601
|
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: 1.1] [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
|
602
|
Santiago-Fernández C, García-Serrano S, Tome M, Valdes S, Ocaña-Wilhelmi L, Rodríguez-Cañete A, Tinahones FJ, García-Fuentes E, Garrido-Sánchez L. Ghrelin levels could be involved in the improvement of insulin resistance after bariatric surgery. ACTA ACUST UNITED AC 2017; 64:355-362. [PMID: 28745606 DOI: 10.1016/j.endinu.2017.05.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 05/04/2017] [Accepted: 05/11/2017] [Indexed: 12/19/2022]
Abstract
BACKGROUND AND OBJECTIVE Ghrelin is a gastrointestinal peptide involved in regulation of body weight and energy balance. However, its behavior after bariatric surgery and its relationship to insulin resistance are still controversial. A simultaneous assessment was made of the association between changes in ghrelin levels and different variables after three types of bariatric surgery. PATIENTS AND METHODS Ghrelin levels were measured in 103 morbidly obese subjects before and 6 months after bariatric surgery (Roux-en-Y gastric bypass (RYGB), biliopancreatic diversion of Scopinaro (BPD), and sleeve gastrectomy (SG)), and in 21 non-obese subjects. RESULTS Ghrelin levels increased after RYGB (p<0.05), were unchanged after BPD, and decreased after SG (p<0.05). The percent change in ghrelin levels (Δ-ghrelin) was associated to the type of surgery in a multiple linear regression model (p=0.017). When the same analysis was only performed in subjects in whom the gastric fundus was maintained (RYGB and BPD), Δ-ghrelin was negatively associated to Δ-HOMA-IR (p=0.001). In morbidly obese subjects who underwent RYGB and BPD, the odds ratio of a lower Δ-HOMA-IR in patients with Δ-ghrelin in the Q1 quartile versus those with Δ-ghrelin in the Q4 quartile was 8.74 (1.73-44.06) (p=0.009). CONCLUSIONS Changes in ghrelin levels after bariatric surgery are associated to the presence or absence of the gastric fundus. After bariatric surgery, the decrease in insulin resistance was associated to increased ghrelin levels in procedures in which the fundus is not excluded.
Collapse
Affiliation(s)
- Concepción Santiago-Fernández
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Spain
| | - Sara García-Serrano
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario, Málaga, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Malaga, Spain
| | - Mónica Tome
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Spain
| | - Sergio Valdes
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario, Málaga, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Malaga, Spain
| | - Luis Ocaña-Wilhelmi
- Unidad de Gestión Clínica de Cirugía General, Digestiva y Transplantes, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Málaga, Spain
| | - Alberto Rodríguez-Cañete
- Unidad de Gestión Clínica de Cirugía General, Digestiva y Transplantes, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario, Málaga, Spain
| | - Francisco J Tinahones
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Málaga, Spain.
| | - Eduardo García-Fuentes
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Málaga, Spain; Unidad de Gestión Clínica de Aparato Digestivo, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Málaga, Spain.
| | - Lourdes Garrido-Sánchez
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Málaga, Spain
| |
Collapse
|
603
|
The role of ghrelin-responsive mediobasal hypothalamic neurons in mediating feeding responses to fasting. Mol Metab 2017; 6:882-896. [PMID: 28752052 PMCID: PMC5518774 DOI: 10.1016/j.molmet.2017.06.011] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 06/15/2017] [Accepted: 06/20/2017] [Indexed: 12/20/2022] Open
Abstract
Objective Ghrelin is a stomach-derived hormone that affects food intake and regulates blood glucose. The best-characterized actions of ghrelin are mediated by its binding to and activation of the growth hormone secretagogue receptor (GHSR; ghrelin receptor). Adequate examination of the identity, function, and relevance of specific subsets of GHSR-expressing neurons has been hampered by the absence of a suitable Cre recombinase (Cre)-expressing mouse line with which to manipulate gene expression in a targeted fashion within GHSR-expressing neurons. The present study aims to characterize the functional significance and neurocircuitry of GHSR-expressing neurons in the mediobasal hypothalamus (MBH), as they relate to ghrelin-induced food intake and fasting-associated rebound hyperphagia, using a novel mouse line in which Cre expression is controlled by the Ghsr promoter. Methods A Ghsr-IRES-Cre mouse line that expresses Cre directed by the Ghsr promoter was generated. The line was validated by comparing Cre activity in reporter mice to the known brain distribution pattern of GHSR. Next, the requirement of MBH GHSR-expressing neuronal activity in mediating food intake in response to administered ghrelin and in response to fasting was assessed after stereotaxic delivery of inhibitory designer receptor exclusively activated by designer drugs (DREADD) virus to the MBH. In a separate cohort of Ghsr-IRES-Cre mice, stereotaxic delivery of stimulatory DREADD virus to the MBH was performed to assess the sufficiency of MBH GHSR-expressing neuronal activity on food intake. Finally, the distribution of MBH GHSR-expressing neuronal axonal projections was assessed in the DREADD virus-injected animals. Results The pattern of Cre activity in the Ghsr-IRES-Cre mouse line mostly faithfully reproduced the known GHSR expression pattern. DREADD-assisted inhibition of MBH GHSR neuronal activity robustly suppressed the normal orexigenic response to ghrelin and fasting-associated rebound food intake. DREADD-assisted stimulation of MBH GHSR neuronal activity was sufficient to induce food intake. Axonal projections of GHSR-expressing MBH neurons were observed in a subset of hypothalamic and extra-hypothalamic regions. Conclusions These results suggest that 1) activation of GHSR-expressing neurons in the MBH is required for the normal feeding responses following both peripheral administration of ghrelin and fasting, 2) activation of MBH GHSR-expressing neurons is sufficient to induce feeding, and 3) axonal projections to a subset of hypothalamic and/or extra-hypothalamic regions likely mediate these responses. The Ghsr-IRES-Cre line should serve as a valuable tool to further our understanding of the functional significance of ghrelin-responsive/GHSR-expressing neurons and the neuronal circuitry within which they act. We generated a novel Ghsr-IRES-Cre knock-in mouse line. Cre activity in the line mirrors the known GHSR expression pattern. Chemogenetic modulation of neuronal activity reveals a required role of MBH GHSR neurons in rebound food intake after a fast. Neuronal projections of mediobasal hypothalamic GHSR neurons are reminiscent of AgRP neuronal projections.
Collapse
|
604
|
Llamas-Covarrubias IM, Llamas-Covarrubias MA, Martinez-López E, Zepeda-Carrillo EA, Rivera-León EA, Palmeros-Sánchez B, Alcalá-Zermeño JL, Sánchez-Enríquez S. Association of A-604G ghrelin gene polymorphism and serum ghrelin levels with the risk of obesity in a mexican population. Mol Biol Rep 2017; 44:289-293. [DOI: 10.1007/s11033-017-4109-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 05/11/2017] [Indexed: 01/27/2023]
|
605
|
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
|
606
|
Endothelial and Perivascular Adipose Tissue Abnormalities in Obesity-Related Vascular Dysfunction: Novel Targets for Treatment. J Cardiovasc Pharmacol 2017; 69:360-368. [DOI: 10.1097/fjc.0000000000000469] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
607
|
Russo C, Russo A, Gulino R, Pellitteri R, Stanzani S. Effects of different musical frequencies on NPY and Ghrelin secretion in the rat hypothalamus. Brain Res Bull 2017; 132:204-212. [DOI: 10.1016/j.brainresbull.2017.06.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Revised: 05/22/2017] [Accepted: 06/02/2017] [Indexed: 01/14/2023]
|
608
|
Ceranowicz P, Warzecha Z, Cieszkowski J, Ceranowicz D, Kuśnierz-Cabala B, Bonior J, Jaworek J, Ambroży T, Gil K, Olszanecki R, Pihut M, Dembiński A. Essential Role of Growth Hormone and IGF-1 in Therapeutic Effect of Ghrelin in the Course of Acetic Acid-Induced Colitis. Int J Mol Sci 2017; 18:ijms18061118. [PMID: 28538694 PMCID: PMC5485942 DOI: 10.3390/ijms18061118] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 05/16/2017] [Accepted: 05/18/2017] [Indexed: 01/03/2023] Open
Abstract
Previous studies have shown that ghrelin exhibits a protective and therapeutic effect in the gut. The aim of the present study was to examine whether administration of ghrelin affects the course of acetic acid-induced colitis and to determine what is the role of growth hormone (GH) and insulin-like growth factor-1 (IGF-1) in this effect. In sham-operated or hypophysectomized male Wistar rats, colitis was induced by enema with 1 mL of 3% solution of acetic acid. Saline or ghrelin (given at the dose of 8 nmol/kg/dose) was administered intraperitoneally twice a day. Seven days after colitis induction, rats were anesthetized and the severity of the colitis was assessed. Treatment with ghrelin reduced the area of colonic mucosa damage in pituitary-intact rat. This effect was associated with increase in serum levels of GH and IGF-1. Moreover, administration of ghrelin improved blood flow in colonic mucosa and mucosal cell proliferation, as well as reduced mucosal concentration of proinflammatory interleukin-1β (IL-1β) and activity of myeloperoxidase. Hypophysectomy reduced serum levels of GH and IGF-1 and increased the area of colonic damage in rats with colitis. These effects were associated with additional reduction in mucosal blood follow and DNA synthesis when compared to pituitary-intact rats. Mucosal concentration of IL-1β and mucosal activity of myeloperoxidase were maximally increased. Moreover, in hypophysectomized rats, administration of ghrelin failed to affect serum levels of GH or IGF-1, as well as the healing rate of colitis, mucosal cell proliferation, and mucosal concentration of IL-1β, or activity of myeloperoxidase. We conclude that administration of ghrelin accelerates the healing of the acetic acid-induced colitis. Therapeutic effect of ghrelin in experimental colitis is mainly mediated by the release of endogenous growth hormone and IGF-1.
Collapse
Affiliation(s)
- Piotr Ceranowicz
- Department of Physiology, Faculty of Medicine, Jagiellonian University Medical College, 31-531 Cracow, Poland.
| | - Zygmunt Warzecha
- Department of Physiology, Faculty of Medicine, Jagiellonian University Medical College, 31-531 Cracow, Poland.
| | - Jakub Cieszkowski
- Department of Physiology, Faculty of Medicine, Jagiellonian University Medical College, 31-531 Cracow, Poland.
| | - Dagmara Ceranowicz
- Department of Physiology, Faculty of Medicine, Jagiellonian University Medical College, 31-531 Cracow, Poland.
- Department of Pediatrics, Gastroenterology and Nutrition, University Children's Hospital, Faculty of Medicine, Jagiellonian University Medical College, 30-663 Cracow, Poland.
| | - Beata Kuśnierz-Cabala
- Department of Diagnostics, Chair of Clinical Biochemistry, Faculty of Medicine Jagiellonian University Medical College, 31-501 Cracow, Poland.
| | - Joanna Bonior
- Department of Medical Physiology Faculty of Health Sciences, Jagiellonian University Medical College, 31-126 Cracow, Poland.
| | - Jolanta Jaworek
- Department of Medical Physiology Faculty of Health Sciences, Jagiellonian University Medical College, 31-126 Cracow, Poland.
| | - Tadeusz Ambroży
- Department of Theory of Sport and Kinesiology, Faculty of Physical Education, University of Physical Education, 31-571 Cracow, Poland.
| | - Krzysztof Gil
- Department of Pathophysiology, Faculty of Medicine, Jagiellonian University Medical College, 31-121 Cracow, Poland.
| | - Rafał Olszanecki
- Department of Pharmacology, Faculty of Medicine, Jagiellonian University Medical College, 31-531 Cracow, Poland.
| | - Małgorzata Pihut
- Department of Prosthetic Dentistry, Faculty of Medicine, Jagiellonian University Medical College, 31-155 Cracow, Poland.
| | - Artur Dembiński
- Department of Physiology, Faculty of Medicine, Jagiellonian University Medical College, 31-531 Cracow, Poland.
| |
Collapse
|
609
|
Koch M. Cannabinoid Receptor Signaling in Central Regulation of Feeding Behavior: A Mini-Review. Front Neurosci 2017; 11:293. [PMID: 28596721 PMCID: PMC5442223 DOI: 10.3389/fnins.2017.00293] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 05/09/2017] [Indexed: 12/16/2022] Open
Abstract
Cannabinoids are lipid messengers that modulate a variety of physiological processes and modify the generation of specific behaviors. In this regard, the cannabinoid receptor type 1 (CB1) represents the most relevant target molecule of cannabinoids so far. One main function of central CB1 signaling is to maintain whole body energy homeostasis. Thus, cannabinoids functionally interact with classical neurotransmitters in neural networks that control energy metabolism and feeding behavior. The promotion of CB1 signaling can increase appetite and stimulate feeding, while blockade of CB1 suppresses hunger and induces hypophagia. However, in order to treat overeating, pharmacological blockade of CB1 by the inverse agonist rimonabant not only suppressed feeding but also resulted in psychiatric side effects. Therefore, research within the last decade focused on deciphering the underlying cellular and molecular mechanisms of central cannabinoid signaling that control feeding and other behaviors, with the overall aim still being the identification of specific targets to develop safe pharmacological interventions for the treatment of obesity. Today, many studies unraveled the subcellular localization of CB1 and the function of cannabinoids in neurons and glial cells within circumscribed brain regions that represent integral parts of neural circuitries controlling feeding behavior. Here, these novel experimental findings will be summarized and recent advances in understanding the mechanisms of CB1-dependent cannabinoid signaling being relevant for central regulation of feeding behavior will be highlighted. Finally, presumed alternative pathways of cannabinoids that are not driven by CB1 activation but also contributing to control of feeding behavior will be introduced.
Collapse
Affiliation(s)
- Marco Koch
- Medical Faculty, Institute of Anatomy, University of LeipzigLeipzig, Germany
| |
Collapse
|
610
|
Luo QQ, Zhou YF, Chen MYJ, Liu L, Ma J, Zhang MW, Zhang FL, Ke Y, Qian ZM. Fasting up-regulates ferroportin 1 expression via a Ghrelin/GHSR/MAPK signaling pathway. J Cell Physiol 2017; 233:30-37. [PMID: 28338217 DOI: 10.1002/jcp.25931] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 03/22/2017] [Indexed: 01/18/2023]
Abstract
The significant positive correlation between ghrelin and iron and hepcidin levels in the plasma of children with iron deficiency anemia prompted us to hypothesize that ghrelin may affect iron metabolism. Here, we investigated the effects of fasting or ghrelin on the expression of hepcidin, ferroportin 1 (Fpn1), transferrin receptor 1 (TfR1), ferritin light chain (Ft-L) proteins, and ghrelin, and also hormone secretagogue receptor 1 alpha (GHSR1α) and ghrelin O-acyltransferase (GOAT) mRNAs in the spleen and/or macrophage. We demonstrated that fasting induces a significant increase in the expression of ghrelin, GHSR1α, GOAT, and hepcidin mRNAs, as well as Ft-L and Fpn1 but not TfR1 proteins in the spleens of mice in vivo. Similar to the effects of fasting on the spleen, ghrelin induced a significant increase in the expression of Ft-L and Fpn1 but not TfR1 proteins in macrophages in vitro. In addition, ghrelin was found to induce a significant enhancement in phosphorylation of ERK as well as translocation of pERK from the cytosol to nuclei. Furthermore, the increased pERK and Fpn1 induced by ghrelin was demonstrated to be preventable by pre-treatment with either GHSR1α antagonist or pERK inhibitor. Our findings support the hypothesis that fasting upregulates Fpn1 expression, probably via a ghrelin/GHSR/MAPK signaling pathway.
Collapse
Affiliation(s)
- Qian-Qian Luo
- Laboratory of Neuropharmacology, Fudan University School of Pharmacy, Shanghai, China.,Pharmacological Evaluation and Research Center, Shanghai Institute of PharmaceuticalIndustry, Shanghai, China.,Department of Biochemistry, Institute for Nautical Medicine, Nantong University, Nantong, China
| | - Yu-Fu Zhou
- Laboratory of Neuropharmacology, Fudan University School of Pharmacy, Shanghai, China
| | - Mesona Yung-Jin Chen
- Laboratory of Neuropharmacology, Fudan University School of Pharmacy, Shanghai, China
| | - Li Liu
- Pharmacological Evaluation and Research Center, Shanghai Institute of PharmaceuticalIndustry, Shanghai, China
| | - Juan Ma
- Laboratory of Neuropharmacology, Fudan University School of Pharmacy, Shanghai, China
| | - Meng-Wan Zhang
- Laboratory of Neuropharmacology, Fudan University School of Pharmacy, Shanghai, China
| | - Fa-Li Zhang
- Laboratory of Neuropharmacology, Fudan University School of Pharmacy, Shanghai, China
| | - Ya Ke
- Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, NT, Hong Kong
| | - Zhong-Ming Qian
- Laboratory of Neuropharmacology, Fudan University School of Pharmacy, Shanghai, China
| |
Collapse
|
611
|
Abstract
Pancreatic islet β cells secrete insulin in response to nutrient secretagogues, like glucose, dependent on calcium influx and nutrient metabolism. One of the most intriguing qualities of β cells is their ability to use metabolism to amplify the amount of secreted insulin independent of further alterations in intracellular calcium. Many years studying this amplifying process have shaped our current understanding of β cell stimulus-secretion coupling; yet, the exact mechanisms of amplification have been elusive. Recent studies utilizing metabolomics, computational modeling, and animal models have progressed our understanding of the metabolic amplifying pathway of insulin secretion from the β cell. New approaches will be discussed which offer in-roads to a more complete model of β cell function. The development of β cell therapeutics may be aided by such a model, facilitating the targeting of aspects of the metabolic amplifying pathway which are unique to the β cell.
Collapse
Affiliation(s)
- Michael A Kalwat
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, United States.
| | - Melanie H Cobb
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
612
|
Ghrelin attenuates vascular calcification in diabetic patients with amputation. Biomed Pharmacother 2017; 91:1053-1064. [PMID: 28525946 DOI: 10.1016/j.biopha.2017.05.031] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Revised: 05/06/2017] [Accepted: 05/06/2017] [Indexed: 02/07/2023] Open
Abstract
Vascular calcification is established to be a critical factor in diabetes mellitus, which causes cardiovascular and amputation complication of diabetic patients. OPG/RANKL/RANK axis serves as a regulatory role in vascular calcification. Ghrelin, an endogenous ligand of growth hormone secretagogue receptor (GHSR), has been reported to exhibit potent cardiovascular protective effects. However, the role of ghrelin in the regulation of diabetic vascular calcification is still elusive. Here, we reported the role of ghrelin and its relationship with OPG/RANKL/RANK system in patients with diabetic foot amputation. In vivo and in vitro investigations were performed. Sixty type 2 diabetic patients with foot amputation were enrolled in vivo investigation, and they were divided into three groups through Doppler ultrasound: mild stenosis group (n=20), moderate stenosis group (n=20), and severe stenosis/occlusion group (n=20). Morphological analysis results showed diffused calcium depositions in the anterior tibial artery of diabetic amputees. Compared with the mild and moderate stenosis group, the severe stenosis/occlusion group had more spotty calcium depositions in atherosclerotic plaques. Western blot analysis indicated the expressions of osteoprotegerin (OPG) and ghrelin were downregulated, while the expression of receptor activator of nuclear factor kappa B ligand (RANKL) was upregulated with the vascular stenosis aggravation. Pearson correlation analysis revealed a negative correlation between calcium content and ghrelin levels (r=-0.58, P<0.001), as well as the ghrelin levels and sRANKL levels (r=-0.57, P<0.001). Meanwhile, OPG levels were positively correlated with ghrelin levels (r=0.63, P<0.001). From in vitro investigation, we found that the high-glucose (HG), high-lipid (HL), and β-glycerophosphate (β-GP) considerably increased the total calcium content, ALP activity, and expression of osteogenic markers in vascular smooth muscle cells (VSMCs). Ghrelin blunted calcification in a dose-dependent manner. In addition, ghrelin upregulated OPG expression and downregulated RANKL expression in VSMC calcification when anti-OPG antibody and RANKL were performed. Collectively, we therefore conclude serum ghrelin level may be a predictor of diabetic vascular calcification. The possible mechanism may be related with OPG/RANKL signal.
Collapse
|
613
|
Velasco C, Moreiras G, Conde-Sieira M, Leao JM, Míguez JM, Soengas JL. Ceramide counteracts the effects of ghrelin on the metabolic control of food intake in rainbow trout. ACTA ACUST UNITED AC 2017; 220:2563-2576. [PMID: 28495865 DOI: 10.1242/jeb.159871] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 05/02/2017] [Indexed: 12/31/2022]
Abstract
In mammals, ceramides are involved in the modulation of the orexigenic effects of ghrelin (GHRL). We previously demonstrated in rainbow trout that intracerebroventricular (ICV) treatment with ceramide (2.5 µg/100 g fish) resulted in an anorexigenic response, i.e. a response opposed to that described in mammals, where ceramide treatment is orexigenic. Therefore, we hypothesized that the putative interaction between GHRL and ceramide must be different in fish. Accordingly, in a first experiment, we observed that ceramide levels in the hypothalamus of rainbow trout did not change after ICV treatment with GHRL. In a second experiment, we assessed whether the effects of GHRL treatment on the regulation of food intake in rainbow trout changed in the presence of ceramide. Thus, we injected ICV GHRL and ceramide alone or in combination to evaluate in hypothalamus and hindbrain changes in parameters related to the metabolic control of food intake. The presence of ceramide generally counteracted the effects elicited by GHRL on fatty acid-sensing systems, the capacity of integrative sensors (AMPK, mTOR and SIRT-1), proteins involved in cellular signalling pathways (Akt and FoxO1) and neuropeptides involved in the regulation of food intake (AgRP, NPY, POMC and CART). The results are discussed in the context of regulation of food intake by metabolic and endocrine inputs.
Collapse
Affiliation(s)
- Cristina Velasco
- Laboratorio de Fisioloxía Animal, Departamento de Bioloxía Funcional e Ciencias da Saúde, Facultade de Bioloxía and Centro Singular de Investigación Mariña-ECIMAT, Universidade de Vigo, 36310 Vigo, Spain
| | - Guillermo Moreiras
- Departamento de Química Analítica e Alimentaria, Facultade de Química, Universidade de Vigo, 36310 Vigo, Spain
| | - Marta Conde-Sieira
- Laboratorio de Fisioloxía Animal, Departamento de Bioloxía Funcional e Ciencias da Saúde, Facultade de Bioloxía and Centro Singular de Investigación Mariña-ECIMAT, Universidade de Vigo, 36310 Vigo, Spain
| | - José M Leao
- Departamento de Química Analítica e Alimentaria, Facultade de Química, Universidade de Vigo, 36310 Vigo, Spain
| | - Jesús M Míguez
- Laboratorio de Fisioloxía Animal, Departamento de Bioloxía Funcional e Ciencias da Saúde, Facultade de Bioloxía and Centro Singular de Investigación Mariña-ECIMAT, Universidade de Vigo, 36310 Vigo, Spain
| | - José L Soengas
- Laboratorio de Fisioloxía Animal, Departamento de Bioloxía Funcional e Ciencias da Saúde, Facultade de Bioloxía and Centro Singular de Investigación Mariña-ECIMAT, Universidade de Vigo, 36310 Vigo, Spain
| |
Collapse
|
614
|
Reano S, Angelino E, Ferrara M, Malacarne V, Sustova H, Sabry O, Agosti E, Clerici S, Ruozi G, Zentilin L, Prodam F, Geuna S, Giacca M, Graziani A, Filigheddu N. Unacylated Ghrelin Enhances Satellite Cell Function and Relieves the Dystrophic Phenotype in Duchenne Muscular Dystrophy mdx Model. Stem Cells 2017; 35:1733-1746. [PMID: 28436144 DOI: 10.1002/stem.2632] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 02/28/2017] [Accepted: 04/06/2017] [Indexed: 12/17/2022]
Abstract
Muscle regeneration depends on satellite cells (SCs), quiescent precursors that, in consequence of injury or in pathological states such as muscular dystrophies, activate, proliferate, and differentiate to repair the damaged tissue. A subset of SCs undergoes self-renewal, thus preserving the SC pool and its regenerative potential. Unacylated ghrelin (UnAG) is a circulating hormone that protects muscle from atrophy, promotes myoblast differentiation, and enhances ischemia-induced muscle regeneration. Here we show that UnAG increases SC activity and stimulates Par polarity complex/p38-mediated asymmetric division, fostering both SC self-renewal and myoblast differentiation. Because of those activities on different steps of muscle regeneration, we hypothesized a beneficial effect of UnAG in mdx dystrophic mice, in which the absence of dystrophin leads to chronic muscle degeneration, defective muscle regeneration, fibrosis, and, at later stages of the pathology, SC pool exhaustion. Upregulation of UnAG levels in mdx mice reduces muscle degeneration, improves muscle function, and increases dystrophin-null SC self-renewal, maintaining the SC pool. Our results suggest that UnAG has significant therapeutic potential for preserving the muscles in dystrophies. Stem Cells 2017;35:1733-1746.
Collapse
Affiliation(s)
- Simone Reano
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy and Istituto Interuniversitario di Miologia (IIM)
| | - Elia Angelino
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy and Istituto Interuniversitario di Miologia (IIM)
| | - Michele Ferrara
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy and Istituto Interuniversitario di Miologia (IIM)
| | - Valeria Malacarne
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy and Istituto Interuniversitario di Miologia (IIM)
| | - Hana Sustova
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy and Istituto Interuniversitario di Miologia (IIM)
| | - Omar Sabry
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy and Istituto Interuniversitario di Miologia (IIM)
| | - Emanuela Agosti
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy and Istituto Interuniversitario di Miologia (IIM)
| | - Sara Clerici
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy and Istituto Interuniversitario di Miologia (IIM)
| | - Giulia Ruozi
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Lorena Zentilin
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Flavia Prodam
- Department of Health Sciences, University of Piemonte Orientale, Novara, Italy
| | - Stefano Geuna
- Department of Clinical and Biological Sciences, University of Torino and Neuroscience Institute Cavalieri Ottolenghi (NICO), Orbassano (TO), Italy
| | - Mauro Giacca
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Andrea Graziani
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy and Istituto Interuniversitario di Miologia (IIM)
| | - Nicoletta Filigheddu
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy and Istituto Interuniversitario di Miologia (IIM)
| |
Collapse
|
615
|
Attenuating the Biologic Drive for Weight Regain Following Weight Loss: Must What Goes Down Always Go Back Up? Nutrients 2017; 9:nu9050468. [PMID: 28481261 PMCID: PMC5452198 DOI: 10.3390/nu9050468] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2017] [Revised: 04/26/2017] [Accepted: 04/28/2017] [Indexed: 01/01/2023] Open
Abstract
Metabolic adaptations occur with weight loss that result in increased hunger with discordant simultaneous reductions in energy requirements—producing the so-called energy gap in which more energy is desired than is required. The increased hunger is associated with elevation of the orexigenic hormone ghrelin and decrements in anorexigenic hormones. The lower total daily energy expenditure with diet-induced weight loss results from (1) a disproportionately greater decrease in circulating leptin and resting metabolic rate (RMR) than would be predicted based on the decline in body mass, (2) decreased thermic effect of food (TEF), and (3) increased energy efficiency at work intensities characteristic of activities of daily living. These metabolic adaptations can readily promote weight regain. While more experimental research is needed to identify effective strategies to narrow the energy gap and attenuate weight regain, some factors contributing to long-term weight loss maintenance have been identified. Less hunger and greater satiation have been associated with higher intakes of protein and dietary fiber, and lower glycemic load diets. High levels of physical activity are characteristic of most successful weight maintainers. A high energy flux state characterized by high daily energy expenditure and matching energy intake may attenuate the declines in RMR and TEF, and may also result in more accurate regulation of energy intake to match daily energy expenditure.
Collapse
|
616
|
Brown JA, Bugescu R, Mayer TA, Gata-Garcia A, Kurt G, Woodworth HL, Leinninger GM. Loss of Action via Neurotensin-Leptin Receptor Neurons Disrupts Leptin and Ghrelin-Mediated Control of Energy Balance. Endocrinology 2017; 158:1271-1288. [PMID: 28323938 PMCID: PMC5460836 DOI: 10.1210/en.2017-00122] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 03/06/2017] [Indexed: 01/30/2023]
Abstract
The hormones ghrelin and leptin act via the lateral hypothalamic area (LHA) to modify energy balance, but the underlying neural mechanisms remain unclear. We investigated how leptin and ghrelin engage LHA neurons to modify energy balance behaviors and whether there is any crosstalk between leptin and ghrelin-responsive circuits. We demonstrate that ghrelin activates LHA neurons expressing hypocretin/orexin (OX) to increase food intake. Leptin mediates anorectic actions via separate neurons expressing the long form of the leptin receptor (LepRb), many of which coexpress the neuropeptide neurotensin (Nts); we refer to these as NtsLepRb neurons. Because NtsLepRb neurons inhibit OX neurons, we hypothesized that disruption of the NtsLepRb neuronal circuit would impair both NtsLepRb and OX neurons from responding to their respective hormonal cues, thus compromising adaptive energy balance. Indeed, mice with developmental deletion of LepRb specifically from NtsLepRb neurons exhibit blunted adaptive responses to leptin and ghrelin that discoordinate the mesolimbic dopamine system and ingestive and locomotor behaviors, leading to weight gain. Collectively, these data reveal a crucial role for LepRb in the proper formation of LHA circuits, and that NtsLepRb neurons are important neuronal hubs within the LHA for hormone-mediated control of ingestive and locomotor behaviors.
Collapse
Affiliation(s)
- Juliette A. Brown
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan 48824
- Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan 48824
| | - Raluca Bugescu
- Department of Physiology, Michigan State University, East Lansing, Michigan 48824
| | - Thomas A. Mayer
- Department of Physiology, Michigan State University, East Lansing, Michigan 48824
| | - Adriana Gata-Garcia
- Division of Metabolism, Endocrinology and Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109
| | - Gizem Kurt
- Department of Physiology, Michigan State University, East Lansing, Michigan 48824
| | - Hillary L. Woodworth
- Department of Physiology, Michigan State University, East Lansing, Michigan 48824
| | - Gina M. Leinninger
- Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan 48824
- Department of Physiology, Michigan State University, East Lansing, Michigan 48824
| |
Collapse
|
617
|
Santos VV, Stark R, Rial D, Silva HB, Bayliss JA, Lemus MB, Davies JS, Cunha RA, Prediger RD, Andrews ZB. Acyl ghrelin improves cognition, synaptic plasticity deficits and neuroinflammation following amyloid β (Aβ1-40) administration in mice. J Neuroendocrinol 2017; 29. [PMID: 28380673 DOI: 10.1111/jne.12476] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 03/16/2017] [Accepted: 04/03/2017] [Indexed: 12/24/2022]
Abstract
Ghrelin is a metabolic hormone that has neuroprotective actions in a number of neurological conditions, including Parkinson's disease (PD), stroke and traumatic brain injury. Acyl ghrelin treatment in vivo and in vitro also shows protective capacity in Alzheimer's disease (AD). In the present study, we used ghrelin knockout (KO) and their wild-type littermates to test whether or not endogenous ghrelin is protective in a mouse model of AD, in which human amyloid β peptide 1-40 (Aβ1-40 ) was injected into the lateral ventricles i.c.v. Recognition memory, using the novel object recognition task, was significantly impaired in ghrelin KO mice and after i.c.v. Aβ1-40 treatment. These deficits could be prevented by acyl ghrelin injections for 7 days. Spatial orientation, as assessed by the Y-maze task, was also significantly impaired in ghrelin KO mice and after i.c.v. Aβ1-40 treatment. These deficits could be prevented by acyl ghrelin injections for 7 days. Ghrelin KO mice had deficits in olfactory discrimination; however, neither i.c.v. Aβ1-40 treatment, nor acyl ghrelin injections affected olfactory discrimination. We used stereology to show that ghrelin KO and Aβ1-40 increased the total number of glial fibrillary acidic protein expressing astrocytes and ionised calcium-binding adapter expressing microglial in the rostral hippocampus. Finally, Aβ1-40 blocked long-term potentiation induced by high-frequency stimulation and this effect could be acutely blocked with co-administration of acyl ghrelin. Collectively, our studies demonstrate that ghrelin deletion affects memory performance and also that acyl ghrelin treatment may delay the onset of early events of AD. This supports the idea that acyl ghrelin treatment may be therapeutically beneficial with respect to restricting disease progression in AD.
Collapse
Affiliation(s)
- V V Santos
- Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, VIC, Australia
| | - R Stark
- Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, VIC, Australia
| | - D Rial
- Department of Pharmacology, Center of Biological Sciences, Universidade Federal de Santa Catarina UFSC, Florianópolis, SC, Brazil
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - H B Silva
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - J A Bayliss
- Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, VIC, Australia
| | - M B Lemus
- Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, VIC, Australia
| | - J S Davies
- Molecular Neurobiology, Institute of Life Science, Swansea University, Swansea, UK
| | - R A Cunha
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - R D Prediger
- Department of Pharmacology, Center of Biological Sciences, Universidade Federal de Santa Catarina UFSC, Florianópolis, SC, Brazil
| | - Z B Andrews
- Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, VIC, Australia
| |
Collapse
|
618
|
Monteiro MP, Batterham RL. The Importance of the Gastrointestinal Tract in Controlling Food Intake and Regulating Energy Balance. Gastroenterology 2017; 152:1707-1717.e2. [PMID: 28193513 DOI: 10.1053/j.gastro.2017.01.053] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 12/31/2016] [Accepted: 01/03/2017] [Indexed: 12/16/2022]
Abstract
The gastrointestinal tract, the key interface between ingested nutrients and the body, plays a critical role in regulating energy homeostasis. Gut-derived signals convey information regarding incoming nutrients to the brain, initiating changes in eating behavior and energy expenditure, to maintain energy balance. Here we review hormonal, neural, and nutrient signals emanating from the gastrointestinal tract and evidence for their role in controlling feeding behavior. Mechanistic studies that have utilized pharmacologic and/or transgenic approaches targeting an individual hormone/mediator have yielded somewhat disappointing body weight changes, often leading to the hormone/mediator in question being dismissed as a potential obesity therapy. However, the recent finding of sustained weight reduction in response to systemic administration of a long-acting analog of the gut-hormone glucagon-like peptide-1 highlights the therapeutic potential of gut-derived signals acting via nonphysiologic mechanisms. Thus, we also review therapeutics strategies being utilized or developed to leverage gastrointestinal signals in order to treat obesity.
Collapse
Affiliation(s)
- Mariana P Monteiro
- Clinical and Experimental Endocrinology, Unit for Multidisciplinary Research in Biomedicine, Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Portugal; 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.
| |
Collapse
|
619
|
Exploring the Behavioral and Metabolic Phenotype Generated by Re-Introduction of the Ghrelin Receptor in the Ventral Tegmental Area. Int J Mol Sci 2017; 18:ijms18050914. [PMID: 28445429 PMCID: PMC5454827 DOI: 10.3390/ijms18050914] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 04/01/2017] [Accepted: 04/20/2017] [Indexed: 11/16/2022] Open
Abstract
Ghrelin receptor (Ghr-R) signaling in neurons of the ventral tegmental area (VTA) can modulate dopaminergic function and the reward-related effects of both palatable foods and drugs of abuse. In this study, we re-introduced the Ghr-R in VTA neurons in Ghr-R knockout mice (Ghr-RVTA mice) to specifically study the importance of the constitutively active Ghr-R for VTA neuronal signaling. Our results showed that re-introduction of the Ghr-R in the VTA had no impact on body weight or food intake under basal conditions. However, during novel environment stress Ghr-RVTA mice showed increased food intake and energy expenditure compared to Ghr-R knockout mice, demonstrating the significance of Ghr-R signaling in the response to stress. Ghr-RVTA mice also showed increased cocaine-induced locomotor activity compared to Ghr-R knockout mice, highlighting the importance of ghrelin signaling for the reward-related effects of activation of VTA neurons. Overall, our data suggest that re-introduction of the Ghr-R in the mesolimbic reward system of Ghr-R knockout mice increases the level of activation induced by both cocaine and novelty stress.
Collapse
|
620
|
Suppression of GHS-R in AgRP Neurons Mitigates Diet-Induced Obesity by Activating Thermogenesis. Int J Mol Sci 2017; 18:ijms18040832. [PMID: 28420089 PMCID: PMC5412416 DOI: 10.3390/ijms18040832] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 04/07/2017] [Accepted: 04/07/2017] [Indexed: 12/21/2022] Open
Abstract
Ghrelin, an orexigenic hormone released primarily from the gut, signals the hypothalamus to stimulate growth hormone release, enhance appetite and promote weight gain. The ghrelin receptor, aka Growth Hormone Secretagogue Receptor (GHS-R), is highly expressed in the brain, with highest expression in Agouti-Related Peptide (AgRP) neurons of the hypothalamus. We recently reported that neuron-specific deletion of GHS-R completely prevents diet-induced obesity (DIO) in mice by activating non-shivering thermogenesis. To further decipher the specific neuronal circuits mediating the metabolic effects of GHS-R, we generated AgRP neuron-specific GHS-R knockout mice (AgRP-Cre;Ghsrf/f). Our data showed that GHS-R in AgRP neurons is required for ghrelin’s stimulatory effects on growth hormone secretion, acute food intake and adiposity, but not for long-term total food intake. Importantly, deletion of GHS-R in AgRP neurons attenuated diet-induced obesity (DIO) and enhanced cold-resistance in mice fed high fat diet (HFD). The HFD-fed knockout mice showed increased energy expenditure, and exhibited enhanced thermogenic activation in both brown and subcutaneous fat; this implies that GHS-R suppression in AgRP neurons enhances sympathetic outflow. In summary, our results suggest that AgRP neurons are key site for GHS-R mediated thermogenesis, and demonstrate that GHS-R in AgRP neurons plays crucial roles in governing energy utilization and pathogenesis of DIO.
Collapse
|
621
|
Colldén G, Tschöp MH, Müller TD. Therapeutic Potential of Targeting the Ghrelin Pathway. Int J Mol Sci 2017; 18:ijms18040798. [PMID: 28398233 PMCID: PMC5412382 DOI: 10.3390/ijms18040798] [Citation(s) in RCA: 101] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 04/03/2017] [Accepted: 04/06/2017] [Indexed: 02/07/2023] Open
Abstract
Ghrelin was discovered in 1999 as the endogenous ligand of the growth-hormone secretagogue receptor 1a (GHSR1a). Since then, ghrelin has been found to exert a plethora of physiological effects that go far beyond its initial characterization as a growth hormone (GH) secretagogue. Among the numerous well-established effects of ghrelin are the stimulation of appetite and lipid accumulation, the modulation of immunity and inflammation, the stimulation of gastric motility, the improvement of cardiac performance, the modulation of stress, anxiety, taste sensation and reward-seeking behavior, as well as the regulation of glucose metabolism and thermogenesis. Due to a variety of beneficial effects on systems’ metabolism, pharmacological targeting of the endogenous ghrelin system is widely considered a valuable approach to treat metabolic complications, such as chronic inflammation, gastroparesis or cancer-associated anorexia and cachexia. The aim of this review is to discuss and highlight the broad pharmacological potential of ghrelin pathway modulation for the treatment of anorexia, cachexia, sarcopenia, cardiopathy, neurodegenerative disorders, renal and pulmonary disease, gastrointestinal (GI) disorders, inflammatory disorders and metabolic syndrome.
Collapse
Affiliation(s)
- Gustav Colldén
- Institute for Diabetes and Obesity & Helmholtz Diabetes Center, Helmholtz Zentrum München German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany.
| | - Matthias H Tschöp
- Institute for Diabetes and Obesity & Helmholtz Diabetes Center, Helmholtz Zentrum München German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany.
- Division of Metabolic Diseases, Department of Medicine, Technische Universität München, 80333 Munich, Germany.
| | - Timo D Müller
- Institute for Diabetes and Obesity & Helmholtz Diabetes Center, Helmholtz Zentrum München German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany.
- Institute for Diabetes and Obesity (IDO), Business Campus Garching-Hochbrück, Parkring 13, 85748 Garching, Germany.
| |
Collapse
|
622
|
Moldovan RP, Els-Heindl S, Worm DJ, Kniess T, Kluge M, Beck-Sickinger AG, Deuther-Conrad W, Krügel U, Brust P. Development of Fluorinated Non-Peptidic Ghrelin Receptor Ligands for Potential Use in Molecular Imaging. Int J Mol Sci 2017; 18:ijms18040768. [PMID: 28379199 PMCID: PMC5412352 DOI: 10.3390/ijms18040768] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 03/23/2017] [Accepted: 03/31/2017] [Indexed: 12/19/2022] Open
Abstract
The ghrelin receptor (GhrR) is a widely investigated target in several diseases. However, the current knowledge of its role and distribution in the brain is limited. Recently, the small and non-peptidic compound (S)-6-(4-bromo-2-fluorophenoxy)-3-((1-isopropylpiperidin-3-yl)methyl)-2-methylpyrido[3,2-d]pyrimidin-4(3H)-one ((S)-9) has been described as a GhrR ligand with high binding affinity. Here, we describe the synthesis of fluorinated derivatives, the in vitro evaluation of their potency as partial agonists and selectivity at GhrRs, and their physicochemical properties. These results identified compounds (S)-9, (R)-9, and (S)-16 as suitable parent molecules for 18F-labeled positron emission tomography (PET) radiotracers to enable future investigation of GhrR in the brain.
Collapse
Affiliation(s)
- Rareş-Petru Moldovan
- Helmholtz-Zentrum Dresden-Rossendorf e. V., Institute of Radiopharmaceutical Cancer Research, 04318 Leipzig, Germany.
| | - Sylvia Els-Heindl
- Institute of Biochemistry, Universität Leipzig, 04103 Leipzig, Germany.
| | - Dennis J Worm
- Institute of Biochemistry, Universität Leipzig, 04103 Leipzig, Germany.
| | - Torsten Kniess
- Helmholtz-Zentrum Dresden-Rossendorf e. V., Institute of Radiopharmaceutical Cancer Research, 04318 Leipzig, Germany.
| | - Michael Kluge
- Department of Psychiatry, Universität Leipzig, 04103 Leipzig, Germany.
| | | | - Winnie Deuther-Conrad
- Helmholtz-Zentrum Dresden-Rossendorf e. V., Institute of Radiopharmaceutical Cancer Research, 04318 Leipzig, Germany.
| | - Ute Krügel
- Rudolf Boehm Institute of Pharmacology and Toxicology, Medical Faculty, Universität Leipzig, 04107 Leipzig, Germany.
| | - Peter Brust
- Helmholtz-Zentrum Dresden-Rossendorf e. V., Institute of Radiopharmaceutical Cancer Research, 04318 Leipzig, Germany.
| |
Collapse
|
623
|
Abstract
The metabolic syndrome is a cluster of risk factors (central obesity, hyperglycaemia, dyslipidaemia and arterial hypertension), indicating an increased risk of diabetes, cardiovascular disease and premature mortality. The gastrointestinal tract is seldom discussed as an organ system of principal importance for metabolic diseases. The present overview connects various metabolic research lines into an integrative physiological context in which the gastrointestinal tract is included. Strong evidence for the involvement of the gut in the metabolic syndrome derives from the powerful effects of weight-reducing (bariatric) gastrointestinal surgery. In fact, gastrointestinal surgery is now recommended as a standard treatment option for type 2 diabetes in obesity. Several gut-related mechanisms that potentially contribute to the metabolic syndrome will be presented. Obesity can be caused by hampered release of satiety-signalling gut hormones, reduced meal-associated energy expenditure and microbiota-assisted harvest of energy from nondigestible food ingredients. Adiposity per se is a well-established risk factor for hyperglycaemia. In addition, a leaky gut mucosa can trigger systemic inflammation mediating peripheral insulin resistance that together with a blunted incretin response aggravates the hyperglycaemic state. The intestinal microbiota is strongly associated with obesity and the related metabolic disease states, although the mechanisms involved remain unclear. Enterorenal signalling has been suggested to be involved in the pathophysiology of hypertension and postprandial triglyceride-rich chylomicrons; in addition, intestinal cholesterol metabolism probably contributes to atherosclerosis. It is likely that in the future, the metabolic syndrome will be treated according to novel pharmacological principles interfering with gastrointestinal functionality.
Collapse
Affiliation(s)
- L Fändriks
- Department of Gastrosurgical Research and Education, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
624
|
Carr DF, Ayehunie S, Davies A, Duckworth CA, French S, Hall N, Hussain S, Mellor HR, Norris A, Park BK, Penrose A, Pritchard DM, Probert CS, Ramaiah S, Sadler C, Schmitt M, Shaw A, Sidaway JE, Vries RG, Wagoner M, Pirmohamed M. Towards better models and mechanistic biomarkers for drug-induced gastrointestinal injury. Pharmacol Ther 2017; 172:181-194. [DOI: 10.1016/j.pharmthera.2017.01.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
625
|
Hopkins AL, Nelson TAS, Guschina IA, Parsons LC, Lewis CL, Brown RC, Christian HC, Davies JS, Wells T. Unacylated ghrelin promotes adipogenesis in rodent bone marrow via ghrelin O-acyl transferase and GHS-R 1a activity: evidence for target cell-induced acylation. Sci Rep 2017; 7:45541. [PMID: 28361877 PMCID: PMC5374529 DOI: 10.1038/srep45541] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 02/21/2017] [Indexed: 11/09/2022] Open
Abstract
Despite being unable to activate the cognate ghrelin receptor (GHS-R), unacylated ghrelin (UAG) possesses a unique activity spectrum that includes promoting bone marrow adipogenesis. Since a receptor mediating this action has not been identified, we re-appraised the potential interaction of UAG with GHS-R in the regulation of bone marrow adiposity. Surprisingly, the adipogenic effects of intra-bone marrow (ibm)-infused acylated ghrelin (AG) and UAG were abolished in male GHS-R-null mice. Gas chromatography showed that isolated tibial marrow adipocytes contain the medium-chain fatty acids utilised in the acylation of UAG, including octanoic acid. Additionally, immunohistochemistry and immunogold electron microscopy revealed that tibial marrow adipocytes show prominent expression of the UAG-activating enzyme ghrelin O-acyl transferase (GOAT), which is located in the membranes of lipid trafficking vesicles and in the plasma membrane. Finally, the adipogenic effect of ibm-infused UAG was completely abolished in GOAT-KO mice. Thus, the adipogenic action of exogenous UAG in tibial marrow is dependent upon acylation by GOAT and activation of GHS-R. This suggests that UAG is subject to target cell-mediated activation – a novel mechanism for manipulating hormone activity.
Collapse
Affiliation(s)
- Anna L Hopkins
- Neuroscience &Mental Health Research Institute, and School of Biosciences, Cardiff University, Museum Avenue, Cardiff, CF10 3AX, UK
| | - Timothy A S Nelson
- Neuroscience &Mental Health Research Institute, and School of Biosciences, Cardiff University, Museum Avenue, Cardiff, CF10 3AX, UK
| | - Irina A Guschina
- Neuroscience &Mental Health Research Institute, and School of Biosciences, Cardiff University, Museum Avenue, Cardiff, CF10 3AX, UK
| | - Lydia C Parsons
- Neuroscience &Mental Health Research Institute, and School of Biosciences, Cardiff University, Museum Avenue, Cardiff, CF10 3AX, UK
| | - Charlotte L Lewis
- Neuroscience &Mental Health Research Institute, and School of Biosciences, Cardiff University, Museum Avenue, Cardiff, CF10 3AX, UK
| | - Richard C Brown
- Neuroscience &Mental Health Research Institute, and School of Biosciences, Cardiff University, Museum Avenue, Cardiff, CF10 3AX, UK
| | - Helen C Christian
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, OX1 3QX, UK
| | - Jeffrey S Davies
- Institute of Life Science, School of Medicine, Swansea University, Swansea, SA2 8PP, UK
| | - Timothy Wells
- Neuroscience &Mental Health Research Institute, and School of Biosciences, Cardiff University, Museum Avenue, Cardiff, CF10 3AX, UK
| |
Collapse
|
626
|
Blanco AM, Bertucci JI, Ramesh N, Delgado MJ, Valenciano AI, Unniappan S. Ghrelin Facilitates GLUT2-, SGLT1- and SGLT2-mediated Intestinal Glucose Transport in Goldfish (Carassius auratus). Sci Rep 2017; 7:45024. [PMID: 28338019 PMCID: PMC5364492 DOI: 10.1038/srep45024] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 02/17/2017] [Indexed: 12/13/2022] Open
Abstract
Glucose homeostasis is an important biological process that involves a variety of regulatory mechanisms. This study aimed to determine whether ghrelin, a multifunctional gut-brain hormone, modulates intestinal glucose transport in goldfish (Carassius auratus). Three intestinal glucose transporters, the facilitative glucose transporter 2 (GLUT2), and the sodium/glucose co-transporters 1 (SGLT1) and 2 (SGLT2), were studied. Immunostaining of intestinal sections found colocalization of ghrelin and GLUT2 and SGLT2 in mucosal cells. Some cells containing GLUT2, SGLT1 and SGLT2 coexpressed the ghrelin/growth hormone secretagogue receptor 1a (GHS-R1a). Intraperitoneal glucose administration led to a significant increase in serum ghrelin levels, as well as an upregulation of intestinal preproghrelin, ghrelin O-acyltransferase and ghs-r1 expression. In vivo and in vitro ghrelin treatment caused a concentration- and time-dependent modulation (mainly stimulatory) of GLUT2, SGLT1 and SGLT2. These effects were abolished by the GHS-R1a antagonist [D-Lys3]-GHRP-6 and the phospholipase C inhibitor U73122, suggesting that ghrelin actions on glucose transporters are mediated by GHS-R1a via the PLC/PKC signaling pathway. Finally, ghrelin stimulated the translocation of GLUT2 into the plasma membrane of goldfish primary intestinal cells. Overall, data reported here indicate an important role for ghrelin in the modulation of glucoregulatory machinery and glucose homeostasis in fish.
Collapse
Affiliation(s)
- Ayelén Melisa Blanco
- Departamento de Fisiología (Fisiología Animal II), Facultad de Biología, Universidad Complutense de Madrid, Madrid, Spain.,Laboratory of Integrative Neuroendocrinology, Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Juan Ignacio Bertucci
- Laboratory of Integrative Neuroendocrinology, Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada.,Instituto de Investigaciones Biotecnológicas-Instituto Tecnológico Chascomús, Buenos Aires, Argentina
| | - Naresh Ramesh
- Laboratory of Integrative Neuroendocrinology, Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - María Jesús Delgado
- Departamento de Fisiología (Fisiología Animal II), Facultad de Biología, Universidad Complutense de Madrid, Madrid, Spain
| | - Ana Isabel Valenciano
- Departamento de Fisiología (Fisiología Animal II), Facultad de Biología, Universidad Complutense de Madrid, Madrid, Spain
| | - Suraj Unniappan
- Laboratory of Integrative Neuroendocrinology, Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| |
Collapse
|
627
|
Aghdam Shahryar H, Lotfi A. Effect of Ghrelin Administration on Serum Corticostrone, T3, T4 and Some Biochemical Indices in the Turkey (Meleagridis gallopova). Int J Pept Res Ther 2017. [DOI: 10.1007/s10989-017-9588-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
628
|
Alamri BN, Shin K, Chappe V, Anini Y. The role of ghrelin in the regulation of glucose homeostasis. Horm Mol Biol Clin Investig 2017; 26:3-11. [PMID: 27235674 DOI: 10.1515/hmbci-2016-0018] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Accepted: 05/08/2016] [Indexed: 12/16/2022]
Abstract
Ghrelin is a 28-amino acid (aa) stomach-derived peptide discovered in 1999 as the endogenous ligand for growth hormone secretagogue-receptor (GHS-R). Ghrelin-producing cells constitute a distinct group of endocrine cells dispersed throughout the gastric mucosa and to a lesser extent in the small intestine and the endocrine pancreas. Ghrelin plasma levels rise during fasting and chronic caloric restriction to stimulate food intake and fat storage and to prevent life-threatening falls in blood glucose. Plasma ghrelin levels decrease after a meal is consumed and in conditions of energy surplus (such as obesity). Ghrelin has emerged as a key player in the regulation of appetite and energy homeostasis. Ghrelin achieves these functions through binding the ghrelin receptor GHS-R in appetite-regulating neurons and in peripheral metabolic organs including the endocrine pancreas. Ghrelin levels are negatively correlated with body mass index (BMI) and insulin resistance. In addition, ghrelin secretion is impaired in obesity and insulin resistance. Several studies highlight an important role for ghrelin in glucose homeostasis. Genetic, immunological, and pharmacological blockade of ghrelin signaling resulted in improved glucose tolerance and insulin sensitivity. Furthermore, exogenous ghrelin administration was shown to decrease glucose-induced insulin release and increase glucose level in both humans and rodents. GHS-R was shown to be expressed in pancreatic β-cells and ghrelin suppressed insulin release via a Ca2+-mediated pathway. In this review, we provide a detailed summary of recent advances in the field that focuses on the role of insulin and insulin resistance in the regulation of ghrelin secretion and on the role of ghrelin in glucose-stimulated insulin secretion (GSIS).
Collapse
|
629
|
Coppens J, Bentea E, Bayliss JA, Demuyser T, Walrave L, Albertini G, Van Liefferinge J, Deneyer L, Aourz N, Van Eeckhaut A, Portelli J, Andrews ZB, Massie A, De Bundel D, Smolders I. Caloric Restriction Protects against Lactacystin-Induced Degeneration of Dopamine Neurons Independent of the Ghrelin Receptor. Int J Mol Sci 2017; 18:ijms18030558. [PMID: 28273852 PMCID: PMC5372574 DOI: 10.3390/ijms18030558] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 02/14/2017] [Accepted: 02/20/2017] [Indexed: 02/06/2023] Open
Abstract
Parkinson’s disease (PD) is a neurodegenerative disorder, characterized by a loss of dopamine (DA) neurons in the substantia nigra pars compacta (SNc). Caloric restriction (CR) has been shown to exert ghrelin-dependent neuroprotective effects in the 1-methyl-4-phenyl-1,2,3,6-tetrathydropyridine (MPTP)-based animal model for PD. We here investigated whether CR is neuroprotective in the lactacystin (LAC) mouse model for PD, in which proteasome disruption leads to the destruction of the DA neurons of the SNc, and whether this effect is mediated via the ghrelin receptor. Adult male ghrelin receptor wildtype (WT) and knockout (KO) mice were maintained on an ad libitum (AL) diet or on a 30% CR regimen. After 3 weeks, LAC was injected unilaterally into the SNc, and the degree of DA neuron degeneration was evaluated 1 week later. In AL mice, LAC injection significanty reduced the number of DA neurons and striatal DA concentrations. CR protected against DA neuron degeneration following LAC injection. However, no differences were observed between ghrelin receptor WT and KO mice. These results indicate that CR can protect the nigral DA neurons from toxicity related to proteasome disruption; however, the ghrelin receptor is not involved in this effect.
Collapse
Affiliation(s)
- Jessica Coppens
- Research Group Experimental Pharmacology (EFAR/FASC), Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussel, Belgium.
| | - Eduard Bentea
- Research Group Pharmaceutical Biotechnology and Molecular Biology (MICH), Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussel, Belgium.
| | - Jacqueline A Bayliss
- Department of Physiology, School of Biomedical and Psychological Sciences, Monash University, Clayton, Melbourne 3800, Australia.
| | - Thomas Demuyser
- Research Group Experimental Pharmacology (EFAR/FASC), Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussel, Belgium.
| | - Laura Walrave
- Research Group Experimental Pharmacology (EFAR/FASC), Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussel, Belgium.
| | - Giulia Albertini
- Research Group Experimental Pharmacology (EFAR/FASC), Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussel, Belgium.
| | - Joeri Van Liefferinge
- Research Group Experimental Pharmacology (EFAR/FASC), Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussel, Belgium.
| | - Lauren Deneyer
- Research Group Pharmaceutical Biotechnology and Molecular Biology (MICH), Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussel, Belgium.
| | - Najat Aourz
- Research Group Experimental Pharmacology (EFAR/FASC), Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussel, Belgium.
| | - Ann Van Eeckhaut
- Research Group Experimental Pharmacology (EFAR/FASC), Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussel, Belgium.
| | - Jeanelle Portelli
- Research Group Experimental Pharmacology (EFAR/FASC), Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussel, Belgium.
| | - Zane B Andrews
- Department of Physiology, School of Biomedical and Psychological Sciences, Monash University, Clayton, Melbourne 3800, Australia.
| | - Ann Massie
- Research Group Pharmaceutical Biotechnology and Molecular Biology (MICH), Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussel, Belgium.
| | - Dimitri De Bundel
- Research Group Experimental Pharmacology (EFAR/FASC), Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussel, Belgium.
| | - Ilse Smolders
- Research Group Experimental Pharmacology (EFAR/FASC), Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussel, Belgium.
| |
Collapse
|
630
|
Frago LM, Chowen JA. Involvement of Astrocytes in Mediating the Central Effects of Ghrelin. Int J Mol Sci 2017; 18:ijms18030536. [PMID: 28257088 PMCID: PMC5372552 DOI: 10.3390/ijms18030536] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Revised: 02/16/2017] [Accepted: 02/25/2017] [Indexed: 12/31/2022] Open
Abstract
Although astrocytes are the most abundant cells in the mammalian brain, much remains to be learned about their molecular and functional features. Astrocytes express receptors for numerous hormones and metabolic factors, including the appetite-promoting hormone ghrelin. The metabolic effects of ghrelin are largely opposite to those of leptin, as it stimulates food intake and decreases energy expenditure. Ghrelin is also involved in glucose-sensing and glucose homeostasis. The widespread expression of the ghrelin receptor in the central nervous system suggests that this hormone is not only involved in metabolism, but also in other essential functions in the brain. In fact, ghrelin has been shown to promote cell survival and neuroprotection, with some studies exploring the use of ghrelin as a therapeutic agent against metabolic and neurodegenerative diseases. In this review, we highlight the possible role of glial cells as mediators of ghrelin's actions within the brain.
Collapse
Affiliation(s)
- Laura M Frago
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación la Princesa, CIBER de Obesidad Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28009 Madrid, Spain.
- Department of Pediatrics, Universidad Autónoma de Madrid, 28049 Madrid, Spain.
| | - Julie A Chowen
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación la Princesa, CIBER de Obesidad Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28009 Madrid, Spain.
| |
Collapse
|
631
|
MacLean PS, Blundell JE, Mennella JA, Batterham RL. Biological control of appetite: A daunting complexity. Obesity (Silver Spring) 2017; 25 Suppl 1:S8-S16. [PMID: 28229538 PMCID: PMC5407690 DOI: 10.1002/oby.21771] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 12/20/2016] [Indexed: 01/01/2023]
Abstract
OBJECTIVE This review summarizes a portion of the discussions of an NIH Workshop (Bethesda, MD, 2015) titled "Self-Regulation of Appetite-It's Complicated," which focused on the biological aspects of appetite regulation. METHODS This review summarizes the key biological inputs of appetite regulation and their implications for body weight regulation. RESULTS These discussions offer an update of the long-held, rigid perspective of an "adipocentric" biological control, taking a broader view that also includes important inputs from the digestive tract, from lean mass, and from the chemical sensory systems underlying taste and smell. It is only beginning to be understood how these biological systems are integrated and how this integrated input influences appetite and food eating behaviors. The relevance of these biological inputs was discussed primarily in the context of obesity and the problem of weight regain, touching on topics related to the biological predisposition for obesity and the impact that obesity treatments (dieting, exercise, bariatric surgery, etc.) might have on appetite and weight loss maintenance. Finally considered is a common theme that pervaded the workshop discussions, which was individual variability. CONCLUSIONS It is this individual variability in the predisposition for obesity and in the biological response to weight loss that makes the biological component of appetite regulation so complicated. When this individual biological variability is placed in the context of the diverse environmental and behavioral pressures that also influence food eating behaviors, it is easy to appreciate the daunting complexities that arise with the self-regulation of appetite.
Collapse
Affiliation(s)
- Paul S. MacLean
- University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - John E. Blundell
- Faculty of Medicine and Health, University of Leeds, Leeds LS2 9JT, UK
| | | | - Rachel L. Batterham
- Centre for Obesity Research, Rayne Institute, University College London, London WC1E 6JJ, UK
- National Institute of Health Research, University College London Hospital Biomedical Research Centre, London W1T 7DN, UK
| |
Collapse
|
632
|
Isorna E, de Pedro N, Valenciano AI, Alonso-Gómez ÁL, Delgado MJ. Interplay between the endocrine and circadian systems in fishes. J Endocrinol 2017; 232:R141-R159. [PMID: 27999088 DOI: 10.1530/joe-16-0330] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 12/20/2016] [Indexed: 12/11/2022]
Abstract
The circadian system is responsible for the temporal organisation of physiological functions which, in part, involves daily cycles of hormonal activity. In this review, we analyse the interplay between the circadian and endocrine systems in fishes. We first describe the current model of fish circadian system organisation and the basis of the molecular clockwork that enables different tissues to act as internal pacemakers. This system consists of a net of central and peripherally located oscillators and can be synchronised by the light-darkness and feeding-fasting cycles. We then focus on two central neuroendocrine transducers (melatonin and orexin) and three peripheral hormones (leptin, ghrelin and cortisol), which are involved in the synchronisation of the circadian system in mammals and/or energy status signalling. We review the role of each of these as overt rhythms (i.e. outputs of the circadian system) and, for the first time, as key internal temporal messengers that act as inputs for other endogenous oscillators. Based on acute changes in clock gene expression, we describe the currently accepted model of endogenous oscillator entrainment by the light-darkness cycle and propose a new model for non-photic (endocrine) entrainment, highlighting the importance of the bidirectional cross-talking between the endocrine and circadian systems in fishes. The flexibility of the fish circadian system combined with the absence of a master clock makes these vertebrates a very attractive model for studying communication among oscillators to drive functionally coordinated outputs.
Collapse
Affiliation(s)
- Esther Isorna
- Departamento de Fisiología (Fisiología Animal II)Facultad de Biología, Universidad Complutense de Madrid, Madrid, Spain
| | - Nuria de Pedro
- Departamento de Fisiología (Fisiología Animal II)Facultad de Biología, Universidad Complutense de Madrid, Madrid, Spain
| | - Ana I Valenciano
- Departamento de Fisiología (Fisiología Animal II)Facultad de Biología, Universidad Complutense de Madrid, Madrid, Spain
| | - Ángel L Alonso-Gómez
- Departamento de Fisiología (Fisiología Animal II)Facultad de Biología, Universidad Complutense de Madrid, Madrid, Spain
| | - María J Delgado
- Departamento de Fisiología (Fisiología Animal II)Facultad de Biología, Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
633
|
Blanco AM, Bertucci JI, Sánchez-Bretaño A, Delgado MJ, Valenciano AI, Unniappan S. Ghrelin modulates gene and protein expression of digestive enzymes in the intestine and hepatopancreas of goldfish (Carassius auratus) via the GHS-R1a: Possible roles of PLC/PKC and AC/PKA intracellular signaling pathways. Mol Cell Endocrinol 2017; 442:165-181. [PMID: 28042022 DOI: 10.1016/j.mce.2016.12.027] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 12/28/2016] [Accepted: 12/28/2016] [Indexed: 12/13/2022]
Abstract
Ghrelin, a multifunctional gut-brain hormone, is involved in the regulation of gastric functions in mammals. This study aimed to determine whether ghrelin modulates digestive enzymes in goldfish (Carassius auratus). Immunofluorescence microscopy found colocalization of ghrelin, GHS-R1a and the digestive enzymes sucrase-isomaltase, aminopeptidase A, trypsin and lipoprotein lipase in intestinal and hepatopancreatic cells. In vitro ghrelin treatment in intestinal and hepatopancreas explant culture led to a concentration- and time-dependent modulation (mainly stimulatory) of most of the digestive enzymes tested. The ghrelin-induced upregulations of digestive enzyme expression were all abolished by preincubation with the GHS-R1a ghrelin receptor antagonist [D-Lys3]-GHRP-6, and most of them by the phospholipase C inhibitor U73122 or the protein kinase A inhibitor H89. This indicates that ghrelin effects on digestive enzymes are mediated by GHS-R1a, partly by triggering the PLC/PKC and AC/PKA intracellular signaling pathways. These data suggest a role for ghrelin on digestive processes in fish.
Collapse
Affiliation(s)
- Ayelén Melisa Blanco
- Laboratory of Integrative Neuroendocrinology, Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, 52 Campus Drive, S7N 5B4 Saskatoon, Saskatchewan, Canada; Departamento de Fisiología (Fisiología Animal II), Facultad de Biología, Universidad Complutense de Madrid, José Antonio Nováis 12, 28040 Madrid, Spain.
| | - Juan Ignacio Bertucci
- Laboratory of Integrative Neuroendocrinology, Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, 52 Campus Drive, S7N 5B4 Saskatoon, Saskatchewan, Canada; Instituto de Investigaciones Biotecnológicas-Instituto Tecnológico Chascomús, Avenida Intendente Marinos Km. 8,2, 7130 Chascomús, Buenos Aires, Argentina.
| | - Aída Sánchez-Bretaño
- Department of Pharmacology and Toxicology, and Neuroscience Institute, Morehouse School of Medicine, 720 Westview Drive, GA 30310 Atlanta, GA, United States.
| | - María Jesús Delgado
- Departamento de Fisiología (Fisiología Animal II), Facultad de Biología, Universidad Complutense de Madrid, José Antonio Nováis 12, 28040 Madrid, Spain.
| | - Ana Isabel Valenciano
- Departamento de Fisiología (Fisiología Animal II), Facultad de Biología, Universidad Complutense de Madrid, José Antonio Nováis 12, 28040 Madrid, Spain.
| | - Suraj Unniappan
- Laboratory of Integrative Neuroendocrinology, Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, 52 Campus Drive, S7N 5B4 Saskatoon, Saskatchewan, Canada.
| |
Collapse
|
634
|
Morin JP, Rodríguez-Durán LF, Guzmán-Ramos K, Perez-Cruz C, Ferreira G, Diaz-Cintra S, Pacheco-López G. Palatable Hyper-Caloric Foods Impact on Neuronal Plasticity. Front Behav Neurosci 2017; 11:19. [PMID: 28261067 PMCID: PMC5306218 DOI: 10.3389/fnbeh.2017.00019] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 01/23/2017] [Indexed: 01/01/2023] Open
Abstract
Neural plasticity is an intrinsic and essential characteristic of the nervous system that allows animals “self-tuning” to adapt to their environment over their lifetime. Activity-dependent synaptic plasticity in the central nervous system is a form of neural plasticity that underlies learning and memory formation, as well as long-lasting, environmentally-induced maladaptive behaviors, such as drug addiction and overeating of palatable hyper-caloric (PHc) food. In western societies, the abundance of PHc foods has caused a dramatic increase in the incidence of overweight/obesity and related disorders. To this regard, it has been suggested that increased adiposity may be caused at least in part by behavioral changes in the affected individuals that are induced by the chronic consumption of PHc foods; some authors have even drawn attention to the similarity that exists between over-indulgent eating and drug addiction. Long-term misuse of certain dietary components has also been linked to chronic neuroimmune maladaptation that may predispose individuals to neurodegenerative conditions such as Alzheimer’s disease. In this review article, we discuss recent evidence that shows how consumption of PHc food can cause maladaptive neural plasticity that converts short-term ingestive drives into compulsive behaviors. We also discuss the neural mechanisms of how chronic consumption of PHc foods may alter brain function and lead to cognitive impairments, focusing on prenatal, childhood and adolescence as vulnerable neurodevelopmental stages to dietary environmental insults. Finally, we outline a societal agenda for harnessing permissive obesogenic environments.
Collapse
Affiliation(s)
- Jean-Pascal Morin
- Department of Health Sciences, Metropolitan Autonomous University (UAM)Lerma, Mexico; Institute of Medical Psychology and Behavioral Immunobiology, University Hospital Essen, University of Duisburg-EssenEssen, Germany
| | - Luis F Rodríguez-Durán
- Department of Health Sciences, Metropolitan Autonomous University (UAM)Lerma, Mexico; Laboratory of Neurobiology of Learning and Memory, Division of Research and Graduate Studies, Faculty of Psychology, National Autonomous University of Mexico (UNAM)Mexico City, Mexico
| | - Kioko Guzmán-Ramos
- Department of Health Sciences, Metropolitan Autonomous University (UAM) Lerma, Mexico
| | - Claudia Perez-Cruz
- Department of Pharmacology, Center of Research and Advance Studies (CINVESTAV) Mexico City, Mexico
| | - Guillaume Ferreira
- Laboratory of Nutrition and Integrative Neurobiology, National Institute of Agricultural Research (INRA), UMR 1286Bordeaux, France; Laboratory of Nutrition and Integrative Neurobiology, Université de BordeauxBordeaux, France
| | - Sofia Diaz-Cintra
- Institute of Neurobiology, National Autonomous University of Mexico (UNAM) Queretaro, Mexico
| | - Gustavo Pacheco-López
- Department of Health Sciences, Metropolitan Autonomous University (UAM)Lerma, Mexico; Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) ZurichSchwerzenbach, Switzerland
| |
Collapse
|
635
|
McGovern-Gooch KR, Mahajani NS, Garagozzo A, Schramm AJ, Hannah LG, Sieburg MA, Chisholm JD, Hougland JL. Synthetic Triterpenoid Inhibition of Human Ghrelin O-Acyltransferase: The Involvement of a Functionally Required Cysteine Provides Mechanistic Insight into Ghrelin Acylation. Biochemistry 2017; 56:919-931. [PMID: 28134508 DOI: 10.1021/acs.biochem.6b01008] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The peptide hormone ghrelin plays a key role in regulating hunger and energy balance within the body. Ghrelin signaling presents a promising and unexploited target for development of small molecule therapeutics for treatment of obesity, diabetes, and other health conditions. Inhibition of ghrelin O-acyltransferase (GOAT), which catalyzes an essential octanoylation step in ghrelin maturation, offers a potential avenue for controlling ghrelin signaling. Through screening a small molecule library, we have identified a class of synthetic triterpenoids that efficiently inhibit ghrelin acylation by the human isoform of GOAT (hGOAT). These compounds function as covalent reversible inhibitors of hGOAT, providing the first evidence of the involvement of a nucleophilic cysteine residue in substrate acylation by a MBOAT family acyltransferase. Surprisingly, the mouse form of GOAT does not exhibit susceptibility to cysteine-modifying electrophiles, revealing an important distinction in the activity and behavior between these closely related GOAT isoforms. This study establishes these compounds as potent small molecule inhibitors of ghrelin acylation and provides a foundation for the development of novel hGOAT inhibitors as therapeutics targeting diabetes and obesity.
Collapse
Affiliation(s)
| | - Nivedita S Mahajani
- Department of Chemistry, Syracuse University , Syracuse, New York 13244, United States
| | - Ariana Garagozzo
- Department of Chemistry, Syracuse University , Syracuse, New York 13244, United States
| | - Anthony J Schramm
- Department of Chemistry, Syracuse University , Syracuse, New York 13244, United States
| | - Lauren G Hannah
- Department of Chemistry, Syracuse University , Syracuse, New York 13244, United States
| | - Michelle A Sieburg
- Department of Chemistry, Syracuse University , Syracuse, New York 13244, United States
| | - John D Chisholm
- Department of Chemistry, Syracuse University , Syracuse, New York 13244, United States
| | - James L Hougland
- Department of Chemistry, Syracuse University , Syracuse, New York 13244, United States
| |
Collapse
|
636
|
Gong J, Hu M, Huang Z, Fang K, Wang D, Chen Q, Li J, Yang D, Zou X, Xu L, Wang K, Dong H, Lu F. Berberine Attenuates Intestinal Mucosal Barrier Dysfunction in Type 2 Diabetic Rats. Front Pharmacol 2017; 8:42. [PMID: 28217099 PMCID: PMC5290458 DOI: 10.3389/fphar.2017.00042] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2016] [Accepted: 01/19/2017] [Indexed: 01/19/2023] Open
Abstract
Background: Intestinal mucosal barrier dysfunction plays an important role in the development of diabetes mellitus (DM). Berberine (BBR), a kind of isoquinoline alkaloid, is widely known to be effective for both DM and diarrhea. Here, we explored whether the anti-diabetic effect of BBR was related to the intestine mucosal barrier. Methods and Results: The rat model of T2DM was established by high glucose and fat diet feeding and intravenous injection of streptozocin. Then, those diabetic rats were treated with BBR at different concentrations for 9 weeks. The results showed, in addition to hyperglycemia and hyperlipidemia, diabetic rats were also characterized by proinflammatory intestinal changes, altered gut-derived hormones, and 2.77-fold increase in intestinal permeability. However, the treatment with BBR significantly reversed the above changes in diabetic rats, presenting as the improvement of the high glucose and triglyceride levels, the relief of the inflammatory changes of intestinal immune system, and the attenuation of the intestinal barrier damage. BBR treatment at a high concentration also decreased the intestinal permeability by 27.5% in diabetic rats. Furthermore, BBR regulated the expressions of the molecules involved in TLR4/MyD88/NF-κB signaling pathways in intestinal tissue of diabetic rats. Conclusion: The hypoglycemic effects of BBR might be related to the improvement in gut-derived hormones and the attenuation of intestinal mucosal mechanic and immune barrier damages.
Collapse
Affiliation(s)
- Jing Gong
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan, China
| | - Meilin Hu
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan, China
| | - Zhaoyi Huang
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan, China
| | - Ke Fang
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan, China
| | - Dingkun Wang
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan, China
| | - Qingjie Chen
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology Wuhan, China
| | - Jingbin Li
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan, China
| | - Desen Yang
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, China; Department of Pharmacy, Hubei University of Traditional Chinese MedicineWuhan, China
| | - Xin Zou
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan, China
| | - Lijun Xu
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan, China
| | - Kaifu Wang
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan, China
| | - Hui Dong
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan, China
| | - Fuer Lu
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan, China
| |
Collapse
|
637
|
Stievenard A, Méquinion M, Andrews ZB, Destée A, Chartier-Harlin MC, Viltart O, Vanbesien-Mailliot CC. Is there a role for ghrelin in central dopaminergic systems? Focus on nigrostriatal and mesocorticolimbic pathways. Neurosci Biobehav Rev 2017; 73:255-275. [DOI: 10.1016/j.neubiorev.2016.11.021] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 11/23/2016] [Accepted: 11/25/2016] [Indexed: 12/21/2022]
|
638
|
Zhang CJ, Bidlingmaier M, Altaye M, Page LC, D'Alessio D, Tschöp MH, Tong J. Acute administration of acyl, but not desacyl ghrelin, decreases blood pressure in healthy humans. Eur J Endocrinol 2017; 176:123-132. [PMID: 27913606 PMCID: PMC5325691 DOI: 10.1530/eje-16-0789] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 11/02/2016] [Accepted: 11/08/2016] [Indexed: 01/13/2023]
Abstract
OBJECTIVE To compare the effects of acyl ghrelin (AG) and desacyl ghrelin (DAG) on blood pressure (BP), heart rate (HR) and other autonomic parameters in healthy humans and to elucidate the hormonal mechanisms through which AG could exert its cardiovascular effects. DESIGN Seventeen healthy participants underwent frequent monitoring of systolic (sBP) and diastolic blood pressure (dBP), HR, respiratory rate (RR) and body surface temperature (Temp) during continuous infusion of AG, DAG, combined AG + DAG or saline control before and during an IV glucose tolerance test on 4 separate days. Plasma catecholamines, renin and aldosterone levels were also measured. Differences in outcome measures between treatment groups were assessed using mixed-model analysis. RESULTS Compared to the saline control, AG and combined AG + DAG infusions decreased sBP, dBP, mean arterial blood pressure (MAP), HR and Temp. In contrast, DAG infusion did not alter BP, RR or Temp, but did decrease HR. The AG and AG + DAG infusions also raised plasma aldosterone levels compared to saline (P < 0.001) without affecting renin or catecholamine levels. CONCLUSIONS The decrease in BP, HR, RR and Temp with AG infusion suggests mediation through the autonomic nervous system. The lack of response to DAG suggests that these autonomic effects require activation of the ghrelin receptor.
Collapse
Affiliation(s)
- Cecilia J Zhang
- Division of EndocrinologyMetabolism and Nutrition, Department of Medicine, Duke University, Durham, North Carolina, USA
| | - Martin Bidlingmaier
- Medizinische Klinik und Poliklinik IVLudwig-Maximilians-Universität, Munich, Germany
| | - Mekibib Altaye
- BiostatisticsClinical Translational Research Center, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Laura C Page
- Division of Pediatric Endocrinology and DiabetesDepartment of Pediatrics, Duke University, Durham, North Carolina, USA
| | - David D'Alessio
- Division of EndocrinologyDiabetes and Metabolism, Department of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
- Division of EndocrinologyMetabolism and Nutrition, Department of Medicine, Duke University, Durham, North Carolina, USA
- Cincinnati Veterans Affairs Medical CenterCincinnati, Ohio, USA
| | - Matthias H Tschöp
- Division of EndocrinologyDiabetes and Metabolism, Department of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
- Institute for Obesity and DiabetesHelmholtz Diabetes Center Munich and Division of Metabolic Diseases, Technical University, Munich, Germany
| | - Jenny Tong
- Division of EndocrinologyDiabetes and Metabolism, Department of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
- Division of EndocrinologyMetabolism and Nutrition, Department of Medicine, Duke University, Durham, North Carolina, USA
| |
Collapse
|
639
|
Hinney A, Kesselmeier M, Jall S, Volckmar AL, Föcker M, Antel J, Heid IM, Winkler TW, Grant SFA, Guo Y, Bergen AW, Kaye W, Berrettini W, Hakonarson H, Herpertz-Dahlmann B, de Zwaan M, Herzog W, Ehrlich S, Zipfel S, Egberts KM, Adan R, Brandys M, van Elburg A, Boraska Perica V, Franklin CS, Tschöp MH, Zeggini E, Bulik CM, Collier D, Scherag A, Müller TD, Hebebrand J. Evidence for three genetic loci involved in both anorexia nervosa risk and variation of body mass index. Mol Psychiatry 2017; 22:192-201. [PMID: 27184124 PMCID: PMC5114162 DOI: 10.1038/mp.2016.71] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 02/22/2016] [Accepted: 03/17/2016] [Indexed: 02/06/2023]
Abstract
The maintenance of normal body weight is disrupted in patients with anorexia nervosa (AN) for prolonged periods of time. Prior to the onset of AN, premorbid body mass index (BMI) spans the entire range from underweight to obese. After recovery, patients have reduced rates of overweight and obesity. As such, loci involved in body weight regulation may also be relevant for AN and vice versa. Our primary analysis comprised a cross-trait analysis of the 1000 single-nucleotide polymorphisms (SNPs) with the lowest P-values in a genome-wide association meta-analysis (GWAMA) of AN (GCAN) for evidence of association in the largest published GWAMA for BMI (GIANT). Subsequently we performed sex-stratified analyses for these 1000 SNPs. Functional ex vivo studies on four genes ensued. Lastly, a look-up of GWAMA-derived BMI-related loci was performed in the AN GWAMA. We detected significant associations (P-values <5 × 10-5, Bonferroni-corrected P<0.05) for nine SNP alleles at three independent loci. Interestingly, all AN susceptibility alleles were consistently associated with increased BMI. None of the genes (chr. 10: CTBP2, chr. 19: CCNE1, chr. 2: CARF and NBEAL1; the latter is a region with high linkage disequilibrium) nearest to these SNPs has previously been associated with AN or obesity. Sex-stratified analyses revealed that the strongest BMI signal originated predominantly from females (chr. 10 rs1561589; Poverall: 2.47 × 10-06/Pfemales: 3.45 × 10-07/Pmales: 0.043). Functional ex vivo studies in mice revealed reduced hypothalamic expression of Ctbp2 and Nbeal1 after fasting. Hypothalamic expression of Ctbp2 was increased in diet-induced obese (DIO) mice as compared with age-matched lean controls. We observed no evidence for associations for the look-up of BMI-related loci in the AN GWAMA. A cross-trait analysis of AN and BMI loci revealed variants at three chromosomal loci with potential joint impact. The chromosome 10 locus is particularly promising given that the association with obesity was primarily driven by females. In addition, the detected altered hypothalamic expression patterns of Ctbp2 and Nbeal1 as a result of fasting and DIO implicate these genes in weight regulation.
Collapse
Affiliation(s)
- A Hinney
- Department of Child and Adolescent Psychiatry, Psychotherapy, and Psychosomatics, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - M Kesselmeier
- Clinical Epidemiology, Integrated Research and Treatment Center, Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany
| | - S Jall
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center & German Diabetes Center (DZD), Helmholtz Zentrum München, Neuherberg, Germany
- Division of Metabolic Diseases, Department of Medicine, Technische Universität München, Munich, Germany
| | - A-L Volckmar
- Department of Child and Adolescent Psychiatry, Psychotherapy, and Psychosomatics, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - M Föcker
- Department of Child and Adolescent Psychiatry, Psychotherapy, and Psychosomatics, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - J Antel
- Department of Child and Adolescent Psychiatry, Psychotherapy, and Psychosomatics, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - I M Heid
- Department of Genetic Epidemiology, University of Regensburg, Regensburg, Germany
| | - T W Winkler
- Department of Genetic Epidemiology, University of Regensburg, Regensburg, Germany
| | - S F A Grant
- Department of Pediatrics, University of Pennsylvania, Philadelphia, PA, USA
- Divisions of Genetics and Endocrinology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- The Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Y Guo
- The Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | | | - W Kaye
- Department of Psychiatry, University of California, San Diego, San Diego, CA, USA
| | - W Berrettini
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
| | - H Hakonarson
- The Division of Human Genetics, Department of Pediatrics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - B Herpertz-Dahlmann
- Department of Child and Adolescent Psychiatry and Psychotherapy, University Hospital of the RWTH Aachen, Aachen, Germany
| | - M de Zwaan
- Department of Psychosomatic Medicine and Psychotherapy, Hannover Medical School, Hannover, Germany
| | - W Herzog
- Department of Internal Medicine II, General Internal and Psychosomatic Medicine, University of Heidelberg, Heidelberg, Germany
| | - S Ehrlich
- Translational Developmental Neuroscience Section, Department of Child and Adolescent Psychiatry, Faculty of Medicine, TU-Dresden, University Hospital Carl Gustav Carus, Dresden University of Technology, Dresden, Germany
| | - S Zipfel
- Department of Psychosomatic Medicine and Psychotherapy, Medical University Hospital, Tübingen, Germany
| | - K M Egberts
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University of Würzburg, Würzburg, Germany
| | - R Adan
- Brain Center Rudolf Magnus, Department of Translational Neuroscience, University Medical Center Utrecht, Utrecht, The Netherlands
- Altrecht Eating Disorders Rintveld, Zeist, The Netherlands
| | - M Brandys
- Brain Center Rudolf Magnus, Department of Translational Neuroscience, University Medical Center Utrecht, Utrecht, The Netherlands
- Altrecht Eating Disorders Rintveld, Zeist, The Netherlands
| | - A van Elburg
- Brain Center Rudolf Magnus, Department of Translational Neuroscience, University Medical Center Utrecht, Utrecht, The Netherlands
| | - V Boraska Perica
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, UK
- University of Split School of Medicine, Split, Croatia
| | - C S Franklin
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, UK
| | - M H Tschöp
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center & German Diabetes Center (DZD), Helmholtz Zentrum München, Neuherberg, Germany
- Division of Metabolic Diseases, Department of Medicine, Technische Universität München, Munich, Germany
| | - E Zeggini
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, UK
| | - C M Bulik
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Nutrition, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - D Collier
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, King's College London, London, UK
- Eli Lilly and Company Ltd, Surrey, UK
| | - A Scherag
- Clinical Epidemiology, Integrated Research and Treatment Center, Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany
| | - T D Müller
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center & German Diabetes Center (DZD), Helmholtz Zentrum München, Neuherberg, Germany
- Division of Metabolic Diseases, Department of Medicine, Technische Universität München, Munich, Germany
| | - J Hebebrand
- Department of Child and Adolescent Psychiatry, Psychotherapy, and Psychosomatics, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
640
|
Churm R, Davies JS, Stephens JW, Prior SL. Ghrelin function in human obesity and type 2 diabetes: a concise review. Obes Rev 2017; 18:140-148. [PMID: 27899023 DOI: 10.1111/obr.12474] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 09/06/2016] [Accepted: 09/06/2016] [Indexed: 12/12/2022]
Abstract
The 28 amino acid hormone, ghrelin, has been found to have various effects on metabolism. This review will focus on the pathways integrated into ghrelin's effect within the hypothalamus, pancreas and adipocytes. The identification of molecules and pathways that regulate ghrelin-mediated lipid retention could establish new mechanisms underlying cellular energy homeostasis. The impact of acyl-ghrelin on glucose metabolism and lipid homeostasis may allow for novel preventative or early intervention therapeutic strategies to treat obesity related type 2 diabetes and associated metabolic dysfunction.
Collapse
Affiliation(s)
- R Churm
- Diabetes Research Group, Institute of Life Science 1, Swansea University, Swansea, UK
| | - J S Davies
- Molecular Neurobiology Research Group, Institute of Life Science 1, Swansea University, Swansea, UK
| | - J W Stephens
- Diabetes Research Group, Institute of Life Science 1, Swansea University, Swansea, UK
| | - S L Prior
- Diabetes Research Group, Institute of Life Science 1, Swansea University, Swansea, UK
| |
Collapse
|
641
|
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: 13.1] [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
|
642
|
Model-Based Discovery of Synthetic Agonists for the Zn 2+-Sensing G-Protein-Coupled Receptor 39 (GPR39) Reveals Novel Biological Functions. J Med Chem 2017; 60:886-898. [PMID: 28045522 DOI: 10.1021/acs.jmedchem.6b00648] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The G-protein-coupled receptor 39 (GPR39) is a G-protein-coupled receptor activated by Zn2+. We used a homology model-based approach to identify small-molecule pharmacological tool compounds for the receptor. The method focused on a putative binding site in GPR39 for synthetic ligands and knowledge of ligand binding to other receptors with similar binding pockets to select iterative series of minilibraries. These libraries were cherry-picked from all commercially available synthetic compounds. A total of only 520 compounds were tested in vitro, making this method broadly applicable for tool compound development. The compounds of the initial library were inactive when tested alone, but lead compounds were identified using Zn2+ as an allosteric enhancer. Highly selective, highly potent Zn2+-independent GPR39 agonists were found in subsequent minilibraries. These agonists identified GPR39 as a novel regulator of gastric somatostatin secretion.
Collapse
|
643
|
Al Massadi O, López M, Tschöp M, Diéguez C, Nogueiras R. Current Understanding of the Hypothalamic Ghrelin Pathways Inducing Appetite and Adiposity. Trends Neurosci 2017; 40:167-180. [PMID: 28108113 DOI: 10.1016/j.tins.2016.12.003] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 12/13/2016] [Accepted: 12/15/2016] [Indexed: 12/21/2022]
Abstract
Ghrelin is a multifaceted regulator of metabolism. Ghrelin regulates energy balance in the short term via induction of appetite and in the long term via increased body weight and adiposity. Recently, several central pathways modulating the metabolic actions of ghrelin were unmasked, and it was shown to act through different hypothalamic nuclei to induce feeding. Ghrelin also modulates glucose homeostasis, but the central mechanisms responsible for this action have not been studied in detail. Although ghrelin also acts through extrahypothalamic areas to promote feeding, this review specifically dissects hypothalamic control of ghrelin's orexigenic and adipogenic actions and presents current understanding of the intracellular ghrelin orexigenic pathways, including their dependence on other relevant systems implicated in energy balance.
Collapse
Affiliation(s)
- Omar Al Massadi
- Department of Physiology, School of Medicine-CiMUS, Instituto de Investigación Sanitaria (IDIS), University of Santiago de Compostela, Av de Barcelona s/n Santiago de Compostela (A Coruña), 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Spain.
| | - Miguel López
- Department of Physiology, School of Medicine-CiMUS, Instituto de Investigación Sanitaria (IDIS), University of Santiago de Compostela, Av de Barcelona s/n Santiago de Compostela (A Coruña), 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Spain
| | - Matthias Tschöp
- Helmholtz Diabetes Center, Helmholtz Zentrum München and German Center for Diabetes Research (DZD), Neuherberg, Germany; Division of Metabolic Diseases, Technische Universität München, Munich, Germany
| | - Carlos Diéguez
- Department of Physiology, School of Medicine-CiMUS, Instituto de Investigación Sanitaria (IDIS), University of Santiago de Compostela, Av de Barcelona s/n Santiago de Compostela (A Coruña), 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Spain
| | - Ruben Nogueiras
- Department of Physiology, School of Medicine-CiMUS, Instituto de Investigación Sanitaria (IDIS), University of Santiago de Compostela, Av de Barcelona s/n Santiago de Compostela (A Coruña), 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Spain.
| |
Collapse
|
644
|
Pereira JADS, da Silva FC, de Moraes-Vieira PMM. The Impact of Ghrelin in Metabolic Diseases: An Immune Perspective. J Diabetes Res 2017; 2017:4527980. [PMID: 29082258 PMCID: PMC5610818 DOI: 10.1155/2017/4527980] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Revised: 07/07/2017] [Accepted: 07/31/2017] [Indexed: 01/22/2023] Open
Abstract
Obesity and insulin resistance have reached epidemic proportions. Obesogenic conditions are associated with increased risk for the development of other comorbidities and obesity-related diseases. In metabolic disorders, there is chronic low-grade inflammation induced by the activation of immune cells, especially in metabolic relevant organs such as white adipose tissue (WAT). These immune cells are regulated by environmental and systemic cues. Ghrelin is a peptide secreted mainly by X/A-like gastric cells and acts through the growth hormone secretagogue receptor (GHS-R). This receptor is broadly expressed in the central nervous system (CNS) and in several cell types, including immune cells. Studies show that ghrelin induces an orexigenic state, and there is increasing evidence implicating an immunoregulatory role for ghrelin. Ghrelin mainly acts on the innate and adaptive immune systems to suppress inflammation and induce an anti-inflammatory profile. In this review, we discuss the immunoregulatory roles of ghrelin, the mechanisms by which ghrelin acts and potential pharmacological applications for ghrelin in the treatment of obesity-associated inflammatory diseases, such as type 2 diabetes (T2D).
Collapse
Affiliation(s)
- Jéssica Aparecida da Silva Pereira
- Laboratory of Immunometabolism, Department of Genetics, Evolution and Bioagents, Institute of Biology, University of Campinas, São Paulo, SP, Brazil
- Department of Immunology, Institute of Biomedical Science, University of São Paulo, São Paulo, SP, Brazil
| | - Felipe Corrêa da Silva
- Laboratory of Immunometabolism, Department of Genetics, Evolution and Bioagents, Institute of Biology, University of Campinas, São Paulo, SP, Brazil
| | - Pedro Manoel Mendes de Moraes-Vieira
- Laboratory of Immunometabolism, Department of Genetics, Evolution and Bioagents, Institute of Biology, University of Campinas, São Paulo, SP, Brazil
- Department of Immunology, Institute of Biomedical Science, University of São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
645
|
Fetissov SO. Role of the gut microbiota in host appetite control: bacterial growth to animal feeding behaviour. Nat Rev Endocrinol 2017; 13:11-25. [PMID: 27616451 DOI: 10.1038/nrendo.2016.150] [Citation(s) in RCA: 222] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The life of all animals is dominated by alternating feelings of hunger and satiety - the main involuntary motivations for feeding-related behaviour. Gut bacteria depend fully on their host for providing the nutrients necessary for their growth. The intrinsic ability of bacteria to regulate their growth and to maintain their population within the gut suggests that gut bacteria can interfere with molecular pathways controlling energy balance in the host. The current model of appetite control is based mainly on gut-brain signalling and the animal's own needs to maintain energy homeostasis; an alternative model might also involve bacteria-host communications. Several bacterial components and metabolites have been shown to stimulate intestinal satiety pathways; at the same time, their production depends on bacterial growth cycles. This short-term bacterial growth-linked modulation of intestinal satiety can be coupled with long-term regulation of appetite, controlled by the neuropeptidergic circuitry in the hypothalamus. Indeed, several bacterial products are detected in the systemic circulation, which might act directly on hypothalamic neurons. This Review analyses the data relevant to possible involvement of the gut bacteria in the regulation of host appetite and proposes an integrative homeostatic model of appetite control that includes energy needs of both the host and its gut bacteria.
Collapse
Affiliation(s)
- Sergueï O Fetissov
- Nutrition, Gut &Brain Laboratory, Inserm UMR 1073, University of Rouen Normandy, 22 Boulevard Gambetta, 76183 Rouen, France
| |
Collapse
|
646
|
Sakata I, Gong Z, Ikenoya C, Takemi S, Sakai T. The study of ghrelin secretion and acyl-modification using mice and ghrelinoma cell lines. Endocr J 2017; 64:S27-S29. [PMID: 28652540 DOI: 10.1507/endocrj.64.s27] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Ghrelin is a peptide hormone with a unique structure comprising a medium chain fatty acid modification. Ghrelin cells are known to be abundantly localized in the gastric mucosa and are released into the blood stream to exert their multifunctional physiological effects. To elucidate the regulatory mechanisms of ghrelin secretion and acyl-modification, we developed novel ghrelin-producing cell lines. Using ghrelinoma cell lines, we focused on the mechanisms of ghrelin secretion and found that several GPCRs were highly expressed in ghrelin cells. Then, we showed that noradrenaline treatment stimulated ghrelin secretion via β1-adrenergic receptor, and fasting-induced ghrelin elevation was completely inhibited by the β1-adrenergic receptor antagonist in mice. In addition, we demonstrated that long chain fatty acids, glucose, and L-glutamate significantly inhibited ghrelin secretion. Furthermore, we recently revealed that the genes involved in fatty acid synthesis and long chain fatty acid metabolism were expressed in ghrelin cells, and that CPT-1 inhibitor treatment dramatically decreased the levels of acyl-modified ghrelin. Here, we introduce the current knowledge of the mechanisms involving ghrelin secretion and its acyl-modification.
Collapse
Affiliation(s)
- Ichiro Sakata
- Area of Regulatory Biology, Division of Life Science, Graduate School of Science and Engineering, Saitama University, Saitama, Japan
| | - Zhi Gong
- Area of Regulatory Biology, Division of Life Science, Graduate School of Science and Engineering, Saitama University, Saitama, Japan
| | - Chika Ikenoya
- Area of Regulatory Biology, Division of Life Science, Graduate School of Science and Engineering, Saitama University, Saitama, Japan
| | - Shota Takemi
- Area of Regulatory Biology, Division of Life Science, Graduate School of Science and Engineering, Saitama University, Saitama, Japan
| | - Takafumi Sakai
- Area of Regulatory Biology, Division of Life Science, Graduate School of Science and Engineering, Saitama University, Saitama, Japan
- Area of Life-NanoBio, Division of Strategy Research, Graduate School of Science and Engineering, Saitama University, Saitama, Japan
| |
Collapse
|
647
|
Plasma kisspeptin and ghrelin levels are independently correlated with physical activity in patients with anorexia nervosa. Appetite 2017; 108:141-150. [DOI: 10.1016/j.appet.2016.09.032] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 09/01/2016] [Accepted: 09/27/2016] [Indexed: 12/22/2022]
|
648
|
Özcan B, Leenen PJM, Delhanty PJD, Baldéon-Rojas LY, Neggers SJ, van der Lely AJ. Unacylated ghrelin modulates circulating angiogenic cell number in insulin-resistant states. Diabetol Metab Syndr 2017; 9:43. [PMID: 28572856 PMCID: PMC5452348 DOI: 10.1186/s13098-017-0239-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 05/17/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Type 2 diabetes (T2D) is associated with reduced numbers and impaired function of circulating angiogenic cells (CAC) which contributes to the progression of atherosclerosis and microvascular disease. Previous studies suggest that short-term infusion of unacylated ghrelin (UAG) normalizes CAC number in patients with T2D. To determine dose-dependent effects of short-term infusion of UAG in T2D patients using a cross-over model, and of long-term infusion of UAG in obese mice, on differentiation of monocyte progenitors into CAC. METHODS Eight overweight T2D patients were infused overnight with 3 and 10 µg/kg/h of UAG in a double-blind, placebo-controlled cross-over study. To assess the effects of long-term UAG treatment, obese mice were infused with UAG for 4 weeks. Monocyte progenitors were assessed for their ability to differentiate into CAC in vitro. RESULTS In T2D patients, UAG treatment caused a reduction in differentiation of CAC, dependent on UAG dose and differentiation method. However, mice treated with UAG showed a significant increase in differentiation of bone marrow progenitors into CAC. CONCLUSION UAG causes a minor suppressive effect on CAC development after short-term treatment in humans, but experiments in mice suggest that long-term treatment has beneficial effects on CAC formation. The Netherlands Trial Register: TC=2487.
Collapse
Affiliation(s)
- Behiye Özcan
- Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
| | | | | | | | | | | |
Collapse
|
649
|
Steinert RE, Feinle-Bisset C, Asarian L, Horowitz M, Beglinger C, Geary N. Ghrelin, CCK, GLP-1, and PYY(3-36): Secretory Controls and Physiological Roles in Eating and Glycemia in Health, Obesity, and After RYGB. Physiol Rev 2017; 97:411-463. [PMID: 28003328 PMCID: PMC6151490 DOI: 10.1152/physrev.00031.2014] [Citation(s) in RCA: 367] [Impact Index Per Article: 52.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The efficacy of Roux-en-Y gastric-bypass (RYGB) and other bariatric surgeries in the management of obesity and type 2 diabetes mellitus and novel developments in gastrointestinal (GI) endocrinology have renewed interest in the roles of GI hormones in the control of eating, meal-related glycemia, and obesity. Here we review the nutrient-sensing mechanisms that control the secretion of four of these hormones, ghrelin, cholecystokinin (CCK), glucagon-like peptide-1 (GLP-1), and peptide tyrosine tyrosine [PYY(3-36)], and their contributions to the controls of GI motor function, food intake, and meal-related increases in glycemia in healthy-weight and obese persons, as well as in RYGB patients. Their physiological roles as classical endocrine and as locally acting signals are discussed. Gastric emptying, the detection of specific digestive products by small intestinal enteroendocrine cells, and synergistic interactions among different GI loci all contribute to the secretion of ghrelin, CCK, GLP-1, and PYY(3-36). While CCK has been fully established as an endogenous endocrine control of eating in healthy-weight persons, the roles of all four hormones in eating in obese persons and following RYGB are uncertain. Similarly, only GLP-1 clearly contributes to the endocrine control of meal-related glycemia. It is likely that local signaling is involved in these hormones' actions, but methods to determine the physiological status of local signaling effects are lacking. Further research and fresh approaches are required to better understand ghrelin, CCK, GLP-1, and PYY(3-36) physiology; their roles in obesity and bariatric surgery; and their therapeutic potentials.
Collapse
Affiliation(s)
- Robert E Steinert
- University of Adelaide Discipline of Medicine and National Health and Medical Research Council of Australia Centre of Research Excellence in Translating Nutritional Science to Good Health, Adelaide, Australia; DSM Nutritional Products, R&D Human Nutrition and Health, Basel, Switzerland; Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland; Department of Biomedicine and Division of Gastroenterology, University Hospital Basel, Basel, Switzerland; and Department of Psychiatry, Weill Medical College of Cornell University, New York, New York
| | - Christine Feinle-Bisset
- University of Adelaide Discipline of Medicine and National Health and Medical Research Council of Australia Centre of Research Excellence in Translating Nutritional Science to Good Health, Adelaide, Australia; DSM Nutritional Products, R&D Human Nutrition and Health, Basel, Switzerland; Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland; Department of Biomedicine and Division of Gastroenterology, University Hospital Basel, Basel, Switzerland; and Department of Psychiatry, Weill Medical College of Cornell University, New York, New York
| | - Lori Asarian
- University of Adelaide Discipline of Medicine and National Health and Medical Research Council of Australia Centre of Research Excellence in Translating Nutritional Science to Good Health, Adelaide, Australia; DSM Nutritional Products, R&D Human Nutrition and Health, Basel, Switzerland; Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland; Department of Biomedicine and Division of Gastroenterology, University Hospital Basel, Basel, Switzerland; and Department of Psychiatry, Weill Medical College of Cornell University, New York, New York
| | - Michael Horowitz
- University of Adelaide Discipline of Medicine and National Health and Medical Research Council of Australia Centre of Research Excellence in Translating Nutritional Science to Good Health, Adelaide, Australia; DSM Nutritional Products, R&D Human Nutrition and Health, Basel, Switzerland; Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland; Department of Biomedicine and Division of Gastroenterology, University Hospital Basel, Basel, Switzerland; and Department of Psychiatry, Weill Medical College of Cornell University, New York, New York
| | - Christoph Beglinger
- University of Adelaide Discipline of Medicine and National Health and Medical Research Council of Australia Centre of Research Excellence in Translating Nutritional Science to Good Health, Adelaide, Australia; DSM Nutritional Products, R&D Human Nutrition and Health, Basel, Switzerland; Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland; Department of Biomedicine and Division of Gastroenterology, University Hospital Basel, Basel, Switzerland; and Department of Psychiatry, Weill Medical College of Cornell University, New York, New York
| | - Nori Geary
- University of Adelaide Discipline of Medicine and National Health and Medical Research Council of Australia Centre of Research Excellence in Translating Nutritional Science to Good Health, Adelaide, Australia; DSM Nutritional Products, R&D Human Nutrition and Health, Basel, Switzerland; Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland; Department of Biomedicine and Division of Gastroenterology, University Hospital Basel, Basel, Switzerland; and Department of Psychiatry, Weill Medical College of Cornell University, New York, New York
| |
Collapse
|
650
|
Jiao Q, Du X, Li Y, Gong B, Shi L, Tang T, Jiang H. The neurological effects of ghrelin in brain diseases: Beyond metabolic functions. Neurosci Biobehav Rev 2016; 73:98-111. [PMID: 27993602 DOI: 10.1016/j.neubiorev.2016.12.010] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2016] [Revised: 12/01/2016] [Accepted: 12/10/2016] [Indexed: 02/08/2023]
Abstract
Ghrelin, a peptide released by the stomach that plays a major role in regulating energy metabolism, has recently been shown to have effects on neurobiological behaviors. Ghrelin enhances neuronal survival by reducing apoptosis, alleviating inflammation and oxidative stress, and accordingly improving mitochondrial function. Ghrelin also stimulates the proliferation, differentiation and migration of neural stem/progenitor cells (NS/PCs). Additionally, the ghrelin is benefit for the recovery of memory, mood and cognitive dysfunction after stroke or traumatic brain injury. Because of its neuroprotective and neurogenic roles, ghrelin may be used as a therapeutic agent in the brain to combat neurodegenerative disease. In this review, we highlight the pre-clinical evidence and the proposed mechanisms underlying the role of ghrelin in physiological and pathological brain function.
Collapse
Affiliation(s)
- Qian Jiao
- 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; Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, 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; Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, China.
| | - Yong Li
- 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; Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, China.
| | - Bing Gong
- 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.
| | - Limin Shi
- 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; Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, China.
| | - Tingting Tang
- 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; Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, 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; Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, China.
| |
Collapse
|