101
|
Jensen M, Ratner C, Rudenko O, Christiansen SH, Skov LJ, Hundahl C, Woldbye DPD, Holst B. Anxiolytic-Like Effects of Increased Ghrelin Receptor Signaling in the Amygdala. Int J Neuropsychopharmacol 2016; 19:pyv123. [PMID: 26578081 PMCID: PMC4886665 DOI: 10.1093/ijnp/pyv123] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Revised: 10/13/2015] [Accepted: 11/02/2015] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Besides the well-known effects of ghrelin on adiposity and food intake regulation, the ghrelin system has been shown to regulate aspects of behavior including anxiety and stress. However, the effect of virus-mediated overexpression of the ghrelin receptor in the amygdala has not previously been addressed directly. METHODS First, we examined the acute effect of peripheral ghrelin administration on anxiety- and depression-like behavior using the open field, elevated plus maze, forced swim, and tail suspension tests. Next, we examined the effect of peripheral ghrelin administration and ghrelin receptor deficiency on stress in a familiar and social environment using the Intellicage system. Importantly, we also used a novel approach to study ghrelin receptor signaling in the brain by overexpressing the ghrelin receptor in the amygdala. We examined the effect of ghrelin receptor overexpression on anxiety-related behavior before and after acute stress and measured the modulation of serotonin receptor expression. RESULTS We found that ghrelin caused an anxiolytic-like effect in both the open field and elevated plus maze tests. Additionally, it attenuated air-puff-induced stress in the social environment, while the opposite was shown in ghrelin receptor deficient mice. Finally, we found that overexpression of the ghrelin receptor in the basolateral division of the amygdala caused an anxiolytic-like effect and decreased the 5HT1a receptor expression. CONCLUSIONS Ghrelin administration and overexpression of the ghrelin receptor in the amygdala induces anxiolytic-like behavior. Since the ghrelin receptor has high constitutive activity, ligand-independent signaling in vivo may be important for the observed anxiolytic-like effects. The anxiolytic effects seem to be mediated independently from the HPA axis, potentially engaging the central serotonin system.
Collapse
MESH Headings
- Amygdala/drug effects
- Amygdala/metabolism
- Amygdala/physiopathology
- Animals
- Anti-Anxiety Agents/pharmacology
- Anxiety/genetics
- Anxiety/metabolism
- Anxiety/prevention & control
- Anxiety/psychology
- Behavior, Animal/drug effects
- Disease Models, Animal
- Dose-Response Relationship, Drug
- Female
- Ghrelin/pharmacology
- Hindlimb Suspension
- Humans
- Locomotion/drug effects
- Male
- Maze Learning/drug effects
- Mice, Inbred C57BL
- Mice, Knockout
- Motor Activity/drug effects
- Receptor, Serotonin, 5-HT1A/metabolism
- Receptors, Ghrelin/agonists
- Receptors, Ghrelin/genetics
- Receptors, Ghrelin/metabolism
- Signal Transduction/drug effects
- Social Behavior
- Stress, Psychological/complications
- Stress, Psychological/metabolism
- Stress, Psychological/psychology
- Swimming
- Time Factors
Collapse
Affiliation(s)
- Morten Jensen
- Laboratory of Molecular Pharmacology, Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark, and The Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark (Mr Jensen, Ms Ratner, Dr Rudenko, Ms Skov, Ms Hundahl, and Dr Holst); Laboratory of Neural Plasticity, Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark (Drs Christiansen and Woldbye)
| | - Cecilia Ratner
- Laboratory of Molecular Pharmacology, Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark, and The Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark (Mr Jensen, Ms Ratner, Dr Rudenko, Ms Skov, Ms Hundahl, and Dr Holst); Laboratory of Neural Plasticity, Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark (Drs Christiansen and Woldbye)
| | - Olga Rudenko
- Laboratory of Molecular Pharmacology, Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark, and The Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark (Mr Jensen, Ms Ratner, Dr Rudenko, Ms Skov, Ms Hundahl, and Dr Holst); Laboratory of Neural Plasticity, Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark (Drs Christiansen and Woldbye)
| | - Søren H Christiansen
- Laboratory of Molecular Pharmacology, Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark, and The Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark (Mr Jensen, Ms Ratner, Dr Rudenko, Ms Skov, Ms Hundahl, and Dr Holst); Laboratory of Neural Plasticity, Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark (Drs Christiansen and Woldbye)
| | - Louise J Skov
- Laboratory of Molecular Pharmacology, Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark, and The Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark (Mr Jensen, Ms Ratner, Dr Rudenko, Ms Skov, Ms Hundahl, and Dr Holst); Laboratory of Neural Plasticity, Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark (Drs Christiansen and Woldbye)
| | - Cecilie Hundahl
- Laboratory of Molecular Pharmacology, Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark, and The Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark (Mr Jensen, Ms Ratner, Dr Rudenko, Ms Skov, Ms Hundahl, and Dr Holst); Laboratory of Neural Plasticity, Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark (Drs Christiansen and Woldbye)
| | - David P D Woldbye
- Laboratory of Molecular Pharmacology, Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark, and The Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark (Mr Jensen, Ms Ratner, Dr Rudenko, Ms Skov, Ms Hundahl, and Dr Holst); Laboratory of Neural Plasticity, Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark (Drs Christiansen and Woldbye)
| | - Birgitte Holst
- Laboratory of Molecular Pharmacology, Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark, and The Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark (Mr Jensen, Ms Ratner, Dr Rudenko, Ms Skov, Ms Hundahl, and Dr Holst); Laboratory of Neural Plasticity, Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark (Drs Christiansen and Woldbye).
| |
Collapse
|
102
|
Al-Hadraawy SK, Al-ghurabi ME, Al-musawi MM, Alzeyadi M. Ghrelin and melatonin as biomarkers in patients with giardiasis. BIOTECHNOL BIOTEC EQ 2016. [DOI: 10.1080/13102818.2016.1149038] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Affiliation(s)
| | | | - Malak Maged Al-musawi
- Department of Biology, College of Education for Girls, University of Kufa, Najaf, Iraq
| | - Mohammad Alzeyadi
- Department of Biology, College of Sciences, University of Kufa, Najaf, Iraq
| |
Collapse
|
103
|
Lv Z, Gao J, Wang L, Chen Z, Yuan H, Ma X, Lu J, Lv J, Wu X, Zhang L, Wei L, Xue R, Fu R, Ma L. Uremia-caused changes of ghrelin system in hippocampus may be associated with impaired cognitive function of hippocampus. Int Urol Nephrol 2016; 48:807-15. [DOI: 10.1007/s11255-016-1228-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 01/25/2016] [Indexed: 12/01/2022]
|
104
|
Ghrelin and Neurodegenerative Disorders-a Review. Mol Neurobiol 2016; 54:1144-1155. [PMID: 26809582 DOI: 10.1007/s12035-016-9729-1] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 01/14/2016] [Indexed: 12/13/2022]
Abstract
Ghrelin, the endogenous ligand of the growth hormone secretagogue receptor 1a (GHS-R1a), is a gut-derived, orexigenic peptide hormone that primarily regulates growth hormone secretion, food intake, and energy homeostasis. With the wide expression of GHS-R1a in extra-hypothalamic regions, the physiological role of ghrelin is more extensive than solely its involvement in metabolic function. Ghrelin has been shown to be involved in numerous higher brain functions, such as memory, reward, mood, and sleep. Some of these functions are disrupted in neurodegenerative disorders, including Parkinson's disease (PD), Alzheimer's disease (AD), and Huntington's disease (HD). This link between ghrelin and these neurodegenerative diseases is supported by numerous studies. This review aims to provide a comprehensive overview of the most recent evidence of the novel neuromodulatory role of ghrelin in PD, AD, and HD. Moreover, the changes in circulating and/or central ghrelin levels that are associated with disease progression are also postulated to be a biomarker for clinical diagnosis and therapy.
Collapse
|
105
|
Carnitine palmitoyltransferase 1C: From cognition to cancer. Prog Lipid Res 2015; 61:134-48. [PMID: 26708865 DOI: 10.1016/j.plipres.2015.11.004] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 11/18/2015] [Accepted: 11/23/2015] [Indexed: 11/21/2022]
Abstract
Carnitine palmitoyltransferase 1 (CPT1) C was the last member of the CPT1 family of genes to be discovered. CPT1A and CPT1B were identified as the gate-keeper enzymes for the entry of long-chain fatty acids (as carnitine esters) into mitochondria and their further oxidation, and they show differences in their kinetics and tissue expression. Although CPT1C exhibits high sequence similarity to CPT1A and CPT1B, it is specifically expressed in neurons (a cell-type that does not use fatty acids as fuel to any major extent), it is localized in the endoplasmic reticulum of cells, and it has minimal CPT1 catalytic activity with l-carnitine and acyl-CoA esters. The lack of an easily measurable biological activity has hampered attempts to elucidate the cellular and physiological role of CPT1C but has not diminished the interest of the biomedical research community in this CPT1 isoform. The observations that CPT1C binds malonyl-CoA and long-chain acyl-CoA suggest that it is a sensor of lipid metabolism in neurons, where it appears to impact ceramide and triacylglycerol (TAG) metabolism. CPT1C global knock-out mice show a wide range of brain disorders, including impaired cognition and spatial learning, motor deficits, and a deregulation in food intake and energy homeostasis. The first disease-causing CPT1C mutation was recently described in humans, with Cpt1c being identified as the gene causing hereditary spastic paraplegia. The putative role of CPT1C in the regulation of complex-lipid metabolism is supported by the observation that it is highly expressed in certain virulent tumor cells, conferring them resistance to glucose- and oxygen-deprivation. Therefore, CPT1C may be a promising target in the treatment of cancer. Here we review the molecular, biochemical, and structural properties of CPT1C and discuss its potential roles in brain function, and cancer.
Collapse
|
106
|
Szakács J, Csabafi K, Lipták N, Szabó G. The effect of obestatin on anxiety-like behaviour in mice. Behav Brain Res 2015; 293:41-5. [DOI: 10.1016/j.bbr.2015.06.042] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Revised: 06/24/2015] [Accepted: 06/28/2015] [Indexed: 01/08/2023]
|
107
|
Morley JE. Peptides and aging: Their role in anorexia and memory. Peptides 2015; 72:112-8. [PMID: 25895851 DOI: 10.1016/j.peptides.2015.04.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Revised: 04/03/2015] [Accepted: 04/04/2015] [Indexed: 12/16/2022]
Abstract
The rapid aging of the world's population has led to a need to increase our understanding of the pathophysiology of the factors leading to frailty and cognitive decline. Peptides have been shown to be involved in the pathophysiology of frailty and cognitive decline. Weight loss is a major component of frailty. In this review, we demonstrate a central role for both peripheral peptides (e.g., cholecystokinin and ghrelin) and neuropeptides (e.g., dynorphin and alpha-MSH) in the pathophysiology of the anorexia of aging. Similarly, peripheral peptides (e.g., ghrelin, glucagon-like peptide 1, and cholecystokinin) are modulators of memory. A number of centrally acting neuropeptides have also been shown to modulate cognitive processes. Amyloid-beta peptide in physiological levels is a memory enhancer, while in high (pathological) levels, it plays a key role in the development of Alzheimer's disease.
Collapse
Affiliation(s)
- John E Morley
- Divisions of Geriatric Medicine and Endocrinology, Saint Louis University School of Medicine, St Louis, MO, United States.
| |
Collapse
|
108
|
Fadó R, Soto D, Miñano-Molina AJ, Pozo M, Carrasco P, Yefimenko N, Rodríguez-Álvarez J, Casals N. Novel Regulation of the Synthesis of α-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid (AMPA) Receptor Subunit GluA1 by Carnitine Palmitoyltransferase 1C (CPT1C) in the Hippocampus. J Biol Chem 2015; 290:25548-60. [PMID: 26338711 DOI: 10.1074/jbc.m115.681064] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Indexed: 01/04/2023] Open
Abstract
The regulation of AMPA-type receptor (AMPAR) abundance in the postsynaptic membrane is an important mechanism involved in learning and memory formation. Recent data suggest that one of the constituents of the AMPAR complex is carnitine palmitoyltransferase 1C (CPT1C), a brain-specific isoform located in the endoplasmic reticulum of neurons. Previous results had demonstrated that CPT1C deficiency disrupted spine maturation in hippocampal neurons and impaired spatial learning, but the role of CPT1C in AMPAR physiology had remained mostly unknown. In the present study, we show that CPT1C binds GluA1 and GluA2 and that the three proteins have the same expression profile during neuronal maturation. Moreover, in hippocampal neurons of CPT1C KO mice, AMPAR-mediated miniature excitatory postsynaptic currents and synaptic levels of AMPAR subunits GluA1 and GluA2 are significantly reduced. We show that AMPAR expression is dependent on CPT1C levels because total protein levels of GluA1 and GluA2 are decreased in CPT1C KO neurons and are increased in CPT1C-overexpressing neurons, whereas other synaptic proteins remain unaltered. Notably, mRNA levels of AMPARs remained unchanged in those cultures, indicating that CPT1C is post-transcriptionally involved. We demonstrate that CPT1C is directly involved in the de novo synthesis of GluA1 and not in protein degradation. Moreover, in CPT1C KO cultured neurons, GluA1 synthesis after chemical long term depression was clearly diminished, and brain-derived neurotrophic factor treatment was unable to phosphorylate the mammalian target of rapamycin (mTOR) and stimulate GluA1 protein synthesis. These data newly identify CPT1C as a regulator of AMPAR translation efficiency and therefore also synaptic function in the hippocampus.
Collapse
Affiliation(s)
- Rut Fadó
- From the Basic Sciences Department, Facultat de Medicina i Ciències de la Salut, Universitat Internacional de Catalunya, Sant Cugat del Vallès 08195, Spain
| | - David Soto
- the Laboratori de Neurobiologia, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Feixa Llarga s/n 08907, L'Hospitalet de Llobregat 08907, Spain, the Department of Pathology and Experimental Therapeutics, Faculty of Medicine, Universitat de Barcelona, Feixa Llarga s/n 08907, L'Hospitalet de Llobregat 08907, Spain
| | - Alfredo J Miñano-Molina
- the Institut de Neurociències and Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Cerdanyola del Vallès 08193, Spain, the Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid 28031, Spain, and
| | - Macarena Pozo
- From the Basic Sciences Department, Facultat de Medicina i Ciències de la Salut, Universitat Internacional de Catalunya, Sant Cugat del Vallès 08195, Spain
| | - Patricia Carrasco
- From the Basic Sciences Department, Facultat de Medicina i Ciències de la Salut, Universitat Internacional de Catalunya, Sant Cugat del Vallès 08195, Spain, the Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), 15706 Santiago de Compostela, Spain
| | - Natalia Yefimenko
- the Laboratori de Neurobiologia, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Feixa Llarga s/n 08907, L'Hospitalet de Llobregat 08907, Spain, the Department of Pathology and Experimental Therapeutics, Faculty of Medicine, Universitat de Barcelona, Feixa Llarga s/n 08907, L'Hospitalet de Llobregat 08907, Spain
| | - José Rodríguez-Álvarez
- the Institut de Neurociències and Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Cerdanyola del Vallès 08193, Spain, the Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid 28031, Spain, and
| | - Núria Casals
- From the Basic Sciences Department, Facultat de Medicina i Ciències de la Salut, Universitat Internacional de Catalunya, Sant Cugat del Vallès 08195, Spain, the Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), 15706 Santiago de Compostela, Spain
| |
Collapse
|
109
|
Beheshti S, Shahrokhi S. Blocking the ghrelin receptor type 1a in the rat brain impairs memory encoding. Neuropeptides 2015; 52:97-102. [PMID: 26072187 DOI: 10.1016/j.npep.2015.05.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Revised: 05/18/2015] [Accepted: 05/21/2015] [Indexed: 12/17/2022]
Abstract
Studies have shown that intracerebral administration of ghrelin hormone affects learning and memory in different experimental models of learning. However, the effect of antagonism of ghrelin receptor type 1a (GHS-R1a) on different stages of learning has not been investigated. In this study the effect of intracerebroventricular (i.c.v) injection of a GHS-R1a selective antagonist (d-Lys-3-GHRP-6) was examined on acquisition and consolidation of learning in the passive avoidance task. In total, 72 male Wistar rats weighing 230-280g were randomly distributed into 9 groups of 8 each. Animals underwent stereotaxic surgery and cannulated in their right ventricle. One week after surgery, the rats received different doses of d-Lys-3-GHRP-6 (0.2, 2, 20 and 80nM/5μl; i.c.v) 10min before, or (2, 20 and 80nM/5μl; i.c.v) immediately after training. The control groups received solvent of the drug. Twenty four hours later in the test day, memory retrieval was assessed. Pre-training injection of d-Lys-3-GHRP-6 decreased step-through latency (STL) and increased number of step-throughs into the dark compartment (NST) in a dose-dependent manner, but failed to be statistically significant. It also increased time spent in the dark compartment (TDC), significantly and in a dose-dependent manner. Post-training injection of d-Lys-3-GHRP-6 decreased step-through latency and increased time spent in the dark compartment and number of step-throughs into the dark compartment, significantly and in a dose-dependent manner. The results indicate that antagonism of the GHS-R1a in the rat brain impairs memory encoding on both acquisition and consolidation stages. Further studies are required to elucidate the main brain regions affected by the antagonist.
Collapse
Affiliation(s)
- Siamak Beheshti
- Division of Animal Sciences, Department of Biology, Faculty of Sciences, University of Isfahan, Isfahan, Iran.
| | - Shahrzad Shahrokhi
- Division of Animal Sciences, Department of Biology, Faculty of Sciences, University of Isfahan, Isfahan, Iran
| |
Collapse
|
110
|
Murgatroyd CA, Peña CJ, Podda G, Nestler EJ, Nephew BC. Early life social stress induced changes in depression and anxiety associated neural pathways which are correlated with impaired maternal care. Neuropeptides 2015; 52:103-11. [PMID: 26049556 PMCID: PMC4537387 DOI: 10.1016/j.npep.2015.05.002] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Revised: 05/06/2015] [Accepted: 05/18/2015] [Indexed: 11/26/2022]
Abstract
Exposures to various types of early life stress can be robust predictors of the development of psychiatric disorders, including depression and anxiety. The objective of the current study was to investigate the roles of the translationally relevant targets of central vasopressin, oxytocin, ghrelin, orexin, glucocorticoid, and the brain-derived neurotrophic factor (BDNF) pathway in an early chronic social stress (ECSS) based rodent model of postpartum depression and anxiety. The present study reports novel changes in gene expression and extracellular signal related kinase (ERK) protein levels in the brains of ECSS exposed rat dams that display previously reported depressed maternal care and increased maternal anxiety. Decreases in oxytocin, orexin, and ERK proteins, increases in ghrelin receptor, glucocorticoid and mineralocorticoid receptor mRNA levels, and bidirectional changes in vasopressin underscore related work on the adverse long-term effects of early life stress on neural activity and plasticity, maternal behavior, responses to stress, and depression and anxiety-related behavior. The differences in gene and protein expression and robust correlations between expression and maternal care and anxiety support increased focus on these targets in animal and clinical studies of the adverse effects of early life stress, especially those focusing on depression and anxiety in mothers and the transgenerational effects of these disorders on offspring.
Collapse
Affiliation(s)
- Christopher A Murgatroyd
- Manchester Metropolitan University School of Healthcare Science, All Saints Building, Manchester M15 6BH, UK
| | - Catherine J Peña
- Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, New York, NY 10029, United States
| | - Giovanni Podda
- Manchester Metropolitan University School of Healthcare Science, All Saints Building, Manchester M15 6BH, UK
| | - Eric J Nestler
- Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, New York, NY 10029, United States
| | - Benjamin C Nephew
- Tufts University Cummings School of Veterinary Medicine, 200 Westboro Road, North Grafton, MA 01536, United States.
| |
Collapse
|
111
|
Cavalier M, Crouzin N, Ben Sedrine A, de Jesus Ferreira MC, Guiramand J, Cohen-Solal C, Fehrentz JA, Martinez J, Barbanel G, Vignes M. Involvement of PKA and ERK pathways in ghrelin-induced long-lasting potentiation of excitatory synaptic transmission in the CA1 area of rat hippocampus. Eur J Neurosci 2015; 42:2568-76. [DOI: 10.1111/ejn.13013] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 07/01/2015] [Accepted: 07/02/2015] [Indexed: 12/21/2022]
Affiliation(s)
- Mélanie Cavalier
- Institut des Biomolécules Max Mousseron; UMR 5247 CNRS-University of Montpellier; Place E Bataillon 34095 Montpellier Cedex 5 France
| | - Nadine Crouzin
- Institut des Biomolécules Max Mousseron; UMR 5247 CNRS-University of Montpellier; Place E Bataillon 34095 Montpellier Cedex 5 France
| | - Azza Ben Sedrine
- Institut des Biomolécules Max Mousseron; UMR 5247 CNRS-University of Montpellier; Place E Bataillon 34095 Montpellier Cedex 5 France
| | - Marie Celeste de Jesus Ferreira
- Institut des Biomolécules Max Mousseron; UMR 5247 CNRS-University of Montpellier; Place E Bataillon 34095 Montpellier Cedex 5 France
| | - Janique Guiramand
- Institut des Biomolécules Max Mousseron; UMR 5247 CNRS-University of Montpellier; Place E Bataillon 34095 Montpellier Cedex 5 France
| | - Catherine Cohen-Solal
- Institut des Biomolécules Max Mousseron; UMR 5247 CNRS-University of Montpellier; Place E Bataillon 34095 Montpellier Cedex 5 France
| | - Jean-Alain Fehrentz
- Institut des Biomolécules Max Mousseron; UMR 5247 CNRS-University of Montpellier; Place E Bataillon 34095 Montpellier Cedex 5 France
| | - Jean Martinez
- Institut des Biomolécules Max Mousseron; UMR 5247 CNRS-University of Montpellier; Place E Bataillon 34095 Montpellier Cedex 5 France
| | - Gérard Barbanel
- Institut des Biomolécules Max Mousseron; UMR 5247 CNRS-University of Montpellier; Place E Bataillon 34095 Montpellier Cedex 5 France
| | - Michel Vignes
- Institut des Biomolécules Max Mousseron; UMR 5247 CNRS-University of Montpellier; Place E Bataillon 34095 Montpellier Cedex 5 France
| |
Collapse
|
112
|
Amin SN, Gamal SM, Esmail RSEN, Aziz TMA, Rashed LA. Cognitive effects of acute restraint stress in male albino rats and the impact of pretreatment with quetiapine versus ghrelin. J Integr Neurosci 2015; 13:669-92. [PMID: 25391717 DOI: 10.1142/s0219635214500253] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Stress is any condition that seriously affects the balance of the organism physiologically and psychologically. Stress activates the hypothalamic-pituitary-adrenal (HPA) releasing glucocorticoid hormones that produce generalized effects on different body systems including the nervous system. This study aimed to investigate the effect of acute restraint stress (ARS) on cognitive performance by measuring spatial working memory in Y-maze, behavior (anxiety and exploratory behavior) in open field test, expression of synaptophysin and glial fibrillary acidic protein (GFAP) in the hippocampus by immunohistochemistry, dopaminergic receptors (D2) in the basal ganglia by gene expression and comparing the effect of ghrelin and quetiapine on the previous parameters. 36 adult male albino rats constituted the animal model of this work and have been divided into six groups: control group, control group exposed to ARS, quetiapine group, quetiapine group exposed to ARS, ghrelin group and ghrelin group exposed to ARS. We demonstrated more neuroprotective effect for quetiapine compared to ghrelin on stress response, anxiety behavior and working spatial memory impairment due to ARS.
Collapse
Affiliation(s)
- Shaimaa Nasr Amin
- Department of Medical Physiology, Kasr Al Ainy Faculty of Medicine, Cairo University, Kasr Al Ainy St. Cairo, Egypt 11562, Egypt
| | | | | | | | | |
Collapse
|
113
|
Ghrelin's Role in the Hypothalamic-Pituitary-Adrenal Axis Stress Response: Implications for Mood Disorders. Biol Psychiatry 2015; 78:19-27. [PMID: 25534754 DOI: 10.1016/j.biopsych.2014.10.021] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Revised: 09/26/2014] [Accepted: 10/15/2014] [Indexed: 12/24/2022]
Abstract
Ghrelin is a stomach hormone normally associated with feeding behavior and energy homeostasis. Recent studies highlight that ghrelin targets the brain to regulate a diverse number of functions, including learning, memory, motivation, stress responses, anxiety, and mood. In this review, we discuss recent animal and human studies showing that ghrelin regulates the hypothalamic-pituitary-adrenal axis and affects anxiety and mood disorders, such as depression and fear. We address the neural sites of action through which ghrelin regulates the hypothalamic-pituitary-adrenal axis and associated stress-induced behaviors, including the centrally projecting Edinger-Westphal nucleus, the hippocampus, amygdala, locus coeruleus, and the ventral tegmental area. Stressors modulate many behaviors associated with motivation, fear, anxiety, depression, and appetite; therefore, we assess the potential role for ghrelin as a stress feedback signal that regulates these associated behaviors. Finally, we briefly discuss important areas for future research that will help us move closer to potential ghrelin-based therapies to treat stress responses and related disorders.
Collapse
|
114
|
Wauson SER, Sarkodie K, Schuette LM, Currie PJ. Midbrain raphe 5-HT1A receptor activation alters the effects of ghrelin on appetite and performance in the elevated plus maze. J Psychopharmacol 2015; 29:836-44. [PMID: 25922422 DOI: 10.1177/0269881115581981] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Prior research suggests that midbrain serotonergic signaling and hypothalamic ghrelinergic signaling both play critical roles in appetitive and emotional behaviors. In the present study, we investigated the effects of median raphe nucleus (MRN) somatodentritic 5-HT1A receptor activation on the feeding-stimulant and anxiogenic action of paraventricular nucleus (PVN) ghrelin. In an initial experiment, adult male Sprague-Dawley rats were injected with either ghrelin (200-800 pmol) into the PVN or 8-OH-DPAT (2.5-10 nmol), a 5-HT1A receptor agonist, into the MRN. Performance on the elevated plus maze (EPM) was then assessed. In separate rats, MRN 8-OH-DPAT (2.5-5 nmol) was administered 5 min prior to PVN injection of ghrelin (400 pmol) followed by EPM testing. The orexigenic effects of MRN 8-OH-DPAT (0.1-1.6 nmol) paired with PVN ghrelin (50 pmol) were also examined. When administered alone into the PVN, ghrelin significantly decreased the number of entries and time spent in the open arms of the EPM. This anxiogenic effect was blocked if rats were allowed to eat immediately after ghrelin administration and then tested in the plus maze. MRN injections of 8-OH-DPAT were anxiolytic, and when rats were pretreated with 8-OH-DPAT prior to ghrelin, the anxiogenic action of the peptide was attenuated. In contrast, MRN administration of 8-OH-DPAT potentiated the eating-stimulant effect of PVN ghrelin. Overall, our findings demonstrate that ghrelinergic and serotonergic circuits interact in the neural control of eating and anxiety-like behaviors, with 5-HT1A receptor mechanisms potentiating the orexigenic action of ghrelin while inhibiting ghrelin-induced anxiogenesis as measured via the EPM.
Collapse
Affiliation(s)
| | - Kwaku Sarkodie
- Department of Psychology, Reed College, Portland, OR, USA
| | | | - Paul J Currie
- Department of Psychology, Reed College, Portland, OR, USA
| |
Collapse
|
115
|
Kunath N, van Groen T, Allison DB, Kumar A, Dozier-Sharpe M, Kadish I. Ghrelin agonist does not foster insulin resistance but improves cognition in an Alzheimer's disease mouse model. Sci Rep 2015; 5:11452. [PMID: 26090621 PMCID: PMC4473679 DOI: 10.1038/srep11452] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 05/15/2015] [Indexed: 12/24/2022] Open
Abstract
The orexigenic hormone ghrelin, a potential antagonist of the insulin system, ensures sufficient serum glucose in times of fasting. In the race for new therapeutics for diabetes, one focus of study has been antagonizing the ghrelin system in order to improve glucose tolerance. We provide evidence for a differential role of a ghrelin agonist on glucose homeostasis in an Alzheimer’s disease mouse model fed a high–glycemic index diet as a constant challenge for glucose homeostasis. The ghrelin agonist impaired glucose tolerance immediately after administration but not in the long term. At the same time, the ghrelin agonist improved spatial learning in the mice, raised their activity levels, and reduced their body weight and fat mass. Immunoassay results showed a beneficial impact of long-term treatment on insulin signaling pathways in hippocampal tissue. The present results suggest that ghrelin might improve cognition in Alzheimer’s disease via a central nervous system mechanism involving insulin signaling.
Collapse
Affiliation(s)
- Nicolas Kunath
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham AL, USA.,Department of Clinical Research, Max-Planck-Institute of Psychiatry, Munich, Germany
| | - Thomas van Groen
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham AL, USA
| | - David B Allison
- Office of Energetics; Nutrition Obesity Research Center; Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ashish Kumar
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham AL, USA
| | - Monique Dozier-Sharpe
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham AL, USA
| | - Inga Kadish
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham AL, USA
| |
Collapse
|
116
|
François M, Schaefer JM, Bole-Feysot C, Déchelotte P, Verhulst FC, Fetissov SO. Ghrelin-reactive immunoglobulins and anxiety, depression and stress-induced cortisol response in adolescents. The TRAILS study. Prog Neuropsychopharmacol Biol Psychiatry 2015; 59:1-7. [PMID: 25562566 DOI: 10.1016/j.pnpbp.2014.12.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Revised: 12/16/2014] [Accepted: 12/28/2014] [Indexed: 12/11/2022]
Abstract
BACKGROUND Ghrelin, a hunger hormone, has been implicated in the regulation of stress-response, anxiety and depression. Ghrelin-reactive immunoglobulins (Ig) were recently identified in healthy and obese humans showing abilities to increase ghrelin's stability and orexigenic effects. Here we studied if ghrelin-reactive Ig are associated with anxiety and depression and with the stress-induced cortisol response in a general population of adolescents. Furthermore, to test the possible infectious origin of ghrelin-reactive Ig, their levels were compared with serum IgG against common viruses. METHODS We measured ghrelin-reactive IgM, IgG and IgA in serum samples of 1199 adolescents from the Dutch TRAILS study and tested their associations with 1) anxiety and depression symptoms assessed with the Youth Self-Report, 2) stress-induced salivary cortisol levels and 3) IgG against human herpesvirus 1, 2, 4 and 6 and Influenza A and B viruses. RESULTS Ghrelin-reactive IgM and IgG correlated positively with levels of antibodies against Influenza A virus. Ghrelin-reactive IgM correlated negatively with antibodies against Influenza B virus. Ghrelin-reactive IgM correlated positively with anxiety scores in girls and ghrelin-reactive IgG correlated with stress-induced cortisol secretion, but these associations were weak and not significant after correction for multiple testing. CONCLUSION These data indicate that production of ghrelin-reactive autoantibodies could be influenced by viral infections. Serum levels of ghrelin-reactive autoantibodies probably do not play a role in regulating anxiety, depression and the stress-response in adolescents from the general population.
Collapse
Affiliation(s)
- Marie François
- Nutrition, Gut and Brain Laboratory, Inserm UMR1073, Institute for Research and Innovation in Biomedicine (IRIB), Rouen University, Normandy University, 76183, France
| | - Johanna M Schaefer
- Department of Child and Adolescent Psychiatry & Psychology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Christine Bole-Feysot
- Nutrition, Gut and Brain Laboratory, Inserm UMR1073, Institute for Research and Innovation in Biomedicine (IRIB), Rouen University, Normandy University, 76183, France
| | - Pierre Déchelotte
- Nutrition, Gut and Brain Laboratory, Inserm UMR1073, Institute for Research and Innovation in Biomedicine (IRIB), Rouen University, Normandy University, 76183, France
| | - Frank C Verhulst
- Department of Child and Adolescent Psychiatry & Psychology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Sergueï O Fetissov
- Nutrition, Gut and Brain Laboratory, Inserm UMR1073, Institute for Research and Innovation in Biomedicine (IRIB), Rouen University, Normandy University, 76183, France.
| |
Collapse
|
117
|
Folch J, Patraca I, Martínez N, Pedrós I, Petrov D, Ettcheto M, Abad S, Marin M, Beas-Zarate C, Camins A. The role of leptin in the sporadic form of Alzheimer's disease. Interactions with the adipokines amylin, ghrelin and the pituitary hormone prolactin. Life Sci 2015; 140:19-28. [PMID: 25998028 DOI: 10.1016/j.lfs.2015.05.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Revised: 05/05/2015] [Accepted: 05/11/2015] [Indexed: 12/11/2022]
Abstract
Leptin (Lep) is emerging as a pivotal molecule involved in both the early events and the terminal phases of Alzheimer's disease (AD). In the canonical pathway, Lep acts as an anorexigenic factor via its effects on hypothalamic nucleus. However, additional functions of Lep in the hippocampus and cortex have been unravelled in recent years. Early events in the sporadic form of AD likely involve cellular level alterations which can have an effect on food intake and metabolism. Thus, AD can be conceivably interpreted as a multiorgan pathology that not only results in a dramatic neuronal loss in brain areas such as the hippocampus and the cortex (ultimately leading to a significant cognitive impairment) but as a disease which also affects body-weight homeostasis. According to this view, body-weight control disruptions are to be expected in both the early- and late-stage AD, concomitant with changes in serum Lep content, alterations in Lep transport across the blood-brain barrier (BBB) and Lep receptor-related signalling abnormalities. Lep is a member of the adipokine family of molecules, while the Lep receptor belongs to the class I cytokine receptors. Since cellular response to adipokine signalling can be either potentiated or diminished as a result of specific ligand-receptor interactions, Lep interactions with other members of the adipokine family including amylin, ghrelin and hormones such as prolactin require further investigation. In this review, we provide a general perspective on the functions of Lep in the brain, with a particular focus on the sporadic AD.
Collapse
Affiliation(s)
- Jaume Folch
- Unitats de Bioquímica i Farmacologia, Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, C./ St. Llorenç 21, 43201 Reus, Tarragona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos Tercero, Madrid, Spain
| | - Iván Patraca
- Unitats de Bioquímica i Farmacologia, Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, C./ St. Llorenç 21, 43201 Reus, Tarragona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos Tercero, Madrid, Spain
| | - Nohora Martínez
- Unitats de Bioquímica i Farmacologia, Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, C./ St. Llorenç 21, 43201 Reus, Tarragona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos Tercero, Madrid, Spain
| | - Ignacio Pedrós
- Unitats de Bioquímica i Farmacologia, Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, C./ St. Llorenç 21, 43201 Reus, Tarragona, Spain; Unitat de Farmacologia i Farmacognòsia Facultat de Farmàcia, Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Dmitry Petrov
- Unitat de Farmacologia i Farmacognòsia Facultat de Farmàcia, Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos Tercero, Madrid, Spain
| | - Miren Ettcheto
- Unitat de Farmacologia i Farmacognòsia Facultat de Farmàcia, Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos Tercero, Madrid, Spain
| | - Sonia Abad
- Unitat de Farmacologia i Farmacognòsia Facultat de Farmàcia, Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos Tercero, Madrid, Spain
| | - Miguel Marin
- Centro de Biotecnología, Universidad Nacional de Loja, Av. Pío Jaramillo Alvarado y Reinaldo Espinosa, La Argelia, Loja, Ecuador
| | - Carlos Beas-Zarate
- Departamento de Biología Celular y Molecular, C.U.C.B.A., Universidad de Guadalajara and División de Neurociencias, Centro de Investigación Biomédica de Occidente (CIBO), Mexico; Instituto Mexicano del Seguro Social (IMSS), Sierra Mojada 800, Col. Independencia, Guadalajara, Jalisco 44340, Mexico
| | - Antoni Camins
- Unitat de Farmacologia i Farmacognòsia Facultat de Farmàcia, Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos Tercero, Madrid, Spain; Centro de Biotecnología, Universidad Nacional de Loja, Av. Pío Jaramillo Alvarado y Reinaldo Espinosa, La Argelia, Loja, Ecuador.
| |
Collapse
|
118
|
Ghrelin increases memory consolidation through hippocampal mechanisms dependent on glutamate release and NR2B-subunits of the NMDA receptor. Psychopharmacology (Berl) 2015; 232:1843-57. [PMID: 25466701 DOI: 10.1007/s00213-014-3817-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 11/10/2014] [Indexed: 12/29/2022]
Abstract
RATIONALE Ghrelin (Ghr) is a peptide that participates in the modulation of several biological processes. Ghr administration into the hippocampus improves learning and memory in different memory tests. However, the possible mechanisms underlying this effect on memory have not yet been clarified. OBJECTIVE The purpose of the present work is to add new insights about the mechanisms by which Ghr modulates long-term memory consolidation in the hippocampus. We examined Ghr effects upon processes related to increased synaptic efficacy as presynaptic glutamate release and changes in the expression of the NR2B-subunits containing n-methyl-d-aspartate receptors (NMDAR), which are critical for LTP induction. We also attempted to determine the temporal window in which Ghr administration induces memory facilitation and if the described effects depend on GHS-R1a stimulation. RESULTS The present research demonstrated that Ghr increased glutamate release from hippocampal synaptosomes; intra-hippocampal Ghr administration increased NR2B-subunits expression in CA1 and DG subareas and also reversed the deleterious effects of the NR2B-subunit-specific antagonist, Ro 25-6981, upon memory consolidation and LTP generation in the hippocampus. These effects are likely to be the consequence of GHS-R1a activation. CONCLUSION According to the results above mentioned and previous findings, we can hypothesize some of the mechanisms by which Ghr modulates memory consolidation. At presynaptic level, Ghr stimulates glutamate release, probably by enhancing [Ca(2+)]i. At postsynaptic level, the glutamate released activates NMDAR while Ghr also mediates effects directly activating its specific receptors and increases NR2B-subunit expression.
Collapse
|
119
|
Müller TD, Nogueiras R, Andermann ML, Andrews ZB, Anker SD, Argente J, Batterham RL, Benoit SC, Bowers CY, Broglio F, Casanueva FF, D'Alessio D, Depoortere I, Geliebter A, Ghigo E, Cole PA, Cowley M, Cummings DE, Dagher A, Diano S, Dickson SL, Diéguez C, Granata R, Grill HJ, Grove K, Habegger KM, Heppner K, Heiman ML, Holsen L, Holst B, Inui A, Jansson JO, Kirchner H, Korbonits M, Laferrère B, LeRoux CW, Lopez M, Morin S, Nakazato M, Nass R, Perez-Tilve D, Pfluger PT, Schwartz TW, Seeley RJ, Sleeman M, Sun Y, Sussel L, Tong J, Thorner MO, van der Lely AJ, van der Ploeg LHT, Zigman JM, Kojima M, Kangawa K, Smith RG, Horvath T, Tschöp MH. Ghrelin. Mol Metab 2015; 4:437-60. [PMID: 26042199 PMCID: PMC4443295 DOI: 10.1016/j.molmet.2015.03.005] [Citation(s) in RCA: 760] [Impact Index Per Article: 76.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Revised: 03/11/2015] [Accepted: 03/11/2015] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The gastrointestinal peptide hormone ghrelin was discovered in 1999 as the endogenous ligand of the growth hormone secretagogue receptor. Increasing evidence supports more complicated and nuanced roles for the hormone, which go beyond the regulation of systemic energy metabolism. SCOPE OF REVIEW In this review, we discuss the diverse biological functions of ghrelin, the regulation of its secretion, and address questions that still remain 15 years after its discovery. MAJOR CONCLUSIONS In recent years, ghrelin has been found to have a plethora of central and peripheral actions in distinct areas including learning and memory, gut motility and gastric acid secretion, sleep/wake rhythm, reward seeking behavior, taste sensation and glucose metabolism.
Collapse
Affiliation(s)
- T D Müller
- Institute for Diabetes and Obesity, Helmholtz Zentrum München, München, Germany
| | - R Nogueiras
- Department of Physiology, Centro de Investigación en Medicina Molecular y Enfermedades Crónicas, University of Santiago de Compostela (CIMUS)-Instituto de Investigación Sanitaria (IDIS)-CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, Spain
| | - M L Andermann
- Division of Endocrinology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Z B Andrews
- Department of Physiology, Faculty of Medicine, Monash University, Melbourne, Victoria, Australia
| | - S D Anker
- Applied Cachexia Research, Department of Cardiology, Charité Universitätsmedizin Berlin, Germany
| | - J Argente
- Department of Pediatrics and Pediatric Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain ; Department of Pediatrics, Universidad Autónoma de Madrid and CIBER Fisiopatología de la obesidad y nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - R L Batterham
- Centre for Obesity Research, University College London, London, United Kingdom
| | - S C Benoit
- Metabolic Disease Institute, Division of Endocrinology, Department of Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - C Y Bowers
- Tulane University Health Sciences Center, Endocrinology and Metabolism Section, Peptide Research Section, New Orleans, LA, USA
| | - F Broglio
- Division of Endocrinology, Diabetes and Metabolism, Dept. of Medical Sciences, University of Torino, Torino, Italy
| | - F F Casanueva
- Department of Medicine, Santiago de Compostela University, Complejo Hospitalario Universitario de Santiago (CHUS), CIBER de Fisiopatologia Obesidad y Nutricion (CB06/03), Instituto Salud Carlos III, Santiago de Compostela, Spain
| | - D D'Alessio
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA
| | - I Depoortere
- Translational Research Center for Gastrointestinal Disorders, University of Leuven, Leuven, Belgium
| | - A Geliebter
- New York Obesity Nutrition Research Center, Department of Medicine, St Luke's-Roosevelt Hospital Center, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - E Ghigo
- Department of Pharmacology & Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - P A Cole
- Monash Obesity & Diabetes Institute, Monash University, Clayton, Victoria, Australia
| | - M Cowley
- Department of Physiology, Faculty of Medicine, Monash University, Melbourne, Victoria, Australia ; Monash Obesity & Diabetes Institute, Monash University, Clayton, Victoria, Australia
| | - D E Cummings
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - A Dagher
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - S Diano
- Dept of Neurobiology, Yale University School of Medicine, New Haven, CT, USA
| | - S L Dickson
- Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - C Diéguez
- Department of Physiology, School of Medicine, Instituto de Investigacion Sanitaria (IDIS), University of Santiago de Compostela, Spain
| | - R Granata
- Division of Endocrinology, Diabetes and Metabolism, Dept. of Medical Sciences, University of Torino, Torino, Italy
| | - H J Grill
- Department of Psychology, Institute of Diabetes, Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, USA
| | - K Grove
- Department of Diabetes, Obesity and Metabolism, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA
| | - K M Habegger
- Comprehensive Diabetes Center, University of Alabama School of Medicine, Birmingham, AL, USA
| | - K Heppner
- Division of Diabetes, Obesity, and Metabolism, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - M L Heiman
- NuMe Health, 1441 Canal Street, New Orleans, LA 70112, USA
| | - L Holsen
- Departments of Psychiatry and Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - B Holst
- Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen N, Denmark
| | - A Inui
- Department of Psychosomatic Internal Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - J O Jansson
- Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - H Kirchner
- Medizinische Klinik I, Universitätsklinikum Schleswig-Holstein Campus Lübeck, Lübeck, Germany
| | - M Korbonits
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London, Queen Mary University of London, London, UK
| | - B Laferrère
- New York Obesity Research Center, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - C W LeRoux
- Diabetes Complications Research Centre, Conway Institute, University College Dublin, Ireland
| | - M Lopez
- Department of Physiology, Centro de Investigación en Medicina Molecular y Enfermedades Crónicas, University of Santiago de Compostela (CIMUS)-Instituto de Investigación Sanitaria (IDIS)-CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, Spain
| | - S Morin
- Institute for Diabetes and Obesity, Helmholtz Zentrum München, München, Germany
| | - M Nakazato
- Division of Neurology, Respirology, Endocrinology and Metabolism, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Kiyotake, Miyazaki, Japan
| | - R Nass
- Division of Endocrinology and Metabolism, University of Virginia, Charlottesville, VA, USA
| | - D Perez-Tilve
- Department of Internal Medicine, Department of Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - P T Pfluger
- Institute for Diabetes and Obesity, Helmholtz Zentrum München, München, Germany
| | - T W Schwartz
- Department of Neuroscience and Pharmacology, Laboratory for Molecular Pharmacology, The Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | - R J Seeley
- Department of Surgery, University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - M Sleeman
- Department of Physiology, Faculty of Medicine, Monash University, Melbourne, Victoria, Australia
| | - Y Sun
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - L Sussel
- Department of Genetics and Development, Columbia University, New York, NY, USA
| | - J Tong
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA
| | - M O Thorner
- Division of Endocrinology and Metabolism, University of Virginia, Charlottesville, VA, USA
| | - A J van der Lely
- Department of Medicine, Erasmus University MC, Rotterdam, The Netherlands
| | | | - J M Zigman
- Departments of Internal Medicine and Psychiatry, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - M Kojima
- Molecular Genetics, Institute of Life Science, Kurume University, Kurume, Japan
| | - K Kangawa
- National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
| | - R G Smith
- The Scripps Research Institute, Florida Department of Metabolism & Aging, Jupiter, FL, USA
| | - T Horvath
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Section of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - M H Tschöp
- Institute for Diabetes and Obesity, Helmholtz Zentrum München, München, Germany ; Division of Metabolic Diseases, Department of Medicine, Technical University Munich, Munich, Germany
| |
Collapse
|
120
|
Munn RGK, Tyree SM, McNaughton N, Bilkey DK. The frequency of hippocampal theta rhythm is modulated on a circadian period and is entrained by food availability. Front Behav Neurosci 2015; 9:61. [PMID: 25814943 PMCID: PMC4356069 DOI: 10.3389/fnbeh.2015.00061] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 02/23/2015] [Indexed: 11/13/2022] Open
Abstract
The hippocampal formation plays a critical role in the generation of episodic memory. While the encoding of the spatial and contextual components of memory have been extensively studied, how the hippocampus encodes temporal information, especially at long time intervals, is less well understood. The activity of place cells in hippocampus has previously been shown to be modulated at a circadian time-scale, entrained by a behavioral stimulus, but not entrained by light. The experimental procedures used in the previous study of this phenomenon, however, necessarily conflated two alternative entraining stimuli, the exposure to the recording environment and the availability of food, making it impossible to distinguish between these possibilities. Here we demonstrate that the frequency of theta-band hippocampal EEG varies with a circadian period in freely moving animals and that this periodicity mirrors changes in the firing rate of hippocampal neurons. Theta activity serves, therefore, as a proxy of circadian-modulated hippocampal neuronal activity. We then demonstrate that the frequency of hippocampal theta driven by stimulation of the reticular formation also varies with a circadian period. Because this effect can be observed without having to feed the animal to encourage movement we were able to identify what stimulus entrains the circadian oscillation. We show that with reticular-activated recordings started at various times of the day the frequency of theta varies quasi-sinusoidally with a 25 h period and phase-aligned when referenced to the animal’s regular feeding time, but not the recording start time. Furthermore, we show that theta frequency consistently varied with a circadian period when the data obtained from repeated recordings started at various times of the day were referenced to the start of food availability in the recording chamber. This pattern did not occur when data were referenced to the start of the recording session or to the actual time of day when this was not also related to feeding time. This double dissociation demonstrates that hippocampal theta is modulated with a circadian timescale, and that this modulation is strongly entrained by food. One interpretation of this finding is that the hippocampus is responsive to a food entrainable oscillator (FEO) that might modulate foraging behavior over circadian periods.
Collapse
Affiliation(s)
- Robert G K Munn
- Department of Psychology, University of Otago Dunedin, New Zealand ; Department of Neurobiology, Stanford University Stanford, CA, USA
| | - Susan M Tyree
- Department of Psychology, University of Otago Dunedin, New Zealand
| | - Neil McNaughton
- Department of Psychology, University of Otago Dunedin, New Zealand
| | - David K Bilkey
- Department of Psychology, University of Otago Dunedin, New Zealand
| |
Collapse
|
121
|
Abstract
Ongoing mouse studies of a proposed therapy for cocaine abuse based on viral gene transfer of butyrylcholinesterase (BChE) mutated for accelerated cocaine hydrolysis have yielded surprising effects on aggression. Further investigation has linked these effects to a reduction in circulating ghrelin, driven by BChE at levels ∼ 100-fold above normal. Tests with human BChE showed ready ghrelin hydrolysis at physiologic concentrations, and multiple low-mass molecular dynamics simulations revealed that ghrelin's first five residues fit sterically and electrostatically into BChE's active site. Consistent with in vitro results, male BALB/c mice with high plasma BChE after gene transfer exhibited sharply reduced plasma ghrelin. Unexpectedly, such animals fought less, both spontaneously and in a resident/intruder provocation model. One mutant BChE was found to be deficient in ghrelin hydrolysis. BALB/c mice transduced with this variant retained normal plasma ghrelin levels and did not differ from untreated controls in the aggression model. In contrast, C57BL/6 mice with BChE gene deletion exhibited increased ghrelin and fought more readily than wild-type animals. Collectively, these findings indicate that BChE-catalyzed ghrelin hydrolysis influences mouse aggression and social stress, with potential implications for humans.
Collapse
|
122
|
Wittekind DA, Kluge M. Ghrelin in psychiatric disorders - A review. Psychoneuroendocrinology 2015; 52:176-94. [PMID: 25459900 DOI: 10.1016/j.psyneuen.2014.11.013] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Revised: 11/13/2014] [Accepted: 11/13/2014] [Indexed: 12/21/2022]
Abstract
Ghrelin is a 28-amino-acid peptide hormone, first described in 1999 and broadly expressed in the organism. As the only known orexigenic hormone secreted in the periphery, it increases hunger and appetite, promoting food intake. Ghrelin has also been shown to be involved in various physiological processes being regulated in the central nervous system such as sleep, mood, memory and reward. Accordingly, it has been implicated in a series of psychiatric disorders, making it subject of increasing investigation, with knowledge rapidly accumulating. This review aims at providing a concise yet comprehensive overview of the role of ghrelin in psychiatric disorders. Ghrelin was consistently shown to exert neuroprotective and memory-enhancing effects and alleviated psychopathology in animal models of dementia. Few human studies show a disruption of the ghrelin system in dementia. It was also shown to play a crucial role in the pathophysiology of addictive disorders, promoting drug reward, enhancing drug seeking behavior and increasing craving in both animals and humans. Ghrelin's exact role in depression and anxiety is still being debated, as it was shown to both promote and alleviate depressive and anxiety-behavior in animal studies, with an overweight of evidence suggesting antidepressant effects. Not surprisingly, the ghrelin system is also implicated in eating disorders, however its exact role remains to be elucidated. Its widespread involvement has made the ghrelin system a promising target for future therapies, with encouraging findings in recent literature.
Collapse
Affiliation(s)
| | - Michael Kluge
- Department of Psychiatry and Psychotherapy, University of Leipzig, Leipzig, Germany
| |
Collapse
|
123
|
Potential ghrelin-mediated benefits and risks of hydrogen water. Med Hypotheses 2015; 84:350-5. [PMID: 25649854 DOI: 10.1016/j.mehy.2015.01.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Accepted: 01/15/2015] [Indexed: 12/12/2022]
Abstract
Molecular hydrogen (H2) can scavenge hydroxyl radical and diminish the toxicity of peroxynitrite; hence, it has interesting potential for antioxidant protection. Recently, a number of studies have explored the utility of inhaled hydrogen gas, or of hydrogen-saturated water, administered parenterally or orally, in rodent models of pathology and in clinical trials, oftentimes with very positive outcomes. The efficacy of orally ingested hydrogen-rich water (HW) has been particularly surprising, given that only transient and rather small increments in plasma hydrogen can be achieved by this method. A recent study in mice has discovered that orally administered HW provokes increased gastric production of the orexic hormone ghrelin, and that this ghrelin mediates the favorable impact of HW on a mouse model of Parkinson's disease. The possibility that most of the benefits observed with HW in experimental studies are mediated by ghrelin merits consideration. Ghrelin is well known to function as an appetite stimulant and secretagogue for growth hormone, but it influences physiological function throughout the body via interaction with the widely express GHS-R1a receptor. Rodent and, to a more limited extent, clinical studies establish that ghrelin has versatile neuroprotective and cognitive enhancing activity, favorably impacts vascular health, exerts anti-inflammatory activity useful in autoimmune disorders, and is markedly hepatoprotective. The stimulatory impact of ghrelin on GH-IGF-I activity, while potentially beneficial in sarcopenia or cachectic disorders, does raise concerns regarding the long-term impact of ghrelin up-regulation on cancer risk. The impact of ingesting HW water on ghrelin production in humans needs to be evaluated; if HW does up-regulate ghrelin in humans, it may have versatile potential for prevention and control of a number of health disorders.
Collapse
|
124
|
Yamawaki H, Futagami S, Shimpuku M, Shindo T, Maruki Y, Nagoya H, Kodaka Y, Sato H, Gudis K, Kawagoe T, Sakamoto C. Leu72Met408 Polymorphism of the Ghrelin Gene Is Associated With Early Phase of Gastric Emptying in the Patients With Functional Dyspepsia in Japan. J Neurogastroenterol Motil 2015; 21:93-102. [PMID: 25540946 PMCID: PMC4288091 DOI: 10.5056/jnm14086] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2014] [Revised: 09/11/2014] [Accepted: 09/13/2014] [Indexed: 12/11/2022] Open
Abstract
Background/Aims There are no available data about the relationship between ghrelin gene genotypes and early phase of gastric emptying in functional dyspepsia (FD) as defined by Rome III classification. Methods We enrolled 74 patients presenting with typical symptoms of FD and 64 healthy volunteers. Gastric motility was evaluated using the 13C-acetate breath test. We used Rome III criteria to evaluate upper abdominal symptoms and self-rating questionnaires for depression (SRQ-D) scores to determine status of depression. The Arg51Gln (346G>A), preproghrelin (3056T>C), Leu72Met (408C>A), Gln90Leu (3412T>A) and G-protein β3 (825C>T) polymorphisms were analyzed in the DNA from blood samples of enrolled subjects. Genotyping was performed by polymerase chain reaction. Results There was a significant relationship between the Gln90Leu3412 genotype and SRQ-D score in FD patients (P = 0.009). Area under the curve at 15 minutes (AUC15) value was significantly associated with the Leu72Met408 genotype (P = 0.015) but not with entire gastric emptying. Conclusions The Leu72Met (408C>A) single nucleotide polymorphism was significantly associated with early phase of gastric emptying in FD patients. Further studies will be necessary to clarify the association between ghrelin gene single nucleotide polymorphisms and early phase of gastric emptying in FD patients.
Collapse
Affiliation(s)
- Hiroshi Yamawaki
- Division of Gastroenterology, Department of Internal Medicine, Nippon Medical School, Sendagi, Tokyo, Japan
| | - Seiji Futagami
- Division of Gastroenterology, Department of Internal Medicine, Nippon Medical School, Sendagi, Tokyo, Japan
| | - Mayumi Shimpuku
- Division of Gastroenterology, Department of Internal Medicine, Nippon Medical School, Sendagi, Tokyo, Japan
| | - Tomotaka Shindo
- Division of Gastroenterology, Department of Internal Medicine, Nippon Medical School, Sendagi, Tokyo, Japan
| | - Yuuta Maruki
- Division of Gastroenterology, Department of Internal Medicine, Nippon Medical School, Sendagi, Tokyo, Japan
| | - Hiroyuki Nagoya
- Division of Gastroenterology, Department of Internal Medicine, Nippon Medical School, Sendagi, Tokyo, Japan
| | - Yasuhiro Kodaka
- Division of Gastroenterology, Department of Internal Medicine, Nippon Medical School, Sendagi, Tokyo, Japan
| | - Hitomi Sato
- Division of Gastroenterology, Department of Internal Medicine, Nippon Medical School, Sendagi, Tokyo, Japan
| | - Katya Gudis
- Division of Gastroenterology, Department of Internal Medicine, Nippon Medical School, Sendagi, Tokyo, Japan
| | - Tetsuro Kawagoe
- Division of Gastroenterology, Department of Internal Medicine, Nippon Medical School, Sendagi, Tokyo, Japan
| | - Choitsu Sakamoto
- Division of Gastroenterology, Department of Internal Medicine, Nippon Medical School, Sendagi, Tokyo, Japan
| |
Collapse
|
125
|
Ghrelin effects expression of several genes associated with depression-like behavior. Prog Neuropsychopharmacol Biol Psychiatry 2015; 56:227-34. [PMID: 25286107 DOI: 10.1016/j.pnpbp.2014.09.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Revised: 09/23/2014] [Accepted: 09/23/2014] [Indexed: 02/03/2023]
Abstract
Ghrelin (Ghr) is an orexigenic peptide that is being investigated for its potential role in development of anxiety-like behavior and modulation of depressive-like symptoms induced by bilateral olfactory bulbectomy (OB) in rodents. Olfactory bulbectomy is an animal model useful to study of depression and Ghr could be an alternative therapeutic tool in depression therapy. We studied the effects of intracerebroventricular (i.c.v.) Ghr administration on the expression of hypothalamic genes related to depression and mood (delta opioid receptor (DOR), mu opioid receptor (MOR) and kappa opioid receptor (KOR), lutropin-choriogonadotropic hormone receptor (LHCGR), serotonin transporter (SERT), interleukin 1 beta (IL-1b), vasopressin (AVP) and corticotrophin releasing hormone (CRH)) in OB animals, as well as changes in plasma levels of AVP, CRH and adenocorticotropic hormone (ACTH). We found that acute Ghr 0.3 nmol/μl administration increases gene expression of DOR, SERT and LHCGR in OB mice and decreased expression of IL-1b, suggesting that these genes could be involved in the antidepressant-like effects of Ghr. In addition, OB animals exhibit high AVP gene expression and elevated plasma concentrations of AVP and ACTH and acute Ghr 0.3 nmol/μl administration reduces AVP gene expression and the concentration of these hormones, suggesting that peptide-effects on depressive-like behavior could be mediated at least in part via AVP. In conclusion, this study provides new evidence about genes, receptors and hormones involved in the antidepressant mechanism/s induced by Ghr in OB animals.
Collapse
|
126
|
Wellman MK, Patterson ZR, MacKay H, Darling JE, Mani BK, Zigman JM, Hougland JL, Abizaid A. Novel Regulator of Acylated Ghrelin, CF801, Reduces Weight Gain, Rebound Feeding after a Fast, and Adiposity in Mice. Front Endocrinol (Lausanne) 2015; 6:144. [PMID: 26441834 PMCID: PMC4585333 DOI: 10.3389/fendo.2015.00144] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 09/01/2015] [Indexed: 01/04/2023] Open
Abstract
Ghrelin is a 28 amino acid hormonal peptide that is intimately related to the regulation of food intake and body weight. Once secreted, ghrelin binds to the growth hormone secretagogue receptor-1a, the only known receptor for ghrelin and is capable of activating a number of signaling cascades, ultimately resulting in an increase in food intake and adiposity. Because ghrelin has been linked to overeating and the development of obesity, a number of pharmacological interventions have been generated in order to interfere with either the activation of ghrelin or interrupting ghrelin signaling as a means to reducing appetite and decrease weight gain. Here, we present a novel peptide, CF801, capable of reducing circulating acylated ghrelin levels and subsequent body weight gain and adiposity. To this end, we show that IP administration of CF801 is sufficient to reduce circulating plasma acylated ghrelin levels. Acutely, intraperitoneal injections of CF801 resulted in decreased rebound feeding after an overnight fast. When delivered chronically, they decreased weight gain and adiposity without affecting caloric intake. CF801, however, did cause a change in diet preference, decreasing preference for a high-fat diet and increasing preference for regular chow diet. Given the complexity of ghrelin receptor function, we propose that CF801, along with other compounds that regulate ghrelin secretion, may prove to be a beneficial tool in the study of the ghrelin system, and potential targets for ghrelin-based obesity treatments without altering the function of ghrelin receptors.
Collapse
Affiliation(s)
| | | | - Harry MacKay
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada
| | | | - Bharath K. Mani
- Department of Internal Medicine, Division of Hypothalamic Research, The University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Internal Medicine, Division of Endocrinology and Metabolism, The University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jeffrey M. Zigman
- Department of Internal Medicine, Division of Hypothalamic Research, The University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Internal Medicine, Division of Endocrinology and Metabolism, The University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | | - Alfonso Abizaid
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada
- *Correspondence: Alfonso Abizaid, Department of Neuroscience, Carleton University, 1125 Colonel By Drive, 329 Life Science Research Building, Ottawa, ON K1S 5B6, Canada,
| |
Collapse
|
127
|
Miller AL, Jong H, Lumeng JC. Obesity-associated biomarkers and executive function in children. Pediatr Res 2015; 77:143-7. [PMID: 25310758 PMCID: PMC4416088 DOI: 10.1038/pr.2014.158] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Accepted: 08/13/2014] [Indexed: 12/12/2022]
Abstract
There is a growing focus on links between obesity and cognitive decline in adulthood, including Alzheimer's disease. It is also increasingly recognized that obesity in youth is associated with poorer cognitive function, specifically executive functioning skills such as inhibitory control and working memory, which are critical for academic achievement. Emerging literature provides evidence for possible biological mechanisms driven by obesity; obesity-associated biomarkers such as adipokines, obesity-associated inflammatory cytokines, and obesity-associated gut hormones have been associated with learning, memory, and general cognitive function. To date, examination of obesity-associated biology with brain function has primarily occurred in animal models. The few studies examining such biologically mediated pathways in adult humans have corroborated the animal data, but this body of work has gone relatively unrecognized by the pediatric literature. Despite the fact that differences in these biomarkers have been found in association with obesity in children, the possibility that obesity-related biology could affect brain development in children has not been actively considered. We review obesity-associated biomarkers that have shown associations with neurocognitive skills, specifically executive functioning skills, which have far-reaching implications for child development. Understanding such gut-brain associations early in the lifespan may yield unique intervention implications.
Collapse
Affiliation(s)
- Alison L. Miller
- Center for Human Growth and Development, University of Michigan, Ann Arbor, Michigan
- Department of Health Behavior and Health Education, University of Michigan School of Public Health, Ann Arbor, Michigan
| | - Hannah Jong
- Center for Human Growth and Development, University of Michigan, Ann Arbor, Michigan
| | - Julie C. Lumeng
- Center for Human Growth and Development, University of Michigan, Ann Arbor, Michigan
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, Michigan
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, Michigan
| |
Collapse
|
128
|
Gastón MS, Schiöth HB, De Barioglio SR, Salvatierra NA. Gabaergic control of anxiety-like behavior, but not food intake, induced by ghrelin in the intermediate medial mesopallium of the neonatal chick. Horm Behav 2015; 67:66-72. [PMID: 25499794 DOI: 10.1016/j.yhbeh.2014.11.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Revised: 11/21/2014] [Accepted: 11/24/2014] [Indexed: 12/20/2022]
Abstract
Ghrelin (Grh) is an endogenous ligand of the growth hormone secretagogue receptor. In neonatal chicks, central Ghr induces anxiogenic-like behavior but strongly inhibits food intake. The intermediate medial mesopallium (IMM) of the chick forebrain has been identified to be a site of the memory formation, and the modulation of the GABAA receptors that are present here modifies the expression of behavior. Thus, the GABAergic system may constitute a central pathway for Ghr action in regulating the processes of food intake and stress-related behaviors. Therefore, we investigated if the effect of systemic administration of bicuculline (GABAA receptor antagonist) and diazepam (benzodiazepine receptor agonist) on the anxiety in an Open Field test and inhibition in food intake induced by Grh (30pmol) when injected into IMM, were mediated by GABAergic transmission. In Open Field test, bicuculline was able to block the anxiogenic-like behavior induced by Ghr, whereas diazepam did not produce it. However, the co-administration of bicuculline or diazepam plus Ghr did not show any change in food intake at 30, 60 and 120min after injection compared to Ghr alone. Our results indicate for the first time that Ghr, injected into the forebrain IMM area, induces an anxiogenic-like behavior, which was blocked by bicuculline but not diazepam, thus suggesting that Ghr plays an important role in the response pattern to acute stressor, involving the possible participation of the GABAergic system. Nevertheless, as neither drug affected the hypophagia induced by intra-IMM Ghr, this suggests that it may be mediated by different mechanisms.
Collapse
Affiliation(s)
- M S Gastón
- Departamento de Química, Facultad de Ciencias Exactas Físicas y Naturales, Universidad Nacional de Córdoba, Instituto de Investigaciones Biológicas y Tecnológicas (IIByT-CONICET), Av. Vélez Sarsfield 1611, 5016 Córdoba, Argentina
| | - H B Schiöth
- Section of Functional Pharmacology, Department of Neuroscience, Uppsala University, Institutionen för Neurovetenskap BMC, Box 593, 751 24 Uppsala, Sweden
| | - S R De Barioglio
- Departamento de Farmacología, Facultad de Ciencias Químicas, Instituto de Farmacología Experimental Córdoba (IFEC-CONICET), Haya de la Torre y Medina Allende, Universidad Nacional de Córdoba, Ciudad Universitaria, 5016 Córdoba, Argentina
| | - N A Salvatierra
- Departamento de Química, Facultad de Ciencias Exactas Físicas y Naturales, Universidad Nacional de Córdoba, Instituto de Investigaciones Biológicas y Tecnológicas (IIByT-CONICET), Av. Vélez Sarsfield 1611, 5016 Córdoba, Argentina.
| |
Collapse
|
129
|
Souza AC, Souza A, Medeiros LF, De Oliveira C, Scarabelot VL, Da Silva RS, Bogo MR, Capiotti KM, Kist LW, Bonan CD, Caumo W, Torres IL. Maternal caffeine exposure alters neuromotor development and hippocampus acetylcholinesterase activity in rat offspring. Brain Res 2015; 1595:10-8. [DOI: 10.1016/j.brainres.2014.10.039] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Revised: 09/29/2014] [Accepted: 10/21/2014] [Indexed: 02/03/2023]
|
130
|
Oliveira CD, Oliveira CMD, de Macedo IC, Quevedo AS, Filho PRM, Silva FRD, Vercelino R, de Souza ICC, Caumo W, Torres ILS. Hypercaloric diet modulates effects of chronic stress: a behavioral and biometric study on rats. Stress 2015; 18:514-23. [PMID: 26364693 DOI: 10.3109/10253890.2015.1079616] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Obesity is a chronic disease that has been associated with chronic stress and hypercaloric diet (HD) consumption. Increased ingestion of food containing sugar and fat ingredients (comfort food) is proposed to "compensate" chronic stress effects. However, this eating habit may increase body fat depositions leading to obesity. This study evaluated behavioral/physiological parameters seeking to establish whether there is an association between the effects of HD intake and stress, and to test the hypothesis that the development of anxious behavior and obesity during chronic stress periods depends on the type of diet. Sixty-day-old male Wistar rats (n = 100) were divided into four groups: standard chow, hypercaloric diet, chronic stress/standard chow and chronic stress/hypercaloric diet. Chronic stress was induced by restraint stress exposure for 1 h/day, for 80 d. At the end of this period, rat behavior was evaluated using open-field and plus-maze tests. The results showed that HD alone increased weight gain and adipose deposition in subcutaneous and mesenteric areas. However, stress reduced weight gain and adipose tissue in these areas. HD also increased naso-anal length and concurrent stress prevented this. Behavioral data indicated that stress increased anxiety-like behaviors and comfort food reduced these anxiogenic effects; locomotor activity increased in rats fed with HD. Furthermore, HD decreased corticosterone levels and stress increased adrenal weight. The data indicate that when rats are given HD and experience chronic stress this association reduces the pro-obesogenic effects of HD, and decreases adrenocortical activity.
Collapse
Affiliation(s)
- Carla de Oliveira
- a Pharmacology of Pain and Neuromodulation Laboratory: Animal Models, Department of Pharmacology , Institute of Basic Health Sciences (ICBS), Federal University of Rio Grande do Sul , ICBS , Porto Alegre , RS , Brazil
- b Medicine School, Federal University of Rio Grande do Sul , Porto Alegre , RS , Brazil
- c Animal Experimentation Unit and Graduate Research Group, Hospital de Clinicas de Porto Alegre , Porto Alegre , RS , Brazil , and
| | - Cleverson Moraes de Oliveira
- a Pharmacology of Pain and Neuromodulation Laboratory: Animal Models, Department of Pharmacology , Institute of Basic Health Sciences (ICBS), Federal University of Rio Grande do Sul , ICBS , Porto Alegre , RS , Brazil
- b Medicine School, Federal University of Rio Grande do Sul , Porto Alegre , RS , Brazil
- c Animal Experimentation Unit and Graduate Research Group, Hospital de Clinicas de Porto Alegre , Porto Alegre , RS , Brazil , and
| | - Isabel Cristina de Macedo
- a Pharmacology of Pain and Neuromodulation Laboratory: Animal Models, Department of Pharmacology , Institute of Basic Health Sciences (ICBS), Federal University of Rio Grande do Sul , ICBS , Porto Alegre , RS , Brazil
- c Animal Experimentation Unit and Graduate Research Group, Hospital de Clinicas de Porto Alegre , Porto Alegre , RS , Brazil , and
- d Institute of Basic Health Sciences (ICBS), Federal University of Rio Grande do Sul , Porto Alegre , RS , Brazil
| | - Alexandre S Quevedo
- a Pharmacology of Pain and Neuromodulation Laboratory: Animal Models, Department of Pharmacology , Institute of Basic Health Sciences (ICBS), Federal University of Rio Grande do Sul , ICBS , Porto Alegre , RS , Brazil
- b Medicine School, Federal University of Rio Grande do Sul , Porto Alegre , RS , Brazil
- c Animal Experimentation Unit and Graduate Research Group, Hospital de Clinicas de Porto Alegre , Porto Alegre , RS , Brazil , and
| | - Paulo Ricardo Marques Filho
- a Pharmacology of Pain and Neuromodulation Laboratory: Animal Models, Department of Pharmacology , Institute of Basic Health Sciences (ICBS), Federal University of Rio Grande do Sul , ICBS , Porto Alegre , RS , Brazil
- b Medicine School, Federal University of Rio Grande do Sul , Porto Alegre , RS , Brazil
- c Animal Experimentation Unit and Graduate Research Group, Hospital de Clinicas de Porto Alegre , Porto Alegre , RS , Brazil , and
| | - Fernanda Ribeiro da Silva
- a Pharmacology of Pain and Neuromodulation Laboratory: Animal Models, Department of Pharmacology , Institute of Basic Health Sciences (ICBS), Federal University of Rio Grande do Sul , ICBS , Porto Alegre , RS , Brazil
- b Medicine School, Federal University of Rio Grande do Sul , Porto Alegre , RS , Brazil
- c Animal Experimentation Unit and Graduate Research Group, Hospital de Clinicas de Porto Alegre , Porto Alegre , RS , Brazil , and
| | - Rafael Vercelino
- a Pharmacology of Pain and Neuromodulation Laboratory: Animal Models, Department of Pharmacology , Institute of Basic Health Sciences (ICBS), Federal University of Rio Grande do Sul , ICBS , Porto Alegre , RS , Brazil
- d Institute of Basic Health Sciences (ICBS), Federal University of Rio Grande do Sul , Porto Alegre , RS , Brazil
| | - Izabel C Custodio de Souza
- a Pharmacology of Pain and Neuromodulation Laboratory: Animal Models, Department of Pharmacology , Institute of Basic Health Sciences (ICBS), Federal University of Rio Grande do Sul , ICBS , Porto Alegre , RS , Brazil
- b Medicine School, Federal University of Rio Grande do Sul , Porto Alegre , RS , Brazil
| | - Wolnei Caumo
- a Pharmacology of Pain and Neuromodulation Laboratory: Animal Models, Department of Pharmacology , Institute of Basic Health Sciences (ICBS), Federal University of Rio Grande do Sul , ICBS , Porto Alegre , RS , Brazil
- b Medicine School, Federal University of Rio Grande do Sul , Porto Alegre , RS , Brazil
| | - Iraci L S Torres
- a Pharmacology of Pain and Neuromodulation Laboratory: Animal Models, Department of Pharmacology , Institute of Basic Health Sciences (ICBS), Federal University of Rio Grande do Sul , ICBS , Porto Alegre , RS , Brazil
- b Medicine School, Federal University of Rio Grande do Sul , Porto Alegre , RS , Brazil
- c Animal Experimentation Unit and Graduate Research Group, Hospital de Clinicas de Porto Alegre , Porto Alegre , RS , Brazil , and
- d Institute of Basic Health Sciences (ICBS), Federal University of Rio Grande do Sul , Porto Alegre , RS , Brazil
| |
Collapse
|
131
|
Cepko LC, Selva JA, Merfeld EB, Fimmel AI, Goldberg SA, Currie PJ. Ghrelin alters the stimulatory effect of cocaine on ethanol intake following mesolimbic or systemic administration. Neuropharmacology 2014; 85:224-31. [DOI: 10.1016/j.neuropharm.2014.05.030] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2014] [Revised: 04/29/2014] [Accepted: 05/18/2014] [Indexed: 01/16/2023]
|
132
|
Effects of Toxoplasma gondii infection on anxiety, depression and ghrelin level in male rats. J Parasit Dis 2014; 40:688-93. [PMID: 27605768 DOI: 10.1007/s12639-014-0561-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 09/03/2014] [Indexed: 10/24/2022] Open
Abstract
The aim of the present study was to determine the effects of Toxoplasma gondii (T. gondii) infection on anxiety, depression and ghrelin level in male rats. Twenty four male rats were assessed in two equal groups. T. gondii tachyzoite (ip) were injected in infected group and control group received (2 ml) physiological serum (ip). Elevated plus Maze and swimming tests were used to assess anxiety and depression in rats respectively. The ghrelin and T. gondii IgG serum levels were measured by enzyme immunoassay kits. The Student's t test and Pearson correlation coefficient were used. The ghrelin serum level was significantly lower in the infected rats than control (P = 0.03). There were no significant differences in the depression and anxiety behavior between two groups. However, here were no significant correlations between ghrelin level and anxiety or depression in rats. It seems that latent T. gondii infection decreases the ghrelin serum level but does not change anxiety and depression like behaviors.
Collapse
|
133
|
Wang L, Song Y, Li F, Liu Y, Ma J, Mao M, Wu F, Wu Y, Li S, Guan B, Liu X. Effects of Wen Dan Tang on insomnia-related anxiety and levels of the brain-gut peptide Ghrelin. Neural Regen Res 2014; 9:205-12. [PMID: 25206802 PMCID: PMC4146165 DOI: 10.4103/1673-5374.125351] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/05/2013] [Indexed: 11/04/2022] Open
Abstract
Ghrelin, a brain-gut peptide that induces anxiety and other abnormal emotions, contributes to the effects of insomnia on emotional behavior. In contrast, the traditional Chinese Medicine remedy Wen Dan Tang reduces insomnia-related anxiety, which may perhaps correspond to changes in the brain-gut axis. This suggests a possible relationship between Wen Dan Tang's pharmacological mechanism and the brain-gut axis. Based on this hypothesis, a sleep-deprived rat model was induced and Wen Dan Tang was administered using oral gavage during model establishment. Wen Dan Tang significantly reduced insomnia-related anxiety and prevented Ghrelin level decreases following sleep deprivation, especially in the hypothalamus. Increased expression of Ghrelin receptor mRNA in the hypothalamus was also observed, suggesting that reduced anxiety may be a result of Wen Dan Tang's regulation of Ghrelin-Ghrelin receptors.
Collapse
Affiliation(s)
- Liye Wang
- Nautical Chinese Medicine Key Subject, Beijing University of Chinese Medicine, Beijing, China
| | - Yuehan Song
- Nautical Chinese Medicine Key Subject, Beijing University of Chinese Medicine, Beijing, China
| | - Feng Li
- Nautical Chinese Medicine Key Subject, Beijing University of Chinese Medicine, Beijing, China
| | - Yan Liu
- Nautical Chinese Medicine Key Subject, Beijing University of Chinese Medicine, Beijing, China
| | - Jie Ma
- Nautical Chinese Medicine Key Subject, Beijing University of Chinese Medicine, Beijing, China
| | - Meng Mao
- Nautical Chinese Medicine Key Subject, Beijing University of Chinese Medicine, Beijing, China
| | - Fengzhi Wu
- Nautical Chinese Medicine Key Subject, Beijing University of Chinese Medicine, Beijing, China
| | - Ying Wu
- Nautical Chinese Medicine Key Subject, Beijing University of Chinese Medicine, Beijing, China
| | - Sinai Li
- Nautical Chinese Medicine Key Subject, Beijing University of Chinese Medicine, Beijing, China
| | - Binghe Guan
- Nautical Chinese Medicine Key Subject, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaolan Liu
- Nautical Chinese Medicine Key Subject, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
134
|
Kent BA. Synchronizing an aging brain: can entraining circadian clocks by food slow Alzheimer's disease? Front Aging Neurosci 2014; 6:234. [PMID: 25225484 PMCID: PMC4150207 DOI: 10.3389/fnagi.2014.00234] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2014] [Accepted: 08/15/2014] [Indexed: 01/21/2023] Open
Abstract
Alzheimer's disease (AD) is a global epidemic. Unfortunately, we are still without effective treatments or a cure for this disease, which is having devastating consequences for patients, their families, and societies around the world. Until effective treatments are developed, promoting overall health may hold potential for delaying the onset or preventing neurodegenerative diseases such as AD. In particular, chronobiological concepts may provide a useful framework for identifying the earliest signs of age-related disease as well as inexpensive and noninvasive methods for promoting health. It is well reported that AD is associated with disrupted circadian functioning to a greater extent than normal aging. However, it is unclear if the central circadian clock (i.e., the suprachiasmatic nucleus) is dysfunctioning, or whether the synchrony between the central and peripheral clocks that control behavior and metabolic processes are becoming uncoupled. Desynchrony of rhythms can negatively affect health, increasing morbidity and mortality in both animal models and humans. If the uncoupling of rhythms is contributing to AD progression or exacerbating symptoms, then it may be possible to draw from the food-entrainment literature to identify mechanisms for re-synchronizing rhythms to improve overall health and reduce the severity of symptoms. The following review will briefly summarize the circadian system, its potential role in AD, and propose using a feeding-related neuropeptide, such as ghrelin, to synchronize uncoupled rhythms. Synchronizing rhythms may be an inexpensive way to promote healthy aging and delay the onset of neurodegenerative disease such as AD.
Collapse
Affiliation(s)
- Brianne A. Kent
- Department of Psychology, University of CambridgeCambridge, UK
| |
Collapse
|
135
|
Sivertsen B, Holliday N, Madsen AN, Holst B. Functionally biased signalling properties of 7TM receptors - opportunities for drug development for the ghrelin receptor. Br J Pharmacol 2014; 170:1349-62. [PMID: 24032557 DOI: 10.1111/bph.12361] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Revised: 06/17/2013] [Accepted: 08/06/2013] [Indexed: 12/23/2022] Open
Abstract
UNLABELLED The ghrelin receptor is a 7 transmembrane (7TM) receptor involved in a variety of physiological functions including growth hormone secretion, increased food intake and fat accumulation as well as modulation of reward and cognitive functions. Because of its important role in metabolism and energy expenditure, the ghrelin receptor has become an important therapeutic target for drug design and the development of anti-obesity compounds. However, none of the compounds developed so far have been approved for commercial use. Interestingly, the ghrelin receptor is able to signal through several different signalling pathways including Gαq , Gαi/o , Gα12/13 and arrestin recruitment. These multiple signalling pathways allow for functionally biased signalling, where one signalling pathway may be favoured over another either by selective ligands or through mutations in the receptor. In the present review, we have described how ligands and mutations in the 7TM receptor may bias the receptors to favour either one G-protein over another or to promote G-protein independent signalling pathways rather than G-protein-dependent pathways. For the ghrelin receptor, both agonist and inverse agonists have been demonstrated to signal more strongly through the Gαq -coupled pathway than the Gα12/13 -coupled pathway. Similarly a ligand that promotes Gαq coupling over Gαi coupling has been described and it has been suggested that several different active conformations of the receptor may exist dependent on the properties of the agonist. Importantly, ligands with such biased signalling properties may allow the development of drugs that selectively modulate only the therapeutically relevant physiological functions, thereby decreasing the risk of side effects. LINKED ARTICLES This article is part of a themed section on Neuropeptides. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2013.170.issue-7.
Collapse
Affiliation(s)
- B Sivertsen
- Laboratory for Molecular Pharmacology, Department of Neuroscience and Pharmacology, the Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | | | | | | |
Collapse
|
136
|
Ghrelin: a link between ageing, metabolism and neurodegenerative disorders. Neurobiol Dis 2014; 72 Pt A:72-83. [PMID: 25173805 DOI: 10.1016/j.nbd.2014.08.026] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Revised: 06/28/2014] [Accepted: 08/20/2014] [Indexed: 12/13/2022] Open
Abstract
Along with the increase in life expectancy over the last century comes the increased risk for development of age-related disorders, including metabolic and neurodegenerative diseases such as Alzheimer's, Parkinson's and Huntington's diseases. These chronic disorders share two main characteristics: 1) neuronal loss in motor, sensory or cognitive systems, leading to cognitive and motor decline; and 2) a strong correlation between metabolic changes and neurodegeneration. In order to treat them, a better understanding of their complexity is required: it is necessary to interpret the neuronal damage in light of the metabolic changes, and to find the disrupted link between the peripheral organs governing energy metabolism and the CNS. This review is an attempt to present ghrelin as part of molecular regulatory interface between energy metabolism, neuroendocrine and neurodegenerative processes. Ghrelin takes part in lipid and glucose metabolism, in higher brain functions such as sleep-wake state, learning and memory consolidation; it influences mitochondrial respiration and shows neuroprotective effect. All these make ghrelin an attractive target for development of biomarkers or therapeutics for prevention or treatment of disorders, in which cell protection and recruitment of new neurons or synapses are needed.
Collapse
|
137
|
Akter S, Pham NM, Nanri A, Kurotani K, Kuwahara K, Jacka FN, Yasuda K, Sato M, Mizoue T. Association of serum leptin and ghrelin with depressive symptoms in a Japanese working population: a cross-sectional study. BMC Psychiatry 2014; 14:203. [PMID: 25079305 PMCID: PMC4261564 DOI: 10.1186/1471-244x-14-203] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Accepted: 07/09/2014] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Leptin and ghrelin have been implicated in the pathogenesis of major depression. However, evidence is lacking among apparently healthy people. This study examined the relationship of these appetite hormones to depressive symptoms in a Japanese working population. METHODS A cross-sectional study was conducted in 2009 among 497 Japanese employees (287 men and 210 women) aged 20-68 years. Fasting serum leptin and ghrelin levels were measured using a Luminex suspension bead-based multiplexed array. Depressive symptoms were assessed using the Center for Epidemiologic Studies Depression (CES-D) scale. Logistic regression analysis was performed to estimate odds ratio (OR) and 95% confidence interval (CI) for depressive symptoms with adjustment for potential confounders. RESULTS The prevalence of depressive symptoms (CES-D ≥16) was 26.5% and 33.3% among men and women, respectively. Women in the middle and highest tertiles of leptin levels showed lower odds for depressive symptoms compared with those in the lowest level, although the trend association was not statistically significant (Ptrend = 0.14). Higher ghrelin levels were associated with increased odds for depressive symptoms in women (Ptrend = 0.02). The multivariable adjusted OR (95% CI) of having depressive symptoms for the lowest through highest tertiles of ghrelin levels were 1.00 (reference), 1.71 (0.76 - 3.86), and 2.69 (1.16 - 6.28), respectively. Neither leptin nor ghrelin was associated with depressive symptoms in men. CONCLUSIONS Results suggest that lower leptin and higher ghrelin levels may be related to higher prevalence of depressive status among Japanese women.
Collapse
Affiliation(s)
- Shamima Akter
- Department of Epidemiology and Prevention, Center for Clinical Sciences, National Center for Global Health and Medicine, Toyama 1-21-1, 162-8655 Shinjuku-ku, Tokyo, Japan.
| | - Ngoc Minh Pham
- Department of Epidemiology and Prevention, Center for Clinical Sciences, National Center for Global Health and Medicine, Toyama 1-21-1, 162-8655 Shinjuku-ku, Tokyo, Japan
| | - Akiko Nanri
- Department of Epidemiology and Prevention, Center for Clinical Sciences, National Center for Global Health and Medicine, Toyama 1-21-1, 162-8655 Shinjuku-ku, Tokyo, Japan
| | - Kayo Kurotani
- Department of Epidemiology and Prevention, Center for Clinical Sciences, National Center for Global Health and Medicine, Toyama 1-21-1, 162-8655 Shinjuku-ku, Tokyo, Japan
| | - Keisuke Kuwahara
- Department of Epidemiology and Prevention, Center for Clinical Sciences, National Center for Global Health and Medicine, Toyama 1-21-1, 162-8655 Shinjuku-ku, Tokyo, Japan
| | - Felice N Jacka
- IMPACT Strategic Research Centre, Deakin University, Melbourne, Australia
| | - Kazuki Yasuda
- Department of Metabolic Disorder, Diabetes Research Center, National Center for Global Health and Medicine, Shinjuku-ku, Tokyo, Japan
| | - Masao Sato
- Department of Applied Biological Chemistry, Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, Fukuoka, Japan
| | - Tetsuya Mizoue
- Department of Epidemiology and Prevention, Center for Clinical Sciences, National Center for Global Health and Medicine, Toyama 1-21-1, 162-8655 Shinjuku-ku, Tokyo, Japan
| |
Collapse
|
138
|
Ulrich-Lai YM, Ryan KK. Neuroendocrine circuits governing energy balance and stress regulation: functional overlap and therapeutic implications. Cell Metab 2014; 19:910-25. [PMID: 24630812 PMCID: PMC4047143 DOI: 10.1016/j.cmet.2014.01.020] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Significant comorbidities between obesity-related metabolic disease and stress-related psychological disorders suggest important functional interactions between energy balance and brain stress integration. Largely overlapping neural circuits control these systems, and this anatomical arrangement optimizes opportunities for mutual influence. Here we first review the current literature identifying effects of metabolic neuroendocrine signals on stress regulation, and vice versa. Next, the contributions of reward-driven food intake to these metabolic and stress interactions are discussed. Lastly, we consider the interrelationships between metabolism, stress, and reward in light of their important implications in the development of therapies for metabolism- or stress-related disease.
Collapse
Affiliation(s)
- Yvonne M Ulrich-Lai
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, OH 45237, USA
| | - Karen K Ryan
- Department of Internal Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Cincinnati College of Medicine, Cincinnati, OH 45237, USA.
| |
Collapse
|
139
|
Sárvári M, Kocsis P, Deli L, Gajári D, Dávid S, Pozsgay Z, Hegedűs N, Tihanyi K, Liposits Z. Ghrelin modulates the fMRI BOLD response of homeostatic and hedonic brain centers regulating energy balance in the rat. PLoS One 2014; 9:e97651. [PMID: 24830778 PMCID: PMC4022590 DOI: 10.1371/journal.pone.0097651] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 04/22/2014] [Indexed: 12/22/2022] Open
Abstract
The orexigenic gut-brain peptide, ghrelin and its G-protein coupled receptor, the growth hormone secretagogue receptor 1a (GHS-R1A) are pivotal regulators of hypothalamic feeding centers and reward processing neuronal circuits of the brain. These systems operate in a cooperative manner and receive a wide array of neuronal hormone/transmitter messages and metabolic signals. Functional magnetic resonance imaging was employed in the current study to map BOLD responses to ghrelin in different brain regions with special reference on homeostatic and hedonic regulatory centers of energy balance. Experimental groups involved male, ovariectomized female and ovariectomized estradiol-replaced rats. Putative modulation of ghrelin signaling by endocannabinoids was also studied. Ghrelin-evoked effects were calculated as mean of the BOLD responses 30 minutes after administration. In the male rat, ghrelin evoked a slowly decreasing BOLD response in all studied regions of interest (ROI) within the limbic system. This effect was antagonized by pretreatment with GHS-R1A antagonist JMV2959. The comparison of ghrelin effects in the presence or absence of JMV2959 in individual ROIs revealed significant changes in the prefrontal cortex, nucleus accumbens of the telencephalon, and also within hypothalamic centers like the lateral hypothalamus, ventromedial nucleus, paraventricular nucleus and suprachiasmatic nucleus. In the female rat, the ghrelin effects were almost identical to those observed in males. Ovariectomy and chronic estradiol replacement had no effect on the BOLD response. Inhibition of the endocannabinoid signaling by rimonabant significantly attenuated the response of the nucleus accumbens and septum. In summary, ghrelin can modulate hypothalamic and mesolimbic structures controlling energy balance in both sexes. The endocannabinoid signaling system contributes to the manifestation of ghrelin's BOLD effect in a region specific manner. In females, the estradiol milieu does not influence the BOLD response to ghrelin.
Collapse
Affiliation(s)
- Miklós Sárvári
- Laboratory of Endocrine Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
- * E-mail:
| | - Pál Kocsis
- Preclinical Imaging Center, Gedeon Richter Ltd., Budapest, Hungary
| | - Levente Deli
- Preclinical Imaging Center, Gedeon Richter Ltd., Budapest, Hungary
| | - Dávid Gajári
- Preclinical Imaging Center, Gedeon Richter Ltd., Budapest, Hungary
| | - Szabolcs Dávid
- Preclinical Imaging Center, Gedeon Richter Ltd., Budapest, Hungary
| | - Zsófia Pozsgay
- Preclinical Imaging Center, Gedeon Richter Ltd., Budapest, Hungary
| | - Nikolett Hegedűs
- Preclinical Imaging Center, Gedeon Richter Ltd., Budapest, Hungary
| | - Károly Tihanyi
- Preclinical Imaging Center, Gedeon Richter Ltd., Budapest, Hungary
| | - Zsolt Liposits
- Laboratory of Endocrine Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
- Faculty of Information Technology, Pázmány Péter Catholic University, Budapest, Hungary
| |
Collapse
|
140
|
Hansson C, Alvarez-Crespo M, Taube M, Skibicka KP, Schmidt L, Karlsson-Lindahl L, Egecioglu E, Nissbrandt H, Dickson SL. Influence of ghrelin on the central serotonergic signaling system in mice. Neuropharmacology 2014; 79:498-505. [DOI: 10.1016/j.neuropharm.2013.12.012] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Revised: 11/22/2013] [Accepted: 12/14/2013] [Indexed: 02/09/2023]
|
141
|
Glucose impairment and ghrelin gene variants are associated to cognitive dysfunction. Aging Clin Exp Res 2014; 26:161-9. [PMID: 24619886 DOI: 10.1007/s40520-014-0203-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Accepted: 09/10/2013] [Indexed: 10/25/2022]
Abstract
BACKGROUND AND AIMS Cognitive state and brain volume have been related to body mass index, abdominal fat, waist-hip ratio, components of metabolic syndrome (MS) and ghrelin. Genetic variations within the ghrelin gene have been recently associated to MS. The aim of our study was to investigate cognitive state by Mini-Mental State Examination (MMSE) in relation to MS components (ATP-III criteria) and ghrelin gene polymorphisms in dwelling individuals aged ≥70. METHODS 280 subjects (137 men/143 women, age 77.03 ± 5.92) from the Mataró Ageing Study were included. Individuals were phenotypically characterized by anthropometric variables, lipids, glucose, blood pressure and MMSE. SNPs -501AC (rs26802), -994CT (rs26312), -604GA (rs27647), M72L (rs696217) and L90G (rs4684677) of the ghrelin gene were studied. Genotypes were determined by polymerase chain reaction and SNapshot minisequencing. RESULTS 22.1 % had MMSE <24. MMSE <24 was associated with age (p < 0.001), female gender (p = 0.016), low education (p < 0.001) and glucose impairment or diabetes (p = 0.040). MMSE was influenced by obesity, central obesity, MS and glucose impairment. This latter association remained significant after adjustment by gender, age, alcohol, educational level, GDS and ApoE genotype (p = 0.009). Ghrelin SNPs were associated to MMSE: M72L C/A genotype showed lower score than C/C (p = 0.032, after adjusting for confounders 0.049); L90G A/T genotype showed lower score than A/A (p = 0.054, after adjusting 0.005). MMSE <24 was associated to L90G (39.1 % in A/T genotype vs 19.3 % in A/A, p = 0.026, after adjusting for confounders p = 0.002, OR 6.18 CI 1.93-21.75). CONCLUSIONS Glucose impairment and L90G Ghrelin gene variant influence cognitive function in old dwelling individuals participating in the Mataró Ageing Study.
Collapse
|
142
|
Cahill SP, Hatchard T, Abizaid A, Holahan MR. An examination of early neural and cognitive alterations in hippocampal-spatial function of ghrelin receptor-deficient rats. Behav Brain Res 2014; 264:105-15. [PMID: 24525421 DOI: 10.1016/j.bbr.2014.02.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Revised: 01/29/2014] [Accepted: 02/03/2014] [Indexed: 12/31/2022]
Abstract
Ghrelin, a hormone implicated in the regulation of feeding and energy balance, has also been associated with neural function underlying learning and memory. These effects are thought to be mediated by ghrelin targeting receptors at extra hypothalamic sites such as the hippocampus. Exogenous ghrelin administration increases dendritic spine density in the hippocampal CA1 region and neurogenesis in the dentate gyrus (DG), while improving memory in rats. In the present study, we sought to determine whether rats lacking the ghrelin receptor would show early neural or cognitive decline measured via hippocampal integrity (spine density and neurogenesis) and spatial learning and memory. As such, we used young and middle-aged adult rats with mutations to the gene encoding for the ghrelin receptor (GHS-R KO) and wildtype (WT) littermates to determine differences in performance on hippocampal-dependent tasks (the water maze and radial arm maze). In addition, we examined the hippocampal dentate gyrus of these rats for differences in dendritic spine density and cell proliferation (doublecortin). Overall, results demonstrated that spine density and doublecortin staining in the dentate gyrus of the young GHS-R KO group was similar to that seen in middle-aged groups (both KO and WT) and lower than the young WT group. Middle-aged GHS-R KO and WT groups showed deficits on the radial arm maze food-motivated task but not the water maze task. These data suggest that impaired ghrelin signaling leads to an early onset decrement in hippocampal structural integrity that may manifest in non- spatial-related behavioral deficits.
Collapse
Affiliation(s)
- Shaina P Cahill
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada
| | - Taylor Hatchard
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada
| | - Alfonso Abizaid
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada
| | - Matthew R Holahan
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada.
| |
Collapse
|
143
|
Paoluzi OA, Blanco DVG, Caruso R, Monteleone I, Monteleone G, Pallone F. Impairment of ghrelin synthesis in Helicobacter pylori-colonized stomach: New clues for the pathogenesis of H. pylori-related gastric inflammation. World J Gastroenterol 2014; 20:639-646. [PMID: 24574737 PMCID: PMC3921473 DOI: 10.3748/wjg.v20.i3.639] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Revised: 11/14/2013] [Accepted: 12/06/2013] [Indexed: 02/06/2023] Open
Abstract
Ghrelin, the ligand of growth hormone secretagogue receptor 1a, takes part in several functions of the digestive system, including regulation of appetite, energy homeostasis, gastric acid secretion and motility. Ghrelin has also immunoregulatory properties and is supposed to inhibit some inflammatory pathways that can mediate gastric damage. Interestingly, ghrelin synthesis is reduced in the gastric mucosa of patients with Helicobacter pylori (H. pylori) infection, a worldwide condition inducing a T helper (Th)1/Th17 cell response-driven gastritis, which may evolve towards gastric atrophy and cancer. In this article, we review the available data on the expression of ghrelin in H. pylori infection and discuss how the defective ghrelin synthesis may contribute to sustain the ongoing inflammatory response in this disease.
Collapse
|
144
|
Gahete MD, Rincón-Fernández D, Villa-Osaba A, Hormaechea-Agulla D, Ibáñez-Costa A, Martínez-Fuentes AJ, Gracia-Navarro F, Castaño JP, Luque RM. Ghrelin gene products, receptors, and GOAT enzyme: biological and pathophysiological insight. J Endocrinol 2014; 220:R1-24. [PMID: 24194510 DOI: 10.1530/joe-13-0391] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Ghrelin is a 28-amino acid acylated hormone, highly expressed in the stomach, which binds to its cognate receptor (GHSR1a) to regulate a plethora of relevant biological processes, including food intake, energy balance, hormonal secretions, learning, inflammation, etc. However, ghrelin is, in fact, the most notorious component of a complex, intricate regulatory system comprised of a growing number of alternative peptides (e.g. obestatin, unacylated ghrelin, and In1-ghrelin, etc.), known (GHSRs) and, necessarily unknown receptors, as well as modifying enzymes (e.g. ghrelin-O-acyl-transferase), which interact among them as well as with other regulatory systems in order to tightly modulate key (patho)-physiological processes. This multiplicity of functions and versatility of the ghrelin system arise from a dual, genetic and functional, complexity. Importantly, a growing body of evidence suggests that dysregulation in some of the components of the ghrelin system can lead to or influence the development and/or progression of highly concerning pathologies such as endocrine-related tumors, inflammatory/cardiovascular diseases, and neurodegeneration, wherein these altered components could be used as diagnostic, prognostic, or therapeutic targets. In this context, the aim of this review is to integrate and comprehensively analyze the multiple components and functions of the ghrelin system described to date in order to define and understand its biological and (patho)-physiological significance.
Collapse
Affiliation(s)
- Manuel D Gahete
- Department of Cell Biology, Physiology and Immunology, Campus Universitario de Rabanales, Edificio Severo Ochoa (C6), Planta 3, University of Córdoba, 14014-Córdoba; Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), University of Córdoba; Reina Sofia University Hospital, Córdoba; and CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
145
|
Lee CY, Abizaid A. The gut-brain-axis as a target to treat stress-induced obesity. Front Endocrinol (Lausanne) 2014; 5:117. [PMID: 25101055 PMCID: PMC4102906 DOI: 10.3389/fendo.2014.00117] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Accepted: 07/03/2014] [Indexed: 12/16/2022] Open
Affiliation(s)
- Chooi Yeng Lee
- School of Pharmacy, Monash University Malaysia, Bandar Sunway, Malaysia
- *Correspondence:
| | - Alfonso Abizaid
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada
| |
Collapse
|
146
|
Gong Z, Yoshimura M, Aizawa S, Kurotani R, Zigman JM, Sakai T, Sakata I. G protein-coupled receptor 120 signaling regulates ghrelin secretion in vivo and in vitro. Am J Physiol Endocrinol Metab 2014; 306:E28-35. [PMID: 24222669 DOI: 10.1152/ajpendo.00306.2013] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Ghrelin, an endogenous ligand for the growth hormone secretagogue receptor, is produced predominantly in the stomach. It has been reported that endogenous ghrelin levels are increased by fasting and decreased immediately after feeding and that fasting-induced ghrelin release is controlled by the sympathetic nervous system. However, the mechanisms of plasma ghrelin decrement after feeding are poorly understood. Here, we studied the control of ghrelin secretion using ghrelin-producing cell lines and found that these cells express high levels of mRNA encoding G-protein coupled receptor 120 (GPR120). Addition of GW-9508 (a GPR120 chemical agonist) and α-linolenic acid (a natural ligand for GPR120) inhibited the secretion of ghrelin by ∼50 and 70%, respectively. However, the expression levels of preproghrelin and ghrelin O-acyltransferase (GOAT) mRNAs were not influenced by GW-9508. In contrast, the expression levels of prohormone convertase 1 were decreased significantly by GW-9508 incubation. Moreover, we observed that the inhibitory effect of GW-9508 on ghrelin secretion was blocked by a small interfering RNA (siRNA) targeting the sequence of GPR120. Furthermore, pretreatment with GW-9508 blocked the effect of the norepinephrine (NE)-induced ghrelin elevation in ghrelin cell lines. In addition, we showed that GW-9508 inhibited ghrelin secretion via extracellular signal-regulated kinase activity in ghrelin cell lines. Finally, we found that GW-9508 decreased plasma ghrelin levels in mice. These results suggest that the decrease of ghrelin secretion after feeding is induced partially by long-chain fatty acids that act directly on gastric GPR120-expressing ghrelin cells.
Collapse
Affiliation(s)
- Zhi Gong
- Area of Regulatory Biology, Division of Life Science, Graduate School of Science and Engineering, Saitama University, Shimo-ohkubo, Sakuraku, Saitama, Japan
| | | | | | | | | | | | | |
Collapse
|
147
|
Tinoco AB, Näslund J, Delgado MJ, de Pedro N, Johnsson JI, Jönsson E. Ghrelin increases food intake, swimming activity and growth in juvenile brown trout (Salmo trutta). Physiol Behav 2014; 124:15-22. [DOI: 10.1016/j.physbeh.2013.10.034] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2013] [Revised: 10/17/2013] [Accepted: 10/23/2013] [Indexed: 01/05/2023]
|
148
|
Ghrelin triggers the synaptic incorporation of AMPA receptors in the hippocampus. Proc Natl Acad Sci U S A 2013; 111:E149-58. [PMID: 24367106 DOI: 10.1073/pnas.1313798111] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Ghrelin is a peptide mainly produced by the stomach and released into circulation, affecting energy balance and growth hormone release. These effects are guided largely by the expression of the ghrelin receptor growth hormone secretagogue type 1a (GHS-R1a) in the hypothalamus and pituitary. However, GHS-R1a is expressed in other brain regions, including the hippocampus, where its activation enhances memory retention. Herein we explore the molecular mechanism underlying the action of ghrelin on hippocampal-dependent memory. Our data show that GHS-R1a is localized in the vicinity of hippocampal excitatory synapses, and that its activation increases delivery of α-amino-3-hydroxy-5-methyl-4-isoxazole propionic-type receptors (AMPARs) to synapses, producing functional modifications at excitatory synapses. Moreover, GHS-R1a activation enhances two different paradigms of long-term potentiation in the hippocampus, activates the phosphatidylinositol 3-kinase, and increases GluA1 AMPAR subunit and stargazin phosphorylation. We propose that GHS-R1a activation in the hippocampus enhances excitatory synaptic transmission and synaptic plasticity by regulating AMPAR trafficking. Our study provides insights into mechanisms that may mediate the cognition-enhancing effect of ghrelin, and suggests a possible link between the regulation of energy metabolism and learning.
Collapse
|
149
|
Ghrelin but not nesfatin-1 affects certain forms of learning and memory in both rats and mice. Brain Res 2013; 1541:42-51. [DOI: 10.1016/j.brainres.2013.10.022] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Revised: 10/09/2013] [Accepted: 10/13/2013] [Indexed: 12/16/2022]
|
150
|
Mora M, Granada ML, Palomera E, Serra-Prat M, Puig-Domingo M. Obestatin is associated to muscle strength, functional capacity and cognitive status in old women. AGE (DORDRECHT, NETHERLANDS) 2013; 35:2515-2523. [PMID: 23604919 PMCID: PMC3824982 DOI: 10.1007/s11357-013-9532-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Accepted: 04/01/2013] [Indexed: 06/02/2023]
Abstract
Obestatin has been proposed to have anorexigenic and anti-ghrelin actions. The objective was to study obestatin concentrations in relation to handgrip strength, functional capacity and cognitive state in old women. The prospective study included 110 women (age, 76.93 ± 6.32) from the Mataró Ageing Study. Individuals were characterized by anthropometric variables, grip strength, Barthel and assessment of cognitive impairment [Mini Cognoscitive Examination (MCE) Spanish version], depressive status by the Geriatric Depression Scale (GDS) and frailty by the Fried criteria. Obestatin was measured by IRMA. Obestatin showed negative correlation to handgrip at basal time point (r = -0.220, p = 0.023) and at 2-year follow-up (r = -0.344, p = 0.002). Obestatin, divided into quartiles, showed a negative lineal association with handgrip: 11.03 ± 4.88 kg in first, 8.75 ± 4.08 kg in second, 8.11 ± 3.66 kg in third and 7.61 ± 4.08 kg in fourth quartile (p = 0.018). Higher obestatin levels were associated to increased weakness (categorized by handgrip of frailty criteria): 2.24 ± 0.42 ng/ml in weak vs. 1.87 ± 0.57 ng/ml in non-weak (p = 0.01). The decrease of either MCE or Barthel scores at 2-year follow-up was significantly higher in individuals in the fourth quartile of obestatin in comparison with individuals in the first quartile (p = 0.046 and p = 0.019, respectively). No association was found between obestatin and GDS score and neither with frailty as a condition. Obestatin is associated to low muscle strength, and impaired functional and cognitive capacity in old women participating in the Mataró Ageing Study.
Collapse
Affiliation(s)
- Mireia Mora
- />Department of Endocrinology and Nutrition, Hospital Clínic i Universitari of Barcelona, Barcelona, Spain
| | - María Luisa Granada
- />Department of Biochemistry, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | | | | | - Manel Puig-Domingo
- />Department of Endocrinology and Nutrition, Hospital Universitari Germans Trias i Pujol, Ctra. del Canyet, s/n, 08916 Badalona, Barcelona Spain
| | | |
Collapse
|