdoba, Spain; ceiA3, Córdoba, Spain
| | - José López-Miranda
- Maimonides Institute of Biomedical Research of Córdoba (IMIBIC), Córdoba, Spain; Hospital Universitario Reina Sofía (HURS), Córdoba, Spain; CIBERobn, Córdoba, Spain; Lipids and Atherosclerosis Unit, HURS, Córdoba, Spain
| | - María J Requena
- Maimonides Institute of Biomedical Research of Córdoba (IMIBIC), Córdoba, Spain; Hospital Universitario Reina Sofía (HURS), Córdoba, Spain; Urology Service, HURS, Córdoba, Spain
| | - Justo P Castaño
- Maimonides Institute of Biomedical Research of Córdoba (IMIBIC), Córdoba, Spain; Department of Cell Biology, Physiology and Immunology, University of Córdoba, Córdoba, Spain; Hospital Universitario Reina Sofía (HURS), Córdoba, Spain; CIBERobn, Córdoba, Spain; ceiA3, Córdoba, Spain.
| | - Raúl M Luque
- Maimonides Institute of Biomedical Research of Córdoba (IMIBIC), Córdoba, Spain; Department of Cell Biology, Physiology and Immunology, University of Córdoba, Córdoba, Spain; Hospital Universitario Reina Sofía (HURS), Córdoba, Spain; CIBERobn, Córdoba, Spain; ceiA3, Córdoba, Spain.
| |
Collapse
|
12
|
Tong J, Davis HW, Gastaldelli A, D'Alessio D. Ghrelin Impairs Prandial Glucose Tolerance and Insulin Secretion in Healthy Humans Despite Increasing GLP-1. J Clin Endocrinol Metab 2016; 101:2405-14. [PMID: 27055279 PMCID: PMC4891805 DOI: 10.1210/jc.2015-4154] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
OBJECTIVES Administration of ghrelin inhibits the acute insulin response to glucose and worsens IV glucose tolerance in healthy subjects. Evidence from preclinical studies suggests that ghrelin may have differential effects on glucose metabolism during fasting and feeding. Our objective was to test the effects of ghrelin on glucose and insulin responses during a meal tolerance test. DESIGN Acyl ghrelin (0.26 and 2.0 μg/kg/h) or saline was infused in 13 healthy subjects on three separate occasions in randomized order. Ghrelin was infused for 45 minutes to achieve steady-state levels and continued for 240 minutes after ingestion of a liquid test meal. Primary outcomes were area under the curve for glucose and insulin secretion. RESULTS We found that ghrelin infusions of 0.26 and 2.0 μg/kg/h raised steady-state plasma total ghrelin levels to 1.7- and 4.8-fold above fasting concentrations, but did not alter fasting plasma glucose or insulin levels. During the meal tolerance test, ghrelin decreased insulin sensitivity, impaired β-cell function, and induced glucose intolerance. The high-dose ghrelin infusion also raised postprandial glucagon like peptide 1 secretion without affecting glucose dependent insulinotropic polypeptide, glucagon, or peptide YY concentrations. CONCLUSIONS We conclude that both physiologic and pharmacologic doses of ghrelin worsen the glucose and β-cell responses to meal ingestion in healthy humans. The increase in postprandial glucagon like peptide 1 secretion by ghrelin suggests a novel enteroendocrine connection, but does not mitigate the glucose intolerance.
Collapse
Affiliation(s)
- Jenny Tong
- Division of Endocrinology, Diabetes and Metabolism (J.T., H.W.D., D.D.), Department of Medicine, University of Cincinnati, Cincinnati, Ohio; Institute of Clinical Physiology (A.G.), National Research Council, Pisa, Italy; Cincinnati VA Medical Center (D.D.), Cincinnati, Ohio
| | - Harold W Davis
- Division of Endocrinology, Diabetes and Metabolism (J.T., H.W.D., D.D.), Department of Medicine, University of Cincinnati, Cincinnati, Ohio; Institute of Clinical Physiology (A.G.), National Research Council, Pisa, Italy; Cincinnati VA Medical Center (D.D.), Cincinnati, Ohio
| | - Amalia Gastaldelli
- Division of Endocrinology, Diabetes and Metabolism (J.T., H.W.D., D.D.), Department of Medicine, University of Cincinnati, Cincinnati, Ohio; Institute of Clinical Physiology (A.G.), National Research Council, Pisa, Italy; Cincinnati VA Medical Center (D.D.), Cincinnati, Ohio
| | - David D'Alessio
- Division of Endocrinology, Diabetes and Metabolism (J.T., H.W.D., D.D.), Department of Medicine, University of Cincinnati, Cincinnati, Ohio; Institute of Clinical Physiology (A.G.), National Research Council, Pisa, Italy; Cincinnati VA Medical Center (D.D.), Cincinnati, Ohio
| |
Collapse
|
13
|
Blanco AM, Sánchez-Bretaño A, Delgado MJ, Valenciano AI. Brain Mapping of Ghrelin O-Acyltransferase in Goldfish (Carassius Auratus): Novel Roles for the Ghrelinergic System in Fish? Anat Rec (Hoboken) 2016; 299:748-58. [PMID: 27064922 DOI: 10.1002/ar.23346] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 02/16/2016] [Accepted: 02/22/2016] [Indexed: 12/19/2022]
Abstract
Ghrelin O-acyltransferase (GOAT) is the enzyme responsible for acylation of ghrelin, a gut-brain hormone with important roles in many physiological functions in vertebrates. Many aspects of GOAT remain to be elucidated, especially in fish, and particularly its anatomical distribution within the different brain areas has never been reported to date. The present study aimed to characterize the brain mapping of GOAT using RT-qPCR and immunohistochemistry in a teleost, the goldfish (Carassius auratus). Results show that goat transcripts are expressed in different brain areas of the goldfish, with the highest levels in the vagal lobe. Using immunohistochemistry, we also report the presence of GOAT immunoreactive cells in different encephalic areas, including the telencephalon, some hypothalamic nuclei, pineal gland, optic tectum and cerebellum, although they are especially abundant in the hindbrain. Particularly, an important signal is observed in the vagal lobe and some fiber tracts of the brainstem, such as the medial longitudinal fasciculus, Mauthneri fasciculus, secondary gustatory tract and spinothalamic tract. Most of the forebrain areas where GOAT is detected, particularly the hypothalamic nuclei, also express the ghs-r1a ghrelin receptor and other appetite-regulating hormones (e.g., orexin and NPY), supporting the role of ghrelin as a modulator of food intake and energy balance in fish. Present results are the first report on the presence of GOAT in the brain using imaging techniques. The high presence of GOAT in the hindbrain is a novelty, and point to possible new functions for the ghrelinergic system in fish. Anat Rec, 299:748-758, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Ayelén M Blanco
- Department of Animal Physiology II, Faculty of Biology, Complutense University, Madrid, 28040, Spain
| | - Aída Sánchez-Bretaño
- Department of Animal Physiology II, Faculty of Biology, Complutense University, Madrid, 28040, Spain
| | - María J Delgado
- Department of Animal Physiology II, Faculty of Biology, Complutense University, Madrid, 28040, Spain
| | - Ana I Valenciano
- Department of Animal Physiology II, Faculty of Biology, Complutense University, Madrid, 28040, Spain
| |
Collapse
|
14
|
Mohan H, Gasner M, Ramesh N, Unniappan S. Ghrelin, ghrelin-O-acyl transferase, nucleobindin-2/nesfatin-1 and prohormone convertases in the pancreatic islets of Sprague Dawley rats during development. J Mol Histol 2016; 47:325-36. [DOI: 10.1007/s10735-016-9673-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2015] [Accepted: 03/29/2016] [Indexed: 12/18/2022]
|
15
|
Li Z, Mulholland M, Zhang W. Ghrelin O-acyltransferase (GOAT) and energy metabolism. SCIENCE CHINA-LIFE SCIENCES 2016; 59:281-91. [PMID: 26732975 DOI: 10.1007/s11427-015-4973-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 10/09/2015] [Indexed: 12/16/2022]
Abstract
Ghrelin O-acyltransferase (GOAT), a member of MBOATs family, is essential for octanoylation of ghrelin, which is required for active ghrelin to bind with and activate its receptor. GOAT is expressed mainly in the stomach, pancreas and hypothalamus. Levels of GOAT are altered by energy status. GOAT contains 11 transmembrane helices and one reentrant loop. Its invariant residue His-338 and conserved Asn-307 are located in the endoplasmic reticulum lumen and cytosol respectively. GOAT contributes to the regulation of food intake and energy expenditure, as well as glucose and lipids homeostasis. Deletion of GOAT blocks the acylation of ghrelin leading to subsequent impairment in energy homeostasis and survival when mice are challenged with high energy diet or severe caloric restriction. GO-CoA-Tat, a peptide GOAT inhibitor, attenuates acyl-ghrelin production and prevents weight gain induced by a medium-chain triglycerides-rich high fat diet. Further, GO-CoA-Tat increases glucose- induced insulin secretion. Overall, inhibition of GOAT is a novel strategy for treatment of obesity and related metabolic disorders.
Collapse
Affiliation(s)
- Ziru Li
- Department of Surgery, University of Michigan Medical Center, Ann Arbor, MI, 48109-0346, USA
| | - Michael Mulholland
- Department of Surgery, University of Michigan Medical Center, Ann Arbor, MI, 48109-0346, USA.
| | - Weizhen Zhang
- Department of Surgery, University of Michigan Medical Center, Ann Arbor, MI, 48109-0346, USA. .,Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, 100191, China.
| |
Collapse
|
16
|
Benso A, Gramaglia E, Olivetti I, Tomelini M, Belcastro S, Calvi E, Dotta A, St-Pierre D, Ghigo E, Broglio F. Acute effects of acylated ghrelin on salbutamol-induced metabolic actions in humans. Endocrine 2015; 48:937-41. [PMID: 25012253 DOI: 10.1007/s12020-014-0343-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Accepted: 06/18/2014] [Indexed: 10/25/2022]
Abstract
The aim of this study is to describe a potential modulatory effect of acute acylated ghrelin (AG) administration on the glucose, insulin, and free fatty acids (FFA) responses to salbutamol (SALBU). Six healthy young male volunteers underwent the following four testing sessions in random order at least 7 days apart: (a) acute AG administration (1.0 μg/kg i.v. as bolus at 0'); (b) SALBU infusion (0.06 μg/kg/min i.v. from -15' to +45'); (c) SALBU infusion+AG; and (d) isotonic saline infusion. Blood samples for glucose, insulin, and FFA levels were collected every 15 min. As expected, with respect to saline, SALBU infusion induced a remarkable increase in glucose (10.8±5.6 mmol/l×min; P<0.05), insulin (2436.8±556.9 pmol/l×min; P<0.05), and FFA (18.9±4.5 mmol/l×min; P<0.01) levels. A significant increase in glucose (7.4±3.9 mmol/l×min; P<0.05) and FFA levels (10.0±2.8 mmol/l×min; P<0.01) without significant variations in insulin levels were recorded after AG administration. Interestingly, the hyperglycemic effect of AG appeared to be significantly potentiated during SALBU infusion (26.7±4.8 mmol/l×min; P<0.05). On the other hand, the stimulatory effect of SALBU on insulin and FFA was not significantly modified by AG administration. The results of this study show that acute AG administration has a synergic effect with β2-adrenergic receptor activation by SALBU on blood glucose increase, suggesting that their pharmacological hyperglycemic action takes place via different mechanisms. On the other hand, AG has a negligible influence on the other pharmacological metabolic effects of SALBU infusion.
Collapse
Affiliation(s)
- A Benso
- Division of Endocrinology, Diabetology and Metabolism, Department of Medical Sciences, University of Turin, A.O. Città della Salute e della Scienza - Molinette, Corso Dogliotti 14, 10126, Turin, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Khatib MN, Gaidhane S, Gaidhane AM, Simkhada P, Zahiruddin QS. Ghrelin O Acyl Transferase (GOAT) as a Novel Metabolic Regulatory Enzyme. J Clin Diagn Res 2015; 9:LE01-5. [PMID: 25859472 PMCID: PMC4378754 DOI: 10.7860/jcdr/2015/9787.5514] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Accepted: 12/06/2014] [Indexed: 01/12/2023]
Abstract
BACKGROUND Obesity and Type 2 Diabetes Mellitus (T2DM) presents a growing threat to the global health. Evidences highlight an important role of ghrelin as a key regulator of glucose metabolism. The physiological functions of ghrelin are mediated by enzyme ghrelin-O-acyltransferase (GOAT) which is capable of generating the active form of this metabolic hormone. However, its exact mechanism of action and influence on energy balance and glucose metabolism is yet to be explored. OBJECTIVES To review the physiological role of GOAT in the regulation of energy balance and glucose metabolism and explore the potential therapeutic avenues of modulators of GOAT to counter the progression of obesity and T2DM. METHODS Publications were sought through electronic searches. The bibliographies of all papers, book, chapters and editorials were scanned and hand searches were also conducted for journals, and conference proceedings. CONCLUSION GOAT peptide modulates the insulin secretion as well as insulin sensitivity. Modulators of GOAT signaling like inhibitors of GOAT increases insulin secretion, enhance peripheral insulin sensitivity and thus counters obesity and T2DM. Modulators of GOAT can be a probable therapy for modifying food intake and for countering obesity and T2DM.
Collapse
Affiliation(s)
- Mahalaqua Nazli Khatib
- Professor, Department of Physiology, JN Medical College, Datta Meghe Institute of Medical Sciences, Wardha, Maharashtra, India
| | - Shilpa Gaidhane
- Associate Professor, Department of Medicine, JN Medical College, Datta Meghe Institute of Medical Sciences, Wardha, Maharashtra, India
| | - Abhay M. Gaidhane
- Professor, Department of Community Medicine, JN Medical College, Datta Meghe Institute of Medical Sciences, Wardha, Maharashtra, India
| | - Padam Simkhada
- Senior Lecturer in International Health ScHARR, University of Sheffield, UK and Centre for public Health Liverpool Johns Moores University, Liverpool, UK
| | - Quazi Syed Zahiruddin
- Professor, Department of Community Medicine, JN Medical College, Datta Meghe Institute of Medical Sciences, Wardha, Maharashtra, India
| |
Collapse
|
18
|
Yada T, Damdindorj B, Rita RS, Kurashina T, Ando A, Taguchi M, Koizumi M, Sone H, Nakata M, Kakei M, Dezaki K. Ghrelin signalling in β-cells regulates insulin secretion and blood glucose. Diabetes Obes Metab 2014; 16 Suppl 1:111-7. [PMID: 25200304 DOI: 10.1111/dom.12344] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2014] [Accepted: 04/20/2014] [Indexed: 12/13/2022]
Abstract
Insulin secretion from pancreatic islet β-cells is stimulated by glucose. Glucose-induced insulin release is potentiated or suppressed by hormones and neural substances. Ghrelin, an acylated 28-amino acid peptide, was isolated from the stomach in 1999 as the endogenous ligand for the growth hormone (GH) secretagogue-receptor (GHS-R). Circulating ghrelin is produced predominantly in the stomach and to a lesser extent in the intestine, pancreas and brain. Ghrelin, initially identified as a potent stimulator of GH release and feeding, has been shown to suppress glucose-induced insulin release. This insulinostatic action is mediated by Gα(i2) subtype of GTP-binding proteins and delayed outward K⁺ (Kv) channels. Interestingly, ghrelin is produced in pancreatic islets. The ghrelin originating from islets restricts insulin release and thereby upwardly regulates the systemic glucose level. Furthermore, blockade or elimination of ghrelin enhances insulin release, which can ameliorate glucose intolerance in high-fat diet fed mice and ob/ob mice. This review focuses on the insulinostatic action of ghrelin, its signal transduction mechanisms in islet β-cells, ghrelin's status as an islet hormone, physiological roles of ghrelin in regulating systemic insulin levels and glycaemia, and therapeutic potential of the ghrelin-GHS-R system as the target to treat type 2 diabetes.
Collapse
Affiliation(s)
- T Yada
- Division of Integrative Physiology, Department of Physiology, Jichi Medical University School of Medicine, Tochigi, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Cameron KO, Bhattacharya SK, Loomis AK. Small Molecule Ghrelin Receptor Inverse Agonists and Antagonists. J Med Chem 2014; 57:8671-91. [DOI: 10.1021/jm5003183] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- Kimberly O. Cameron
- Worldwide
Medicinal Chemistry, Pfizer Worldwide Research and Development, 610
Main Street, Cambridge, Massachusetts 02139, United States
| | - Samit K. Bhattacharya
- Worldwide
Medicinal Chemistry, Pfizer Worldwide Research and Development, 610
Main Street, Cambridge, Massachusetts 02139, United States
| | - A. Katrina Loomis
- Pharmatherapeutics
Precision Medicine, Pfizer Worldwide Research and Development, Eastern
Point Road, Groton, Connecticut 06340, United States
| |
Collapse
|
20
|
Seim I, Jeffery PL, de Amorim L, Walpole CM, Fung J, Whiteside EJ, Lourie R, Herington AC, Chopin LK. Ghrelin O-acyltransferase (GOAT) is expressed in prostate cancer tissues and cell lines and expression is differentially regulated in vitro by ghrelin. Reprod Biol Endocrinol 2013; 11:70. [PMID: 23879975 PMCID: PMC3724588 DOI: 10.1186/1477-7827-11-70] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Accepted: 07/05/2013] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Ghrelin is a 28 amino acid peptide hormone that is expressed in the stomach and a range of peripheral tissues, where it frequently acts as an autocrine/paracrine growth factor. Ghrelin is modified by a unique acylation required for it to activate its cognate receptor, the growth hormone secretagogue receptor (GHSR), which mediates many of the actions of ghrelin. Recently, the enzyme responsible for adding the fatty acid residue (octanoyl/acyl group) to the third amino acid of ghrelin, GOAT (ghrelin O-acyltransferase), was identified. METHODS We used cell culture, quantitative real-time reverse transcription (RT)-PCR and immunohistochemistry to demonstrate the expression of GOAT in prostate cancer cell lines and tissues from patients. Real-time RT-PCR was used to demonstrate the expression of prohormone convertase (PC)1/3, PC2 and furin in prostate cancer cell lines. Prostate-derived cell lines were treated with ghrelin and desacyl ghrelin and the effect on GOAT expression was measured using quantitative RT-PCR. RESULTS We have demonstrated that GOAT mRNA and protein are expressed in the normal prostate and human prostate cancer tissue samples. The RWPE-1 and RWPE-2 normal prostate-derived cell lines and the LNCaP, DU145, and PC3 prostate cancer cell lines express GOAT and at least one other enzyme that is necessary to produce mature, acylated ghrelin from proghrelin (PC1/3, PC2 or furin). Finally, ghrelin, but not desacyl ghrelin (unacylated ghrelin), can directly regulate the expression of GOAT in the RWPE-1 normal prostate derived cell line and the PC3 prostate cancer cell line. Ghrelin treatment (100nM) for 6 hours significantly decreased GOAT mRNA expression two-fold (P < 0.05) in the PC3 prostate cancer cell line, however, ghrelin did not regulate GOAT expression in the DU145 and LNCaP prostate cancer cell lines. CONCLUSIONS This study demonstrates that GOAT is expressed in prostate cancer specimens and cell lines. Ghrelin regulates GOAT expression, however, this is likely to be cell-type specific. The expression of GOAT in prostate cancer supports the hypothesis that the ghrelin axis has autocrine/paracrine roles. We propose that the RWPE-1 prostate cell line and the PC3 prostate cancer cell line may be useful for investigating GOAT regulation and function.
Collapse
Affiliation(s)
- Inge Seim
- Ghrelin Research Group, Translational Research Institute - Institute of Health and Biomedical Innovation, Queensland University of Technology, 37 Kent St, Woolloongabba, Queensland, 4102, Australia
- Australian Prostate Cancer Research Centre, Queensland, Princess Alexandra Hospital, 199 Ipswich Road, Brisbane, Queensland, 4102, Australia
| | - Penny L Jeffery
- Ghrelin Research Group, Translational Research Institute - Institute of Health and Biomedical Innovation, Queensland University of Technology, 37 Kent St, Woolloongabba, Queensland, 4102, Australia
- Australian Prostate Cancer Research Centre, Queensland, Princess Alexandra Hospital, 199 Ipswich Road, Brisbane, Queensland, 4102, Australia
- Mater Medical Research Institute, Mater Health Services, University of Queensland, South Brisbane, Queensland,, 4103, Australia
| | - Laura de Amorim
- Ghrelin Research Group, Translational Research Institute - Institute of Health and Biomedical Innovation, Queensland University of Technology, 37 Kent St, Woolloongabba, Queensland, 4102, Australia
| | - Carina M Walpole
- Ghrelin Research Group, Translational Research Institute - Institute of Health and Biomedical Innovation, Queensland University of Technology, 37 Kent St, Woolloongabba, Queensland, 4102, Australia
| | - Jenny Fung
- Ghrelin Research Group, Translational Research Institute - Institute of Health and Biomedical Innovation, Queensland University of Technology, 37 Kent St, Woolloongabba, Queensland, 4102, Australia
| | - Eliza J Whiteside
- Ghrelin Research Group, Translational Research Institute - Institute of Health and Biomedical Innovation, Queensland University of Technology, 37 Kent St, Woolloongabba, Queensland, 4102, Australia
| | - Rohan Lourie
- Mater Medical Research Institute, Mater Health Services, University of Queensland, South Brisbane, Queensland,, 4103, Australia
- Department of Pathology, Mater Health Services, South Brisbane, Queensland, 4103, Australia
| | - Adrian C Herington
- Ghrelin Research Group, Translational Research Institute - Institute of Health and Biomedical Innovation, Queensland University of Technology, 37 Kent St, Woolloongabba, Queensland, 4102, Australia
- Australian Prostate Cancer Research Centre, Queensland, Princess Alexandra Hospital, 199 Ipswich Road, Brisbane, Queensland, 4102, Australia
| | - Lisa K Chopin
- Ghrelin Research Group, Translational Research Institute - Institute of Health and Biomedical Innovation, Queensland University of Technology, 37 Kent St, Woolloongabba, Queensland, 4102, Australia
- Australian Prostate Cancer Research Centre, Queensland, Princess Alexandra Hospital, 199 Ipswich Road, Brisbane, Queensland, 4102, Australia
| |
Collapse
|
21
|
Yagi T, Asakawa A, Ueda H, Miyawaki S, Inui A. The role of ghrelin in patients with functional dyspepsia and its potential clinical relevance (Review). Int J Mol Med 2013; 32:523-31. [PMID: 23778458 DOI: 10.3892/ijmm.2013.1418] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Accepted: 06/03/2013] [Indexed: 12/31/2022] Open
Abstract
Functional dyspepsia (FD) is a functional gastrointestinal disorder (FGID). According to the Rome III consensus, FD is divided into 2 subgroups: epigastric pain syndrome (EPS) and postprandial distress syndrome (PDS). Although multiple mechanisms of FD pathogenesis have been suggested, its underlying etiology and pharmacological therapy remain unclear. Ghrelin is a gut-derived peptide found in the stomach. It plays a role in the regulation of gastric motility and appetite. The ghrelin gene encodes 3 molecular forms, acyl ghrelin, des-acyl ghrelin and obestatin. Acyl ghrelin acts as an endogenous ligand for growth hormone secretagogue receptor; furthermore, it is orexigenic, with effects on food intake, energy homeostasis and gastrointestinal motility. Des-acyl ghrelin exerts an opposite effect to acyl ghrelin. Obestatin exerts an inhibitory effect on the motor activity of the antrum and duodenum in fed animals. These peptides exert differential effects on gut motility and food intake. The therapeutic potential of ghrelin has attracted attention due to its varied bioactivities. Certain studies have shown that total ghrelin levels are significantly lower in patients with FD compared with healthy volunteers and that the acyl ghrelin levels of patients with FD are higher compared with healthy volunteers. However, a recent study demonstrated that acyl ghrelin levels in patients with PDS were lower compared with healthy volunteers; the association between FD and other ghrelin family gene products also remains unclear. Although certain studies have demonstrated the beneficial effects of acyl ghrelin administration and its agonist in patients with FD, only a few clinical reports exist. Further studies are required in order to examine the effects of ghrelin on FD.
Collapse
Affiliation(s)
- Takakazu Yagi
- Department of Orthodontics and Dentofacial Orthopedics, Medical and Dental Hospital, Kagoshima University, Kagoshima 890-8544, Japan
| | | | | | | | | |
Collapse
|
22
|
Tong J, Prigeon RL, Davis HW, Bidlingmaier M, Tschöp MH, D'Alessio D. Physiologic concentrations of exogenously infused ghrelin reduces insulin secretion without affecting insulin sensitivity in healthy humans. J Clin Endocrinol Metab 2013; 98:2536-43. [PMID: 23589527 PMCID: PMC3667259 DOI: 10.1210/jc.2012-4162] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Infusion of ghrelin to supraphysiologic levels inhibits glucose-stimulated insulin secretion, reduces insulin sensitivity, and worsens glucose tolerance in humans. OBJECTIVE The purpose of this study was to determine the effects of lower doses of ghrelin on insulin secretion and insulin sensitivity in healthy men and women. METHODS Acyl ghrelin (0.2 and 0.6 nmol kg(-1) h(-1)) or saline was infused for 225 minutes in 16 healthy subjects on 3 separate occasions in randomized order. An i.v. glucose tolerance test was performed, and the insulin sensitivity index (SI) was derived from the minimal model. Insulin secretion was measured as the acute insulin response to glucose (AIRg) and the disposition index was computed as AIRg × SI. RESULTS Ghrelin infusions at 0.2 and 0.6 nmol kg(-1) h(-1) raised steady-state plasma total ghrelin levels 2.2- and 6.1-fold above fasting concentrations. Neither dose of ghrelin altered fasting plasma insulin, glucose, or SI, but both doses reduced insulin secretion compared with the saline control, computed either as AIRg (384 ± 75 and 354 ± 65 vs 520 ± 110 pM · min [mean ± SEM], respectively; P < .01 for both low- and high-dose vs saline) or disposition index (2238 ± 421 and 2067 ± 396 vs 3339 ± 705, respectively; P < .02 for both comparisons). The high-dose ghrelin infusion also decreased glucose tolerance. CONCLUSIONS Ghrelin infused to levels occurring in physiologic states such as starvation decreases insulin secretion without affecting insulin sensitivity. These findings are consistent with a role for endogenous ghrelin in the regulation of insulin secretion and suggest that ghrelin antagonism could improve β-cell function.
Collapse
Affiliation(s)
- Jenny Tong
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Cincinnati, Cincinnati, Ohio 45267, USA.
| | | | | | | | | | | |
Collapse
|
23
|
Chanclón B, Luque RM, Córdoba-Chacón J, Gahete MD, Pozo-Salas AI, Castaño JP, Gracia-Navarro F, Martínez-Fuentes AJ. Role of endogenous cortistatin in the regulation of ghrelin system expression at pancreatic level under normal and obese conditions. PLoS One 2013; 8:e57834. [PMID: 23469081 PMCID: PMC3585174 DOI: 10.1371/journal.pone.0057834] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2012] [Accepted: 01/26/2013] [Indexed: 12/11/2022] Open
Abstract
Ghrelin-system components [native ghrelin, In1-ghrelin, Ghrelin-O-acyltransferase enzyme (GOAT) and receptors (GHS-Rs)] are expressed in a wide variety of tissues, including the pancreas, where they exert different biological actions including regulation of neuroendocrine secretions, food intake and pancreatic function. The expression of ghrelin system is regulated by metabolic conditions (fasting/obesity) and is associated with the progression of obesity and insulin resistance. Cortistatin (CORT), a neuropeptide able to activate GHS-R, has emerged as an additional link in gut-brain interplay. Indeed, we recently reported that male CORT deficient mice (cort−/−) are insulin-resistant and present a clear dysregulation in the stomach ghrelin-system. The present work was focused at analyzing the expression pattern of ghrelin-system components at pancreas level in cort−/− mice and their control littermates (cort +/+) under low- or high-fat diet. Our data reveal that all the ghrelin-system components are expressed at the mouse pancreatic level, where, interestingly, In1-ghrelin was expressed at higher levels than native-ghrelin. Thus, GOAT mRNA levels were significantly lower in cort−/− mice compared with controls while native ghrelin, In1-ghrelin and GHS-R transcript levels remained unaltered under normal metabolic conditions. Moreover, under obese condition, a significant increase in pancreatic expression of native-ghrelin, In1-ghrelin and GHS-R was observed in obese cort+/+ but not in cort−/− mice. Interestingly, insulin expression and release was elevated in obese cort+/+, while these changes were not observed in obese cort−/− mice. Altogether, our results indicate that the ghrelin-system expression is clearly regulated in the pancreas of cort+/+ and cort −/− under normal and/or obesity conditions suggesting that this system may play relevant roles in the endocrine pancreas. Most importantly, our data demonstrate, for the first time, that endogenous CORT is essential for the obesity-induced changes in insulin expression/secretion observed in mice, suggesting that CORT is a key regulatory component of the pancreatic function.
Collapse
Affiliation(s)
- Belén Chanclón
- Department of Cell Biology, Physiology and Immunology, University of Cordoba and Reina Sofia University Hospital, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), and CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain
| | - Raúl M. Luque
- Department of Cell Biology, Physiology and Immunology, University of Cordoba and Reina Sofia University Hospital, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), and CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain
| | - José Córdoba-Chacón
- Department of Cell Biology, Physiology and Immunology, University of Cordoba and Reina Sofia University Hospital, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), and CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain
| | - Manuel D. Gahete
- Department of Cell Biology, Physiology and Immunology, University of Cordoba and Reina Sofia University Hospital, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), and CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain
| | - Ana I. Pozo-Salas
- Department of Cell Biology, Physiology and Immunology, University of Cordoba and Reina Sofia University Hospital, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), and CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain
| | - Justo P. Castaño
- Department of Cell Biology, Physiology and Immunology, University of Cordoba and Reina Sofia University Hospital, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), and CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain
| | - Francisco Gracia-Navarro
- Department of Cell Biology, Physiology and Immunology, University of Cordoba and Reina Sofia University Hospital, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), and CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain
| | - Antonio J. Martínez-Fuentes
- Department of Cell Biology, Physiology and Immunology, University of Cordoba and Reina Sofia University Hospital, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), and CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain
- * E-mail:
| |
Collapse
|
24
|
Rodríguez A, Gómez-Ambrosi J, Catalán V, Rotellar F, Valentí V, Silva C, Mugueta C, Pulido MR, Vázquez R, Salvador J, Malagón MM, Colina I, Frühbeck G. The ghrelin O-acyltransferase-ghrelin system reduces TNF-α-induced apoptosis and autophagy in human visceral adipocytes. Diabetologia 2012; 55:3038-50. [PMID: 22869322 DOI: 10.1007/s00125-012-2671-5] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Accepted: 06/25/2012] [Indexed: 12/19/2022]
Abstract
AIMS/HYPOTHESIS Proinflammatory and proapoptotic cytokines such as TNF-α are upregulated in human obesity. We evaluated the association between ghrelin isoforms (acylated and desacyl ghrelin) and TNF-α in obesity and obesity-associated type 2 diabetes, as well as the potential role of ghrelin in the control of apoptosis and autophagy in human adipocytes. METHODS Plasma concentrations of the ghrelin isoforms and TNF-α were measured in 194 participants. Ghrelin and ghrelin O-acyltransferase (GOAT) levels were analysed by western-blot, immunohistochemistry and real-time PCR in 53 biopsies of human omental adipose tissue. We also determined the effect of acylated and desacyl ghrelin (10 to 1,000 pmol/l) on TNF-α-induced apoptosis and autophagy-related molecules in omental adipocytes. RESULTS Circulating concentrations of acylated ghrelin and TNF-α were increased, whereas desacyl ghrelin levels were decreased in obesity-associated type 2 diabetes. Ghrelin and GOAT were produced in omental and subcutaneous adipose tissue. Visceral adipose tissue from obese patients with type 2 diabetes showed higher levels of GOAT, increased adipocyte apoptosis and increased expression of the autophagy-related genes ATG5, BECN1 and ATG7. In differentiating human omental adipocytes, incubation with acylated and desacyl ghrelin reduced TNF-α-induced activation of caspase-8 and caspase-3, and cell death. In addition, acylated ghrelin reduced the basal expression of the autophagy-related genes ATG5 and ATG7, while desacyl ghrelin inhibited the TNF-α-induced increase of ATG5, BECN1 and ATG7 expression. CONCLUSIONS/INTERPRETATION Apoptosis and autophagy are upregulated in human visceral adipose tissue of patients with type 2 diabetes. Acylated and desacyl ghrelin reduce TNF-α-induced apoptosis and autophagy in human visceral adipocytes.
Collapse
Affiliation(s)
- A Rodríguez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Vörös K, Prohászka Z, Kaszás E, Alliquander A, Terebesy A, Horváth F, Janik L, Sima A, Forrai J, Cseh K, Kalabay L. Serum ghrelin level and TNF-α/ghrelin ratio in patients with previous myocardial infarction. Arch Med Res 2012; 43:548-54. [PMID: 23079033 DOI: 10.1016/j.arcmed.2012.09.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Accepted: 09/06/2012] [Indexed: 01/23/2023]
Abstract
BACKGROUND AND AIMS Studies investigating serum ghrelin level in atherosclerosis yielded contradictory results. Interaction of ghrelin with adipocytokines is obscure in cardiovascular diseases. We undertook this study to determine which molecules influence ghrelin level and to see whether post-myocardial infarction (MI) patients have decreased ghrelin levels. METHODS In this cross-sectional study, acyl-ghrelin concentration was determined by radioimmunoassay in sera of 171 patients (ages 62 ± 6 years, mean ± SD) with previous MI and 81 age-matched referent subjects. We evaluated the associations of ghrelin with insulin, adiponectin, leptin, resistin, fetuin-A and tumor necrosis factor-alpha (TNF-α). RESULTS Patients had lower ghrelin levels compared to referent subjects (240.55 ± 59.33 vs. 337.96 ± 30.75 pg/mL, p <0.001) even after excluding diabetic and obese patients (240.63 ± 54.08 vs. 337.96 ± 30.75, p <0.001). In multivariate analysis, insulin (β = -0.327, p <0.001) and adiponectin (β = 0.301, p <0.001) determined ghrelin level (R(2) = 0.199, p <0.001). There was no association between ghrelin and TNF-α levels. In discriminant analysis using ghrelin, adiponectin, leptin, fetuin-A, resistin and TNF-α, the structure matrix revealed ghrelin and TNF-α as strongest predictors for belonging to the patient group (0.760 and -0.569, respectively). Using these two parameters, 89.7% of cases were correctly classified. Subjects with high TNF-α/ghrelin ratio had 11.25 times higher chance for belonging to the patient group (95% CI 5.80-21.80; χ(2) (1) = 215.6, p <0.001) CONCLUSIONS Acylated ghrelin levels are decreased in patients with coronary atherosclerosis, independently of body weight and the presence of type 2 diabetes mellitus. Ghrelin level is determined by elevated insulin and decreased adiponectin levels. Ghrelin alone or in combination with TNF-α may prove to be a novel indicator of coronary atherosclerosis.
Collapse
Affiliation(s)
- Krisztián Vörös
- Department of Family Medicine, Semmelweis University, Budapest, Hungary.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Damdindorj B, Dezaki K, Kurashina T, Sone H, Rita R, Kakei M, Yada T. Exogenous and endogenous ghrelin counteracts GLP-1 action to stimulate cAMP signaling and insulin secretion in islet β-cells. FEBS Lett 2012; 586:2555-62. [PMID: 22750144 DOI: 10.1016/j.febslet.2012.06.034] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Revised: 06/20/2012] [Accepted: 06/21/2012] [Indexed: 10/28/2022]
Abstract
We studied interactive effects of insulinotropic GLP-1 and insulinostatic ghrelin on rat pancreatic islets. GLP-1 potentiated glucose-induced insulin release and cAMP production in isolated islets and [Ca(2+)](i) increases in single β-cells, and these potentiations were attenuated by ghrelin. Ghrelin suppressed [Ca(2+)](i) responses to an adenylate cyclase activator forskolin. Moreover, GLP-1-induced insulin release and cAMP production were markedly enhanced by [D-lys(3)]-GHRP-6, a ghrelin receptor antagonist, in isolated islets. These results indicate that both exogenous and endogenous islet-derived ghrelin counteracts glucose-dependent GLP-1 action to increase cAMP production, [Ca(2+)](i) and insulin release in islet β-cells, positioning ghrelin as a modulator of insulinotropic GLP-1.
Collapse
Affiliation(s)
- Boldbaatar Damdindorj
- Division of Integrative Physiology, Department of Physiology, Jichi Medical University School of Medicine, Yakushiji 3311-1, Shimotsuke, Tochigi 329-0498, Japan
| | | | | | | | | | | | | |
Collapse
|
27
|
Xu G, Wang Z, Li Y, Li Z, Tang H, Zhao J, Xiang X, Ding L, Ma L, Yuan F, Fei J, Wang W, Wang N, Guan Y, Tang C, Mulholland M, Zhang W. Ghrelin contributes to derangements of glucose metabolism induced by rapamycin in mice. Diabetologia 2012; 55:1813-23. [PMID: 22391948 PMCID: PMC3496261 DOI: 10.1007/s00125-012-2509-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2011] [Accepted: 01/16/2012] [Indexed: 02/06/2023]
Abstract
AIMS/HYPOTHESIS Rapamycin impairs glucose tolerance and insulin sensitivity. Our previous study demonstrated that rapamycin significantly increases the production of gastric ghrelin, which is critical in the regulation of glucose metabolism. Here, we investigated whether ghrelin contributes to derangements of glucose metabolism induced by rapamycin. METHODS The effects of rapamycin on glucose metabolism were examined in mice receiving ghrelin receptor antagonist or with Ghsr1a gene knockout. Changes in GLUT4, c-Jun N-terminal kinase (JNK) and phosphorylated ribosomal protein S6 (pS6) were investigated by immunofluorescent staining or western blotting. Related hormones were detected by radioimmunoassay kits. RESULTS Rapamycin impaired glucose metabolism and insulin sensitivity not only in normal C57BL/6J mice but also in both obese mice induced by a high fat diet and db/db mice. This was accompanied by elevation of plasma acylated ghrelin. Rapamycin significantly increased the levels of plasma acylated ghrelin in normal C57BL/6J mice, high-fat-diet-induced obese mice and db/db mice. Elevation in plasma acylated ghrelin and derangements of glucose metabolism upon administration of rapamycin were significantly correlated. The deterioration in glucose homeostasis induced by rapamycin was blocked by D: -Lys3-GHRP-6, a ghrelin receptor antagonist, or by deletion of the Ghsr1a gene. Ghrelin receptor antagonism and Ghsr1a knockout blocked the upregulation of JNK activity and downregulation of GLUT4 levels and translocation in the gastrocnemius muscle induced by rapamycin. CONCLUSIONS/INTERPRETATION The current study demonstrates that ghrelin contributes to derangements of glucose metabolism induced by rapamycin via altering the content and translocation of GLUT4 in muscles.
Collapse
Affiliation(s)
- G Xu
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing 100191, China
| | - Z Wang
- Department of General Surgery, The Affiliated Sixth Hospital of Medical School, Shanghai Jiaotong University, Shanghai 200233, China
| | - Y Li
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing 100191, China
| | - Z Li
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing 100191, China
| | - H Tang
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing 100191, China
| | - J Zhao
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing 100191, China
| | - X Xiang
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing 100191, China
| | - L Ding
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing 100191, China
| | - L Ma
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing 100191, China
| | - F Yuan
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing 100191, China
| | - J Fei
- School of Life Science and Technology, Tongji University, Shanghai, China, Shanghai 200092, China
| | - W Wang
- School of Life Science and Technology, Tongji University, Shanghai, China, Shanghai 200092, China
| | - N Wang
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing 100191, China
| | - Y Guan
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing 100191, China
| | - C Tang
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing 100191, China
| | - M Mulholland
- Department of Surgery, University of Michigan Medical Center, Ann Arbor, MI 48109-0346, USA
- Corresponding author: Weizhen Zhang, Department of Physiology and Pathophysiology, Peking University Health Science Center, No 38, Xueyuan Rd, Haidian District, Beijing 100191, China, Tel: 0086-10-82802183; Fax: 0086-10-82802183; Or Michael W. Mulholland, Department of Surgery, University of Michigan Medical Center, 1500 W Medical Center Dr. Ann Arbor, MI 48109-0346, USA, Tel: 734-936-3236;
| | - W Zhang
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing 100191, China
- Department of Surgery, University of Michigan Medical Center, Ann Arbor, MI 48109-0346, USA
- Corresponding author: Weizhen Zhang, Department of Physiology and Pathophysiology, Peking University Health Science Center, No 38, Xueyuan Rd, Haidian District, Beijing 100191, China, Tel: 0086-10-82802183; Fax: 0086-10-82802183; Or Michael W. Mulholland, Department of Surgery, University of Michigan Medical Center, 1500 W Medical Center Dr. Ann Arbor, MI 48109-0346, USA, Tel: 734-936-3236;
| |
Collapse
|
28
|
Beléen C, Martínez-Fuentes AJ, Gracia-Navarro F. Role of SST, CORT and ghrelin and its receptors at the endocrine pancreas. Front Endocrinol (Lausanne) 2012; 3:114. [PMID: 23162532 PMCID: PMC3444847 DOI: 10.3389/fendo.2012.00114] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Accepted: 09/03/2012] [Indexed: 12/21/2022] Open
Abstract
Somatostatin (SST), cortistatin (CORT), and its receptors (sst1-5), and ghrelin and its receptors (GHS-R) are two highly interrelated neuropeptide systems with a broad range of overlapping biological actions at central, cardiovascular, and immune levels among others. Besides their potent regulatory role on GH release, its endocrine actions are highlighted by SST/CORT and ghrelin influence on insulin secretion, glucose homeostasis, and insulin resistance. Interestingly, most components of these systems are expressed at the endocrine pancreas and are actively involved in the modulation of pancreatic islet function and, consequently influence glucose homeostasis. In addition, some of them also participate in islet survival and regeneration. Furthermore, under severe metabolic condition as well as in endocrine pathologies, their expression profile is severely deregulated. These findings suggest that SST/CORT and ghrelin systems could play a relevant role in pancreatic function under metabolic and endocrine pathologies. Accordingly, these systems have been therapeutically targeted for the prevention or amelioration of certain metabolic conditions (obesity) as well as for tumor growth inhibition and/or hormonal regulation in endocrine pathologies (neuroendocrine tumors). This review focuses on the interrelationship between SST/CORT and ghrelin systems and their role in severe metabolic conditions and some endocrine disorders.
Collapse
Affiliation(s)
- Chanclón Beléen
- Department of Cell Biology, Physiology and Immunology, University of CórdobaCórdoba, Spain
- Instituto Maimónides de Investigación Biomédica de CórdobaCórdoba, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y NutriciónCórdoba, Spain
| | - Antonio J. Martínez-Fuentes
- Department of Cell Biology, Physiology and Immunology, University of CórdobaCórdoba, Spain
- Instituto Maimónides de Investigación Biomédica de CórdobaCórdoba, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y NutriciónCórdoba, Spain
| | - Francisco Gracia-Navarro
- Department of Cell Biology, Physiology and Immunology, University of CórdobaCórdoba, Spain
- Instituto Maimónides de Investigación Biomédica de CórdobaCórdoba, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y NutriciónCórdoba, Spain
- *Correspondence: Francisco Gracia-Navarro, Department of Cell Biology, Physiology and Immunology, University of Córdoba, Campus de Rabanales, Edificio Severo-Ochoa, Planta 3, E-14014 Córdoba, Spain. e-mail:
| |
Collapse
|
29
|
Abstract
Ghrelin, an acylated 28-amino acid peptide, was isolated from the stomach, where circulating ghrelin is produced predominantly. In addition to its unique role in regulating growth-hormone release, mealtime hunger, lipid metabolism, and the cardiovascular system, ghrelin is involved in the regulation of glucose metabolism. Ghrelin is expressed in pancreatic islets and released into pancreatic microcirculations. Ghrelin inhibits insulin release in mice, rats, and humans. Pharmacological and genetic blockades of islet-derived ghrelin markedly augment glucose-induced insulin release. The signal transduction mechanisms of ghrelin in islet β-cells are very unique, being distinct from those utilized for growth-hormone release. Ghrelin attenuates the glucose-induced cAMP production and PKA activation, which drives activation of Kv channels and suppression of the glucose-induced [Ca(2+)](i) increase and insulin release in β-cells. Insulinostatic function of the ghrelin-GHS-R system in islets is a potential therapeutic target for type 2 diabetes.
Collapse
Affiliation(s)
- Katsuya Dezaki
- Division of Integrative Physiology, Department of Physiology, Jichi Medical University School of Medicine, Shimotsuke, Japan.
| | | |
Collapse
|
30
|
Al Massadi O, Tschöp MH, Tong J. Ghrelin acylation and metabolic control. Peptides 2011; 32:2301-8. [PMID: 21893140 DOI: 10.1016/j.peptides.2011.08.020] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2011] [Revised: 08/19/2011] [Accepted: 08/22/2011] [Indexed: 01/26/2023]
Abstract
Since its discovery, many physiologic functions have been ascribed to ghrelin, a gut derived hormone. The presence of a median fatty acid side chain on the ghrelin peptide is required for the binding and activation of the classical ghrelin receptor, the growth hormone secretagogue receptor (GHSR)-1a. Ghrelin O-acyl transferase (GOAT) was recently discovered as the enzyme responsible for this acylation process. GOAT is expressed in all tissues that have been found to express ghrelin and has demonstrated actions on several complex endocrine organ systems such as the hypothalamus-pituitary-gonadal, insular and adrenal axis as well as the gastrointestinal (GI) tract, bone and gustatory system. Ghrelin acylation is dependent on the function of GOAT and the availability of substrates such as proghrelin and short- to medium-chain fatty acids (MCFAs). This process is governed by GOAT activity and has been shown to be modified by dietary lipids. In this review, we provided evidence that support an important role of GOAT in the regulation of energy homeostasis and glucose metabolism by modulating acyl ghrelin (AG) production. The relevance of GOAT and AG during periods of starvation remains to be defined. In addition, we summarized the recent literature on the metabolic effects of GOAT specific inhibitors and shared our view on the potential of targeting GOAT for the treatment of metabolic disorders such as obesity and type 2 diabetes.
Collapse
Affiliation(s)
- O Al Massadi
- Division of Endocrinology, Department of Medicine, Metabolic Diseases Institute, University of Cincinnati, Cincinnati, OH 45237, USA.
| | | | | |
Collapse
|
31
|
Kang K, Zmuda E, Sleeman MW. Physiological role of ghrelin as revealed by the ghrelin and GOAT knockout mice. Peptides 2011; 32:2236-41. [PMID: 21600256 DOI: 10.1016/j.peptides.2011.04.028] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2011] [Revised: 03/26/2011] [Accepted: 04/29/2011] [Indexed: 11/27/2022]
Abstract
Ghrelin is a gastric hormone that has been shown to regulate food intake and energy metabolism. One unique feature of ghrelin is that its activity is regulated post transcriptionally by ghrelin O-acyltransferase (GOAT) through the addition of fatty acid to the serine residue in the N terminal region. Despite much biochemical characterization, to date no other proteins have been shown to be specifically octonylated by GOAT, suggesting a unique matching of the acyl transferase for a single ligand, ghrelin. If this is indeed correct, then genetic deletion of ghrelin or GOAT should produce near identical phenotypes and there should be extensive overlap in expression patterns. This review summarizes the similarities and differences in the phenotypes with the genetic deletion of ghrelin and GOAT in the various knockout mouse lines reported to date. While there is considerable overlap in expression pattern between ghrelin and GOAT, the latter does exhibit some unique tissue expression that could suggest that additional peptides may be acylated and await discovery and characterization.
Collapse
Affiliation(s)
- Kihwa Kang
- Regeneron Pharmaceuticals, Inc, 777 Old Saw Mill River Road, Tarrytown, NY 10591, USA
| | | | | |
Collapse
|
32
|
Delhanty PJD, van der Lely AJ. Ghrelin and glucose homeostasis. Peptides 2011; 32:2309-18. [PMID: 21396419 DOI: 10.1016/j.peptides.2011.03.001] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2011] [Revised: 03/01/2011] [Accepted: 03/01/2011] [Indexed: 12/25/2022]
Abstract
Ghrelin plays an important physiological role in modulating GH secretion, insulin secretion and glucose metabolism. Ghrelin has direct effects on pancreatic islet function. Also, ghrelin is part of a mechanism that integrates the physiological response to fasting. However, pharmacologic studies indicate the important obesogenic/diabetogenic properties of ghrelin. This is very likely of physiological relevance, deriving from a requirement to protect against seasonal periods of food scarcity by building energy reserves, predominantly in the form of fat. Available data indicate the potential of ghrelin blockade as a means to prevent its diabetogenic effects. Several studies indicate a negative correlation between ghrelin levels and the incidence of type 2 diabetes and insulin resistance. However, it is unclear if low ghrelin levels are a risk factor or a compensatory response. Direct antagonism of the receptor does not always have the desired effects, however, since it can cause increased body weight gain. Pharmacological suppression of the ghrelin/des-acyl ghrelin ratio by treatment with des-acyl ghrelin may also be a viable alternative approach which appears to improve insulin sensitivity. A promising recently developed approach appears to be through the blockade of GOAT activity, although the longer term effects of this treatment remain to be investigated.
Collapse
Affiliation(s)
- P J D Delhanty
- Department of Internal Medicine, Erasmus MC, 3000 CA Rotterdam, The Netherlands.
| | | |
Collapse
|
33
|
Stengel A, Wang L, Taché Y. Stress-related alterations of acyl and desacyl ghrelin circulating levels: mechanisms and functional implications. Peptides 2011; 32:2208-17. [PMID: 21782868 PMCID: PMC3220774 DOI: 10.1016/j.peptides.2011.07.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2011] [Revised: 07/04/2011] [Accepted: 07/05/2011] [Indexed: 12/11/2022]
Abstract
Ghrelin is the only known peripherally produced and centrally acting peptide hormone that stimulates food intake and digestive functions. Ghrelin circulates as acylated and desacylated forms and recently the acylating enzyme, ghrelin-O-acyltransferase (GOAT) and the de-acylating enzyme, thioesterase 1/lysophospholipase 1 have been identified adding new layers of complexity to the regulation of ghrelin. Stress is known to alter gastrointestinal motility and food intake and was recently shown to modify circulating ghrelin and GOAT levels with differential responses related to the type of stressors including a reduction induced by physical stressors (abdominal surgery and immunological/endotoxin injection, exercise) and elevation by metabolic (cold exposure, acute fasting and caloric restriction) and psychological stressors. However, the pathways underlying the alterations of ghrelin under these various stress conditions are still largely to be defined and may relate to stress-associated autonomic changes. There is evidence that alterations of circulating ghrelin may contribute to the neuroendocrine and behavioral responses along with sustaining the energetic requirement needed upon repeated exposure to stressors. A better understanding of these mechanisms will allow targeting components of ghrelin signaling that may improve food intake and gastric motility alterations induced by stress.
Collapse
Affiliation(s)
- Andreas Stengel
- CURE: Digestive Diseases Research Center, David Geffen School of Medicine, University of California Los Angeles and Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA 90073, United States.
| | | | | |
Collapse
|
34
|
Ghrelin o-acyl transferase: bridging ghrelin and energy homeostasis. INTERNATIONAL JOURNAL OF PEPTIDES 2011; 2011:217957. [PMID: 21941572 PMCID: PMC3175403 DOI: 10.1155/2011/217957] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/03/2011] [Accepted: 07/20/2011] [Indexed: 11/30/2022]
Abstract
Ghrelin O-acyl transferase (GOAT) is a recently identified enzyme responsible for the unique n-acyl modification of ghrelin, a multifunctional metabolic hormone. GOAT structure and activity appears to be conserved from fish to man. Since the acyl modification is critical for most of the biological actions of ghrelin, especially metabolic functions, GOAT emerged as a very important molecule of interest. The research on GOAT is on the rise, and several important results reiterating its significance have been reported. Notable among these discoveries are the identification of GOAT tissue expression patterns, effects on insulin secretion, blood glucose levels, feeding, body weight, and metabolism. Several attempts have been made to design and test synthetic compounds that can modulate endogenous GOAT, which could turn beneficial in favorably regulating whole body energy homeostasis. This paper will focus to provide an update on recent advances in GOAT research and its broader implications in the regulation of energy balance.
Collapse
|
35
|
Dezaki K, Damdindorj B, Sone H, Dyachok O, Tengholm A, Gylfe E, Kurashina T, Yoshida M, Kakei M, Yada T. Ghrelin attenuates cAMP-PKA signaling to evoke insulinostatic cascade in islet β-cells. Diabetes 2011; 60:2315-24. [PMID: 21788571 PMCID: PMC3161328 DOI: 10.2337/db11-0368] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE Ghrelin reportedly restricts insulin release in islet β-cells via the Gα(i2) subtype of G-proteins and thereby regulates glucose homeostasis. This study explored whether ghrelin regulates cAMP signaling and whether this regulation induces insulinostatic cascade in islet β-cells. RESEARCH DESIGN AND METHODS Insulin release was measured in rat perfused pancreas and isolated islets and cAMP production in isolated islets. Cytosolic cAMP concentrations ([cAMP](i)) were monitored in mouse MIN6 cells using evanescent-wave fluorescence imaging. In rat single β-cells, cytosolic protein kinase-A activity ([PKA](i)) and Ca(2+) concentration ([Ca(2+)](i)) were measured by DR-II and fura-2 microfluorometry, respectively, and whole cell currents by patch-clamp technique. RESULTS Ghrelin suppressed glucose (8.3 mmol/L)-induced insulin release in rat perfused pancreas and isolated islets, and these effects of ghrelin were blunted in the presence of cAMP analogs or adenylate cyclase inhibitor. Glucose-induced cAMP production in isolated islets was attenuated by ghrelin and enhanced by ghrelin receptor antagonist and anti-ghrelin antiserum, which counteract endogenous islet-derived ghrelin. Ghrelin inhibited the glucose-induced [cAMP](i) elevation and [PKA](i) activation in MIN6 and rat β-cells, respectively. Furthermore, ghrelin potentiated voltage-dependent K(+) (Kv) channel currents without altering Ca(2+) channel currents and attenuated glucose-induced [Ca(2+)](i) increases in rat β-cells in a PKA-dependent manner. CONCLUSIONS Ghrelin directly interacts with islet β-cells to attenuate glucose-induced cAMP production and PKA activation, which lead to activation of Kv channels and suppression of glucose-induced [Ca(2+)](i) increase and insulin release.
Collapse
Affiliation(s)
- Katsuya Dezaki
- Division of Integrative Physiology, Department of Physiology, Jichi Medical University School of Medicine, Tochigi, Japan
- Corresponding authors: Katsuya Dezaki, , and Toshihiko Yada,
| | - Boldbaatar Damdindorj
- Division of Integrative Physiology, Department of Physiology, Jichi Medical University School of Medicine, Tochigi, Japan
| | - Hideyuki Sone
- Division of Integrative Physiology, Department of Physiology, Jichi Medical University School of Medicine, Tochigi, Japan
| | - Oleg Dyachok
- Department of Medical Cell Biology, Uppsala University, Biomedical Centre, Uppsala, Sweden
| | - Anders Tengholm
- Department of Medical Cell Biology, Uppsala University, Biomedical Centre, Uppsala, Sweden
| | - Erik Gylfe
- Department of Medical Cell Biology, Uppsala University, Biomedical Centre, Uppsala, Sweden
| | - Tomoyuki Kurashina
- Division of Integrative Physiology, Department of Physiology, Jichi Medical University School of Medicine, Tochigi, Japan
| | - Masashi Yoshida
- First Department of Comprehensive Medicine, Saitama Medical Center, Jichi Medical University School of Medicine, Saitama, Japan
| | - Masafumi Kakei
- First Department of Comprehensive Medicine, Saitama Medical Center, Jichi Medical University School of Medicine, Saitama, Japan
| | - Toshihiko Yada
- Division of Integrative Physiology, Department of Physiology, Jichi Medical University School of Medicine, Tochigi, Japan
- Department of Developmental Physiology, Division of Adaptation Development, National Institute for Physiological Sciences, Aichi, Japan
- Corresponding authors: Katsuya Dezaki, , and Toshihiko Yada,
| |
Collapse
|
36
|
Gurriarán-Rodríguez U, Al-Massadi O, Crujeiras AB, Mosteiro CS, Amil-Diz M, Beiroa D, Nogueiras R, Seoane LM, Gallego R, Pazos Y, Casanueva FF, Camiña JP. Preproghrelin expression is a key target for insulin action on adipogenesis. J Endocrinol 2011; 210:R1-7. [PMID: 21690170 DOI: 10.1530/joe-11-0233] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
This study aimed to investigate the role of preproghrelin-derived peptides in adipogenesis. Immunocytochemical analysis of 3T3-L1 adipocyte cells showed stronger preproghrelin expression compared with that observed in 3T3-L1 preadipocyte cells. Insulin promoted this expression throughout adipogenesis identifying mTORC1 as a critical downstream substrate for this profile. The role of preproghrelin-derived peptides on the differentiation process was supported by preproghrelin knockdown experiments, which revealed its contribution to adipogenesis. Neutralization of endogenous O-acyl ghrelin (acylated ghrelin), unacylated ghrelin, and obestatin by specific antibodies supported their adipogenic potential. Furthermore, a parallel increase in the expression of ghrelin-associated enzymatic machinery, prohormone convertase 1/3 (PC1/3) and membrane-bound O-acyltransferase 4 (MBOAT4), was dependent on the expression of preproghrelin in the course of insulin-induced adipogenesis. The coexpression of preproghrelin system and their receptors, GHSR1a and GPR39, during adipogenesis supports an autocrine/paracrine role for these peptides. Preproghrelin, PC1/3, and MBOAT4 exhibited dissimilar expression depending on the white fat depot, revealing their regulation in a positive energy balance situation in mice. The results underscore a key role for preproghrelin-derived peptides on adipogenesis through an autocrine/paracrine mechanism.
Collapse
Affiliation(s)
- Uxía Gurriarán-Rodríguez
- Área de Endocrinología Molecular y Celular, Instituto de Investigación Sanitaria de Santiago (IDIS), Hospital Clínico Universitario de Santiago, Servicio Gallego de Salud (SERGAS), Santiago de Compostela, Spain
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Diet-induced obesity suppresses ghrelin in rat gastrointestinal tract and serum. Mol Cell Biochem 2011; 355:299-308. [DOI: 10.1007/s11010-011-0867-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2011] [Accepted: 04/28/2011] [Indexed: 12/14/2022]
|