1
|
Kehagias D, Georgopoulos N, Habeos I, Lampropoulos C, Mulita F, Kehagias I. The role of the gastric fundus in glycemic control. Hormones (Athens) 2023; 22:151-163. [PMID: 36705877 DOI: 10.1007/s42000-023-00429-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 01/10/2023] [Indexed: 01/28/2023]
Abstract
PURPOSE Ghrelin, one of the most studied gut hormones, is mainly produced by the gastric fundus. Abundant evidence exists from preclinical and clinical studies underlining its contribution to glucose regulation. In the following narrative review, the role of the gastric fundus in glucose regulation is summarized and we investigate whether its resection enhances glycemic control. METHODS An electronic search was conducted in the PubMed® database and in Google Scholar® using a combination of medical subject headings (MeSH). We examined types of metabolic surgery, including, in particular, gastric fundus resection, either as part of laparoscopic sleeve gastrectomy (LSG) or modified laparoscopic gastric bypass with fundus resection (LRYGBP + FR), and the contribution of ghrelin reduction to glucose regulation. RESULTS Fourteen human studies were judged to be eligible and included in this narrative review. Reduction of ghrelin levels after fundus resection might be related to early glycemic improvement before significant weight loss is achieved. Long-term data regarding the role of ghrelin reduction in glucose homeostasis are sparse. CONCLUSION The exact role of ghrelin in achieving glycemic control is still ambiguous. Data from human studies reveal a potential contribution of ghrelin reduction to early glycemic improvement, although further well-designed studies are needed.
Collapse
Affiliation(s)
- Dimitrios Kehagias
- Department of General Surgery, General University Hospital of Patras, 26504, Rio, Greece.
| | - Neoklis Georgopoulos
- Division of Reproductive Endocrinology, Department of Obstetrics and Gynecology, University of Patras Medical School, 26504, Rio, Greece
| | - Ioannis Habeos
- Division of Endocrinology and Diabetes, Department of Internal Medicine, University Hospital of Patras, 26504, Rio, Greece
| | | | - Francesk Mulita
- Department of General Surgery, General University Hospital of Patras, 26504, Rio, Greece
| | - Ioannis Kehagias
- Division of Bariatric and Metabolic Surgery, Department of Surgery, General University Hospital of Patras, 26504, Rio, Greece
| |
Collapse
|
2
|
Protective and Healing Effects of Ghrelin and Risk of Cancer in the Digestive System. Int J Mol Sci 2021; 22:ijms221910571. [PMID: 34638910 PMCID: PMC8509076 DOI: 10.3390/ijms221910571] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 09/24/2021] [Accepted: 09/27/2021] [Indexed: 01/19/2023] Open
Abstract
Ghrelin is an endogenous ligand for the ghrelin receptor, previously known as the growth hormone secretagogue receptor. This hormone is mainly produced by endocrine cells present in the gastric mucosa. The ghrelin-producing cells are also present in other organs of the body, mainly in the digestive system, but in much smaller amount. Ghrelin exhibits a broad spectrum of physiological effects, such as stimulation of growth hormone secretion, gastric secretion, gastrointestinal motility, and food intake, as well as regulation of glucose homeostasis and bone formation, and inhibition of inflammatory processes. This review summarizes the recent findings concerning animal and human data showing protective and therapeutic effects of ghrelin in the gut, and also presents the role of growth hormone and insulin-like growth factor-1 in these effects. In addition, the current data on the possible influence of ghrelin on the carcinogenesis, its importance in predicting the risk of developing gastrointestinal malignances, as well as the potential usefulness of ghrelin in the treatment of cancer, have been presented.
Collapse
|
3
|
Mathur N, Mehdi SF, Anipindi M, Aziz M, Khan SA, Kondakindi H, Lowell B, Wang P, Roth J. Ghrelin as an Anti-Sepsis Peptide: Review. Front Immunol 2021; 11:610363. [PMID: 33584688 PMCID: PMC7876230 DOI: 10.3389/fimmu.2020.610363] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 12/09/2020] [Indexed: 12/15/2022] Open
Abstract
Sepsis continues to produce widespread inflammation, illness, and death, prompting intensive research aimed at uncovering causes and therapies. In this article, we focus on ghrelin, an endogenous peptide with promise as a potent anti-inflammatory agent. Ghrelin was discovered, tracked, and isolated from stomach cells based on its ability to stimulate release of growth hormone. It also stimulates appetite and is shown to be anti-inflammatory in a wide range of tissues. The anti-inflammatory effects mediated by ghrelin are a result of both the stimulation of anti-inflammatory processes and an inhibition of pro-inflammatory forces. Anti-inflammatory processes are promoted in a broad range of tissues including the hypothalamus and vagus nerve as well as in a broad range of immune cells. Aged rodents have reduced levels of growth hormone (GH) and diminished immune responses; ghrelin administration boosts GH levels and immune response. The anti-inflammatory functions of ghrelin, well displayed in preclinical animal models of sepsis, are just being charted in patients, with expectations that ghrelin and growth hormone might improve outcomes in patients with sepsis.
Collapse
Affiliation(s)
- Nimisha Mathur
- Laboratory of Diabetes, Obesity, and Other Metabolic Disorders, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
| | - Syed F. Mehdi
- Laboratory of Diabetes, Obesity, and Other Metabolic Disorders, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
| | - Manasa Anipindi
- Laboratory of Diabetes, Obesity, and Other Metabolic Disorders, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
| | - Monowar Aziz
- Laboratory of Diabetes, Obesity, and Other Metabolic Disorders, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
| | - Sawleha A. Khan
- Laboratory of Diabetes, Obesity, and Other Metabolic Disorders, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
| | - Hema Kondakindi
- Laboratory of Diabetes, Obesity, and Other Metabolic Disorders, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
| | - Barbara Lowell
- Laboratory of Diabetes, Obesity, and Other Metabolic Disorders, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
| | - Ping Wang
- Laboratory of Diabetes, Obesity, and Other Metabolic Disorders, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
| | - Jesse Roth
- Laboratory of Diabetes, Obesity, and Other Metabolic Disorders, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
| |
Collapse
|
4
|
Intermittent Fasting Aggravates Lupus Nephritis through Increasing Survival and Autophagy of Antibody Secreting Cells in MRL/lpr Mice. Int J Mol Sci 2020; 21:ijms21228477. [PMID: 33187196 PMCID: PMC7696283 DOI: 10.3390/ijms21228477] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 11/05/2020] [Accepted: 11/09/2020] [Indexed: 11/17/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease in which the main contributors to organ damage are antibodies against autoantigens, such as double-stranded DNA (dsDNA). Calorie restriction and intermittent fasting (IF) have been shown to improve autoimmune disease symptoms in patients and animal models. Here, we tested the hypothesis that IF might improve symptoms in MRL/lpr mice, which spontaneously develop an SLE-like disease. Groups of mice were fed every other day (IF) or provided food ad libitum (controls), and various lupus-associated clinicopathological parameters were analyzed for up to 28 weeks. Contrary to expectations, anti-dsDNA antibody levels, immune complex deposition in the kidney, and glomerular injury were higher in the IF group than the control group, although there were no differences in spleen and lymph node weights between groups. Proteinuria was also worsened in the IF group. IF also increased the abundance of B cells, plasmablasts, and plasma cells and elevated autophagy in plasma cells in the spleen and lymph nodes. Secretion of anti-dsDNA antibody by splenocytes in vitro was reduced by chloroquine-induced inhibition of autophagy. These results suggest that IF exacerbates lupus nephritis in MRL/lpr mice by increasing autoantibody immune complex formation.
Collapse
|
5
|
Blanco AM, Cortés R, Bertucci JI, Soletto L, Sánchez E, Valenciano AI, Cerdá-Reverter JM, Delgado MJ. Brain transcriptome profile after CRISPR-induced ghrelin mutations in zebrafish. FISH PHYSIOLOGY AND BIOCHEMISTRY 2020; 46:1-21. [PMID: 31673996 DOI: 10.1007/s10695-019-00687-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 07/17/2019] [Indexed: 06/10/2023]
Abstract
Ghrelin (GRL) is a gut-brain hormone with a role in a wide variety of physiological functions in mammals and fish, which points out the ghrelinergic system as a key element for the appropriate biological functioning of the organism. However, many aspects of the multifunctional nature of GRL remain to be better explored, especially in fish. In this study, we used the CRISPR/Cas9 genome editing technique to generate F0 zebrafish in which the expression of grl is compromised. Then, we employed high-throughput mRNA sequencing (RNA-seq) to explore changes in the brain transcriptome landscape associated with the silencing of grl. The CRISPR/Cas9 technique successfully edited the genome of F0 zebrafish resulting in individuals with considerably lower levels of GRL mRNAs and protein and ghrelin O-acyl transferase (goat) mRNAs in the brain, intestine, and liver compared to wild-type (WT) zebrafish. Analysis of brain transcriptome revealed a total of 1360 differentially expressed genes (DEGs) between the grl knockdown (KD) and WT zebrafish, with 664 up- and 696 downregulated DEGs in the KD group. Functional enrichment analysis revealed that DEGs are highly enriched for terms related to morphogenesis, metabolism (especially of lipids), entrainment of circadian clocks, oxygen transport, apoptosis, and response to stimulus. The present study offers valuable information on the central genes and pathways implicated in functions of GRL, and points out the possible involvement of this peptide in some novel functions in fish, such as apoptosis and oxygen transport.
Collapse
Affiliation(s)
- Ayelén Melisa Blanco
- Departamento de Genética, Fisiología y Microbiología, Facultad de Biología, Universidad Complutense de Madrid, José Antonio Nováis 12, 28040, Madrid, Spain
- Laboratorio de Fisioloxía Animal, Departamento de Bioloxía Funcional e Ciencias da Saúde, Facultade de Bioloxía and Centro de Investigación Mariña, Universidade de Vigo, Vigo, Spain
| | - Raúl Cortés
- Departamento de Fisiología de Peces y Biotecnología, Instituto de Acuicultura Torre de la Sal, Consejo Superior de Investigaciones Científicas, Ribera de Cabanes, 12595, Torre de la Sal, Castellón, Spain
- Centro de Investigación en Recursos Naturales y Sustentabilidad, Universidad Bernardo O'Higgins, Fábrica, 1990, Santiago, Chile
| | | | - Lucia Soletto
- Departamento de Fisiología de Peces y Biotecnología, Instituto de Acuicultura Torre de la Sal, Consejo Superior de Investigaciones Científicas, Ribera de Cabanes, 12595, Torre de la Sal, Castellón, Spain
| | - Elisa Sánchez
- Departamento de Fisiología de Peces y Biotecnología, Instituto de Acuicultura Torre de la Sal, Consejo Superior de Investigaciones Científicas, Ribera de Cabanes, 12595, Torre de la Sal, Castellón, Spain
| | - Ana Isabel Valenciano
- Departamento de Genética, Fisiología y Microbiología, Facultad de Biología, Universidad Complutense de Madrid, José Antonio Nováis 12, 28040, Madrid, Spain
| | - José Miguel Cerdá-Reverter
- Departamento de Fisiología de Peces y Biotecnología, Instituto de Acuicultura Torre de la Sal, Consejo Superior de Investigaciones Científicas, Ribera de Cabanes, 12595, Torre de la Sal, Castellón, Spain.
| | - María Jesús Delgado
- Departamento de Genética, Fisiología y Microbiología, Facultad de Biología, Universidad Complutense de Madrid, José Antonio Nováis 12, 28040, Madrid, Spain.
| |
Collapse
|
6
|
Li B, Lin Q, Guo H, Liu L, Li Y. Ghrelin regulates sepsis‑induced rat acute gastric injury. Mol Med Rep 2019; 19:5424-5432. [PMID: 31059095 PMCID: PMC6522907 DOI: 10.3892/mmr.2019.10208] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 03/26/2019] [Indexed: 12/25/2022] Open
Abstract
Ghrelin, a peptide expressed in the gastric mucosa, has an essential role in sustaining the normal function of the digestive system. Sepsis is one of the primary causes of mortality in intensive care units and can lead to multiple organ dysfunction, especially in the gastrointestinal system. The aim of the present study was to explore the effect of ghrelin on gastric blood flow in a rat model of sepsis, as well as the effect of ghrelin on the expression of the apoptotic markers, B-cell lymphoma 2 (Bcl-2) and Bcl-2-associated X (Bax), in gastric tissues. The sepsis model was established using cecal ligation and puncture (CLP). The expression levels of apoptosis-related factors in gastric epithelial cell were determined by immunohistochemistry, reverse transcription quantitative-PCR and western blotting. Collectively, the present results suggested that ghrelin administration attenuated sepsis symptoms induced by CLP. Blood flow in the stomach greater curvature was significantly higher in the CLP-induced sepsis group rats (284.3±95.7 perfusion units) compared with the sham operation group (317.8±5.2 perfusion units; P<0.05), whereas there was no difference between the CLP group treated with ghrelin (377.8±99.0 perfusion units) and the sham rats. Ghrelin administration also reduced the secretion of pro-inflammatory cytokines compared with the CLP-induced sepsis group rats. In addition, CLP significantly reduced the expression of Bcl-2 and enhanced the expression of the pro-apoptotic proteins, Bax and cleaved caspase-3; whereas, ghrelin application reversed the effects of CLP on these apoptosis-associated proteins. In conclusion, the present study revealed that ghrelin has the ability to increase blood flow in the gastrointestinal tract in a sepsis model and can also regulate the expressions of apoptosis-associated factors in gastric tissues. These results suggest that ghrelin could be a novel treatment for sepsis-induced gastric injury.
Collapse
Affiliation(s)
- Bin Li
- Department of General Surgery, The Second Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Qingling Lin
- Department of Intensive Medicine, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Hong Guo
- Department of Intensive Medicine, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Liping Liu
- Department of Intensive Medicine, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Yumin Li
- Department of General Surgery, The Second Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| |
Collapse
|
7
|
Nilaweera KN, Speakman JR. Regulation of intestinal growth in response to variations in energy supply and demand. Obes Rev 2018; 19 Suppl 1:61-72. [PMID: 30511508 PMCID: PMC6334514 DOI: 10.1111/obr.12780] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 09/11/2018] [Indexed: 12/14/2022]
Abstract
The growth of the intestine requires energy, which is known to be met by catabolism of ingested nutrients. Paradoxically, during whole body energy deficit including calorie restriction, the intestine grows in size. To understand how and why this happens, we reviewed data from several animal models of energetic challenge. These were bariatric surgery, cold exposure, lactation, dietary whey protein intake and calorie restriction. Notably, these challenges all reduced the adipose tissue mass, altered hypothalamic neuropeptide expression and increased intestinal size. Based on these data, we propose that the loss of energy in the adipose tissue promotes the growth of the intestine via a signalling mechanism involving the hypothalamus. We discuss possible candidates in this pathway including data showing a correlative change in intestinal (ileal) expression of the cyclin D1 gene with adipose tissue mass, adipose derived-hormone leptin and hypothalamic expression of leptin receptor and the pro-opiomelanocortin gene. The ability of the intestine to grow in size during depletion of energy stores provides a mechanism to maximize assimilation of ingested energy and in turn sustain critical functions of tissues important for survival.
Collapse
Affiliation(s)
- K N Nilaweera
- Department of Food Biosciences, Teagasc Food Research Centre, Fermoy, County Cork, Ireland
| | - J R Speakman
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China.,Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, UK
| |
Collapse
|
8
|
Abdanipour A, Dadkhah M, Alipour M, Feizi H. Effect of Ghrelin on Caspase 3 and Bcl2 Gene Expression in H2O2 Treated Rat's Bone Marrow Stromal Cells. Adv Pharm Bull 2018; 8:429-435. [PMID: 30276139 PMCID: PMC6156489 DOI: 10.15171/apb.2018.050] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Revised: 07/14/2018] [Accepted: 07/19/2018] [Indexed: 12/18/2022] Open
Abstract
Purpose: The antiapoptotic effect of ghrelin in various cell lines including bone marrow stromal cells (BMSCs) has been proved. However, the real mechanism of this effect is not clear. Caspase3 and Bcl2 are well-known pro- and antiapoptotic regulatory genes in eukaryotes. The aim of the study was to find out the effect of ghrelin on Caspase 3 and Bcl2 change in BMSCs. Methods: Rat BMSCs were cultivated in DMEM. Passage 3 BMSCs were treated with ghrelin 100 μM for 48 h. Real-time PCR for Caspase 3 and Bcl2 was carried out from B (untreated BMSCs), BH (BMSCs treated with 125 µM H2O2), BGH (BMSCs treated with 100 µM ghrelin then 125 µM H2O2) and BG (BMSCs treated with 100 µM ghrelin) groups. For immunofluorescence, cells were incubated with anti Caspase 3 and Bcl2monoclonal antibodies. Primary antibodies were visualized using the FITC method. All data are presented as means ± SEM. Values of P<0.05 were considered statistically significant. Results: Ghrelin decreased mRNA expressions of Caspase-3 significantly as compared to the BH group (P<0.05). Also, Bcl-2 gene expression showed an increment in BG group as compare with BH and BGH groups (P<0.05). A high present of Bcl-2 positive cells were observed in the BGH group while Caspase-3 positive cells were significantly decreased in the BGH group compared with the BH group (P<0.05). Conclusion: Ghrelin probably enhances BMSCs viability through regulation of pro- and antiapoptotic genes Caspase 3 and Bcl2. However the signaling pathway of this effect should be elucidated in the future.
Collapse
Affiliation(s)
- Alireza Abdanipour
- Department of Anatomical Sciences, Faculty of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Masoud Dadkhah
- Department of Physiology and Pharmacology, Faculty of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mohsen Alipour
- Department of Physiology and Pharmacology, Faculty of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Hadi Feizi
- Department of Physiology and Pharmacology, Faculty of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
9
|
Onishi S, Kaji T, Yamada W, Nakame K, Machigashira S, Kawano M, Yano K, Harumatsu T, Yamada K, Masuya R, Kawano T, Mukai M, Hamada T, Souda M, Yoshioka T, Tanimoto A, Ieiri S. Ghrelin stimulates intestinal adaptation following massive small bowel resection in parenterally fed rats. Peptides 2018; 106:59-67. [PMID: 29966680 DOI: 10.1016/j.peptides.2018.06.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 06/25/2018] [Accepted: 06/26/2018] [Indexed: 12/19/2022]
Abstract
BACKGROUND Since short bowel syndrome (SBS) patients face life-threatening conditions, the development of therapeutic strategies to induce intestinal adaptation has been investigated. Ghrelin, a ligand of growth hormone (GH) secretagogue-receptor that stimulates the release of GH and insulin like growth factor-1 (IGF-1), has several pleiotropic effects. We investigated whether ghrelin induces intestinal adaptation in parenterally fed rats with SBS. METHODS Sprague-Dawley rats underwent venous catheterization and were divided into 3 groups: those receiving 90% small bowel resection while leaving the proximal jejunum and distal ileum (90% SBR) with TPN (SBS/TPN group), those receiving 90% SBR with TPN + ghrelin (SBS/TPN/ghrelin group), and those receiving sham operation and fed chow (sham group). Ghrelin was administered intravenously at 10 μg/kg/day. On Day 13, the rats were euthanized and the small intestine harvested, and the histology and crypt cell proliferation rates (CCPR), apoptosis, and nutrient transporter protein levels were analyzed and the plasma hormones were measured. RESULTS The villus height and crypt depth of the ileum in the SBS/TPN/ghrelin group were significantly higher than in the SBS/TPN group. The CCPR of the jejunum and the ileum significantly increased by the administration of ghrelin; however, the apoptosis rates did not significantly differ between the SBS/TPN and SBS/TPN/ghrelin groups. Significant differences did not exist in the plasma IGF-1 and nutrient transporter protein levels among three groups. CONCLUSIONS The intravenous administration of ghrelin stimulated the morphological intestinal adaptation of the ileum to a greater degree than the jejunum due to the direct effect of ghrelin.
Collapse
Affiliation(s)
- Shun Onishi
- Department of Pediatric Surgery, Research Field in Medicine and Health Sciences, Medical and Dental Sciences Area, Research and Education Assembly, Kagoshima University, Japan
| | - Tatsuru Kaji
- Department of Pediatric Surgery, Research Field in Medicine and Health Sciences, Medical and Dental Sciences Area, Research and Education Assembly, Kagoshima University, Japan
| | - Waka Yamada
- Department of Pediatric Surgery, Research Field in Medicine and Health Sciences, Medical and Dental Sciences Area, Research and Education Assembly, Kagoshima University, Japan
| | - Kazuhiko Nakame
- Department of Pediatric Surgery, Research Field in Medicine and Health Sciences, Medical and Dental Sciences Area, Research and Education Assembly, Kagoshima University, Japan
| | - Seiro Machigashira
- Department of Pediatric Surgery, Research Field in Medicine and Health Sciences, Medical and Dental Sciences Area, Research and Education Assembly, Kagoshima University, Japan
| | - Masato Kawano
- Department of Pediatric Surgery, Research Field in Medicine and Health Sciences, Medical and Dental Sciences Area, Research and Education Assembly, Kagoshima University, Japan
| | - Keisuke Yano
- Department of Pediatric Surgery, Research Field in Medicine and Health Sciences, Medical and Dental Sciences Area, Research and Education Assembly, Kagoshima University, Japan
| | - Toshio Harumatsu
- Department of Pediatric Surgery, Research Field in Medicine and Health Sciences, Medical and Dental Sciences Area, Research and Education Assembly, Kagoshima University, Japan
| | - Koji Yamada
- Department of Pediatric Surgery, Research Field in Medicine and Health Sciences, Medical and Dental Sciences Area, Research and Education Assembly, Kagoshima University, Japan
| | - Ryuta Masuya
- Department of Pediatric Surgery, Research Field in Medicine and Health Sciences, Medical and Dental Sciences Area, Research and Education Assembly, Kagoshima University, Japan
| | - Takafumi Kawano
- Department of Pediatric Surgery, Research Field in Medicine and Health Sciences, Medical and Dental Sciences Area, Research and Education Assembly, Kagoshima University, Japan
| | - Motoi Mukai
- Department of Pediatric Surgery, Research Field in Medicine and Health Sciences, Medical and Dental Sciences Area, Research and Education Assembly, Kagoshima University, Japan
| | - Taiji Hamada
- Department of Pathology, Graduate School of Medical and Dental Sciences, Kagoshima University, Japan
| | - Masakazu Souda
- Department of Pathology, Graduate School of Medical and Dental Sciences, Kagoshima University, Japan; Department of Pathology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Takako Yoshioka
- National Center for Children Health and Development, Pathology, Japan
| | - Akihide Tanimoto
- Department of Pathology, Graduate School of Medical and Dental Sciences, Kagoshima University, Japan
| | - Satoshi Ieiri
- Department of Pediatric Surgery, Research Field in Medicine and Health Sciences, Medical and Dental Sciences Area, Research and Education Assembly, Kagoshima University, Japan.
| |
Collapse
|
10
|
Sista F, Abruzzese V, Clementi M, Guadagni S, Montana L, Carandina S. Resolution of type 2 diabetes after sleeve gastrectomy: a 2-step hypothesis. Surg Obes Relat Dis 2018; 14:284-290. [PMID: 29339031 DOI: 10.1016/j.soard.2017.12.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 11/27/2017] [Accepted: 12/08/2017] [Indexed: 01/19/2023]
Abstract
BACKGROUND Weight loss (WL) and altered gut hormonal levels are involved in glucose homeostasis after laparoscopic sleeve gastrectomy (LSG). OBJECTIVES The aim of this study was to evaluate the time-related effects of WL, ghrelin, and glucacon-like peptide-1 (GLP-1) plasma concentrations on type 2 diabetes resolution after LSG. SETTING University hospital, Italy. METHODS Ninety-one patients who underwent LSG were investigated. Insulin secretion (insulinogenic index [IGI]), insulin resistance, plasma glucose level and percentage glycated hemoglobin using the oral glucose tolerance test were assessed before surgery, on postoperative day 3, and then at 6, 12, 24, and 36 months after LSG. At the same time points, WL, ghrelin, and GLP-1 levels were determined. RESULTS During follow-up, the resolution rate of type 2 diabetes was 9.4%, 42.3%, 71.8%, 81.2%, and 91.8%, respectively. Ghrelin plasma concentrations decreased significantly after LSG (271.5 ± 24.5 pg/mL versus 122.4 ± 23.4 pg/mL, P = .04). GLP-1 plasma concentrations increased significantly after LSG (1.7 ± 2.6 pg/mL versus 2.5 ± 3.4 pg/mL, P = .04). The percentage of excess weight loss and IGI presented a positive linear correlation (r) at all follow-up time points with a strong positive correlation at 12 and 24 months. A strong negative correlation between ghrelin and IGI was recorded during the first 3 days after LSG (r = -.9). GLP-1 and IGI presented a strong positive correlation at day 3 and 6 months (i.e., .8 and .8, respectively). CONCLUSION LSG may affect glucose homeostasis by 2 different time-related modes: a first step in which the hormonal changes play a predominant role in glucose homeostasis and a second step in which the percentage excess weight loss determines the metabolic results.
Collapse
Affiliation(s)
- Federico Sista
- Department of General Surgery, Ospedale civile San Salvatore, University of L'Aquila, L'Aquila, Italy
| | - Valentina Abruzzese
- Department of General Surgery, Ospedale civile San Salvatore, University of L'Aquila, L'Aquila, Italy
| | - Marco Clementi
- Department of General Surgery, Ospedale civile San Salvatore, University of L'Aquila, L'Aquila, Italy
| | - Stefano Guadagni
- Department of General Surgery, Ospedale civile San Salvatore, University of L'Aquila, L'Aquila, Italy
| | - Laura Montana
- Assistance Publique-Hôpitaux de Paris (AP-HP), Department of digestive and metabolic surgery, Avicenne university hospital, Université Paris XIII-UFR SMBH "Léonard de Vinci", AP-HP, Bobigny, France
| | - Sergio Carandina
- ELSAN, Surgical obesity Center (CCO), Clinique Saint Michel, Toulon, France.
| |
Collapse
|
11
|
Ercan S, Şahin P, Kencebay C, Derin N, Çelik Özenci Ç. Evaluation of mTOR signaling pathway proteins in rat gastric mucosa exposed to sulfite and ghrelin. TURKISH JOURNAL OF GASTROENTEROLOGY 2017; 29:94-100. [PMID: 29082888 DOI: 10.5152/tjg.2017.17294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
BACKGROUND/AIMS Mammalian target of rapamycin (mTOR) signaling serves as a central regulator of cell growth, proliferation, and survival. In this study, we planned to evaluate the expressions of mTOR signaling constituents (p-p70S6K, p-mTOR, and p-Tuberin) in rat gastric mucosa and to compare the results in sulfite- and sulfite+ghrelin-exposed groups. MATERIALS AND METHODS Rats were divided into three groups: the control group (C), the sodium metabisulfite (Na2S2O5) (S) group, and sulfite+ghrelin (SG) group. Sodium metabisulfite at 100 mg/kg/day was administered via gavage, and ghrelin at 20 μg/kg/day was administered intraperitoneally for 35 days. We have used immunohistochemistry for mTOR signaling pathway components. RESULTS There were no significant differences for p-p70S6K and p-mTOR expression among the C, S, and SG groups. Tuberin expression was significantly increased in the S group compared to the C group. Furthermore, tuberin expression was found to be significantly decreased in the SG group. CONCLUSION This study is the first one in the literature that shows the expression of mTOR signaling proteins in gastric mucosa of rats exposed to sulfite and ghrelin. Furthermore, it demonstrates that ghrelin treatment reduces p-Tuberin expression induced by ingested sulfite.
Collapse
Affiliation(s)
- Sevim Ercan
- Department of Medical Services and Techniques, Akdeniz University Vocational School of Health Services, Antalya, Turkey
| | - Pınar Şahin
- Department of Histology and Embryology, Akdeniz University School of Medicine, Antalya, Turkey
| | - Ceren Kencebay
- Department of Biophysics, Akdeniz University School of Medicine, Antalya, Turkey
| | - Narin Derin
- Department of Biophysics, Akdeniz University School of Medicine, Antalya, Turkey
| | - Çiler Çelik Özenci
- Department of Histology and Embryology, Akdeniz University School of Medicine, Antalya, Turkey
| |
Collapse
|
12
|
Ghrelin improves intestinal mucosal atrophy during parenteral nutrition: An experimental study. J Pediatr Surg 2016; 51:2039-2043. [PMID: 27832865 DOI: 10.1016/j.jpedsurg.2016.09.035] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 09/12/2016] [Indexed: 02/02/2023]
Abstract
BACKGROUND/PURPOSE Total parenteral nutrition (TPN) has been reported to be associated with mucosal atrophy of the small intestine. Ghrelin has hormonal, orexigenic, and metabolic activities. We investigated whether ghrelin improved intestinal mucosal atrophy using a TPN-supported rat model. METHODS Rats underwent jugular vein catheterization and were divided into four groups: TPN alone (TPN), TPN plus low-dose ghrelin (TPNLG), TPN plus high-dose ghrelin (TPNHG), and oral feeding with normal chow (OF). Ghrelin was administered continuously at dosages of 10 or 50 μg/kg/day. On day 6 rats were euthanized, and the small intestine was harvested and divided into the jejunum and ileum. Then the villus height (VH) and crypt depth (CD) were evaluated. RESULTS The jejunal and ileal VH and CD in the TPN group were significantly decreased compared with those in the OF group. TPNHG improved only VH of the jejunum. TPNLG improved VH and CD of the jejunum and CD of the ileum. The improvement of TPNLG was significantly stronger than that in CD of the jejunum and ileum. CONCLUSIONS TPN was more strongly associated with mucosal atrophy in the jejunum than in the ileum. Low-dose intravenous administration of ghrelin improved TPN-associated intestinal mucosal atrophy more effectively than high-dose administration.
Collapse
|
13
|
Villa TG, Feijoo-Siota L, Rama JLR, Sánchez-Pérez A, de Miguel-Bouzas T. Fecal Matter Implantation as a Way to Fight Diarrhea-Causing Microorganisms. NEW WEAPONS TO CONTROL BACTERIAL GROWTH 2016:315-352. [DOI: 10.1007/978-3-319-28368-5_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
14
|
Bittar NMVR, Zulian JG, Ogias D, Gama P. Ghrelin and GHS-R in the rat gastric mucosa: Are they involved in regulation of growth during early weaning? Nutrition 2015; 32:101-7. [PMID: 26520918 DOI: 10.1016/j.nut.2015.06.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 06/10/2015] [Accepted: 06/28/2015] [Indexed: 12/25/2022]
Abstract
OBJECTIVES Based on previous evidence showing that early weaning disturbs the ontogenesis of rat gastric glands, which are the major site of ghrelin synthesis, we investigated the distribution of ghrelin and its receptor (GHS-R) in the rat gastric epithelium during postnatal development and evaluated the effects of early weaning on their levels. Additionally, we studied the contribution of ghrelin to gastric growth during the abrupt nutrient transition. METHODS Wistar rats were submitted to early weaning at 15 d and suckling counterparts were taken as controls. RESULTS By running quantitative reverse transcription polymerase chain reaction, immunoblots, and immunohistochemistry, we detected a variation of ghrelin levels and an increase of expression and number of immunolabeled cells, 3 d after treatment (P < 0.05). Through confocal microscopy, we identified GHS-R in the neck region of the gland and did not observe changes in protein levels. Growth was evaluated after ghrelin antagonist ([D-Lys-3]-GHRP-6) administration, which reduced DNA synthesis index in early-weaned rats (P < 0.05) as determined by bromodeoxyuridine incorporation. CONCLUSION The present study demonstrated that ghrelin and GHS-R are distributed in gastric mucosa during the postnatal development, indicating that they can signal and function in epithelial cells. We concluded that early weaning increased ghrelin levels in the stomach, and it takes part of cell proliferation control that is essential for stomach growth. Therefore, among the many effects previously described for early weaning, this abrupt nutrient transition also changed ghrelin levels, which might represent an additional element in the complex mechanism that coordinates stomach development.
Collapse
Affiliation(s)
| | - Juliana Guimarães Zulian
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Daniela Ogias
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Patrícia Gama
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
15
|
MAKOVICKY PETER, TUMOVA EVA, VOLEK ZDENEK, MAKOVICKY PAVOL, VODICKA PAVEL. Histological aspects of the small intestine under variable feed restriction: The effects of short and intense restriction on a growing rabbit model. Exp Ther Med 2014; 8:1623-1627. [PMID: 25289070 PMCID: PMC4186336 DOI: 10.3892/etm.2014.1924] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2014] [Accepted: 06/19/2014] [Indexed: 12/23/2022] Open
Abstract
The objective of this study was to investigate the effect of seven days of feed restriction (between days 42 and 49) on the morphology of the small intestine in experimental rabbit models. Sixty weaned Hyplus rabbits (35 days old) were included in the experiment and split into three groups of 20 rabbits. The first control group (n=20) received feed ad libitum (ADL group), the second (R1) experimental group (n=20) was fed 50 g feed per rabbit per day and the third (R2) experimental group (n=20) received 65 g feed per rabbit per day. Duodenal samples were collected when the rabbits were aged 49, 56, 63 and 70 days. The mean villus height, crypt depth and small intestine length were measured. Significant interactions (P<0.001) between group and age were identified in the villi height and crypt depths. The maximum mean villus height was found in the R2 group in 56-day-old rabbits (643.14 μm), while the minimum was found in the ADL group in 49-day-old rabbits (460.29 μm). The longest (P<0.001) small intestine was measured in the R1 group in 63-day-old rabbits (347.60 cm), while the shortest was measured in the ADL group in 49-day-old rabbits (263.60 cm). The models show that villus height, crypt depth and the length of the small intestine change with the intensity of feed restriction and age.
Collapse
Affiliation(s)
- PETER MAKOVICKY
- Laboratory of Veterinary Histopathology, Komarno, Slovak Republic
| | - EVA TUMOVA
- Department of Animal Husbandry, Czech University of Life Sciences in Prague, Prague, Czech Republic
| | - ZDENEK VOLEK
- Laboratory of Physiology of Nutrition and Quality of Animal Product, Institute of Animal Science, Prague, Czech Republic
| | - PAVOL MAKOVICKY
- Department of Biology, Selye Janos University, Komarno, Slovak Republic
| | - PAVEL VODICKA
- Department of Molecular Biology of Cancer, Institute of Experimental Medicine, Academy of Science of the Czech Republic, Prague, Czech Republic
| |
Collapse
|
16
|
Waseem T, Duxbury M, Ashley SW, Robinson MK. Ghrelin promotes intestinal epithelial cell proliferation through PI3K/Akt pathway and EGFR trans-activation both converging to ERK 1/2 phosphorylation. Peptides 2014; 52:113-21. [PMID: 24365237 DOI: 10.1016/j.peptides.2013.11.021] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2013] [Revised: 11/26/2013] [Accepted: 11/26/2013] [Indexed: 12/16/2022]
Abstract
Little is known about ghrelin's effects on intestinal epithelial cells even though it is known to be a mitogen for a variety of other cell types. Because ghrelin is released in close proximity to the proliferative compartment of the intestinal tract, we hypothesized that ghrelin may have potent pro-proliferative effect on intestinal epithelial cells as well. To test this hypothesis, we characterized the effects of ghrelin on FHs74Int and Caco-2 intestinal epithelial cell lines in vitro. We found that ghrelin has potent dose dependent proliferative effects in both cell lines through a yet to be characterized G protein coupled growth hormone secretagogue receptor (GHS-R) subtype. Consistent with above findings, cell cycle flowcytometric analyses demonstrated that ghrelin shifts cells from the G1 to S phase and thereby promotes cell cycle progression. Further characterization of subcellular events, suggested that ghrelin mediates its pro-proliferative effect through Adenylate cyclase (AC)-independent epidermal growth factor receptor (EGFR) trans-activation and PI3K-Akt phosphorylation. Both these pathways converge to stimulate MAPK, ERK 1/2 downstream. The role of ghrelin in states where intestinal mucosal injury and rapid mucosal repair occur warrants further investigation.
Collapse
Affiliation(s)
- Talat Waseem
- Department of Surgery, Brigham & Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, United States
| | - Mark Duxbury
- Department of Surgery, Brigham & Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, United States
| | - Stanley W Ashley
- Department of Surgery, Brigham & Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, United States
| | - Malcolm K Robinson
- Department of Surgery, Brigham & Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, United States.
| |
Collapse
|
17
|
Muto M, Kaji T, Mukai M, Nakame K, Yoshioka T, Tanimoto A, Matsufuji H. Ghrelin and glucagon-like peptide-2 increase immediately following massive small bowel resection. Peptides 2013; 43:160-6. [PMID: 23517879 DOI: 10.1016/j.peptides.2013.03.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Revised: 03/01/2013] [Accepted: 03/01/2013] [Indexed: 01/17/2023]
Abstract
Children with short bowel syndrome face life-threatening complications. Therefore, there is an urgent need for a new therapy to induce effective adaptation of the remnant intestine. Adaptation occurs only during feeding. We focused on preprandial acyl ghrelin and des-acyl ghrelin, and postprandial glucagon-like peptide-2 (GLP-2), which are known to have active orexigenic and trophic actions. This study aims to clarify the secretion trends of these hormones after massive small bowel resection and to obtain basic data for developing a new treatment. Sixty-three growing male rats were used: 3 were designated as controls receiving no operation and 60 were randomized into the 80% small bowel resection (80% SBR) group and the transection and re-anastomosis group. Changes in body weight, food intake, and remnant intestine morphology were also assessed for 15 days after the operation. Acyl ghrelin and des-acyl ghrelin levels increased immediately, equivalently in both operation groups (P=0.09 and 0.70). Interestingly, in 80% SBR animals, des-acyl ghrelin peaked on day 1 and acyl ghrelin peaked on day 4 (P=0.0007 and P=0.049 vs controls). GLP-2 secretion was obvious in 80% SBR animals (P=2.25×10(-6)), which increased immediately and peaked on day 4 (P=0.009 vs. controls). Body weight and food intake in 80% SBR animals recovered to preoperative levels on day 4. Morphological adaptations were evident after day 4. Our results may suggest a management strategy to reinforce these physiological hormone secretion patterns in developing a new therapy for short bowel syndrome.
Collapse
Affiliation(s)
- Mitsuru Muto
- Department of Pediatric Surgery, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima 8908520, Japan
| | | | | | | | | | | | | |
Collapse
|
18
|
Gao J, Guo J, Li H, Bai S, Li H, Wu B, Wang L, Xi Y, Tian Y, Yang G, Wang R, Wu L, Xu C, Li H. Involvement of dopamine D2 receptors activation in ischemic post-conditioning-induced cardioprotection through promoting PKC-ε particulate translocation in isolated rat hearts. Mol Cell Biochem 2013; 379:267-76. [DOI: 10.1007/s11010-013-1648-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2013] [Accepted: 04/11/2013] [Indexed: 11/30/2022]
|
19
|
Yu H, Xu G, Fan X. The effect of ghrelin on cell proliferation in small intestinal IEC-6 cells. Biomed Pharmacother 2013; 67:235-9. [DOI: 10.1016/j.biopha.2013.01.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2012] [Accepted: 01/24/2013] [Indexed: 11/16/2022] Open
|
20
|
Ercan S, Basaranlar G, Gungor NE, Kencebay C, Sahin P, Celik-Ozenci C, Derin N. Ghrelin inhibits sodium metabisulfite induced oxidative stress and apoptosis in rat gastric mucosa. Food Chem Toxicol 2013; 56:154-61. [PMID: 23439480 DOI: 10.1016/j.fct.2013.02.019] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Revised: 02/05/2013] [Accepted: 02/06/2013] [Indexed: 12/19/2022]
Abstract
This study aimed to investigate the effect of ghrelin administration on sulfite induced oxidative and apoptotic changes in rat gastric mucosa. Forty male albino Wistar rats were randomized into control (C), sodium metabisulfite (Na2S2O5) treated (S), ghrelin treated (G) and, Na2S2O5+ghrelin treated (SG) groups. Sodium metabisulfite (100 mg/kg/day) was given by gastric gavage and, ghrelin (20 μg/kg/day) was given intraperitoneally for 5 weeks. Plasma-S-sulfonate level was increased in S and SG groups. Na2S2O5 administration significantly elevated total oxidant status (TOS) levels while depleting total antioxidant status (TAS) levels in gastric mucosa. Ghrelin significantly decreased gastric TOS levels in the SG group compared with the S group. Additionally, TAS levels were found to be higher in SG group in reference to S group. Na2S2O5 administration also markedly increased the number of apoptotic cells, cleaved caspase-3 and PAR expression (PARP activity indicator) and, decreased Ki67 expression (cell proliferation index) in gastric mucosal cells. Ghrelin treatment decreased the number apoptotic cells, cytochrome C release, PAR and, caspase-3 expressions while increasing Ki67 expression in gastric mucosa exposed to Na2S2O5. In conclusion, we suggest that ghrelin treatment might ameliorate ingested-Na2S2O5 induced gastric mucosal injury stemming from apoptosis and oxidative stress in rats.
Collapse
Affiliation(s)
- Sevim Ercan
- Akdeniz University, Vocational School of Health Services, Antalya 07070, Turkey.
| | | | | | | | | | | | | |
Collapse
|
21
|
Chopin LK, Seim I, Walpole CM, Herington AC. The ghrelin axis--does it have an appetite for cancer progression? Endocr Rev 2012; 33:849-91. [PMID: 22826465 DOI: 10.1210/er.2011-1007] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Ghrelin, the endogenous ligand for the GH secretagogue receptor (GHSR), is a peptide hormone with diverse physiological roles. Ghrelin regulates GH release, appetite and feeding, gut motility, and energy balance and also has roles in the cardiovascular, immune, and reproductive systems. Ghrelin and the GHSR are expressed in a wide range of normal and tumor tissues, and a fluorescein-labeled, truncated form of ghrelin is showing promise as a biomarker for prostate cancer. Plasma ghrelin levels are generally inversely related to body mass index and are unlikely to be useful as a biomarker for cancer, but may be useful as a marker for cancer cachexia. Some single nucleotide polymorphisms in the ghrelin and GHSR genes have shown associations with cancer risk; however, larger studies are required. Ghrelin regulates processes associated with cancer, including cell proliferation, apoptosis, cell migration, cell invasion, inflammation, and angiogenesis; however, the role of ghrelin in cancer is currently unclear. Ghrelin has predominantly antiinflammatory effects and may play a role in protecting against cancer-related inflammation. Ghrelin and its analogs show promise as treatments for cancer-related cachexia. Further studies using in vivo models are required to determine whether ghrelin has a role in cancer progression.
Collapse
Affiliation(s)
- Lisa K Chopin
- Ghrelin Research Group, Institute of Health and Biomedical Innovation, Queensland University of Technology and Australian Prostate Cancer Research Centre-Queensland, Brisbane, Queensland 4001, Australia.
| | | | | | | |
Collapse
|
22
|
Zhang Q, Huang C, Meng B, Tang T, Shi Q, Yang H. Acute effect of Ghrelin on ischemia/reperfusion injury in the rat spinal cord. Int J Mol Sci 2012; 13:9864-9876. [PMID: 22949835 PMCID: PMC3431833 DOI: 10.3390/ijms13089864] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 07/17/2012] [Accepted: 07/19/2012] [Indexed: 01/25/2023] Open
Abstract
Ghrelin, a 28-amino acid peptide, is mainly secreted by the stomach. Ghrelin has been shown to have neuroprotective effects. However, whether ghrelin protects the spinal cord from ischemia/reperfusion (I/R) injury is unknown. To investigate this, 60 rats were randomly divided into three different groups: the sham group (n = 20), the vehicle group (n = 20), and the Ghrelin group (100 μg/kg, n = 20). Rats were sacrificed 12, 24, 48 and 72 h after ischemia. After the evaluation of neurologic function (48 h), the spinal cords were immediately removed for the determination of myeloperoxidase (MPO) activity (12-72 h). Apoptosis was quantitatively measured using the terminal transferase UTP nick end-labeling (TUNEL) method (24 h). The expression of bax and bcl-2 were evaluated by Western blot analysis (1 h), and GHSR-1a mRNA expression was detected using reverse transcriptase polymerase chain reaction (24 h). The neurological motor function was evaluated by 'Tarlov's score'. The neurologic outcomes in the ghrelin-group were significantly better than those in the vehicle group (p < 0.05). Serum tumor necrosis factor (TNF-α) levels were assessed in the peripheral venous blood. Ghrelin decreased the serum TNF-α levels and ameliorated the down regulation of spinal cord MPO activity. The expression of ghrelin receptors (GHSR-1a) in the rat spinal cord was decreased by I/R injury and increased by ghrelin. Ghrelin reduced the TUNEL-positive rate. Greater bcl-2, HSP27, HSP70, and attenuated bax expression were observed in the ghrelin-treated rats. Our results suggest that ghrelin administration may inhibit spinal I/R injury. Moreover, the improvement of neurologic function in rats was increased after the ghrelin treatment.
Collapse
Affiliation(s)
- Qin Zhang
- Department of Orthopedics, Yuncheng Central Hospital, Yuncheng 044000, China; E-Mail:
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Soochow 215007, China; E-Mails: (C.H.); (B.M.); (T.T.); (Q.S.)
| | - Chen Huang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Soochow 215007, China; E-Mails: (C.H.); (B.M.); (T.T.); (Q.S.)
| | - Bin Meng
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Soochow 215007, China; E-Mails: (C.H.); (B.M.); (T.T.); (Q.S.)
| | - Tiansi Tang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Soochow 215007, China; E-Mails: (C.H.); (B.M.); (T.T.); (Q.S.)
| | - Qin Shi
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Soochow 215007, China; E-Mails: (C.H.); (B.M.); (T.T.); (Q.S.)
| | - Huilin Yang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Soochow 215007, China; E-Mails: (C.H.); (B.M.); (T.T.); (Q.S.)
| |
Collapse
|
23
|
Zanuzzi CN, Nishida F, Portiansky EL, Fontana PA, Gimeno EJ, Barbeito CG. Effects of Solanum glaucophyllum toxicity on cell proliferation and apoptosis in the small and large intestine of rabbits. Res Vet Sci 2012; 93:336-42. [PMID: 21862088 PMCID: PMC7125838 DOI: 10.1016/j.rvsc.2011.07.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2011] [Revised: 06/28/2011] [Accepted: 07/21/2011] [Indexed: 11/04/2022]
Abstract
Vitamin D regulates mineral homeostases and enterocyte proliferation and differentiation. Hypervitaminosis D generates changes in cell proliferation, differentiation and apoptosis in several organs. We analysed morphometric parameters and proliferative and apoptotic indices in the intestinal epithelium of rabbits with hypervitaminosis D induced by the chronic treatment with the calcinogenic plant Solanum glaucophyllum. Rabbits were treated for 15 or 30 days. A group was treated for 15 days and led to possible recovery for 30 days. Another group was nutritionally restricted for 30 days. Morphological, morphometric, proliferative and apoptotic changes were found in the treated animals. Mild atrophy and reduced proliferation was found in the jejunum and ileum. Apoptosis increased in the crypts of the ileum and in the superficial epithelium and crypts of the rectum. Most of the alterations were partially recovered. The possible involvement in these changes of the hypervitaminosis D-like state induced by S. glaucophyllum is discussed.
Collapse
Affiliation(s)
- C N Zanuzzi
- Instituto de Patología, Facultad de Ciencias Veterinarias, Universidad Nacional de La Plata, La Plata, Buenos Aires, Argentina.
| | | | | | | | | | | |
Collapse
|
24
|
Kasai A, Gama P, Alvares EP. Protein restriction inhibits gastric cell proliferation during rat postnatal growth in parallel to ghrelin changes. Nutrition 2012; 28:707-12. [DOI: 10.1016/j.nut.2011.10.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2011] [Revised: 10/07/2011] [Accepted: 10/11/2011] [Indexed: 12/14/2022]
|
25
|
Karatug A, Sacan O, Coskun ZM, Bolkent S, Yanardag R, Turk N, Bolkent S. Regulation of gene expression and biochemical changes in small intestine of newborn diabetic rats by exogenous ghrelin. Peptides 2012; 33:101-8. [PMID: 22138721 DOI: 10.1016/j.peptides.2011.11.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2011] [Revised: 11/02/2011] [Accepted: 11/02/2011] [Indexed: 01/27/2023]
Abstract
The aim of this study was to investigate (i) the cholecystokinin, somatostatin and apelin mRNA levels, (ii) the changes in levels and localization of these peptides, (iii) relation between these peptides, (iv) antiapoptotic effects and (v) antioxidant effects of ghrelin. The rats were divided into four groups second day after birth. These groups were respectively treated with physiological saline, ghrelin (100μg/kg/day), streptozotocin (100mg/kg), ghrelin and streptozotocin. After four weeks, small intestine and blood samples were taken from rats. Cholecystokinin mRNA and peptide, somatostatin mRNA, release to duodenal lumen of apelin peptide and apelin mRNA signals decreased in ghrelin-treated diabetic rats compared to the diabetic group. There was no statistically significant difference among the four groups for somatostatin and apelin peptides. Caspase-3 signals were not observed only in diabetic group treated with ghrelin. Caspase-8 signals were increased while PCNA signals were decreased in diabetic group given ghrelin compared to diabetic group. Small intestine CAT, SOD, GP(x) and GST activities and GSH levels were decreased and LPO, PC levels were increased in diabetic rats. Administration of ghrelin to diabetic rats caused an increase in intestinal CAT, SOD, GP(x) and GST activities and GSH levels, while PC levels decreased. As a result, we observed positive changes in diabetic rats treated with ghrelin in both microscopic and biochemical studies. We can suggest that ghrelin may be an important hormone for the treatment of diabetes.
Collapse
Affiliation(s)
- Ayse Karatug
- Istanbul University, Faculty of Science, Department of Biology, 34134 Vezneciler, Istanbul, Turkey
| | | | | | | | | | | | | |
Collapse
|
26
|
Abstract
BACKGROUND AND AIM The aim of the present study was to investigate if ghrelin inhibits apoptosis in colonic cancer cells. METHODS Cell viability in HT-29 cells was measured using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. Apoptosis was measured using 4',6-diamidino-2-phenylindole staining and flow cytometry. The protein expression of Bcl-2, Bax, and caspase-3 activation was examined using Western blotting. RESULTS Ghrelin dose dependently decreased the growth inhibition of HT-29 cells induced by 5-fluorouracil (5-FU). Cells treated with 5-FU displayed chromatin condensation and nuclear fragmentation, which are typical changes of apoptosis. However, co-treatment with ghrelin reduced these changes. Flow cytometry after staining with Annexin V and propidium iodide showed that ghrelin decreased the apoptotic rate of HT-29 cells induced by 5-FU. Caspase-3 activation was significantly lower in the co-treated group than in the group treated with 5-FU alone. In addition, ghrelin reversed the 5-FU-induced Bcl-2/Bax protein ratio. CONCLUSION Ghrelin inhibits 5-FU-induced apoptosis in colon cancer cells through the regulation of the Bcl-2/Bax protein ratio.
Collapse
Affiliation(s)
- Xiao-Tong He
- Department of Gastroenterology, Jinshan Hospital of Fudan University, Shanghai, China
| | | | | |
Collapse
|
27
|
Yarandi SS, Hebbar G, Sauer CG, Cole CR, Ziegler TR. Diverse roles of leptin in the gastrointestinal tract: modulation of motility, absorption, growth, and inflammation. Nutrition 2011; 27:269-75. [PMID: 20947298 PMCID: PMC3066025 DOI: 10.1016/j.nut.2010.07.004] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2010] [Revised: 07/08/2010] [Accepted: 07/09/2010] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Leptin was discovered in 1994 as a hormone produced by adipose tissue with a modulatory effect on feeding behavior and weight control. Recently, the stomach has been identified as an important source of leptin and growing evidence has shown diverse functions for leptin in the gastrointestinal tract. METHODS Using leptin as a keyword in PubMed, more than 17 000 articles were identified, of which more than 500 articles were related to the role of leptin in the gastrointestinal tract. Available abstracts were reviewed and more than 200 original articles were reviewed in detail. RESULTS The available literature demonstrated that leptin can modulate several important functions of the gastrointestinal tract. Leptin interacts with the vagus nerve and cholecystokinin to delay gastric emptying and has a complex effect on motility of the small bowel. Leptin modulates absorption of macronutrients in the gastrointestinal tract differentially in physiologic and pathologic states. In physiologic states, exogenous leptin has been shown to decrease carbohydrate absorption and to increase the absorption of small peptides by the PepT1 di-/tripeptide transporter. In certain pathologic states, leptin has been shown to increase absorption of carbohydrates, proteins, and fat. Leptin has been shown to be upregulated in the colonic mucosa in patients with inflammatory bowel disease. Leptin stimulates gut mucosal cell proliferation and inhibits apoptosis. These functions have led to speculation about the role of leptin in tumorigenesis in the gastrointestinal tract, which is complicated by the multiple immunoregulatory effects of leptin. CONCLUSION Leptin is an important modulator of major aspects of gastrointestinal tract functions, independent of its more well-described roles in appetite regulation and obesity.
Collapse
Affiliation(s)
- Shadi S. Yarandi
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine, Emory University, Atlanta, Georgia, USA
| | - Gautam Hebbar
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine, Emory University, Atlanta, Georgia, USA
| | - Cary G. Sauer
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Emory University, Atlanta, Georgia, USA
| | - Conrad R. Cole
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Emory University, Atlanta, Georgia, USA
| | - Thomas R. Ziegler
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
28
|
Ferrini F, Salio C, Lossi L, Merighi A. Ghrelin in central neurons. Curr Neuropharmacol 2010; 7:37-49. [PMID: 19721816 PMCID: PMC2724662 DOI: 10.2174/157015909787602779] [Citation(s) in RCA: 153] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2008] [Revised: 08/15/2008] [Accepted: 09/01/2008] [Indexed: 12/20/2022] Open
Abstract
Ghrelin, an orexigenic peptide synthesized by endocrine cells of the gastric mucosa, is released in the bloodstream in response to a negative energetic status. Since discovery, the hypothalamus was identified as the main source of ghrelin in the CNS, and effects of the peptide have been mainly observed in this area of the brain. In recent years, an increasing number of studies have reported ghrelin synthesis and effects in specific populations of neurons also outside the hypothalamus. Thus, ghrelin activity has been described in midbrain, hindbrain, hippocampus, and spinal cord. The spectrum of functions and biological effects produced by the peptide on central neurons is remarkably wide and complex. It ranges from modulation of membrane excitability, to control of neurotransmitter release, neuronal gene expression, and neuronal survival and proliferation. There is not at present a general consensus concerning the source of ghrelin acting on central neurons. Whereas it is widely accepted that the hypothalamus represents the most important endogenous source of the hormone in CNS, the existence of extra-hypothalamic ghrelin-synthesizing neurons is still controversial. In addition, circulating ghrelin can theoretically be another natural ligand for central ghrelin receptors. This paper gives an overview on the distribution of ghrelin and its receptor across the CNS and critically analyses the data available so far as regarding the effects of ghrelin on central neurotransmission.
Collapse
Affiliation(s)
- F Ferrini
- Dipartimento di Morfofisiologia Veterinaria, Università di Torino, Via Leonardo da Vinci 44, 10095, Grugliasco, Italy
| | | | | | | |
Collapse
|
29
|
de Segura IAG, Vallejo-Cremades MT, Lomas J, Sánchez MF, Caballero MI, Largo C, De Miguel E. Exogenous ghrelin regulates proliferation and apoptosis in the hypotrophic gut mucosa of the rat. Exp Biol Med (Maywood) 2010; 235:463-9. [DOI: 10.1258/ebm.2009.009247] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Ghrelin is the natural endogenous ligand for growth hormone secretagogue receptors. This peptide regulates energy homeostasis and expenditure and is a potential link between gut absorptive function and growth. We hypothesized that ghrelin may induce a proliferative and antiapoptotic action promoting the recovery of the hypotrophic gut mucosa. Therefore, the aim of the study was to determine the action of exogenous ghrelin following gut mucosal hypotrophia in rats fed an elemental diet. An elemental diet provides readily absorbable simple nutrients and is usually given to patients with absorptive dysfunction. Male Wistar rats ( n = 48) were fed the elemental diet for one week to induce mucosal hypotrophy and then treated for another week with systemic ghrelin and pair-fed with either a normoproteic or hyperproteic isocaloric liquid diet. Another group received a standard diet instead of the elemental diet and served as control (normotrophy). The elemental diet induced intestinal hypotrophia characterized by decreased proliferation in the ileum and increased apoptosis in jejunum and ileum. Ghrelin administration restored normal levels of proliferation in the ileum and apoptosis in the jejunum, with partial apoptosis restoration in the ileum. Ghrelin levels in plasma and fundus were increased in all groups, although the highest levels were found in rats treated with exogenous ghrelin. Ghrelin administration has a positive effect in the hypotrophic gut, regulating both proliferation and apoptosis towards a physiological balance counteracting the negative changes induced by an elemental diet in the intestines.
Collapse
Affiliation(s)
- Ignacio A Gómez de Segura
- Department of Experimental Surgery, Hospital Universitario La Paz, Paseo de la Castellana, 261, 28046 Madrid
- Department of Medicine and Surgery, Veterinary Faculty, University Complutense of Madrid, Avda. Puerta de Hierro s/n, 28040 Madrid
| | - María Teresa Vallejo-Cremades
- Research Unit, Biomedical Research Foundation, Hospital Universitario La Paz, Madrid, Paseo de la Castellana, 261, 28046 Madrid, Spain
| | - Jesús Lomas
- Department of Experimental Surgery, Hospital Universitario La Paz, Paseo de la Castellana, 261, 28046 Madrid
| | - Miriam F Sánchez
- Department of Experimental Surgery, Hospital Universitario La Paz, Paseo de la Castellana, 261, 28046 Madrid
| | - María Isabel Caballero
- Department of Experimental Surgery, Hospital Universitario La Paz, Paseo de la Castellana, 261, 28046 Madrid
| | - Carlota Largo
- Department of Experimental Surgery, Hospital Universitario La Paz, Paseo de la Castellana, 261, 28046 Madrid
| | - Enrique De Miguel
- Department of Experimental Surgery, Hospital Universitario La Paz, Paseo de la Castellana, 261, 28046 Madrid
| |
Collapse
|
30
|
Bossenmeyer-Pourié C, Blaise S, Pourié G, Tomasetto C, Audonnet S, Ortiou S, Koziel V, Rio MC, Daval JL, Guéant JL, Beck B. Methyl donor deficiency affects fetal programming of gastric ghrelin cell organization and function in the rat. THE AMERICAN JOURNAL OF PATHOLOGY 2009; 176:270-7. [PMID: 19948829 DOI: 10.2353/ajpath.2010.090153] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Methyl donor deficiency (MDD) during pregnancy influences intrauterine development. Ghrelin is expressed in the stomach of fetuses and influences fetal growth, but MDD influence on gastric ghrelin is unknown. We examined the gastric ghrelin system in MDD-induced intrauterine growth retardation. By using specific markers and approaches (such as periodic acid-Schiff, bromodeoxyuridine, homocysteine, terminal deoxynucleotidyl transferase biotin-dUTP nick end labeling, immunostaining, reverse transcription-polymerase chain reaction), we studied the gastric oxyntic mucosa cellular organization and ghrelin gene expression in the mucosa in 20-day-old fetuses and weanling pups, and plasma ghrelin concentration in weanling rat pups of dams either normally fed or deprived of choline, folate, vitamin B6, and vitamin B12 during gestation and suckling periods. MDD fetuses weighed less than controls; the weight deficit reached 57% at weaning (P < 0.001). Both at the end of gestation and at weaning, they presented with an aberrant gastric oxyntic mucosa formation with loss of cell polarity, anarchic cell migration, abnormal progenitor differentiation, apoptosis, and signs of surface layer erosion. Ghrelin cells were abnormally located in the pit region of oxyntic glands. At weaning, plasma ghrelin levels were decreased (-28%; P < 0.001) despite unchanged mRNA expression in the stomach. This decrease was associated with lower body weight. Taken together, these data indicate that one mechanism through which MDD influences fetal programming is the remodeling of gastric cellular organization, leading to dysfunction of the ghrelin system and dramatic effects on growth.
Collapse
Affiliation(s)
- Carine Bossenmeyer-Pourié
- INSERM U954, Nutrition, Génétique et Expositions aux Risques Environnementaux, 54505 Vandoeuvre Cedex, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Involvement of the ornithine decarboxylase/polyamine system in precondition-induced cardioprotection through an interaction with PKC in rat hearts. Mol Cell Biochem 2009; 332:135-44. [DOI: 10.1007/s11010-009-0183-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2009] [Accepted: 06/09/2009] [Indexed: 12/13/2022]
|
32
|
Hattori N. Expression, regulation and biological actions of growth hormone (GH) and ghrelin in the immune system. Growth Horm IGF Res 2009; 19:187-197. [PMID: 19144554 DOI: 10.1016/j.ghir.2008.12.001] [Citation(s) in RCA: 138] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2008] [Revised: 11/05/2008] [Accepted: 12/08/2008] [Indexed: 12/20/2022]
Abstract
Immune and neuroendocrine systems have bidirectional communications. Growth hormone (GH) and an orexigenic hormone ghrelin are expressed in various immune cells such as T lymphocytes, B lymphocytes, monocytes and neutrophils. These immune cells also bear receptors for hormones: growth hormone receptor (GHR) for GH and growth hormone secretagogue receptor (GHS-R) for ghrelin. The expression of GH in immune cells is stimulated by ghrelin as in anterior pituitary cells, whereas the regulation of GH secretion in the immune system by other peptides seems to be different from that in the anterior pituitary gland. Cytokines and mitogens enhance GH secretion from immune cells. GH has several biological actions in the immune system: enhancing thymopoiesis and T cell development, modulating cytokine production, enhancing B cell development and antibody production, priming neutrophils and monocytes for superoxide anion secretion, enhancing neutrophil adhesion and monocyte migration and anti-apoptotic action. Biological actions of ghrelin include attenuation of septic shock and anti-inflammatory actions, modulating phagocytosis, and enhancing thymopoiesis. The effect of ghrelin may be direct or through GH production, and that of GH may be direct or through insulin like growth factor-I (IGF-I) production. Elucidation of the roles of GH and ghrelin in the immune system may shed light on the treatment and prevention of immunological disorders such as AIDS and organ damages due to obesity/ageing-related chronic inflammation.
Collapse
Affiliation(s)
- Naoki Hattori
- Department of Pharmacology, Kansai Medical University, Moriguchi-city, Osaka, Japan.
| |
Collapse
|
33
|
Soares JB, Leite-Moreira AF. Ghrelin, des-acyl ghrelin and obestatin: three pieces of the same puzzle. Peptides 2008; 29:1255-70. [PMID: 18396350 DOI: 10.1016/j.peptides.2008.02.018] [Citation(s) in RCA: 130] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2008] [Revised: 02/24/2008] [Accepted: 02/26/2008] [Indexed: 12/26/2022]
Abstract
The major active product of ghrelin gene is a 28-amino acid peptide acylated at the serine 3 position with an octanoyl group, called simply ghrelin. Ghrelin has a multiplicity of physiological functions, affecting GH release, food intake, energy and glucose homeostasis, gastrointestinal, cardiovascular, pulmonary and immune function, cell proliferation and differentiation and bone physiology. Nevertheless, recent developments have shown that ghrelin gene can generate various bioactive molecules besides ghrelin, mainly des-acyl ghrelin and obestatin, obtained from alternative splicing or from extensive post-translational modification. Although their receptors have not yet been identified, they have already proven to be active, having intriguingly subtle but opposite physiological actions to ghrelin. This suggests the existence of a novel endocrine system with multiple effector elements which not only may have opposite actions but may regulate the action of each other. In this review, we summarize the steps which lead to the production of the different ghrelin gene products and examine the most significant differences between them in terms of structure and actions.
Collapse
Affiliation(s)
- João-Bruno Soares
- Department of Physiology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | | |
Collapse
|
34
|
Indigestible Material Attenuated Changes in Apoptosis in the Fasted Rat Jejunal Mucosa. Exp Biol Med (Maywood) 2008; 233:310-6. [DOI: 10.3181/0708-rm-228] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
We have previously demonstrated that fasting induced apoptosis and decreased cell proliferation in the rat intestinal mucosa. The aim was to investigate the effect of expanded polystyrene as indigestible material on apoptosis and cell proliferation in rat small intestinal mucosa during fasting. Male SD rats were divided into 3 groups. The first group was fed with chow and water ad libitum. The second group fasted for 72 hrs. The third group was fasted for 24 hrs and was fed expanded polystyrene. Intestinal apoptosis was evaluated by percent fragmented DNA assay, terminal deoxynucleotidyl transferase–mediated dUDP-biotin nick end-labeling (TUNEL) staining, and caspase-3 assay. Cell proliferation was analyzed by 5-bromo-2′-deoxyuridine (5-BrdU) uptake. Truncal vagotomy was performed to evaluate a role of the central nervous system. In the 72-hr fasted rat, mucosal height of the rat jejunum was decreased to 73% of that in rats fed ad libitum, and this decrease was partly restored to 90% in rats fed expanded polystyrene. The fragmented DNA was increased in fasted rats (28.0%) when compared with that in rats fed ad libitum (2.6%). The increase in fragmented DNA in fasted rats was recovered by feeding them expanded polystyrene (8.3%). TUNEL staining confirmed this result. The effect of polystyrene on apoptosis was decreased by truncal vagotomy. Expression of cleaved caspase-3 was increased in fasted rats, which was then decreased by feeding of expanded polystyrene. In contrast to apoptosis, feeding of expanded polystyrene had no reconstructive effect on 5-BrdU uptake in the intestinal epithelium, which was decreased by fasting to 60% of that in rats fed ad libitum. In conclusion, feeding of indigestible material partly restored the decrease in intestinal mucosal length in the fasted rats through the apoptotic pathway without any influence on BrdU uptake. Further exploration focused on the mechanism of this effect of indigestible material is required.
Collapse
|