51
|
Tan Q, Orsso CE, Deehan EC, Triador L, Field CJ, Tun HM, Han JC, Müller TD, Haqq AM. Current and emerging therapies for managing hyperphagia and obesity in Prader-Willi syndrome: A narrative review. Obes Rev 2020; 21:e12992. [PMID: 31889409 DOI: 10.1111/obr.12992] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 12/07/2019] [Accepted: 12/09/2019] [Indexed: 12/16/2022]
Abstract
In early childhood, individuals with Prader-Willi syndrome (PWS) experience excess weight gain and severe hyperphagia with food compulsivity, which often leads to early onset morbid obesity. Effective treatments for appetite suppression and weight control are currently unavailable for PWS. Our aim to further understand the pathogenesis of PWS led us to carry out a comprehensive search of the current and emerging therapies for managing hyperphagia and extreme weight gain in PWS. A literature search was performed using PubMed and the following keywords: "PWS" AND "therapy" OR "[drug name]"; reference lists, pharmaceutical websites, and the ClinicalTrials.gov registry were also reviewed. Articles presenting data from current standard treatments in PWS and also clinical trials of pharmacological agents in the pipeline were selected. Current standard treatments include dietary restriction/modifications, exercise, and growth hormone replacement, which appear to have limited efficacy for appetite and weight control in patients with PWS. The long-term safety and effectiveness of bariatric surgery in PWS remains unknown. However, many promising pharmacotherapies are in development and, if approved, will bring much needed choices into the PWS pharmacological armamentarium. With the progress that is currently being made in our understanding of PWS, an effective treatment may not be far off.
Collapse
Affiliation(s)
- Qiming Tan
- Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Camila E Orsso
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Edward C Deehan
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Lucila Triador
- Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Catherine J Field
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Hein Min Tun
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, China
| | - Joan C Han
- Departments of Pediatrics and Physiology, College of Medicine, University of Tennessee Health Science Center and Children's Foundation Research Institute, Le Bonheur Children's Hospital, Memphis, Tennessee, USA
| | - Timo D Müller
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München, Neuherberg, Germany.,Department of Pharmacology and Experimental Therapy, Institute of Experimental and Clinical Pharmacology and Toxicology, Eberhard Karls University Hospitals and Clinics, Tübingen, Germany
| | - Andrea M Haqq
- Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada.,Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
52
|
Exogenous Ghrelin Increases Plasma Insulin Level in Diabetic Rats. Biomolecules 2020; 10:biom10040633. [PMID: 32325912 PMCID: PMC7226305 DOI: 10.3390/biom10040633] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 04/09/2020] [Accepted: 04/15/2020] [Indexed: 12/13/2022] Open
Abstract
Ghrelin, a 28-amino acid peptide, is a strong growth hormone secretagogue and a regulator of food intake. In addition, ghrelin is thought to play a role in insulin secretion and in glucose homeostasis. A lot of contradictory data have been reported in the literature regarding the co-localization of ghrelin with other hormones in the islet of Langerhans, its role in insulin secretion and attenuation of type 2 diabetes mellitus. In this study, we investigate the effect of chronic ghrelin treatment on glucose, body weight and insulin level in normal and streptozotocin-induced diabetic male Wistar rats. We have also examined the distribution pattern and co-localization of ghrelin with insulin in pancreatic islet cells using immunohistochemistry and immune-electron microscopy and the ability of ghrelin to stimulate insulin release from the CRL11065 beta cell line. Control, non-diabetic groups received intraperitoneal injection of normal saline, while treated groups received intraperitoneal injection of 5 µg/kg body weight of ghrelin (amino acid chain 24–51) on a daily basis for a duration of four weeks. Our results show that the administration of ghrelin increases the number of insulin-secreting beta cells and serum insulin level in both normal and diabetic rats. We also demonstrated that ghrelin co-localizes with insulin in pancreatic islet cells and that the pattern of ghrelin distribution is altered after the onset of diabetes. Moreover, ghrelin at a dose of 10−6 M and 10−12 M increased insulin release from the CRL11065 beta cell line. In summary, ghrelin co-localizes with insulin in the secretory granules of pancreatic beta cells and enhances insulin production.
Collapse
|
53
|
Perna S, Spadaccini D, Gasparri C, Peroni G, Infantino V, Iannello G, Riva A, Petrangolini G, Alalwan TA, Al-Thawadi S, Rondanelli M. Association between des-acyl ghrelin at fasting and predictive index of muscle derangement, metabolic markers and eating disorders: a cross-sectional study in overweight and obese adults. Nutr Neurosci 2020; 25:336-342. [DOI: 10.1080/1028415x.2020.1752997] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Simone Perna
- Department of Biology, College of Science, University of Bahrain, Sakhir, Kingdom of Bahrain
| | - Daniele Spadaccini
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona ‘Istituto Santa Margherita’, University of Pavia, Pavia, Italy
| | - Clara Gasparri
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona ‘Istituto Santa Margherita’, University of Pavia, Pavia, Italy
| | - Gabriella Peroni
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona ‘Istituto Santa Margherita’, University of Pavia, Pavia, Italy
| | - Vittoria Infantino
- Department of Public Health, Experimental and Forensic Medicine, Unit of Human and Clinical Nutrition, University of Pavia, Pavia, Italy
| | - Giancarlo Iannello
- General Management, Azienda di Servizi alla Persona ‘Istituto Santa Margherita’, Pavia, Italy
| | - Antonella Riva
- Research and Development Department, Indena SpA, Milan, Italy
| | | | - Tariq A. Alalwan
- Department of Biology, College of Science, University of Bahrain, Sakhir, Kingdom of Bahrain
| | - Salwa Al-Thawadi
- Department of Biology, College of Science, University of Bahrain, Sakhir, Kingdom of Bahrain
| | - Mariangela Rondanelli
- Department of Public Health, Experimental and Forensic Medicine, Unit of Human and Clinical Nutrition, University of Pavia, Pavia, Italy
- IRCCS Mondino Foundation, Pavia, Italy
| |
Collapse
|
54
|
Karl JP, Berryman CE, Harris MN, Lieberman HR, Gadde KM, Rood JC, Pasiakos SM. Effects of Testosterone Supplementation on Ghrelin and Appetite During and After Severe Energy Deficit in Healthy Men. J Endocr Soc 2020; 4:bvaa024. [PMID: 32258956 DOI: 10.1210/jendso/bvaa024] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 02/26/2020] [Indexed: 12/17/2022] Open
Abstract
Background Severe energy deficits cause interrelated reductions in testosterone and fat free mass. Testosterone supplementation may mitigate those decrements, but could also reduce circulating concentrations of the orexigenic hormone ghrelin, thereby exacerbating energy deficit by suppressing appetite. Objective To determine whether testosterone supplementation during severe energy deficit influences fasting and postprandial ghrelin concentrations and appetite. Design and methods Secondary analysis of a randomized, double-blind trial that determined the effects of testosterone supplementation on body composition changes during and following severe energy deficit in nonobese, eugonadal men. Phase 1 (PRE-ED): 14-day run-in; phase 2: 28 days, 55% energy deficit with 200 mg testosterone enanthate weekly (TEST; n = 24) or placebo (PLA; n = 26); phase 3: free-living until body mass recovered (end-of-study; EOS). Fasting and postprandial acyl ghrelin and des-acyl ghrelin concentrations and appetite were secondary outcomes measured during the final week of each phase. Results Fasting acyl ghrelin concentrations, and postprandial acyl and des-acyl ghrelin concentrations increased in PLA during energy deficit then returned to PRE-ED values by EOS, but did not change in TEST (phase-by-group, P < 0.05). Correlations between changes in free testosterone and changes in fasting acyl ghrelin concentrations during energy deficit (ρ = -0.42, P = 0.003) and body mass recovery (ρ = -0.38; P = 0.01) were not mediated by changes in body mass or body composition. Transient increases in appetite during energy deficit were not affected by testosterone treatment. Conclusions Testosterone supplementation during short-term, severe energy deficit in healthy men prevents deficit-induced increases in circulating ghrelin without blunting concomitant increases in appetite. Clinical Trials Registration www.clinicaltrials.gov NCT02734238 (registered 12 April 2016).
Collapse
Affiliation(s)
- J Philip Karl
- Military Nutrition Division, US Army Research Institute of Environmental Medicine, Natick, MA, USA
| | - Claire E Berryman
- Military Nutrition Division, US Army Research Institute of Environmental Medicine, Natick, MA, USA.,Oak Ridge Institute for Science and Education, Belcamp, MD, USA.,Department of Nutrition, Food, and Exercise Sciences, Florida State University, Tallahassee, FL, USA
| | - Melissa N Harris
- Louisiana State University's Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Harris R Lieberman
- Military Nutrition Division, US Army Research Institute of Environmental Medicine, Natick, MA, USA
| | - Kishore M Gadde
- Louisiana State University's Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Jennifer C Rood
- Louisiana State University's Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Stefan M Pasiakos
- Military Nutrition Division, US Army Research Institute of Environmental Medicine, Natick, MA, USA
| |
Collapse
|
55
|
Galmiche M, Lucas N, Déchelotte P, Deroissart C, Le Solliec MA, Rondeaux J, Azhar S, Grigioni S, Colange G, Delay J, Achamrah N, Folope V, Belmonte L, Lamarre A, Rimbert A, Saillard T, Petit A, Quillard M, Coeffier M, Gillibert A, Lambert G, Legrand R, Tavolacci MP. Plasma Peptide Concentrations and Peptide-Reactive Immunoglobulins in Patients with Eating Disorders at Inclusion in the French EDILS Cohort (Eating Disorders Inventory and Longitudinal Survey). Nutrients 2020; 12:nu12020522. [PMID: 32085628 PMCID: PMC7071399 DOI: 10.3390/nu12020522] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 02/11/2020] [Accepted: 02/14/2020] [Indexed: 12/12/2022] Open
Abstract
Eating disorders (EDs) are increasingly frequent. Their pathophysiology involves disturbance of peptide signaling and the microbiota–gut–brain axis. This study analyzed peptides and corresponding immunoglobulin (Ig) concentrations in groups of ED. In 120 patients with restrictive (R), bulimic (B), and compulsive (C) ED, the plasma concentrations of leptin, glucagon-like peptide-1 (GLP-1), peptide YY (PYY), and insulin were analyzed by Milliplex and those of acyl ghrelin (AG), des-acyl ghrelin (DAG), and α-melanocyte-stimulating hormone (α-MSH) by ELISA kits. Immunoglobulin G (in response to an antigen) concentrations were analyzed by ELISA, and their affinity for the respective peptide was measured by surface plasmon resonance. The concentrations of leptin, insulin, GLP-1, and PYY were higher in C patients than in R patients. On the contrary, α-MSH, DAG, and AG concentrations were higher in R than in C patients. After adjustment for body mass index (BMI), differences among peptide concentrations were no longer different. No difference in the concentrations of the IgG was found, but the IgG concentrations were correlated with each other. Although differences of peptide concentrations exist among ED subtypes, they may be due to differences in BMI. Changes in the concentration and/or affinity of several anti-peptide IgG may contribute to the physiopathology of ED or may be related to fat mass.
Collapse
Affiliation(s)
- Marie Galmiche
- Inserm UMR1073, 76000 Rouen, France; (M.G.); (S.G.); (N.A.); (V.F.); (L.B.); (A.R.); (A.P.); (M.C.); (M.-P.T.)
- TargEDys SA, 91160 Longjumeau, France; (N.L.); (C.D.); (M.-A.L.S.); (J.R.); (S.A.); (G.L.); (R.L.)
- Institute for Research and Innovation in Biomedicine (IRIB), University of Rouen, 76000 Rouen, France
| | - Nicolas Lucas
- TargEDys SA, 91160 Longjumeau, France; (N.L.); (C.D.); (M.-A.L.S.); (J.R.); (S.A.); (G.L.); (R.L.)
| | - Pierre Déchelotte
- Inserm UMR1073, 76000 Rouen, France; (M.G.); (S.G.); (N.A.); (V.F.); (L.B.); (A.R.); (A.P.); (M.C.); (M.-P.T.)
- Institute for Research and Innovation in Biomedicine (IRIB), University of Rouen, 76000 Rouen, France
- Nutrition unit, University Hospital of Rouen, 76000 Rouen, France; (G.C.); (J.D.); (A.L.); (T.S.)
- Correspondence: ; Tel.: +06-08-49-66-26
| | - Camille Deroissart
- TargEDys SA, 91160 Longjumeau, France; (N.L.); (C.D.); (M.-A.L.S.); (J.R.); (S.A.); (G.L.); (R.L.)
| | - Marie-Anne Le Solliec
- TargEDys SA, 91160 Longjumeau, France; (N.L.); (C.D.); (M.-A.L.S.); (J.R.); (S.A.); (G.L.); (R.L.)
| | - Julie Rondeaux
- TargEDys SA, 91160 Longjumeau, France; (N.L.); (C.D.); (M.-A.L.S.); (J.R.); (S.A.); (G.L.); (R.L.)
| | - Saida Azhar
- TargEDys SA, 91160 Longjumeau, France; (N.L.); (C.D.); (M.-A.L.S.); (J.R.); (S.A.); (G.L.); (R.L.)
| | - Sébastien Grigioni
- Inserm UMR1073, 76000 Rouen, France; (M.G.); (S.G.); (N.A.); (V.F.); (L.B.); (A.R.); (A.P.); (M.C.); (M.-P.T.)
- Institute for Research and Innovation in Biomedicine (IRIB), University of Rouen, 76000 Rouen, France
- Nutrition unit, University Hospital of Rouen, 76000 Rouen, France; (G.C.); (J.D.); (A.L.); (T.S.)
| | - Guillaume Colange
- Nutrition unit, University Hospital of Rouen, 76000 Rouen, France; (G.C.); (J.D.); (A.L.); (T.S.)
| | - Julie Delay
- Nutrition unit, University Hospital of Rouen, 76000 Rouen, France; (G.C.); (J.D.); (A.L.); (T.S.)
| | - Najate Achamrah
- Inserm UMR1073, 76000 Rouen, France; (M.G.); (S.G.); (N.A.); (V.F.); (L.B.); (A.R.); (A.P.); (M.C.); (M.-P.T.)
- Institute for Research and Innovation in Biomedicine (IRIB), University of Rouen, 76000 Rouen, France
- Nutrition unit, University Hospital of Rouen, 76000 Rouen, France; (G.C.); (J.D.); (A.L.); (T.S.)
| | - Vanessa Folope
- Inserm UMR1073, 76000 Rouen, France; (M.G.); (S.G.); (N.A.); (V.F.); (L.B.); (A.R.); (A.P.); (M.C.); (M.-P.T.)
- Institute for Research and Innovation in Biomedicine (IRIB), University of Rouen, 76000 Rouen, France
- Nutrition unit, University Hospital of Rouen, 76000 Rouen, France; (G.C.); (J.D.); (A.L.); (T.S.)
| | - Liliana Belmonte
- Inserm UMR1073, 76000 Rouen, France; (M.G.); (S.G.); (N.A.); (V.F.); (L.B.); (A.R.); (A.P.); (M.C.); (M.-P.T.)
- Institute for Research and Innovation in Biomedicine (IRIB), University of Rouen, 76000 Rouen, France
- Nutrition unit, University Hospital of Rouen, 76000 Rouen, France; (G.C.); (J.D.); (A.L.); (T.S.)
| | - Adèle Lamarre
- Nutrition unit, University Hospital of Rouen, 76000 Rouen, France; (G.C.); (J.D.); (A.L.); (T.S.)
| | - Agnès Rimbert
- Inserm UMR1073, 76000 Rouen, France; (M.G.); (S.G.); (N.A.); (V.F.); (L.B.); (A.R.); (A.P.); (M.C.); (M.-P.T.)
- Institute for Research and Innovation in Biomedicine (IRIB), University of Rouen, 76000 Rouen, France
- Nutrition unit, University Hospital of Rouen, 76000 Rouen, France; (G.C.); (J.D.); (A.L.); (T.S.)
| | - Tiphaine Saillard
- Nutrition unit, University Hospital of Rouen, 76000 Rouen, France; (G.C.); (J.D.); (A.L.); (T.S.)
| | - André Petit
- Inserm UMR1073, 76000 Rouen, France; (M.G.); (S.G.); (N.A.); (V.F.); (L.B.); (A.R.); (A.P.); (M.C.); (M.-P.T.)
- Institute for Research and Innovation in Biomedicine (IRIB), University of Rouen, 76000 Rouen, France
- Nutrition unit, University Hospital of Rouen, 76000 Rouen, France; (G.C.); (J.D.); (A.L.); (T.S.)
| | - Muriel Quillard
- CIC-CRB 1404 INSERM, University Hospital of Rouen, 76000 Rouen, France;
| | - Moise Coeffier
- Inserm UMR1073, 76000 Rouen, France; (M.G.); (S.G.); (N.A.); (V.F.); (L.B.); (A.R.); (A.P.); (M.C.); (M.-P.T.)
- Institute for Research and Innovation in Biomedicine (IRIB), University of Rouen, 76000 Rouen, France
- Nutrition unit, University Hospital of Rouen, 76000 Rouen, France; (G.C.); (J.D.); (A.L.); (T.S.)
| | - André Gillibert
- Department of Biostatistics, Rouen University Hospital, F 76000 Rouen, France;
| | - Grégory Lambert
- TargEDys SA, 91160 Longjumeau, France; (N.L.); (C.D.); (M.-A.L.S.); (J.R.); (S.A.); (G.L.); (R.L.)
| | - Romain Legrand
- TargEDys SA, 91160 Longjumeau, France; (N.L.); (C.D.); (M.-A.L.S.); (J.R.); (S.A.); (G.L.); (R.L.)
| | - Marie-Pierre Tavolacci
- Inserm UMR1073, 76000 Rouen, France; (M.G.); (S.G.); (N.A.); (V.F.); (L.B.); (A.R.); (A.P.); (M.C.); (M.-P.T.)
- Institute for Research and Innovation in Biomedicine (IRIB), University of Rouen, 76000 Rouen, France
- CIC-CRB 1404 INSERM, University Hospital of Rouen, 76000 Rouen, France;
| |
Collapse
|
56
|
Behavioural characterization of ghrelin ligands, anamorelin and HM01: Appetite and reward-motivated effects in rodents. Neuropharmacology 2020; 168:108011. [PMID: 32067989 DOI: 10.1016/j.neuropharm.2020.108011] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 02/12/2020] [Accepted: 02/13/2020] [Indexed: 12/14/2022]
Abstract
The ghrelinergic system has been steadily investigated as a therapeutic target in the treatment of metabolic disorders and modulation of appetite. While endogenous ghrelin activates the full complement of the growth hormone secretagogue receptor (GHSR-1a) pathways, synthetic GHSR-1a ligands display biased signalling and functional selectivity, which have a significant impact on the intended and indeed, unintended, therapeutic effects. The widespread expression of the GHSR-1a receptor in vivo also necessitates an imperative consideration of the biodistribution of GHSR-1a ligands. Here, we investigate anamorelin and HM01, two recently described synthetic GHSR-1a ligands which have shown promising effects on food intake in preclinical and clinical studies. We compare the downstream signalling pathways in cellular in vitro assays, including calcium mobilization, IP-one, internalization and β-arrestin recruitment assays. We describe a novel divergent activation of central reward circuitry by anamorelin and HM01 using c-Fos immunostaining as well as behavioural effects in food intake and reward paradigms. Interestingly, we found a paradoxical reduction in reward-related behaviour for anamorelin and HM01 treated animals in our chosen paradigms. The work highlights the critical importance to consider signalling bias in relation to future ghrelin-based therapies. In addition, central access of GHSR-1a ligands, particularly to reward areas of the brain, remains a crucial factor in eliciting potent appetite-stimulating effects. The precise characterization of downstream ghrelinergic signalling and biodistribution of novel GHSR-1a ligands will be decisive in their successful development and will allow predictive modelling and design of future synthetic ligands to combat metabolic and appetite disorders involving the ghrelinergic system. This article is part of the special issue on 'Neuropeptides'.
Collapse
|
57
|
Wu TH, Chiu CC, Goh KK, Chen PY, Huang MC, Chen CH, Lu ML. Relationship between metabolic syndrome and acylated/desacylated ghrelin ratio in patients with schizophrenia under olanzapine medication. J Psychopharmacol 2020; 34:86-92. [PMID: 31692408 DOI: 10.1177/0269881119885260] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Ghrelin is a peptide hormone that mediates glucose homeostasis and lipid metabolism. Acylated ghrelin (AG) and desacylated ghrelin (DAG) are the two main forms of ghrelin, which have opposing roles in energy homeostasis. The AG/DAG ratio has been proposed to be associated with metabolic syndrome (MetS) in the general population. This study compared the relationships between MetS and ghrelin parameters in patients with schizophrenia. METHODS Patients diagnosed with schizophrenia and under olanzapine monotherapy were recruited. Fasting blood samples were collected for the analyses of metabolic and ghrelin parameters. The serum levels of total ghrelin and AG were measured by enzyme-linked immunosorbent assay kits. DAG level was calculated by subtracting the AG level from the total ghrelin level. RESULTS We recruited 151 subjects with schizophrenia, and classified them into those with MetS (n = 41) and those without MetS (n = 110). Subjects with MetS had a significantly higher AG/DAG ratio, as well as lower total ghrelin and DAG levels. There were no sex differences in ghrelin parameters. The AG/DAG ratio was significantly and positively correlated with weight, body mass index, waist circumference, insulin level, homeostasis model assessment of insulin resistance and number of MetS components. Multiple linear regression analysis indicated that the number of MetS components remained significantly associated with the AG/DAG ratio. CONCLUSIONS Our results revealed that lower AG/DAG ratios were associated with better metabolic profiles in olanzapine-treated patients with schizophrenia. These observations suggest that the balance between AG and DAG plays a crucial role in the metabolic homeostasis among patients with schizophrenia.
Collapse
Affiliation(s)
- Tzu-Hua Wu
- Department of Clinical Pharmacy, School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan.,Psychiatric Research Centre, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Chih-Chiang Chiu
- Department of Psychiatry, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Psychiatry, Taipei City Psychiatric Centre, Taipei City Hospital, Taipei, Taiwan
| | - Kah Kheng Goh
- Psychiatric Research Centre, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.,Department of Psychiatry, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Po-Yu Chen
- Department of Psychiatry, Taipei City Psychiatric Centre, Taipei City Hospital, Taipei, Taiwan
| | - Ming-Chyi Huang
- Department of Psychiatry, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Psychiatry, Taipei City Psychiatric Centre, Taipei City Hospital, Taipei, Taiwan
| | - Chun-Hsin Chen
- Psychiatric Research Centre, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.,Department of Psychiatry, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Psychiatry, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Mong-Liang Lu
- Psychiatric Research Centre, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.,Department of Psychiatry, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Psychiatry, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
58
|
Magherini F, Fiaschi T, Marzocchini R, Mannelli M, Gamberi T, Modesti PA, Modesti A. Oxidative stress in exercise training: the involvement of inflammation and peripheral signals. Free Radic Res 2019; 53:1155-1165. [PMID: 31762356 DOI: 10.1080/10715762.2019.1697438] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The evidence about the health benefits of regular physical activity is well established. Exercise intensity is a significant variable and structured high-intensity interval training (HIIT) has been demonstrated to improve both whole-body and skeletal muscle metabolic health in different populations. Conversely, fatigue accumulation, if not resolved, leads to overwork, chronic fatigue syndrome (CFS), overtraining syndrome up to alterations of endocrine function, immune, systemic inflammation, and organic diseases with health threat. In response to temporary increases in stress during training, some athletes are unable to maintain sufficient caloric intake, thus suffering a negative energy balance that causes further stress. The regulation of the energy balance is controlled by the central nervous system through an elaborate interaction of the signalling that involves different tissues such as leptin, adiponectin and ghrelin whose provide important feedback to the hypothalamus to regulate the energy balance. Although exercise-induced reactive oxygen species are required for normal force production in muscle, high levels of ROS appear to promote contractile dysfunction. However, a high level of oxidative stress in may induce a rise in inflammatory markers and a disregulation in expression of adiponectin, leptin and grelin.
Collapse
Affiliation(s)
- Francesca Magherini
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Tania Fiaschi
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Riccardo Marzocchini
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Michele Mannelli
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Tania Gamberi
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Pietro Amedeo Modesti
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Alessandra Modesti
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", University of Florence, Florence, Italy
| |
Collapse
|
59
|
Lucka A, Wysokiński A. Association between adiposity and fasting serum levels of appetite-regulating peptides: Leptin, neuropeptide Y, desacyl ghrelin, peptide YY(1-36), obestatin, cocaine and amphetamine-regulated transcript, and agouti-related protein in nonobese participants. CHINESE J PHYSIOL 2019; 62:217-225. [PMID: 31670286 DOI: 10.4103/cjp.cjp_29_19] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
The objective of this study was to evaluate the association between adiposity parameters and fasting serum levels of appetite-regulating peptides: leptin, neuropeptide Y (NPY), desacyl ghrelin, peptide YY(1-36), obestatin, cocaine- and amphetamine-regulated transcript (CART), and agouti-related protein in 30 healthy, non-obese subjects. Thirty European Caucasian adult participants were included in the study (17 men and 13 women). Body composition (body fat and lean body mass) was determined using bioelectrical impedance analysis. Concentrations of peptides in serum were assessed using the enzyme-linked immunosorbent assay. Women had higher level of leptin (P < 0.001), with no other differences for analyzed peptides. We have found a significant correlation between serum concentrations of CART and NPY (P < 0.001). Fasting leptin level was associated with age (P = 0.002), waist circumference (P < 0.001), and lean body mass (P < 0.001). Levels of ghrelin were lower in participants with dyslipidemia (P = 0.009). Levels of obestatin (P = 0.008) and leptin (P = 0.02) were higher in participants with insulin resistance. Associations between body fat and appetite-regulating peptides are more complex than simple feedback loops. Leptin is probably the first signal in the pathway that regulates body fat content, as of all analyzed peptides leptin was the only one that was associated with body composition or anthropometric measurements.
Collapse
Affiliation(s)
- Anna Lucka
- Department of Old Age Psychiatry and Psychotic Disorders, Medical University of Lodz, Łódź, Poland
| | - Adam Wysokiński
- Department of Old Age Psychiatry and Psychotic Disorders, Medical University of Lodz, Łódź, Poland
| |
Collapse
|
60
|
Dorling JL, Clayton DJ, Jones J, Carter WG, Thackray AE, King JA, Pucci A, Batterham RL, Stensel DJ. A randomized crossover trial assessing the effects of acute exercise on appetite, circulating ghrelin concentrations, and butyrylcholinesterase activity in normal-weight males with variants of the obesity-linked FTO rs9939609 polymorphism. Am J Clin Nutr 2019; 110:1055-1066. [PMID: 31504106 DOI: 10.1093/ajcn/nqz188] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 07/18/2019] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND The fat mass and obesity-associated gene (FTO) rs9939609 A-allele is associated with higher acyl-ghrelin (AG) concentrations, higher energy intake, and obesity, although exercise may mitigate rs9939609 A-allele-linked obesity risk. Butyrylcholinesterase (BChE) hydrolyzes AG to des-acyl-ghrelin (DAG), potentially decreasing appetite. However, the effects of the FTO rs9939609 genotype and exercise on BChE activity, AG, DAG, and energy intake are unknown. OBJECTIVE We hypothesized that individuals homozygous for the obesity-risk A-allele (AAs) would exhibit higher postprandial AG and energy intake than individuals homozygous for the low obesity-risk T-allele (TTs), but that exercise would increase BChE activity and diminish these differences. METHODS Twelve AA and 12 TT normal-weight males completed a control (8 h rest) and an exercise (1 h of exercise at 70% peak oxygen uptake, 7 h rest) trial in a randomized crossover design. A fixed meal was consumed at 1.5 h and an ad libitum buffet meal at 6.5 h. Appetite, appetite-related hormones, BChE activity, and energy intake were assessed. RESULTS AAs displayed lower baseline BChE activity, higher baseline AG:DAG ratio, attenuated AG suppression after a fixed meal, and higher ad libitum energy intake compared with TTs [effect sizes (ESs) ≥ 0.72, P ≤ 0.049]. Exercise increased Δ BChE activity in both genotypes (ESs = 0.37, P = 0.004); however, exercise lowered AG and the AG:DAG ratio to a greater extent in AAs (P ≤ 0.023), offsetting the higher AG profile observed in AAs during the control trial (ESs ≥ 1.25, P ≤ 0.048). Exercise did not elevate energy intake in either genotype (P = 0.282). CONCLUSIONS Exercise increases BChE activity, suppresses AG and the AG:DAG ratio, and corrects the higher AG profile observed in obesity-risk AA individuals. These findings suggest that exercise or other methods targeting BChE activity may offer a preventative and/or therapeutic strategy for AA individuals. This trial was registered at clinicaltrials.gov as NCT03025347.
Collapse
Affiliation(s)
- James L Dorling
- National Centre for Sport and Exercise Medicine, School of Sport, Exercise, and Health Sciences, Loughborough University, Loughborough, United Kingdom.,Ingestive Behavior Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - David J Clayton
- National Centre for Sport and Exercise Medicine, School of Sport, Exercise, and Health Sciences, Loughborough University, Loughborough, United Kingdom.,School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
| | - Jenny Jones
- Centre for Obesity Research, University College London, London, United Kingdom
| | - Wayne G Carter
- School of Medicine, University of Nottingham Medical School, Royal Derby Hospital Centre, Derby, United Kingdom
| | - Alice E Thackray
- National Centre for Sport and Exercise Medicine, School of Sport, Exercise, and Health Sciences, Loughborough University, Loughborough, United Kingdom.,University Hospitals of Leicester National Health Service Trust, Leicester, United Kingdom
| | - James A King
- National Centre for Sport and Exercise Medicine, School of Sport, Exercise, and Health Sciences, Loughborough University, Loughborough, United Kingdom.,University Hospitals of Leicester National Health Service Trust, Leicester, United Kingdom
| | - Andrea Pucci
- Centre for Obesity Research, University College London, London, United Kingdom.,University College London Hospitals Bariatric Centre for Weight Management and Metabolic Surgery, London, United Kingdom
| | - Rachel L Batterham
- Centre for Obesity Research, University College London, London, United Kingdom.,University College London Hospitals Bariatric Centre for Weight Management and Metabolic Surgery, London, United Kingdom.,National Institute of Health Research, University College London Hospitals Biomedical Research Centre, London, United Kingdom
| | - David J Stensel
- National Centre for Sport and Exercise Medicine, School of Sport, Exercise, and Health Sciences, Loughborough University, Loughborough, United Kingdom.,University Hospitals of Leicester National Health Service Trust, Leicester, United Kingdom
| |
Collapse
|
61
|
Shiomi Y, Yoshimura M, Hori Y, Ohira Y, Nagahama K, Ozaki T, Takei M, Tanaka T, Uno T. Z-505, an Oral Ghrelin Receptor Agonist, Attenuates Anorexia After Total Gastrectomy in Rats. J Surg Res 2019; 246:527-534. [PMID: 31668932 DOI: 10.1016/j.jss.2019.09.035] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 09/13/2019] [Accepted: 09/18/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Anorexia is a serious problem in patients with gastric cancer who have undergone gastrectomy. Ghrelin, an orexigenic hormone primarily secreted from the stomach, has been proposed to prevent anorexia. Significant reduction in plasma ghrelin levels after gastrectomy may contribute to lack of appetite and weight loss. In this study, we investigated the effects of Z-505, a ghrelin receptor agonist, on anorexia after total gastrectomy (TG) in a rat model. METHODS AND MATERIALS Male Sprague-Dawley rats were used to establish a TG model, and then sham-operated (control) and TG rats were randomly assigned to four subgroups receiving administration of Z-505 (100 mg/kg, p.o., once daily) or vehicle for 14 d from day 14 to day 27 after TG. The food intake, body weight, and fat weight were evaluated during the test period. Moreover, the neuronal activity in the hypothalamus was evaluated on day 21 to investigate the mechanism of action of Z-505. RESULTS In TG rats, Z-505 significantly improved the decrease in cumulative food intake induced by the surgery over 14 d (TG + vehicle; 213.8 ± 15.3 g, n = 12 versus TG + Z-505; 258.2 ± 13.1 g, n = 14, P < 0.05). Z-505 also significantly increased fat weight and had a milder effect on body weight over 14 d. In addition, Z-505 significantly increased the number of c-Fos-positive cells in the hypothalamic arcuate nucleus (TG + vehicle; 17.8 ± 2.0, n = 12 versus TG + Z-505; 72.2 ± 11.8, n = 12, P < 0.001). CONCLUSIONS Z-505 may be a useful therapeutic treatment for anorexia after TG.
Collapse
Affiliation(s)
- Yoshihiro Shiomi
- Central Research Laboratories, Zeria Pharmaceutical Co, Ltd, Kumagaya-shi, Saitama, Japan; Agrobioscience, Graduate School of Agricultural Science, Kobe University, Nada-ku, Hyogo, Japan.
| | - Makoto Yoshimura
- Legal Affairs, Zeria Pharmaceutical Co, Ltd, Chuo-ku, Tokyo, Japan
| | - Yuko Hori
- Central Research Laboratories, Zeria Pharmaceutical Co, Ltd, Kumagaya-shi, Saitama, Japan
| | - Yuta Ohira
- Central Research Laboratories, Zeria Pharmaceutical Co, Ltd, Kumagaya-shi, Saitama, Japan
| | - Kenji Nagahama
- Central Research Laboratories, Zeria Pharmaceutical Co, Ltd, Kumagaya-shi, Saitama, Japan
| | - Tomoko Ozaki
- Clinical Administration, Zeria Pharmaceutical Co, Ltd, Chuo-ku, Tokyo, Japan
| | - Mineo Takei
- Central Research Laboratories, Zeria Pharmaceutical Co, Ltd, Kumagaya-shi, Saitama, Japan
| | - Takao Tanaka
- Central Research Laboratories, Zeria Pharmaceutical Co, Ltd, Kumagaya-shi, Saitama, Japan
| | - Tomohide Uno
- Agrobioscience, Graduate School of Agricultural Science, Kobe University, Nada-ku, Hyogo, Japan
| |
Collapse
|
62
|
Soleyman-Jahi S, Sadeghi F, Pastaki Khoshbin A, Khani L, Roosta V, Zendehdel K. Attribution of Ghrelin to Cancer; Attempts to Unravel an Apparent Controversy. Front Oncol 2019; 9:1014. [PMID: 31681567 PMCID: PMC6805778 DOI: 10.3389/fonc.2019.01014] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 09/20/2019] [Indexed: 12/21/2022] Open
Abstract
Ghrelin is an endogenous peptide hormone mainly produced in the stomach. It has been known to regulate energy homeostasis, stimulate secretion of growth hormone, and mediate many other physiologic effects. Various effects attributed to ghrelin contribute to many aspects of cancer development and progression. Accordingly, a large body of evidence has emerged about the association of ghrelin with several types of cancer in scales of cell-line, animal, and human studies. However, existing data are controversial. This controversy occurs in two main domains: one is the controversial results in local effects of ghrelin on different types of human cancer cell-lines; the second is the apparent disagreement in the results of in-vitro and clinical studies that investigated ghrelin association to one type of cancer. These inconsistencies have hampered the indications to consider ghrelin as a potential tumor biomarker or therapeutic agent in cancer patients. Previous studies have reviewed different parts of current literature about the ghrelin-cancer relationship. Although they have highlighted these controversial results in various ways, no specific recommendations have been given to address it. In this study, we comprehensively reviewed in-vitro, in-vivo, and clinical studies and attempted to use the following approaches to unravel the inconsistencies detected: (a) to distinguish local and systemic effects of ghrelin in interpreting its summary clinical role in each cancer; (b) scrutinizing factors that regulate local effects of ghrelin and could justify different effects of ghrelin on different cancer cell-lines. These approaches could have notable implications for future in-vitro and clinical studies.
Collapse
Affiliation(s)
- Saeed Soleyman-Jahi
- Division of Gastroenterology, School of Medicine, Washington University in St. Louis, St. Louis, MO, United States.,Cancer Immunology Project, Universal Scientific Education and Research Network, St. Louis, MO, United States.,Cancer Research Center, Cancer Institute of Iran, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Sadeghi
- Cancer Research Center, Cancer Institute of Iran, Tehran University of Medical Sciences, Tehran, Iran.,Cancer Immunology Project, Universal Scientific Education and Research Network, Tehran, Iran.,Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Amin Pastaki Khoshbin
- Cancer Immunology Project, Universal Scientific Education and Research Network, Tehran, Iran.,School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Leila Khani
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Venus Roosta
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Kazem Zendehdel
- Cancer Research Center, Cancer Institute of Iran, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
63
|
Dunn DP, Bastacky JM, Gray CC, Abtahi S, Currie PJ. Role of mesolimbic ghrelin in the acquisition of cocaine reward. Neurosci Lett 2019; 709:134367. [DOI: 10.1016/j.neulet.2019.134367] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 07/01/2019] [Accepted: 07/02/2019] [Indexed: 12/27/2022]
|
64
|
Walker WH, Borniger JC. Molecular Mechanisms of Cancer-Induced Sleep Disruption. Int J Mol Sci 2019; 20:E2780. [PMID: 31174326 PMCID: PMC6600154 DOI: 10.3390/ijms20112780] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 06/03/2019] [Accepted: 06/04/2019] [Indexed: 02/07/2023] Open
Abstract
Sleep is essential for health. Indeed, poor sleep is consistently linked to the development of systemic disease, including depression, metabolic syndrome, and cognitive impairments. Further evidence has accumulated suggesting the role of sleep in cancer initiation and progression (primarily breast cancer). Indeed, patients with cancer and cancer survivors frequently experience poor sleep, manifesting as insomnia, circadian misalignment, hypersomnia, somnolence syndrome, hot flushes, and nightmares. These problems are associated with a reduction in the patients' quality of life and increased mortality. Due to the heterogeneity among cancers, treatment regimens, patient populations and lifestyle factors, the etiology of cancer-induced sleep disruption is largely unknown. Here, we discuss recent advances in understanding the pathways linking cancer and the brain and how this leads to altered sleep patterns. We describe a conceptual framework where tumors disrupt normal homeostatic processes, resulting in aberrant changes in physiology and behavior that are detrimental to health. Finally, we discuss how this knowledge can be leveraged to develop novel therapeutic approaches for cancer-associated sleep disruption, with special emphasis on host-tumor interactions.
Collapse
Affiliation(s)
- William H Walker
- Department of Neuroscience, West Virginia University, Morgantown, WV 26506, USA.
| | - Jeremy C Borniger
- Department of Psychiatry & Behavioral Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
65
|
Meta-analysis of ghrelin alterations in schizophrenia: Effects of olanzapine. Schizophr Res 2019; 206:21-26. [PMID: 30528312 DOI: 10.1016/j.schres.2018.11.036] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 08/21/2018] [Accepted: 11/30/2018] [Indexed: 01/04/2023]
Abstract
OBJECTIVE Schizophrenia is associated with an increased prevalence of the metabolic syndrome. Patients receiving antipsychotic medications, including olanzapine, are at further risk. Ghrelin is an appetite-stimulating peptide hormone, although whether blood levels are altered by antipsychotic treatment, remains unclear. We performed a systematic review and meta-analysis comparing blood ghrelin levels in patients with schizophrenia before and after treatment with olanzapine. METHOD Two authors independently searched major electronic databases from inception until February 2018 for studies measuring blood ghrelin levels among patients with schizophrenia before and after olanzapine therapy. Random effects meta-analysis calculating standardized mean difference (SMD) and 95% confidence intervals (CI) and meta-regression analyses were performed. RESULTS Six studies met the inclusion criteria. Across these studies, there were 111 patients with schizophrenia (mean age 40, 85% male, baseline BMI 22, and endpoint BMI 23). Olanzapine treatment (mean [standard deviation] duration = 12.3 [7.6] weeks) was associated with a significant decrease in blood ghrelin levels with a medium effect size (SMD = -0.48, 95% CI -0.88 to -0.08, p = 0.018). Age, sex, baseline BMI, geography, olanzapine dose and duration, year of publication, study quality, inpatient status, and antipsychotic washout did not moderate this association. CONCLUSION Our results suggest that in patients with schizophrenia, olanzapine therapy is associated with decreased blood ghrelin levels, a paradoxical phenomenon known to occur in obesity. Future studies should investigate the contribution of dietary factors (e.g., caloric intake) and physical activity to this association, as well as the effects of other antipsychotics on ghrelin levels.
Collapse
|
66
|
Cao X, Zhu M, He Y, Chu W, Du Y, Du H. Increased Serum Acylated Ghrelin Levels in Patients with Mild Cognitive Impairment. J Alzheimers Dis 2019; 61:545-552. [PMID: 29226871 DOI: 10.3233/jad-170721] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Ghrelin is a stomach-derived circulating hormone. In addition to its function as an orexigenic stimulant, the role of ghrelin in the consolidation of learning and memory has been implicated in recent years. However, the status of circulating acylated ghrelin (AG, that is, the functional form of ghrelin) in the symptomatic predementia stage of Alzheimer's disease (AD) has rarely been investigated. In the current study, we examined the serum levels of acylated and total ghrelin in 22 patients with mild cognitive impairment (MCI) and 30 cognitively normal controls. We have found that patients with MCI had significantly increased serum AG levels, which were inversely associated with defected short- and long-term memory as well as language skills. Of note, the levels of total circulating ghrelin were similar between the two groups. Intriguingly, serum AG but not total ghrelin was associated with AD risk factors including the age, hypertension, and hyperlipidemia. Therefore, circulating AG may serve as a potential early systemic biomarker for AD-related cognitive impairments. Nevertheless, the simplest interpretation of the results is that the levels of circulating AG are associated with cognitive impairments in patients with MCI, thereby forming the groundwork for our future studies on the systemic mechanisms of AD pertaining to the ghrelin system.
Collapse
Affiliation(s)
- Xi Cao
- Department of Neurology, Alzheimer's Disease Center, Qianfoshan Hospital affiliated to Shandong University, Jinan, Shandong, PRC
| | - Min Zhu
- Department of Neurology, Alzheimer's Disease Center, Qianfoshan Hospital affiliated to Shandong University, Jinan, Shandong, PRC
| | - Yan He
- Department of Neurology, Alzheimer's Disease Center, Qianfoshan Hospital affiliated to Shandong University, Jinan, Shandong, PRC
| | - Wenzheng Chu
- Department of Neurology, Alzheimer's Disease Center, Qianfoshan Hospital affiliated to Shandong University, Jinan, Shandong, PRC
| | - Yifeng Du
- Department of Neurology, Shandong Provincial Hospital, Jinan, Shandong, PRC
| | - Heng Du
- Department of Neurology, Alzheimer's Disease Center, Qianfoshan Hospital affiliated to Shandong University, Jinan, Shandong, PRC.,Department of Biological Sciences, the University of Texas, Dallas, Richardson, TX, USA
| |
Collapse
|
67
|
Bucur-Grosu ML, Avasiloaiei A, Moscalu M, Dimitriu DC, Păduraru L, Stamatin M. DESACYLATED GHRELIN AND LEPTIN IN THE CORD BLOOD OF SMALL-FOR-GESTATIONAL-AGE NEWBORNS WITH INTRAUTERINE GROWTH RESTRICTION. ACTA ENDOCRINOLOGICA-BUCHAREST 2019; 15:305-310. [PMID: 32010348 DOI: 10.4183/aeb.2019.305] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Context Ghrelin, in both its acylated and desacylated forms, and leptin can modulate fetal energy balance and development. Objective The aim of our study is to assess desacylated ghrelin (DAG) and leptin values and influence on intrauterine and postnatal growth in infants with intrauterine growth restriction. Design subjects and methods We performed a prospective study on 39 infants recruited over five months, 20 appropriate - for - gestational - age (AGA) infants and 19 small-for-gestational-age (SGA) infants, in which we measured DAG and leptin in the umbilical cord blood and we compared their respective values between the two groups, along with auxological parameters at birth and at 10 months of postnatal age. Results Our results show that both DAG and leptin have lower values in SGA infants and correlate with most of the anthropometrical parameters at birth. Both hormones correlate with weight at 10 months in SGA infants, but this correlation lacks in AGA infants. Whereas DAG in the cord blood can be considered a predictor for weight at 10 months (β=0.207, p=0.001), the same cannot be stated about leptin (β=0.078, p=0.195). Conclusion DAG and leptin are involved in both intrauterine and postnatal development, but the extent of their role is still to be determined.
Collapse
Affiliation(s)
- M L Bucur-Grosu
- "Grigore T. Popa" University of Medicine and Pharmacy, Department of Mother and Child Health, Iasi, Romania.,"Cuza-Voda" Clinical Hospital of Obstetrics and Gynecology, Neonatal Intensive Care Unit, Iasi, Romania
| | - A Avasiloaiei
- "Grigore T. Popa" University of Medicine and Pharmacy, Department of Mother and Child Health, Iasi, Romania.,"Cuza-Voda" Clinical Hospital of Obstetrics and Gynecology, Neonatal Intensive Care Unit, Iasi, Romania
| | - M Moscalu
- "Grigore T. Popa" University of Medicine and Pharmacy, Department of Medical Informatics and Biostatistics, Iasi, Romania
| | - D C Dimitriu
- "Grigore T. Popa" University of Medicine and Pharmacy, Department of Biochemistry, Iasi, Romania
| | - L Păduraru
- "Grigore T. Popa" University of Medicine and Pharmacy, Department of Mother and Child Health, Iasi, Romania.,"Cuza-Voda" Clinical Hospital of Obstetrics and Gynecology, Neonatal Intensive Care Unit, Iasi, Romania
| | - M Stamatin
- "Grigore T. Popa" University of Medicine and Pharmacy, Department of Mother and Child Health, Iasi, Romania.,"Cuza-Voda" Clinical Hospital of Obstetrics and Gynecology, Neonatal Intensive Care Unit, Iasi, Romania
| |
Collapse
|
68
|
Sleep Deprivation Selectively Upregulates an Amygdala-Hypothalamic Circuit Involved in Food Reward. J Neurosci 2018; 39:888-899. [PMID: 30559151 DOI: 10.1523/jneurosci.0250-18.2018] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 09/18/2018] [Accepted: 10/15/2018] [Indexed: 01/30/2023] Open
Abstract
Sleep loss is associated with increased obesity risk, as demonstrated by correlations between sleep duration and change in body mass index or body fat percentage. Whereas previous studies linked this weight gain to disturbed endocrine parameters after sleep deprivation or restriction, neuroimaging studies revealed upregulated neural processing of food rewards after sleep loss in reward-processing areas such as the anterior cingulate cortex, ventral striatum, and insula. To address this ongoing debate between hormonal versus hedonic factors underlying sleep-loss-associated weight gain, we rigorously tested the association between sleep deprivation and food cue processing using high-resolution fMRI and assessment of hormones. After taking blood samples from 32 lean, healthy, human male participants, they underwent fMRI while performing a neuroeconomic, value-based decision-making task with snack food and trinket rewards following a full night of habitual sleep and a night of sleep deprivation in a repeated-measures crossover design. We found that des-acyl ghrelin concentrations were increased after sleep deprivation compared with habitual sleep. Despite similar hunger ratings due to fasting in both conditions, participants were willing to spend more money on food items only after sleep deprivation. Furthermore, fMRI data paralleled this behavioral finding, revealing a food-reward-specific upregulation of hypothalamic valuation signals and amygdala-hypothalamic coupling after a single night of sleep deprivation. Behavioral and fMRI results were not significantly correlated with changes in acyl, des-acyl, or total ghrelin concentrations. Our results suggest that increased food valuation after sleep loss might be due to hedonic rather than hormonal mechanisms.SIGNIFICANCE STATEMENT Epidemiological studies suggest an association between overweight and reduced nocturnal sleep, but the relative contributions of hedonic and hormonal factors to overeating after sleep loss are a matter of ongoing controversy. Here, we tested the association between sleep deprivation and food cue processing in a repeated-measures crossover design using fMRI. We found that willingness to pay increased for food items only after sleep deprivation. fMRI data paralleled this behavioral finding, revealing a food-reward-specific upregulation of hypothalamic valuation signals and amygdala-hypothalamic coupling after a single night of sleep deprivation. However, there was no evidence for hormonal modulations of behavioral or fMRI findings. Our results suggest that increased food valuation after sleep loss is due to hedonic rather than hormonal mechanisms.
Collapse
|
69
|
Leptin and ghrelin in chronic kidney disease: their associations with protein-energy wasting. Pediatr Nephrol 2018; 33:2113-2122. [PMID: 29980850 DOI: 10.1007/s00467-018-4002-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 05/22/2018] [Accepted: 06/13/2018] [Indexed: 10/28/2022]
Abstract
BACKGROUND This study aimed to evaluate plasma concentrations of leptin and total ghrelin in children with chronic kidney disease (CKD) and assess their roles in protein-energy wasting (PEW). METHODS This study consisted of three different CKD populations [CKD group (20 patients with non-dialysis CKD), dialysis group (39 patients on dialysis), and kidney transplant (KTx) group (35 KTx recipients)] and control group (18 healthy children). Plasma leptin and total ghrelin levels were measured. Multi-frequency bioimpedance analysis was used for the assessment of fat and lean mass. PEW was defined using criteria including body mass, muscle mass, growth, serum albumin level, and protein intake. RESULTS While plasma leptin levels did not differ among the study groups, total ghrelin levels were significantly higher in the dialysis group (P < 0.001). Seven dialysis patients (18%) and one CKD patient (5%) but none of the KTx recipients met the criteria of PEW. Dialysis patients with PEW had lower plasma leptin levels compared to their counterparts (P = 0.018); however, total ghrelin levels did not differ between the two groups (P = 0.10). Low leptin level in dialysis patients was independently associated with lower fat mass index (P < 0.001) and lower height-specific SD scores of BMI (P = 0.019). CONCLUSIONS PEW is prevalent in dialysis patients. Low levels of leptin seem to be associated with PEW. Our result suggests that low leptin levels may be a consequence rather than a cause of PEW. Longitudinal studies are required to investigate this complex relationship between leptin and PEW in pediatric dialysis patients.
Collapse
|
70
|
[DTPA-(PABn)-Leu5]-des-acyl ghrelin(1-5) as a new carrier of radionuclides and potential precursor of radiopharmaceuticals. Nucl Med Commun 2018; 39:140-146. [PMID: 29315139 DOI: 10.1097/mnm.0000000000000790] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Ghrelin is a peptide consisting of 28 aminoacids and an octadecyl side chain (acyl group) binding to the growth hormone secretagogue receptor type 1a (GHS-R1a). Its des-acylated form, des-acyl ghrelin (DAG) binds to the corticotropin releasing factor receptor type 2a (CRF2a) located on endocrine cancer cells such as the prostate carcinoma cell line DU 145. AIM The aim of this study is to develop a new DAG-based carrier of radionuclides with potential application in therapy. MATERIALS AND METHODS Trunctated C-terminal five aminoacids chain of the DAG peptide (H2N-Gly-Ser-Ser-Phe-Leu-COOH) was linked to DTPA to obtain [DTPA-(PABn)-Leu5]-DAG(1-5). For therapeutic application the lutetium-177 (177Lu) radionuclide was coordinated to the peptide. To determine biological and chemical properties of newly synthesized radiopharmaceutical, two iodine-131 (131I)-labelled compounds were used: [131I]-Tyr4-DAG(1-5) and full length [131I]-DAG(1-28) together with their nonradioactive forms: DAG(1-28) and DAG(1-5). RESULTS Identical HPLC elution profiles of [177Lu-DTPA-(PABn)-Leu5]-DAG(1-5) before and after incubation with human serum proved its stability. The lipophilicity profile of [177Lu-DTPA-(PABn)-Leu5]-DAG(1-5) was log DO/W=-2.68±0.05, pH 7.4. Receptor affinity of the nonradioactive conjugate [Lu-DTPA-(PABn)-Leu5]-DAG(1-5) was IC50 (21.06 nmol/l), as shown against the [131I]-DAG(1-28) used as a competitor. The 3-(4,5-dimethyldiazol-2-yl)-2,5-diphenyltetrazolium bromide assay indicated the significant cytotoxicity of the newly synthesized compounds, similar to that of [131I]-Tyr4-DAG(1-5). CONCLUSION The results obtained suggest the potency of the [DTPA-(PABn)-Leu5]-DAG(1-5) as a new carrier of radionuclides in radiopharmacy.
Collapse
|
71
|
Liu SY, Huang CH, Shieh JC, Lee TL. Cinnamomum osmophloeum Kanehira ethanol extracts prevents human liver-derived HepG2 cell death from oxidation stress by induction of ghrelin gene expression. J Biosci 2018; 42:439-448. [PMID: 29358557 DOI: 10.1007/s12038-017-9697-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Diabetes patients associated with liver disease carry a significant risk of morbidity and mortality. Cinnamon has been reported to reduce fructose-induced oxidative stress in the rat liver. However, the mechanism by which cinnamon protects the liver in a high-saccharide environment remains to be investigated. HepG2 cells were cultured with 30 mM D-ribose to mimic the high-oxidative-stress environment, typical of a liver in a diabetic patient. Three different chemical types of C. osmophloeum ethanol extracts (CEEs) were added in HepG2 culture media and the administration of all three CEEs protected HepG2 cells from D-ribose damage and increased cell survival by approximately 20 percent. Exclusively, the transcript variant 1 of the ghrelin gene, but not variant 3, was 2-3 times induced by the addition of these CEEs. Moreover, the mRNAs of ghrelin processing enzyme, furin, and mboat4 were detected in HepG2 cells. The ghrelin hormones in the culture media were increased 4-9 times by the addition of CEEs. The protective effects of ghrelin on HepG2 cells in D-ribose environment were further confirmed by recombinant ghrelin transfection. We conclude that the CEEs induce ghrelin gene expression and protect HepG2 cells from D-ribose-induced oxidative damage through ghrelin signalling.
Collapse
Affiliation(s)
- Shu-Ying Liu
- Department of Molecular Biotechnology, Da-Yeh University, Changhua, Taiwan
| | | | | | | |
Collapse
|
72
|
MK-0677, a Ghrelin Agonist, Alleviates Amyloid Beta-Related Pathology in 5XFAD Mice, an Animal Model of Alzheimer's Disease. Int J Mol Sci 2018; 19:ijms19061800. [PMID: 29912176 PMCID: PMC6032329 DOI: 10.3390/ijms19061800] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 05/31/2018] [Accepted: 06/06/2018] [Indexed: 12/11/2022] Open
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disorder characterized by cognitive deficits, neuroinflammation, and neuronal death. The primary pathogenic cause is believed to be the accumulation of pathogenic amyloid beta (Aβ) assemblies in the brain. Ghrelin, which is a peptide hormone predominantly secreted from the stomach, is an endogenous ligand for the growth hormone secretagogue-receptor type 1a (GHS-R1a). MK-0677 is a ghrelin agonist that potently stimulates the GHS-R1a ghrelin receptor. Interestingly, previous studies have shown that ghrelin improves cognitive impairments and attenuates neuronal death and neuroinflammation in several neurological disorders. However, it is unknown whether MK-0677 can affect Aβ accumulation or Aβ-mediated pathology in the brains of patients with AD. Therefore, we examined the effects of MK-0677 administration on AD-related pathology in 5XFAD mice, an Aβ-overexpressing transgenic mouse model of AD. MK-0677 was intraperitoneally administered to three-month-old 5XFAD mice. To visualize Aβ accumulation, neuroinflammation, and neurodegeneration, thioflavin-S staining and immunostaining with antibodies against Aβ (4G8), ionized calcium-binding adaptor molecule 1 (Iba-1), glial fibrillary acidic protein (GFAP), neuronal nuclear antigen (NeuN), and synaptophysin were conducted in the neocortex of 5XFAD and wild-type mice, and to evaluate changes of phosphorylated cyclic adenosine monophosphate (cAMP) response element binding protein (pCREB) levels, immunostaining with antibody against pCREB was performed in dentate gyrus of the hippocampus of 5XFAD and wild-type mice. The histological analyses indicated that MK-0677-treated 5XFAD mice showed reduced Aβ deposition, gliosis, and neuronal and synaptic loss in the deep cortical layers, and inhibited the decrement of pCREB levels in dentate gyrus of the hippocampus compared to vehicle-treated 5XFAD mice. Our results showed that activation of the ghrelin receptor with MK-0677 inhibited the Aβ burden, neuroinflammation, and neurodegeneration, which suggested that MK-0677 might have potential as a treatment of the early phase of AD.
Collapse
|
73
|
Ikenoya C, Takemi S, Kaminoda A, Aizawa S, Ojima S, Gong Z, Chacrabati R, Kondo D, Wada R, Tanaka T, Tsuda S, Sakai T, Sakata I. β-Oxidation in ghrelin-producing cells is important for ghrelin acyl-modification. Sci Rep 2018; 8:9176. [PMID: 29907775 PMCID: PMC6003948 DOI: 10.1038/s41598-018-27458-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 06/01/2018] [Indexed: 12/30/2022] Open
Abstract
Ghrelin is a unique fatty acid-modified peptide hormone produced in the stomach and has important roles in energy homeostasis and gastrointestinal motility. However, the medium-chain fatty acid source for ghrelin acyl-modification is not known. We found that a fat-free diet and the removal of intestinal microbiota did not decrease acyl-ghrelin production in the stomach or plasma acyl-ghrelin levels in mice. RT-PCR analysis showed that genes involving fatty acid synthesis, metabolism, and transport were expressed in pancreas-derived ghrelinoma (PG-1) cells. Treatment with an irreversible inhibitor of carnitine palmitoyltransferase-1 (CPT-1) strongly decreased acylated ghrelin levels but did not affect ghrelin or ghrelin o-acyl transferase (GOAT) mRNA levels in PG-1 cells. Our results suggest that the medium-chain fatty acid used for the acyl-modification of ghrelin is produced in ghrelin-producing cells themselves by β-oxidation of long-chain fatty acids provided from the circulation.
Collapse
Affiliation(s)
- Chika Ikenoya
- Area of Regulatory Biology, Division of Life Science, Graduate School of Science and Engineering, Saitama University, 255 Shimo-ohkubo, Sakuraku, Saitama, 338-8570, Japan
| | - Shota Takemi
- Area of Regulatory Biology, Division of Life Science, Graduate School of Science and Engineering, Saitama University, 255 Shimo-ohkubo, Sakuraku, Saitama, 338-8570, Japan
| | - Arisa Kaminoda
- Department of Biology, Graduate School of Natural Science and Technology, Okayama University, 3-1-1, Tsushimanaka, Kita-ku, Okayama, 700-8530, Japan
| | - Sayaka Aizawa
- Department of Biology, Graduate School of Natural Science and Technology, Okayama University, 3-1-1, Tsushimanaka, Kita-ku, Okayama, 700-8530, Japan
| | - Shiomi Ojima
- Area of Regulatory Biology, Division of Life Science, Graduate School of Science and Engineering, Saitama University, 255 Shimo-ohkubo, Sakuraku, Saitama, 338-8570, Japan
| | - Zhi Gong
- Area of Regulatory Biology, Division of Life Science, Graduate School of Science and Engineering, Saitama University, 255 Shimo-ohkubo, Sakuraku, Saitama, 338-8570, Japan
| | - Rakhi Chacrabati
- Area of Regulatory Biology, Division of Life Science, Graduate School of Science and Engineering, Saitama University, 255 Shimo-ohkubo, Sakuraku, Saitama, 338-8570, Japan
| | - Daisuke Kondo
- Area of Regulatory Biology, Division of Life Science, Graduate School of Science and Engineering, Saitama University, 255 Shimo-ohkubo, Sakuraku, Saitama, 338-8570, Japan
| | - Reiko Wada
- Area of Regulatory Biology, Division of Life Science, Graduate School of Science and Engineering, Saitama University, 255 Shimo-ohkubo, Sakuraku, Saitama, 338-8570, Japan
| | - Toru Tanaka
- Faculty of Pharmaceutical Sciences, Department of Pharmaceutical and Health Sciences, Josai University, 1-1 Keiyaki dai, Sakado, Saitama, 350-0295, Japan
| | - Sachiko Tsuda
- Area of Regulatory Biology, Division of Life Science, Graduate School of Science and Engineering, Saitama University, 255 Shimo-ohkubo, Sakuraku, Saitama, 338-8570, Japan.,Research and Development Bureau, Saitama University, 255 Shimo-ohkubo, Sakuraku, Saitama, 338-8570, Japan
| | - Takafumi Sakai
- Area of Regulatory Biology, Division of Life Science, Graduate School of Science and Engineering, Saitama University, 255 Shimo-ohkubo, Sakuraku, Saitama, 338-8570, Japan.,Area of Life-NanoBio, Division of Strategy Research, Graduate School of Science and Engineering, Saitama University, 255 Shimo-okubo, Sakura-ku, Saitama, 338-8570, Japan
| | - Ichiro Sakata
- Area of Regulatory Biology, Division of Life Science, Graduate School of Science and Engineering, Saitama University, 255 Shimo-ohkubo, Sakuraku, Saitama, 338-8570, Japan.
| |
Collapse
|
74
|
Uchida Y, Nagashima K, Yuri K. Fasting or systemic des-acyl ghrelin administration to rats facilitates thermoregulatory behavior in a cold environment. Brain Res 2018; 1696:10-21. [PMID: 29859973 DOI: 10.1016/j.brainres.2018.05.038] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 05/19/2018] [Accepted: 05/24/2018] [Indexed: 10/14/2022]
Abstract
Fasted rats place their tails underneath their body trunks in the cold (tail-hiding behavior), which is a thermoregulatory behavior. The aim of the present study was to investigate the effect of fasting and des-acyl ghrelin, a hormone related to fasting, on tail-hiding behavior and neural activity in the cold. Wistar rats were divided into 'fed', '42-h fasting' and des-acyl ghrelin groups. The rats received an intraperitoneal saline or 30-μg des-acyl ghrelin injection, and were then exposed to 27 °C or 15 °C for 2-h with continuous body temperature (Tb), tail skin temperature (Ttail), and tail-hiding behavior measurements. cFos immunoreactive (cFos-IR) cells in the insula, secondary somatosensory cortex, medial preoptic nucleus, parastrial nucleus, amygdala, and lateral parabrachial nucleus were counted in four segments: seg1, 2, 3, and 4 (bregma -0.36, -1.44, -2.64, and -9.00 mm), respectively. At 15 °C, Tb and Ttail were lower in the 42-h fasting group than in the fed and des-acyl ghrelin groups, and the duration of tail-hiding behavior was longer in the 42-h fasting and des-acyl ghrelin groups than in the fed group. The onset of tail-hiding behavior more advanced in the des-acyl ghrelin group than in the fed group at 15 °C. Only at the insula in seg3 at 15 °C, the number of cFos-IR cells was greater in the 42-h fasting group than in the fed group. Both the 42-h fasting and des-acyl ghrelin groups might modulate the tail-hiding behavior of rats in a cold, and a part of the insula might be involved this response during fasting.
Collapse
Affiliation(s)
- Yuki Uchida
- Department of Neurobiology and Anatomy, Kochi Medical School, Kochi University, Kochi, Japan.
| | - Kei Nagashima
- Laboratory of Integrative Physiology (Body Temperature and Fluid Laboratory), Faculty of Human Sciences, Japan; Institute of Applied Brain Sciences, Waseda University, Saitama, Japan
| | - Kazunari Yuri
- Department of Neurobiology and Anatomy, Kochi Medical School, Kochi University, Kochi, Japan
| |
Collapse
|
75
|
Morris LS, Voon V, Leggio L. Stress, Motivation, and the Gut-Brain Axis: A Focus on the Ghrelin System and Alcohol Use Disorder. Alcohol Clin Exp Res 2018; 42:10.1111/acer.13781. [PMID: 29797564 PMCID: PMC6252147 DOI: 10.1111/acer.13781] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 05/17/2018] [Indexed: 01/04/2023]
Abstract
Since its discovery, the gut hormone, ghrelin, has been implicated in diverse functional roles in the central nervous system. Central and peripheral interactions between ghrelin and other hormones, including the stress-response hormone cortisol, govern complex behavioral responses to external cues and internal states. By acting at ventral tegmental area dopaminergic projections and other areas involved in reward processing, ghrelin can induce both general and directed motivation for rewards, including craving for alcohol and other alcohol-seeking behaviors. Stress-induced increases in cortisol seem to increase ghrelin in the periphery, suggesting a pathway by which ghrelin influences how stressful life events trigger motivation for rewards. However, in some states, ghrelin may be protective against the anxiogenic effects of stressors. This critical review brings together a dynamic and growing literature, that is, at times inconsistent, on the relationships between ghrelin, central reward-motivation pathways, and central and peripheral stress responses, with a special focus on its emerging role in the context of alcohol use disorder.
Collapse
Affiliation(s)
- Laurel S. Morris
- Behavioural and Clinical Neuroscience Institute, University of Cambridge, UK
- Department of Psychology, University of Cambridge, UK
- Mood and Anxiety Disorders Program, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Valerie Voon
- Behavioural and Clinical Neuroscience Institute, University of Cambridge, UK
- Department of Psychiatry, University of Cambridge, UK
| | - Lorenzo Leggio
- Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology, National Institute on Alcohol Abuse and Alcoholism and National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD, USA
- Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, Brown University, Providence, RI, USA
| |
Collapse
|
76
|
Fasting and postprandial acyl and desacyl ghrelin and the acyl/desacyl ratio in obese patients before and after different types of bariatric surgery. Wideochir Inne Tech Maloinwazyjne 2018; 13:366-375. [PMID: 30302150 PMCID: PMC6174170 DOI: 10.5114/wiitm.2018.75868] [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: 12/19/2017] [Accepted: 03/04/2018] [Indexed: 12/19/2022] Open
Abstract
Introduction The mechanism underlying beneficial outcomes of bariatric surgery still remains unclear. Especially little is known about hormonal and metabolic changes induced by the novel bariatric procedure mini gastric bypass (MGB). Aim To evaluate pre- and post-prandial changes in both ghrelin isoforms in obese patients without diabetes and cardiovascular complications treated with MGB, sleeve gastrectomy (SG) or Roux-en-Y gastric bypass (RYGB) surgery. Material and methods From 45 patients initially enrolled in the study, 23 persons completed a one-year follow-up period. Venous blood for acyl and desacyl ghrelin (AG and DAG) as well as other metabolic assays was collected 3 months before and 6 and 12 months after bariatric surgery (MGB, RYGB, SG) – in the fasting state and 2 h after the consumption of a standard 300 kcal-mixed meal (Nutridrink standard, Nutricia). Results AG and DAG levels (both fasting and prandial) as well as AG/DAG ratio did not change after 6 and 12 months in MGB and RYGB groups. In the SG group we observed a significant decrease in fasting and postprandial DAG levels and consecutively an increase in the fasting AG/DAG ratio after 6 and 12 months. Six months after surgery we observed some differences between carbohydrate metabolism measures in the MGB group (lower HbA1c, HOMA-IR and fasting insulinaemia) in comparison to the rest of the participants, but 12 months after each type of surgery body mass index and indices of carbohydrate and lipid metabolism did not differ. Conclusions The results of our study demonstrate that all studied bariatric procedures can successfully reduce overall body weight and suggest also that the mechanisms of weight loss and improvement in carbohydrate and lipid metabolism after all three types of surgery are independent of ghrelin and the acyl/desacyl ghrelin ratio.
Collapse
|
77
|
Monzani A, Perrone M, Prodam F, Moia S, Genoni G, Testa S, Paglialonga F, Rapa A, Bona G, Montini G, Edefonti A. Unacylated ghrelin and obestatin: promising biomarkers of protein energy wasting in children with chronic kidney disease. Pediatr Nephrol 2018; 33:661-672. [PMID: 29150712 DOI: 10.1007/s00467-017-3840-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 08/17/2017] [Accepted: 10/23/2017] [Indexed: 12/01/2022]
Abstract
BACKGROUND Impairment in orexigenic/anorexigenic hormone balance may be key in the pathogenesis of protein energy wasting in children with chronic kidney disease (CKD). Measurement of ghrelin and obestatin concentrations in children with CKD would help assess the potential contribution of these hormones to uremic protein energy wasting. METHODS This was a cross-sectional case-control study. Acylated and unacylated ghrelin and obestatin were measured in 42 children on conservative treatment (CT), 20 children on hemodialysis, 48 pediatric renal transplant (RTx) recipients and 43 controls (CTR) (mean age 11.9, range 5-20 years). Weight, height and bicipital, tricipital, subscapular and suprailiac folds were measured, and the body mass index-standard deviation score (BMI-SDS), percentage of fat mass and fat-free mass were calculated. Urea and creatinine were measured and the glomerular filtration rate (GFR) calculated. RESULTS Unacylated ghrelin level was higher in patients than controls (p = 0.0001), with the highest levels found in hemodialysis patients (p = 0.001 vs. CKD-CT, p = 0.0001 vs. RTx, p < 0.0001 vs. CTR). Obestatin level was significantly higher in patients on hemodialysis than those on conservative treatment, RTx recipients and controls (p < 0.0001 in each case). Unacylated ghrelin negatively correlated with weight-SDS (p < 0.0001), BMI-SDS (p = 0.0005) and percentage fat mass (p = 0.004) and positively correlated with percentage fat-free mass (p = 0.004). Obestatin concentration negatively correlated with weight-SDS (p = 0.007). Unacylated ghrelin and obestatin concentrations positively correlated with creatinine and urea and inversely with eGFR, even after adjustments for gender, age, puberty and BMI-SDS (p < 0.0001 for each model). CONCLUSIONS Unacylated ghrelin and obestatin, negatively related to renal function, seem to be promising inverse indicators of nutritional status in children with CKD. Potential therapeutic implications in terms of optimization of their removal in patients on hemodialysis could be hypothesized.
Collapse
Affiliation(s)
- Alice Monzani
- Division of Pediatrics, Department of Health Sciences, Università del Piemonte Orientale, V. Solaroli 17, 28100, Novara, Italy
| | - Michela Perrone
- Pediatric Nephrology, Dialysis and Transplant Unit, Fondazione Cà Granda IRCCS, Ospedale Maggiore Policlinico, Milan, Italy
| | - Flavia Prodam
- Division of Pediatrics, Department of Health Sciences, Università del Piemonte Orientale, V. Solaroli 17, 28100, Novara, Italy
| | - Stefania Moia
- Division of Pediatrics, Department of Health Sciences, Università del Piemonte Orientale, V. Solaroli 17, 28100, Novara, Italy
| | - Giulia Genoni
- Division of Pediatrics, Department of Health Sciences, Università del Piemonte Orientale, V. Solaroli 17, 28100, Novara, Italy.
| | - Sara Testa
- Pediatric Nephrology, Dialysis and Transplant Unit, Fondazione Cà Granda IRCCS, Ospedale Maggiore Policlinico, Milan, Italy
| | - Fabio Paglialonga
- Pediatric Nephrology, Dialysis and Transplant Unit, Fondazione Cà Granda IRCCS, Ospedale Maggiore Policlinico, Milan, Italy
| | - Anna Rapa
- Division of Pediatrics, Department of Health Sciences, Università del Piemonte Orientale, V. Solaroli 17, 28100, Novara, Italy
| | - Gianni Bona
- Division of Pediatrics, Department of Health Sciences, Università del Piemonte Orientale, V. Solaroli 17, 28100, Novara, Italy
| | - Giovanni Montini
- Pediatric Nephrology, Dialysis and Transplant Unit, Fondazione Cà Granda IRCCS, Ospedale Maggiore Policlinico, Milan, Italy.,Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Alberto Edefonti
- Pediatric Nephrology, Dialysis and Transplant Unit, Fondazione Cà Granda IRCCS, Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
78
|
Viltart O, Duriez P, Tolle V. Metabolic and neuroendocrine adaptations to undernutrition in anorexia nervosa: from a clinical to a basic research point of view. Horm Mol Biol Clin Investig 2018; 36:hmbci-2018-0010. [PMID: 29804101 DOI: 10.1515/hmbci-2018-0010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 03/13/2018] [Indexed: 02/07/2023]
Abstract
The exact mechanisms linking metabolic and neuroendocrine adaptations to undernutrition and the pathophysiology of anorexia nervosa (AN) are not fully understood. AN is a psychiatric disorder of complex etiology characterized by extreme starvation while the disease is progressing into a chronic state. Metabolic and endocrine alterations associated to this disorder are part of a powerful response to maintain whole body energy homeostasis. But these modifications may also contribute to associated neuropsychiatric symptoms (reward abnormalities, anxiety, depression) and thus participate to sustain the disease. The current review presents data with both a clinical and basic research point of view on the role of nutritional and energy sensors with neuroendocrine actions in the pathophysiology of the disease, as they modulate metabolic responses, reproductive functions, stress responses as well as physical activity. While clinical data present a full description of changes occurring in AN, animal models that integrate either spontaneous genetic mutations or experimentally-induced food restriction with hyperactivity and/or social stress recapitulate the main metabolic and endocrine alterations of AN and provide mechanistic information between undernutrition state and symptoms of the disease. Further progress on the central and peripheral mechanism involved in the pathophysiology of eating disorders partly relies on the development and/or refinement of existing animal models to include recently identified genetic traits and better mimic the complex and multifactorial dimensions of the disease.
Collapse
Affiliation(s)
- Odile Viltart
- Centre de Psychiatrie et Neurosciences, INSERM UMR 894, Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Université de Lille (Sciences et technologies), Lille, France
| | - Philibert Duriez
- Centre de Psychiatrie et Neurosciences, INSERM UMR 894, Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Clinique des Maladies Mentales et de l'Encéphale (CMME), Hôpital Sainte-Anne, Paris, France
| | - Virginie Tolle
- Centre de Psychiatrie et Neurosciences, INSERM UMR 894, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
79
|
Ralevski E, Horvath TL, Shanabrough M, Hayden R, Newcomb J, Petrakis I. Ghrelin is Supressed by Intravenous Alcohol and is Related to Stimulant and Sedative Effects of Alcohol. Alcohol Alcohol 2018; 52:431-438. [PMID: 28481974 DOI: 10.1093/alcalc/agx022] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 03/10/2017] [Indexed: 12/30/2022] Open
Abstract
Aims Evidence indicates that feeding-related peptides, such as ghrelin, have a role in the rewarding properties of addictive substances like alcohol. Oral alcohol administration significantly suppresses ghrelin. This study was designed to evaluate the effects of two doses of alcohol on ghrelin and examine if ghrelin levels predict the subjective effects of alcohol. Methods Healthy social drinkers (N = 20) participated in three, randomly assigned, and counterbalanced laboratory sessions. During each session they received a continuous IV infusion of either placebo (saline), low dose (40 mg%) or high dose (100 mg%) of alcohol. Participants were given a standardized, light breakfast 90 min before the start of the infusion. Ghrelin levels [acyl ghrelin (AG) and total ghrelin (TG)] were collected at four time points before, during and after the infusion. Subjective effects of alcohol, using the BAES, were evaluated before, during and after alcohol infusion. Results IV alcohol significantly reduced AG but not TG levels with no difference between the two doses of alcohol. The percent change (%∆) in AG suppression was substantial in both high dose (43.4%∆), and low dose (39.5%∆) of alcohol. Also, fasting AG and TG levels were significant predictors of alcohol stimulant and sedative effects. Higher fasting ghrelin levels were associated with longer and more intense subjective effects. Conclusions The results provide evidence that IV alcohol suppresses ghrelin levels similarly to oral alcohol. This study is the first to show that ghrelin predicts subjective effects of alcohol, suggesting that ghrelin may have a role in the rewarding mechanisms for alcohol. Short summary Intravenous alcohol infusion (low dose and high dose of alcohol) when compared to placebo (saline) significantly suppresses ghrelin in healthy social drinkers. Fasting ghrelin levels also predict subjective behavioral effects of alcohol. Those with higher fasting ghrelin levels tend to experience alcohol effects longer and more intensely.
Collapse
Affiliation(s)
- Elizabeth Ralevski
- Department of Psychiatry, Yale University School of Medicine, 333 Cedar St., New Haven, CT, USA.,Department of Veterans Affairs, VA Connecticut Healthcare System, 950 Campbell Ave, West Haven, CT, USA.,Mental Illness Research and Clinical Center, VA Connecticut Healthcare System, 950 Campbell Ave, West Haven, CT, USA
| | - Tamas L Horvath
- Program of Integrative Cell Signaling and Neurobiology of Metabolism, Section of Comparative Medicine, Yale University School of Medicine, 310 Cedar St., Ste 330 BML, New Haven, CT, USA.,Department of Obstetrics/Gynecology and Reproductive Sciences, Yale University School of Medicine, 310 Cedar St., Ste 330 BML, New Haven, CT, USA
| | - Marya Shanabrough
- Program of Integrative Cell Signaling and Neurobiology of Metabolism, Section of Comparative Medicine, Yale University School of Medicine, 310 Cedar St., Ste 330 BML, New Haven, CT, USA
| | - Ryan Hayden
- Department of Psychiatry, Yale University School of Medicine, 333 Cedar St., New Haven, CT, USA
| | - Jenelle Newcomb
- Department of Psychiatry, Yale University School of Medicine, 333 Cedar St., New Haven, CT, USA.,Department of Veterans Affairs, VA Connecticut Healthcare System, 950 Campbell Ave, West Haven, CT, USA.,Mental Illness Research and Clinical Center, VA Connecticut Healthcare System, 950 Campbell Ave, West Haven, CT, USA
| | - Ismene Petrakis
- Department of Psychiatry, Yale University School of Medicine, 333 Cedar St., New Haven, CT, USA.,Department of Veterans Affairs, VA Connecticut Healthcare System, 950 Campbell Ave, West Haven, CT, USA.,Mental Illness Research and Clinical Center, VA Connecticut Healthcare System, 950 Campbell Ave, West Haven, CT, USA
| |
Collapse
|
80
|
Margulska A, Kozłowska E, Wysokiński A. Effect of clozapine dose and concentration on fasting concentration of appetite regulating peptides. Psychiatry Res 2018; 260:473-477. [PMID: 29275180 DOI: 10.1016/j.psychres.2017.12.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 11/17/2017] [Accepted: 12/08/2017] [Indexed: 01/08/2023]
Abstract
The aim of this study is to analyze whether clozapine serum concentration may affect fasting serum levels of several appetite regulating peptides: CART, PYY(1-36), NPY, AgRP, des-acylated ghrelin, leptin and obestatin. Serum concentration of clozapine and fasting serum levels of des-acylated ghrelin, neuropeptide Y (NPY), agouti-related protein (AgRP), peptide YY (PYY(1-36)), cocaine- and amphetamine-regulated transcript (CART), leptin and obestatin were measured in 30 subjects with schizophrenia on clozapine monotherapy. Leptin concentration was negatively correlated with clozapine dose (r = -0.53, p = 0.002), while NPY concentration was negatively correlated with clozapine concentration (r = -0.55, p = 0.01). Correlations with other peptides were not significant. We cannot conclude that serum concentration of clozapine is directly associated with increased or decreased level of appetite-regulating peptides.
Collapse
Affiliation(s)
- Aleksandra Margulska
- Department of Old Age Psychiatry and Psychotic Disorders, Medical University of Lodz, ul.Czechosłowacka 8/10, 92-216 Lodz, Poland.
| | - Elżbieta Kozłowska
- Department of Experimental Immunology, Medical University of Lodz, ul. Pomorska 251, 92-213 Lodz, Poland.
| | - Adam Wysokiński
- Department of Old Age Psychiatry and Psychotic Disorders, Medical University of Lodz, ul.Czechosłowacka 8/10, 92-216 Lodz, Poland.
| |
Collapse
|
81
|
Yu AP, Ugwu FN, Tam BT, Lee PH, Lai CW, Wong CSC, Siu PM. Ghrelin Axis Reveals the Interacting Influence of Central Obesity and Hypertension. Front Endocrinol (Lausanne) 2018; 9:534. [PMID: 30258404 PMCID: PMC6145011 DOI: 10.3389/fendo.2018.00534] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 08/22/2018] [Indexed: 02/06/2023] Open
Abstract
Objective: This study aimed to investigate how central obesity and hypertension modulate unacylated ghrelin (UnAG), acylated ghrelin (AG), obestatin, growth hormone (GH), and the ratios of UnAG/obestatin, AG/obestatin, and total ghrelin/obestatin. Methods: Circulatory abundances of UnAG, AG, obestatin and GH were determined in 387 Hong Kong Chinese female adults with age between 24 to 86 years based on a 2 × 2 factorial design of hypertension (blood pressure ≥140/90 mmHg) and central obesity (waist circumference or WC ≥80 cm). Participants were categorized as neither hypertensive nor centrally obese (NHNO; n = 105), hypertensive but not centrally obese (HNO; n = 102), centrally obese but not hypertensive (NHO; n = 74) and hypertensive and centrally obese (NO; n = 106). Pearson's correlation analyses were performed to detect the association between the peptides examined with WC and blood pressure. The main and interaction effects of hypertension and central obesity were examined by generalized estimating equations analyses. Results: Correlation analyses revealed that systolic blood pressure was negatively correlated with AG/obestatin, UnAG/obestatin and total ghrelin/obestatin ratios, AG, total ghrelin, and GH, while diastolic blood pressure was negatively correlated with UnAG/obestatin, total ghrelin/obestatin ratios, and GH. WC was negatively correlated with AG/obestatin, UnAG/obestatin, and total ghrelin/obestatin ratios, UnAG, AG, total ghrelin, GH, and obestatin. Interaction effects of hypertension and central obesity were observed on UnAG/obestatin, AG/obestatin and total ghrelin/obestatin ratios, and obestatin. Obestatin in NHO group was significantly higher compared to NHNO and HO groups. UnAG/obestatin, AG/obestatin, and total ghrelin/obestatin ratios were higher in NHNO group compared to HNO and HO groups. Main effects of central obesity and hypertension were observed in UnAG, total ghrelin and GH. The HO group manifested the lowest level of UnAG, total ghrelin and GH among all the groups studied. Main effect of hypertension was observed on AG, suggesting that hypertensive individuals exhibited lower levels of AG regardless of central obesity. Conclusion: Circulatory ghrelin gene products and GH exhibit different modes of modulation in response to the co-manifestation of multiple cardiovascular risk factors compared with a single risk factor alone.
Collapse
Affiliation(s)
- Angus P. Yu
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Felix N. Ugwu
- Department of Health Technology and Informatics, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Bjorn T. Tam
- Department of Health, Kinesiology and Applied Physiology, Concordia University, Montreal, QC, Canada
| | - Paul H. Lee
- School of Nursing, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Christopher W. Lai
- Department of Health Technology and Informatics, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Cesar S. C. Wong
- Department of Health Technology and Informatics, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Parco M. Siu
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
- *Correspondence: Parco M. Siu
| |
Collapse
|
82
|
Yam KY, Ruigrok SR, Ziko I, De Luca SN, Lucassen PJ, Spencer SJ, Korosi A. Ghrelin and hypothalamic NPY/AgRP expression in mice are affected by chronic early-life stress exposure in a sex-specific manner. Psychoneuroendocrinology 2017; 86:73-77. [PMID: 28917185 DOI: 10.1016/j.psyneuen.2017.09.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 08/26/2017] [Accepted: 09/03/2017] [Indexed: 10/18/2022]
Abstract
Early-life stress (ES) is a risk factor for metabolic disorders (e.g. obesity) with a notoriously higher prevalence in women compared to men. However, mechanisms underlying these effects remain elusive. The development of the hypothalamic feeding and metabolic regulatory circuits occurs mostly in the early sensitive postnatal phase in rodents and is tightly regulated by the metabolic hormones leptin and ghrelin. We have previously demonstrated that chronic ES reduces circulating leptin and alters adipose tissue metabolism early and later in life similarly in both sexes. However, it is unknown whether chronic ES might also affect developmental ghrelin and insulin levels, and if it induces changes in hypothalamic feeding circuits, possibly in a sex-dependent manner. We here show that chronic ES, in the form of exposure to limited nesting and bedding material from postnatal day (P)2 to P9 in mice, affects ghrelin levels differently, depending on the form of ghrelin (acylated vs desacylated), on age (P9 vs P14) and on sex, while insulin levels were similarly increased in both sexes after ES at P9. Even though ghrelin levels were more strongly affected in ES-exposed females, hypothalamic neuropeptide Y (NPY) and agouti-related peptide (AgRP) fiber density at P14 were similarly altered in both sexes by ES. In the paraventricular nucleus of the hypothalamus, both NPY and AgRP fiber density were increased, while in the arcuate nucleus of the hypothalamus, NPY was increased and AgRP unaltered. Additionally, the hypothalamic mRNA expression of ghrelin's receptor (i.e. growth hormone secretagogue receptor) was not affected by ES. Taken together, the specific alterations found in these important regulatory circuits after ES might contribute to an altered energy balance and feeding behavior in adulthood and thereby to an increased vulnerability to develop metabolic disorders.
Collapse
Affiliation(s)
- K Y Yam
- Swammerdam Institute for Life Sciences, Centre for Neuroscience, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, The Netherlands
| | - S R Ruigrok
- Swammerdam Institute for Life Sciences, Centre for Neuroscience, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, The Netherlands
| | - I Ziko
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Victoria, Australia
| | - S N De Luca
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Victoria, Australia
| | - P J Lucassen
- Swammerdam Institute for Life Sciences, Centre for Neuroscience, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, The Netherlands
| | - S J Spencer
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Victoria, Australia
| | - A Korosi
- Swammerdam Institute for Life Sciences, Centre for Neuroscience, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, The Netherlands.
| |
Collapse
|
83
|
Bouhlal S, Ellefsen KN, Sheskier MB, Singley E, Pirard S, Gorelick DA, Huestis MA, Leggio L. Acute effects of intravenous cocaine administration on serum concentrations of ghrelin, amylin, glucagon-like peptide-1, insulin, leptin and peptide YY and relationships with cardiorespiratory and subjective responses. Drug Alcohol Depend 2017; 180:68-75. [PMID: 28881319 PMCID: PMC5654385 DOI: 10.1016/j.drugalcdep.2017.07.033] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 07/27/2017] [Accepted: 07/28/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND Food intake and use of drugs of abuse like cocaine share common central and peripheral physiological pathways. Appetitive hormones play a major role in regulating food intake; however, little is known about the effects of acute cocaine administration on the blood concentrations of these hormones in cocaine users. METHODS We evaluated serum concentrations of six appetitive hormones: ghrelin (total and acyl-ghrelin), amylin, glucagon-like peptide-1 (GLP-1), insulin, leptin and peptide YY (PYY), as well as acute cardiorespiratory and subjective responses of 8 experienced cocaine users who received 25mg intravenous (IV) cocaine. RESULTS Serum concentrations of GLP-1 (p=0.014) and PYY (p=0.036) were significantly decreased one hour following IV cocaine administration; there was a trend towards a decrease for insulin (p=0.055) and amylin (p=0.063) concentrations, while no significant IV cocaine effect was observed for ghrelin (total or acyl-ghrelin) or leptin concentrations (p's≫>0.5). We also observed associations between hormone concentrations acutely affected by IV cocaine (GLP-1, PYY, insulin, amylin) and some cocaine-related cardiorespiratory and subjective responses (e.g., increased heart and respiratory rates; feeling high and anxious). DISCUSSION These findings show a significant effect of acute IV cocaine administration on some appetitive hormones and suggest potential associations between these hormones and cocaine-related cardiorespiratory and subjective responses. Additional research is needed to further investigate the potential mechanisms underlining these associations.
Collapse
Affiliation(s)
- Sofia Bouhlal
- Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology, National Institute on Alcohol Abuse and Alcoholism and National Institute on Drug Abuse, National Institutes of Health, 10 Center Drive (10CRC/15330), Bethesda, MD 20892, United States
| | - Kayla N. Ellefsen
- Chemistry and Drug Metabolism Section, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, United States
| | - Mikela B. Sheskier
- Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology, National Institute on Alcohol Abuse and Alcoholism and National Institute on Drug Abuse, National Institutes of Health, 10 Center Drive (10CRC/15330), Bethesda, MD 20892, United States
| | - Erick Singley
- Clinical Core Laboratory, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD 20892, United States
| | - Sandrine Pirard
- Chemistry and Drug Metabolism Section, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, United States
| | - David A. Gorelick
- Chemistry and Drug Metabolism Section, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, United States
| | - Marilyn A. Huestis
- Chemistry and Drug Metabolism Section, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, United States
| | - Lorenzo Leggio
- Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology, National Institute on Alcohol Abuse and Alcoholism and National Institute on Drug Abuse, National Institutes of Health, 10 Center Drive (10CRC/15330), Bethesda, MD 20892, United States; Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, Brown University, Providence, RI 02906, United States.
| |
Collapse
|
84
|
Abstract
Ghrelin and motilin are released from gastrointestinal endocrine cells during hunger, to act through G protein-coupled receptors that have closely related amino acid sequences. The actions of ghrelin are more complex than motilin because ghrelin also exists outside the GI tract, it is processed to des-acyl ghrelin which has activity, ghrelin can exist in truncated forms and retain activity, the ghrelin receptor can have constitutive activity and is subject to biased agonism and finally additional ghrelin-like and des-acyl ghrelin receptors are proposed. Both ghrelin and motilin can stimulate gastric emptying, acting via different pathways, perhaps influenced by biased agonism at the receptors, but research is revealing additional pathways of activity. For example, it is becoming apparent that reduction of nausea may be a key therapeutic target for ghrelin receptor agonists and perhaps for compounds that modulate the constitutive activity of the ghrelin receptor. Reduction of nausea may be the mechanism through which gastroparesis symptoms are reduced. Intriguingly, a potential ability of motilin to influence nausea is also becoming apparent. Ghrelin interacts with digestive function through its effects on appetite, and ghrelin antagonists may have a place in treating Prader-Willi syndrome. Unlike motilin, ghrelin receptor agonists also have the potential to treat constipation by acting at the lumbosacral defecation centres. In conclusion, agonists of both ghrelin and motilin receptors hold potential as treatments for specific subsets of digestive system disorders.
Collapse
|
85
|
Chen VP, Gao Y, Geng L, Brimijoin S. Butyrylcholinesterase gene transfer in obese mice prevents postdieting body weight rebound by suppressing ghrelin signaling. Proc Natl Acad Sci U S A 2017; 114:10960-10965. [PMID: 28973869 PMCID: PMC5642694 DOI: 10.1073/pnas.1706517114] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The worldwide prevalence of obesity is increasing at an alarming rate but treatment options remain limited. Despite initial success, weight loss by calorie restriction (CR) often fails because of rebound weight gain. Postdieting hyperphagia along with altered hypothalamic neuro-architecture appears to be one direct cause of this undesirable outcome. In response to calorie deficiency the circulating levels of the appetite-promoting hormone, acyl-ghrelin, rise sharply. We hypothesize that proper modulation of acyl-ghrelin and its receptor's sensitivity will favorably impact energy intake and reprogram the body weight set point. Here we applied viral gene transfer of the acyl-ghrelin hydrolyzing enzyme, butyrylcholinesterase (BChE), in a mouse model of diet-induced obesity. Our results confirmed that BChE overexpression decreased circulating acyl-ghrelin levels, suppressed CR-provoked ghrelin signaling, and restored central ghrelin sensitivity. In addition to maintaining healthy body weights, BChE treated mice had modest postdieting food intake and showed normal glucose homeostasis. Spontaneous activity and energy expenditure did not differ significantly between treated and untreated mice after body weight rebound, suggesting that BChE gene transfer did not alter energy expenditure in the long term. These findings indicate that combining BChE treatment with CR could be an effective approach in treating human obesity and aiding lifelong weight management.
Collapse
Affiliation(s)
- Vicky Ping Chen
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905
| | - Yang Gao
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905
| | - Liyi Geng
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905
| | - Stephen Brimijoin
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905
| |
Collapse
|
86
|
Sominsky L, Hodgson DM, McLaughlin EA, Smith R, Wall HM, Spencer SJ. Linking Stress and Infertility: A Novel Role for Ghrelin. Endocr Rev 2017; 38:432-467. [PMID: 28938425 DOI: 10.1210/er.2016-1133] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 07/24/2017] [Indexed: 12/23/2022]
Abstract
Infertility affects a remarkable one in four couples in developing countries. Psychological stress is a ubiquitous facet of life, and although stress affects us all at some point, prolonged or unmanageable stress may become harmful for some individuals, negatively impacting on their health, including fertility. For instance, women who struggle to conceive are twice as likely to suffer from emotional distress than fertile women. Assisted reproductive technology treatments place an additional physical, emotional, and financial burden of stress, particularly on women, who are often exposed to invasive techniques associated with treatment. Stress-reduction interventions can reduce negative affect and in some cases to improve in vitro fertilization outcomes. Although it has been well-established that stress negatively affects fertility in animal models, human research remains inconsistent due to individual differences and methodological flaws. Attempts to isolate single causal links between stress and infertility have not yet been successful due to their multifaceted etiologies. In this review, we will discuss the current literature in the field of stress-induced reproductive dysfunction based on animal and human models, and introduce a recently unexplored link between stress and infertility, the gut-derived hormone, ghrelin. We also present evidence from recent seminal studies demonstrating that ghrelin has a principal role in the stress response and reward processing, as well as in regulating reproductive function, and that these roles are tightly interlinked. Collectively, these data support the hypothesis that stress may negatively impact upon fertility at least in part by stimulating a dysregulation in ghrelin signaling.
Collapse
Affiliation(s)
- Luba Sominsky
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Victoria 3083, Australia
| | - Deborah M Hodgson
- School of Psychology, Faculty of Science and IT, The University of Newcastle, New South Wales 2308, Australia
| | - Eileen A McLaughlin
- School of Biological Sciences, Faculty of Science, The University of Auckland, Auckland 1010, New Zealand.,School of Environmental & Life Sciences, Faculty of Science and IT, The University of Newcastle, New South Wales 2308, Australia
| | - Roger Smith
- Mothers and Babies Research Centre, Hunter Medical Research Institute, Lookout Road, New Lambton Heights, New South Wales 2305, Australia.,Priority Research Centre in Reproductive Science, The University of Newcastle, New South Wales 2308, Australia
| | - Hannah M Wall
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Victoria 3083, Australia
| | - Sarah J Spencer
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Victoria 3083, Australia
| |
Collapse
|
87
|
Wu J, Wang K, Xu J, Ruan G, Zhu Q, Cai J, Ren J, Zheng S, Zhu Z, Otahal P, Ding C. Associations between serum ghrelin and knee symptoms, joint structures and cartilage or bone biomarkers in patients with knee osteoarthritis. Osteoarthritis Cartilage 2017; 25:1428-1435. [PMID: 28602782 DOI: 10.1016/j.joca.2017.05.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 05/21/2017] [Accepted: 05/27/2017] [Indexed: 02/02/2023]
Abstract
OBJECTIVE The roles of ghrelin in knee osteoarthritis (OA) are unclear. This study aimed to examine cross-sectional associations of ghrelin with knee symptoms, joint structures and cartilage or bone biomarkers in patients with knee OA. METHODS This study included 146 patients with symptomatic knee OA. Serum levels of ghrelin and cartilage or bone biomarkers including cartilage oligomeric matrix protein (COMP), cross linked C-telopeptide of type I collagen (CTXI), cross linked N-telopeptide of type I collagen (NTXI), N-terminal procollagen III propeptide (PIIINP), and matrix metalloproteinase (MMP)-3, 10, 13 were measured using Enzyme-linked immunosorbent assay (ELISA). Knee symptoms were assessed using the Western Ontario and McMaster Universities Arthritis Index (WOMAC). Infrapatellar fat pad (IPFP) volume, IPFP signal intensity alternation, cartilage defects, bone marrow lesions (BMLs) and effusion-synovitis were assessed using the (MRI). Osteophytes and joint space narrowing (JSN) were assessed using the Osteoarthritis Research Society International atlas. RESULTS After adjustment for potential confounders, ghrelin quartiles were positively associated with knee symptoms including pain, stiffness, dysfunction and total score (quartile 4 vs 1: β 24.19, 95% CI 8.13-40.25). Ghrelin quartiles were also significantly associated with increased IPFP signal intensity alteration (quartile 4 vs 1: OR 3.57, 95% CI 1.55-8.25) and NTXI, PIIINP, MMP3 and MMP13. Ghrelin was not significantly associated with other joint structures and biomarkers. CONCLUSIONS Serum levels of ghrelin were significantly associated with increased knee symptoms, IPFP signal intensity alteration and serum levels of MMP3, MMP13, NTXI and PIIINP, suggesting that ghrelin may have a role to play in knee OA.
Collapse
Affiliation(s)
- J Wu
- Department of Rheumatology and Immunology, Arthritis Research Institute, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Street, Hefei, China.
| | - K Wang
- Department of Rheumatology and Immunology, Arthritis Research Institute, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Street, Hefei, China; Menzies Institute for Medical Research, University of Tasmania, Private Bag 23, Hobart, Tasmania 7000, Australia.
| | - J Xu
- Department of Rheumatology and Immunology, Arthritis Research Institute, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Street, Hefei, China.
| | - G Ruan
- Department of Rheumatology and Immunology, Arthritis Research Institute, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Street, Hefei, China.
| | - Q Zhu
- Department of Rheumatology and Immunology, Arthritis Research Institute, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Street, Hefei, China.
| | - J Cai
- Department of Rheumatology and Immunology, Arthritis Research Institute, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Street, Hefei, China.
| | - J Ren
- Department of Rheumatology and Immunology, Arthritis Research Institute, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Street, Hefei, China.
| | - S Zheng
- Menzies Institute for Medical Research, University of Tasmania, Private Bag 23, Hobart, Tasmania 7000, Australia.
| | - Z Zhu
- Menzies Institute for Medical Research, University of Tasmania, Private Bag 23, Hobart, Tasmania 7000, Australia.
| | - P Otahal
- Menzies Institute for Medical Research, University of Tasmania, Private Bag 23, Hobart, Tasmania 7000, Australia.
| | - C Ding
- Department of Rheumatology and Immunology, Arthritis Research Institute, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Street, Hefei, China; Menzies Institute for Medical Research, University of Tasmania, Private Bag 23, Hobart, Tasmania 7000, Australia; Institute of Bone & Joint Translational Research, Southern Medical University, Guangzhou, China.
| |
Collapse
|
88
|
Bischoff SC, Boirie Y, Cederholm T, Chourdakis M, Cuerda C, Delzenne NM, Deutz NE, Fouque D, Genton L, Gil C, Koletzko B, Leon-Sanz M, Shamir R, Singer J, Singer P, Stroebele-Benschop N, Thorell A, Weimann A, Barazzoni R. Towards a multidisciplinary approach to understand and manage obesity and related diseases. Clin Nutr 2017; 36:917-938. [DOI: 10.1016/j.clnu.2016.11.007] [Citation(s) in RCA: 110] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Revised: 11/03/2016] [Accepted: 11/03/2016] [Indexed: 12/13/2022]
|
89
|
Song N, Wang W, Jia F, Du X, Xie A, He Q, Shen X, Zhang J, Rogers JT, Xie J, Jiang H. Assessments of plasma ghrelin levels in the early stages of parkinson's disease. Mov Disord 2017; 32:1487-1491. [PMID: 28681931 DOI: 10.1002/mds.27095] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 06/05/2017] [Accepted: 06/07/2017] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Gastrointestinal symptoms are early events in Parkinson's disease (PD). The gastrointestinal hormone ghrelin was neuroprotective in the nigrostriatal dopamine system. The objective of this study was to assess ghrelin levels in the early stages of PD. METHODS Plasma was collected in the fasting state in 291 PD patients in stages 1-3 and 303 age- and sex-matched healthy controls. Additional samples were taken in the glucose response test to assess nutrition-related ghrelin levels in 20 PD patients and 20 healthy controls. The enzyme-linked immunosorbent assay was used to measure total and active plasma ghrelin levels. RESULTS We reported that total and active plasma ghrelin levels were decreased in PD, although there was no difference across progressive PD stages. Postprandial ghrelin suppression and preprandial peak responses were both attenuated in PD. CONCLUSIONS Plasma ghrelin levels were decreased in PD; however, this event might be irrelevant to PD progression. Ghrelin responses to meals were also impaired in PD. © 2017 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Ning Song
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao, China
| | - Weiwei Wang
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao, China
| | - Fengjv Jia
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao, China
| | - Xixun Du
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao, China
| | - Anmu Xie
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao, China.,Department of Neurology, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Qing He
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao, China
| | - Xiaoli Shen
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao, China
| | - Jing Zhang
- Department of Pathology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Jack T Rogers
- Neurochemistry Laboratory, Department of Psychiatry-Neuroscience, Massachusetts General Hospital (East), Harvard Medical School, Charlestown, Masssachusetts, USA
| | - Junxia Xie
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao, China
| | - Hong Jiang
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao, China
| |
Collapse
|
90
|
Chowen JA, Argente J. Ghrelin: A Link Between Energy Homeostasis and the Immune System. Endocrinology 2017; 158:2077-2081. [PMID: 28881864 DOI: 10.1210/en.2017-00350] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 04/21/2017] [Indexed: 12/12/2022]
Affiliation(s)
- Julie A Chowen
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación la Princesa, 28009 Madrid, Spain
- Centro de Investigación Biomédica en Red: Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 28009 Madrid, Spain
| | - Jesús Argente
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación la Princesa, 28009 Madrid, Spain
- Centro de Investigación Biomédica en Red: Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 28009 Madrid, Spain
- Department of Pediatrics, Facultad de Medicina, Universidad Autónoma de Madrid (UAM), 28049 Madrid, Spain
- IMDEA Food Institute, CEI UAM + CSIC, 28049 Madrid, Spain
| |
Collapse
|
91
|
Chen SR, Chen H, Zhou JJ, Pradhan G, Sun Y, Pan HL, Li DP. Ghrelin receptors mediate ghrelin-induced excitation of agouti-related protein/neuropeptide Y but not pro-opiomelanocortin neurons. J Neurochem 2017; 142:512-520. [DOI: 10.1111/jnc.14080] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 05/19/2017] [Accepted: 05/22/2017] [Indexed: 12/21/2022]
Affiliation(s)
- Shao-Rui Chen
- Department of Anesthesiology and Perioperative Medicine; Center for Neuroscience and Pain Research; The University of Texas MD Anderson Cancer Center; Houston Texas USA
| | - Hong Chen
- Department of Anesthesiology and Perioperative Medicine; Center for Neuroscience and Pain Research; The University of Texas MD Anderson Cancer Center; Houston Texas USA
| | - Jing-Jing Zhou
- Department of Anesthesiology and Perioperative Medicine; Center for Neuroscience and Pain Research; The University of Texas MD Anderson Cancer Center; Houston Texas USA
| | - Geetali Pradhan
- Department of Pediatrics; USDA/ARS Children's Nutrition Research Center; Baylor College of Medicine; Houston Texas USA
| | - Yuxiang Sun
- Department of Pediatrics; USDA/ARS Children's Nutrition Research Center; Baylor College of Medicine; Houston Texas USA
- Department of Nutrition and Food Science (NFSC); Texas A&M University; College Station Texas USA
| | - Hui-Lin Pan
- Department of Anesthesiology and Perioperative Medicine; Center for Neuroscience and Pain Research; The University of Texas MD Anderson Cancer Center; Houston Texas USA
| | - De-Pei Li
- Department of Anesthesiology and Perioperative Medicine; Center for Neuroscience and Pain Research; The University of Texas MD Anderson Cancer Center; Houston Texas USA
| |
Collapse
|
92
|
Abstract
Obesity, a major risk factor for the development of diabetes mellitus, cardiovascular diseases and certain types of cancer, arises from a chronic positive energy balance that is often due to unlimited access to food and an increasingly sedentary lifestyle on the background of a genetic and epigenetic vulnerability. Our understanding of the humoral and neuronal systems that mediate the control of energy homeostasis has improved dramatically in the past few decades. However, our ability to develop effective strategies to slow the current epidemic of obesity has been hampered, largely owing to the limited knowledge of the mechanisms underlying resistance to the action of metabolic hormones such as leptin and ghrelin. The development of resistance to leptin and ghrelin, hormones that are crucial for the neuroendocrine control of energy homeostasis, is a hallmark of obesity. Intensive research over the past several years has yielded tremendous progress in our understanding of the cellular pathways that disrupt the action of leptin and ghrelin. In this Review, we discuss the molecular mechanisms underpinning resistance to leptin and ghrelin and how they can be exploited as targets for pharmacological management of obesity.
Collapse
Affiliation(s)
- Huxing Cui
- Department of Pharmacology, University of Iowa, Iowa City, Iowa 52246, USA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa 52242, USA
| | - Miguel López
- Department of Physiology, Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain
- Centro de Investigación Biomédica en Red-Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela 15706, Spain
| | - Kamal Rahmouni
- Department of Pharmacology, University of Iowa, Iowa City, Iowa 52246, USA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa 52242, USA
| |
Collapse
|
93
|
Cho YH, Lee SY, Jeong DW, Cho AR, Jeon JS, Kim YJ, Lee JG, Yi YH, Tak YJ, Hwang HR, Lee SH, Lee SM. Metabolic Syndrome is Associated with Lower Plasma Levels of Desacyl Ghrelin and Total Ghrelin in Asymptomatic Middle-aged Korean Men. J Obes Metab Syndr 2017; 26:114-121. [PMID: 31089505 PMCID: PMC6484903 DOI: 10.7570/jomes.2017.26.2.114] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 10/27/2016] [Accepted: 12/12/2016] [Indexed: 01/08/2023] Open
Abstract
Background Desacyl ghrelin is acylated by ghrelin O-acyltransferase (GOAT) and converted to acyl ghrelin. To date, little is known about the relationship among the levels of these two forms of ghrelin, GOAT level, and insulin resistance in Asian individuals. The purpose of this study was to determine the relationship between insulin resistance and the levels of plasma acyl ghrelin, desacyl ghrelin, and GOAT in asymptomatic middle-aged Korean men. Methods This cross-sectional study evaluated 78 asymptomatic middle-aged Korean men with metabolic syndrome (MS). We examined the correlation between the plasma levels of acyl ghrelin, desacyl ghrelin, and GOAT and sociodemographic, dietary, anthropometric, and metabolic parameters, as well as the association between insulin resistance and plasma levels of acyl ghrelin, desacyl ghrelin, and GOAT. Results The levels of desacyl ghrelin and total ghrelin were significantly lower in the MS group than in the non-MS group (P<0.017, P=0.01, respectively). HOMA-IR values showed a significant negative correlation with desacyl ghrelin (r=−0.271, P=0.017) and total ghrelin (r=−0.271, P=0.016) levels. Acyl ghrelin and GOAT were not significantly correlated with HOMA-IR, and no correlation was found between the plasma levels of the two ghrelin types and GOAT. Conclusion The plasma levels of desacyl ghrelin and total ghrelin in middle-aged Korean men with MS were lower than in those without MS. A significant negative correlation was observed between desacyl ghrelin level and HOMA-IR; however, no correlation was found between plasma levels of acyl ghrelin and GOAT and HOMA-IR.
Collapse
Affiliation(s)
- Young Hye Cho
- Family Medicine Clinic, Obesity, Metabolism and Nutrition Center and Research Institute of Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Korea
| | - Sang Yeoup Lee
- Family Medicine Clinic, Obesity, Metabolism and Nutrition Center and Research Institute of Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Korea.,Medical Education Unit, Pusan National University, School of Medicine, Yangsan, Korea
| | - Dong Wook Jeong
- Family Medicine Clinic, Obesity, Metabolism and Nutrition Center and Research Institute of Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Korea
| | - A Ra Cho
- Family Medicine Clinic, Obesity, Metabolism and Nutrition Center and Research Institute of Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Korea
| | - Jeong Suk Jeon
- Family Medicine Clinic, Obesity, Metabolism and Nutrition Center and Research Institute of Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Korea
| | - Yun Jin Kim
- Department of Family Medicine, Pusan National University Hospital, Busan, Korea
| | - Jeong Gyu Lee
- Department of Family Medicine, Pusan National University Hospital, Busan, Korea
| | - Yu Hyeon Yi
- Department of Family Medicine, Pusan National University Hospital, Busan, Korea
| | - Young Jin Tak
- Department of Family Medicine, Pusan National University Hospital, Busan, Korea
| | - Hye Rim Hwang
- Department of Family Medicine, Pusan National University Hospital, Busan, Korea
| | - Seung-Hun Lee
- Department of Family Medicine, Pusan National University Hospital, Busan, Korea
| | - Sun Min Lee
- Department of Laboratory Medicine and Molecular Genetics and Research Institute of Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Korea
| |
Collapse
|
94
|
Rigamonti AE, Bini S, Piscitelli F, Lauritano A, Di Marzo V, Vanetti C, Agosti F, De Col A, Lucchetti E, Grugni G, Sartorio A. Hedonic eating in Prader-Willi syndrome is associated with blunted PYY secretion. Food Nutr Res 2017; 61:1297553. [PMID: 28659728 PMCID: PMC5475322 DOI: 10.1080/16546628.2017.1297553] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 02/13/2017] [Indexed: 11/14/2022] Open
Abstract
Hedonic and homeostatic hunger represent two different forms of eating: just for pleasure or following energy deprivation, respectively. Consumption of food for pleasure was reported to be associated with increased circulating levels of both the orexigenic peptide ghrelin and some specific endocannabinoids in normal-weight subjects and patients with morbid obesity. To date, the effects of palatable food on these mediators in Prader–Willi syndrome (PWS) are still unknown. To explore the role of some gastrointestinal orexigenic and anorexigenic peptides and endocannabinoids (and some related congeners) in chocolate consumption, we measured changes in circulating levels of ghrelin, cholecystokinin (CCK), peptide YY (PYY), anandamide (AEA), 2-arachidonoyl-glycerol (2-AG), palmitoylethanolamide (PEA) and oleoylethanolamide (OEA) in eight satiated adult PWS patients after consumption of chocolate and, on a separate day, of a non-palatable isocaloric food with the same macronutrient composition. Evaluation of hunger and satiety was also performed by visual analogic scale. The anticipatory phase and the consumption of food for pleasure were associated with decreased circulating levels of PYY. An increase in PEA levels was also observed. By contrast, circulating levels of ghrelin, CCK, AEA, 2-AG and OEA did not differ before and after the exposure/ingestion of either chocolate or non-palatable foods. Hunger and satiety were similar in the hedonic and non-palatable sessions. In conclusion, when motivation to eat is promoted by highly palatable foods, a depressed post-prandial PYY secretion is observed in PWS. Although preliminary, these findings seem to hypothesize a possible role of PYY agonists in the management of PWS patients. Abbreviations: AEA, Anandamide; 2-AG, 2-arachidonoyl-glycerol; CB1, cannabinoid receptor type 1; OEA, oleoylethanolamide; PEA, palmitoylethanolamide; PWS: Prader-Willi syndrome; VAS, visual analog scales
Collapse
Affiliation(s)
- A E Rigamonti
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - S Bini
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - F Piscitelli
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry, Consiglio Nazionale delle Ricerche, Pozzuoli, Italy
| | - A Lauritano
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry, Consiglio Nazionale delle Ricerche, Pozzuoli, Italy
| | - V Di Marzo
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry, Consiglio Nazionale delle Ricerche, Pozzuoli, Italy
| | - C Vanetti
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - F Agosti
- Experimental Laboratory for Auxo-Endocrinological Research, Istituto Auxologico Italiano, IRCCS, Milan and Piancavallo (VB), Italy
| | - A De Col
- Experimental Laboratory for Auxo-Endocrinological Research, Istituto Auxologico Italiano, IRCCS, Milan and Piancavallo (VB), Italy
| | - E Lucchetti
- Experimental Laboratory for Auxo-Endocrinological Research, Istituto Auxologico Italiano, IRCCS, Milan and Piancavallo (VB), Italy
| | - G Grugni
- Experimental Laboratory for Auxo-Endocrinological Research, Istituto Auxologico Italiano, IRCCS, Milan and Piancavallo (VB), Italy.,Division of Auxology, Istituto Auxologico Italiano, IRCCS, Piancavallo (VB), Italy
| | - A Sartorio
- Experimental Laboratory for Auxo-Endocrinological Research, Istituto Auxologico Italiano, IRCCS, Milan and Piancavallo (VB), Italy.,Division of Auxology, Istituto Auxologico Italiano, IRCCS, Piancavallo (VB), Italy
| |
Collapse
|
95
|
Labarthe A, Tolle V. [Ghrelin: a gastric hormone at the crossroad between growth and appetite regulation]. Biol Aujourdhui 2017; 210:237-257. [PMID: 28327282 DOI: 10.1051/jbio/2016027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Indexed: 06/06/2023]
Abstract
Ghrelin is a 28 amino acid peptide hormone synthesized within the gastrointestinal tract. Initially identified as the endogenous ligand of the GHS-R1a (Growth Hormone Secretagogue Receptor 1a), ghrelin is a powerful stimulator of growth hormone (GH) secretion. At the crossroad between nutrition, growth and long-term energy metabolism, ghrelin also plays a unique role as the first identified gastric hormone increasing appetite and adiposity. However, the role of the ghrelin/GHS-R system in the physiology of growth, feeding behaviour and energy homeostasis needs to be better understood. Utilization of pharmacological tools and complementary animal models with deficiency in preproghrelin, ghrelin-O-acyl-transferase (GOAT - the enzyme that acylates ghrelin -) or GHS-R in situations of chronic undernutrition or high fat diet gives a more precise overview of the role of ghrelin in the pathophysiology of eating and metabolic disorders.
Collapse
|
96
|
Consumption of vitamin D-fortified yogurt drink increased leptin and ghrelin levels but reduced leptin to ghrelin ratio in type 2 diabetes patients: a single blind randomized controlled trial. Eur J Nutr 2017; 56:2029-2036. [DOI: 10.1007/s00394-017-1397-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 02/02/2017] [Indexed: 01/21/2023]
|
97
|
Sominsky L, Ziko I, Nguyen TX, Andrews ZB, Spencer SJ. Early life disruption to the ghrelin system with over-eating is resolved in adulthood in male rats. Neuropharmacology 2017; 113:21-30. [DOI: 10.1016/j.neuropharm.2016.09.023] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 09/21/2016] [Accepted: 09/22/2016] [Indexed: 12/11/2022]
|
98
|
Lee SP, Lee OY, Lee KN, Lee HL, Choi HS, Yoon BC, Jun DW. Effect of DA-9701, a Novel Prokinetic Agent, on Post-operative Ileus in Rats. J Neurogastroenterol Motil 2017; 23:109-116. [PMID: 27832682 PMCID: PMC5216641 DOI: 10.5056/jnm16003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 05/23/2016] [Accepted: 10/11/2016] [Indexed: 12/13/2022] Open
Abstract
Background/Aims Post-operative ileus (POI) is a common complication of abdominal surgery. DA-9701, an extract of Pharbitis Semen and Corydalis Tuber, is a new prokinetic agent that also alleviates visceral pain. The aim of this study was to investigate whether DA-9701 can ameliorate POI in rats. Methods A total of 32 rats were divided into 4 groups: no surgery/no medication (NSNM), no surgery/medication (NSM), surgery/no medication (SNM), and surgery/medication (SM). Gastrointestinal transit (GIT), which is assessed by migration of charcoal, and cumulative stool weight were measured at 24 hours after surgery. Results GIT was significantly more delayed in the SNM group than in the other groups (SNM vs NSNM, P < 0.001; SNM vs NSM, P < 0.001; SNM vs SM, P = 0.005). Cumulative stool weight in that group was also lower than in the no surgery groups (SNM vs NSNM, P = 0.007; SNM vs NSM, P = 0.033), and there was no significant difference between the SM group and the no surgery groups (SM vs NSM, P = 0.703; SM vs NSNM, P = 0.347). Conclusion DA-9701 can ameliorate POI by reducing delayed GIT and improving defecation in a rat model of POI.
Collapse
Affiliation(s)
- Sang Pyo Lee
- Department of Internal Medicine, Konkuk University School of Medicine, Seoul, Korea
| | - Oh Young Lee
- Department of Internal Medicine, Hanyang University College of Medicine, Seoul, Korea
| | - Kang Nyeong Lee
- Department of Internal Medicine, Hanyang University College of Medicine, Seoul, Korea
| | - Hang Lak Lee
- Department of Internal Medicine, Hanyang University College of Medicine, Seoul, Korea
| | - Ho Soon Choi
- Department of Internal Medicine, Hanyang University College of Medicine, Seoul, Korea
| | - Byung Chul Yoon
- Department of Internal Medicine, Hanyang University College of Medicine, Seoul, Korea
| | - Dae Won Jun
- Department of Internal Medicine, Hanyang University College of Medicine, Seoul, Korea
| |
Collapse
|
99
|
From Belly to Brain: Targeting the Ghrelin Receptor in Appetite and Food Intake Regulation. Int J Mol Sci 2017; 18:ijms18020273. [PMID: 28134808 PMCID: PMC5343809 DOI: 10.3390/ijms18020273] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 01/19/2017] [Indexed: 12/20/2022] Open
Abstract
Ghrelin is the only known peripherally-derived orexigenic hormone, increasing appetite and subsequent food intake. The ghrelinergic system has therefore received considerable attention as a therapeutic target to reduce appetite in obesity as well as to stimulate food intake in conditions of anorexia, malnutrition and cachexia. As the therapeutic potential of targeting this hormone becomes clearer, it is apparent that its pleiotropic actions span both the central nervous system and peripheral organs. Despite a wealth of research, a therapeutic compound specifically targeting the ghrelin system for appetite modulation remains elusive although some promising effects on metabolic function are emerging. This is due to many factors, ranging from the complexity of the ghrelin receptor (Growth Hormone Secretagogue Receptor, GHSR-1a) internalisation and heterodimerization, to biased ligand interactions and compensatory neuroendocrine outputs. Not least is the ubiquitous expression of the GHSR-1a, which makes it impossible to modulate centrally-mediated appetite regulation without encroaching on the various peripheral functions attributable to ghrelin. It is becoming clear that ghrelin’s central signalling is critical for its effects on appetite, body weight regulation and incentive salience of food. Improving the ability of ghrelin ligands to penetrate the blood brain barrier would enhance central delivery to GHSR-1a expressing brain regions, particularly within the mesolimbic reward circuitry.
Collapse
|
100
|
Au CC, Furness JB, Brown KA. Ghrelin and Breast Cancer: Emerging Roles in Obesity, Estrogen Regulation, and Cancer. Front Oncol 2017; 6:265. [PMID: 28119851 PMCID: PMC5220482 DOI: 10.3389/fonc.2016.00265] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 12/12/2016] [Indexed: 01/26/2023] Open
Abstract
Local and systemic factors have been shown to drive the growth of breast cancer cells in postmenopausal obese women, who have increased risk of estrogen receptor-positive breast cancer. Estrogens, produced locally in the breast fat by the enzyme aromatase, have an important role in promoting cancer cell proliferation. Ghrelin, a 28-amino acid peptide hormone, may also influence cancer growth. This peptide is produced in the stomach and acts centrally to regulate appetite and growth hormone release. Circulating levels of ghrelin, and its unacylated form, des-acyl ghrelin, are almost always inversely correlated with obesity, and these peptide hormones have recently been shown to inhibit adipose tissue aromatase expression. Ghrelin and des-acyl ghrelin have also been shown to be produced by some tumor cells and influence tumor growth. The ghrelin/des-acyl ghrelin–cancer axis is complex, one reason being that tumor cells have been shown to express splice variants of ghrelin, and ghrelin and des-acyl ghrelin might act at receptors other than the cognate ghrelin receptor, growth hormone secretagogue receptor 1a, in tumors. Effects of ghrelin and des-acyl ghrelin on energy homeostasis may also affect tumor development and growth. This review will summarize our current understanding of the role of ghrelin and des-acyl ghrelin in hormone-dependent cancers, breast cancer in particular.
Collapse
Affiliation(s)
- CheukMan Cherie Au
- Metabolism and Cancer Laboratory, Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC, Australia; Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia
| | - John B Furness
- Department of Anatomy and Neuroscience, University of Melbourne and Florey Institute of Neuroscience and Mental Health , Parkville, VIC , Australia
| | - Kristy A Brown
- Metabolism and Cancer Laboratory, Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC, Australia; Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia; Department of Physiology, Monash University, Clayton, VIC, Australia
| |
Collapse
|