1
|
Prins K, Mutsters N, Volker F, Huisman M, Mies R, Delhanty PJD, Visser JA. Syndecans modulate ghrelin receptor signaling. J Mol Endocrinol 2025; 74:e240070. [PMID: 39565158 PMCID: PMC11729051 DOI: 10.1530/jme-24-0070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 10/21/2024] [Accepted: 11/20/2024] [Indexed: 11/21/2024]
Abstract
Ghrelin is a gut hormone that enhances food intake and growth hormone secretion through its G-protein coupled receptor, the growth hormone secretagogue receptor (GHSR). Recently, we have shown that ghrelin interacts with syndecans (SDCs), a family of membrane proteins known to modulate hypothalamic appetite signaling. Here, we investigated whether SDCs impact ghrelin signaling at GHSR by assessing ghrelin-induced intracellular Ca2+ mobilization (iCa2+) and inositol phosphate 1 (IP1) production in HEK293 cells. Compared with controls, the overexpression of SDCs dose-dependently increased the maximum iCa2+ response two- to four-fold, without affecting EC50. The IP1 response was similarly amplified by SDCs, but it also indicated that they reduce constitutive (ghrelin-independent) activity of GHSR. These enhanced responses occurred despite a SDC dose-dependent reduction in plasma membrane GHSR levels. Although ghrelin-stimulated Gαq activation was unaltered by SDC1 expression, it failed to restore iCa2+ responsiveness in GNAQ/11 knockout cells, indicating dependence on Gαq/11, not another Gα subunit. This suggests that SDCs modulate either signaling downstream of Gαq/11 or quenching of β-arrestin2 recruitment to GHSR. Indeed, expression of SDCs at levels that only modestly suppress cell surface receptor reduced ghrelin-induced β-arrestin2 recruitment by ∼80%. SDC co-expression also delayed the peak β-arrestin2 response. However, peak β-arrestin2 recruitment follows the peak iCa2+ response, making it unclear whether reduced β-arrestin2 recruitment potentiated Ca2+ signaling. Altogether, SDCs enhanced iCa2+/IP1 and reduced β-arrestin2 recruitment by GHSR in response to ghrelin, likely by modulating signaling downstream of Gαq. This could be a novel mechanism through which SDCs affect metabolism and obesity.
Collapse
|
2
|
Saenz C, Fernandez G, Llovera R, Tolosa MJ, Cantel S, Fehrentz JA, Mackie K, Leggio L, Zigman J, De Francesco PN, Perello M. Growth hormone secretagogue receptor and cannabinoid receptor type 1 intersection in the mouse brain. Brain Struct Funct 2024; 230:15. [PMID: 39702649 PMCID: PMC11659360 DOI: 10.1007/s00429-024-02876-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 10/14/2024] [Indexed: 12/21/2024]
Abstract
The growth hormone secretagogue receptor (GHSR) and the cannabinoid receptor type 1 (CB1R) are G-protein coupled receptors highly expressed in the brain and involved in critical regulatory processes, such as energy homeostasis, appetite control, reward, and stress responses. GHSR mediates the effects of both ghrelin and liver-expressed antimicrobial peptide 2, while CB1R is targeted by cannabinoids. Strikingly, both receptors mediate their effects by acting on common brain areas and their individual roles have been well characterized. However, the potential for their co-expression in the same neuronal subsets remains largely unexplored. Here, we aim to map the cell populations where GHSR and CB1R might converge, hypothesizing that their co-expression in specific brain circuits could mediate integrated physiological responses. By utilizing two complementary labeling techniques-GHSR-eGFP mice and Fr-ghrelin labeling of GHSR+ cells-along with specific CB1R immunostaining, we sought to visualize and quantify potential areas of overlap. Also, we analyzed several cell RNA sequencing datasets to estimate the fraction of brain cells expressing both GPCRs and their phenotype. Our neuroanatomical studies revealed evident overlap of GHSR+ and CB1R+ signals in specific neuronal subsets mainly located in the cerebral cortex, hippocampus and the amygdala. Transcriptomic analysis revealed specific subsets of Ghsr+/Cnr1+ glutamatergic neurons in the hippocampus and amygdala, as well as different subtypes of Ghsr+/Cnr1+ neurons in the midbrain, hypothalamus, pons, and medulla. Thus, we revealed that GHSR and CB1R interact differentially across specific regions of the mouse brain, providing new insights into how these receptors' actions are integrated. Current findings may open new avenues for dual therapeutic interventions in metabolic disorders, obesity, and psychiatric conditions.
Collapse
Affiliation(s)
- Camila Saenz
- Laboratory of Neurophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], La Plata, Buenos Aires, Argentina
| | - Gimena Fernandez
- Laboratory of Neurophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], La Plata, Buenos Aires, Argentina
| | - Ramiro Llovera
- Laboratory of Neurophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], La Plata, Buenos Aires, Argentina
| | - María J Tolosa
- Laboratory of Neurophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], La Plata, Buenos Aires, Argentina
| | - Sonia Cantel
- Institut des Biomolécules Max Mousseron, Univ Montpellier, CNRS, ENSCM, Montpellier, France
| | - Jean-Alain Fehrentz
- Institut des Biomolécules Max Mousseron, Univ Montpellier, CNRS, ENSCM, Montpellier, France
| | - Kenneth Mackie
- Department of Psychological & Brain Sciences, Indiana University, Bloomington Indiana, USA
| | - Lorenzo Leggio
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, USA
| | - Jeffrey Zigman
- Center for Hypothalamic Research and Division of Endocrinology & Metabolism, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Pablo N De Francesco
- Laboratory of Neurophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], La Plata, Buenos Aires, Argentina.
| | - Mario Perello
- Laboratory of Neurophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], La Plata, Buenos Aires, Argentina.
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, University of Uppsala, Uppsala, Sweden.
| |
Collapse
|
3
|
Jerlhag E. Ghrelin system and GLP-1 as potential treatment targets for alcohol use disorder. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2024; 178:401-432. [PMID: 39523062 DOI: 10.1016/bs.irn.2024.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Peptides of the gut-brain axis have gained recent attention as potential treatment targets for addiction. While the number of gut-brain peptides is vast, ghrelin and glucagon-like peptide-1 (GLP-1) have been suggested as important players. Ghrelin is traditionally considered an orexigenic peptide, but recent studies found that it increases alcohol intake in rodents and craving for alcohol in humans. Additionally, suppression of the ghrelin receptor attenuates alcohol-related responses in animal models reflecting alcohol use disorder (AUD). For instance, a lower alcohol intake, suppressed motivation to consume alcohol, and attenuated reward from alcohol is observed after ghrelin receptor antagonism treatment. On a similar note, a partial ghrelin receptor agonist prevents hangover symptoms in humans. When it comes to the anorexigenic peptide GLP-1, agonists of its receptor are approved to treat diabetes type 2 and obesity. Extensive preclinical studies have revealed that these GLP-1 receptor agonists reduce alcohol intake, suppress the motivation to consume alcohol, and prevent relapse drink, with effects tentatively associated with a reduced alcohol-induced reward. These preclinical findings have to some extent been varied in humans, as GLP-1 receptor agonists decrease alcohol intake in overweight patients with AUD. Furthermore, genetic variations in either the genes encoding for pre-pro-ghrelin, GHSR, GLP-1, or its receptor, are associated with AUD and heavy alcohol drinking. While central mechanisms appear to modulate the ability of either ghrelin or GLP-1 to regulate alcohol-related responses the exact mechanisms have not been defined. Taken together these preclinical and clinical data imply that gut-brain peptides participate in the addiction process and should be considered as potential targets for AUD treatment.
Collapse
Affiliation(s)
- Elisabet Jerlhag
- Institute of Neuroscience and Physiology, Department of Pharmacology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
4
|
Casado S, Varela-Miguéns M, de Oliveira Diz T, Quintela-Vilariño C, Nogueiras R, Diéguez C, Tovar S. The effects of ghrelin and LEAP-2 in energy homeostasis are modulated by thermoneutrality, high-fat diet and aging. J Endocrinol Invest 2024; 47:2061-2074. [PMID: 38337094 PMCID: PMC11266414 DOI: 10.1007/s40618-024-02307-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 01/09/2024] [Indexed: 02/12/2024]
Abstract
PURPOSE Liver-expressed antimicrobial peptide 2 (LEAP-2) has been recently identified as the endogenous non-competitive allosteric antagonist of the growth hormone secretagogue receptor 1a (GHSR1a). In rodents, LEAP-2 blunts ghrelin-induced feeding and its plasma levels are modulated in response to nutritional status, being decreased upon fasting and increased in high-fat diet (HFD) fed mice. Clinical data support the regulation of circulating LEAP-2 by nutrient availability in humans. In this work, our primary objective was to examine the chronic effects of ghrelin and LEAP-2 administration on food intake, adiposity, and energy expenditure in young mice subjected to standard and HFD at both room temperature and at thermoneutrality. Furthermore, we aimed to assess the impact of these two hormones on aging mice. RESULTS Our results indicate that LEAP-2 produces a significant decrease of body weight and adiposity, an increase in energy expenditure, and activation of the thermogenic program in white and brown adipose tissue depots. However, this effect is not maintained under HFD or under thermoneutral conditions and is only partially observed in aging mice. CONCLUSION In summary our studies describe the central effects of LEAP-2 within distinct experimental contexts, and contribute to the comprehension of LEAP-2's role in energy metabolism.
Collapse
Affiliation(s)
- S Casado
- Departamento de Fisioloxía and Centro de Investigación en Medicina Molecular (CIMUS), Universidade de Santiago de Compostela, Instituto de Investigaciones Sanitarias de Santiago de Compostela (IDIS), 15782, Santiago de Compostela, Spain
| | - M Varela-Miguéns
- Departamento de Fisioloxía and Centro de Investigación en Medicina Molecular (CIMUS), Universidade de Santiago de Compostela, Instituto de Investigaciones Sanitarias de Santiago de Compostela (IDIS), 15782, Santiago de Compostela, Spain
| | - T de Oliveira Diz
- Departamento de Fisioloxía and Centro de Investigación en Medicina Molecular (CIMUS), Universidade de Santiago de Compostela, Instituto de Investigaciones Sanitarias de Santiago de Compostela (IDIS), 15782, Santiago de Compostela, Spain
| | - C Quintela-Vilariño
- Departamento de Fisioloxía and Centro de Investigación en Medicina Molecular (CIMUS), Universidade de Santiago de Compostela, Instituto de Investigaciones Sanitarias de Santiago de Compostela (IDIS), 15782, Santiago de Compostela, Spain
| | - R Nogueiras
- Departamento de Fisioloxía and Centro de Investigación en Medicina Molecular (CIMUS), Universidade de Santiago de Compostela, Instituto de Investigaciones Sanitarias de Santiago de Compostela (IDIS), 15782, Santiago de Compostela, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 28029, Madrid, Spain
| | - C Diéguez
- Departamento de Fisioloxía and Centro de Investigación en Medicina Molecular (CIMUS), Universidade de Santiago de Compostela, Instituto de Investigaciones Sanitarias de Santiago de Compostela (IDIS), 15782, Santiago de Compostela, Spain.
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 28029, Madrid, Spain.
| | - S Tovar
- Departamento de Fisioloxía and Centro de Investigación en Medicina Molecular (CIMUS), Universidade de Santiago de Compostela, Instituto de Investigaciones Sanitarias de Santiago de Compostela (IDIS), 15782, Santiago de Compostela, Spain.
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 28029, Madrid, Spain.
| |
Collapse
|
5
|
Bonifazi A, Ellenberger M, Farino ZJ, Aslanoglou D, Rais R, Pereira S, Mantilla-Rivas JO, Boateng CA, Eshleman AJ, Janowsky A, Hahn MK, Schwartz GJ, Slusher BS, Newman AH, Freyberg Z. Development of novel tools for dissection of central versus peripheral dopamine D 2-like receptor signaling in dysglycemia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.21.581451. [PMID: 38529497 PMCID: PMC10962703 DOI: 10.1101/2024.02.21.581451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/27/2024]
Abstract
Dopamine (DA) D2-like receptors in both the central nervous system (CNS) and the periphery are key modulators of metabolism. Moreover, disruption of D2-like receptor signaling is implicated in dysglycemia. Yet, the respective metabolic contributions of CNS versus peripheral D2-like receptors including D2 (D2R) and D3 (D3R) receptors remain poorly understood. To address this, we developed new pharmacological tools, D2-like receptor agonists with diminished and delayed blood-brain barrier capability, to selectively manipulate D2R/D3R signaling in the periphery. We designated bromocriptine methiodide (BrMeI), a quaternary methiodide analogue of D2/3R agonist and diabetes drug bromocriptine, as our lead compound based on preservation of D2R/D3R binding and functional efficacy. We then used BrMeI and unmodified bromocriptine to dissect relative contributions of CNS versus peripheral D2R/D3R signaling in treating dysglycemia. Systemic administration of bromocriptine, with unrestricted access to CNS and peripheral targets, significantly improved both insulin sensitivity and glucose tolerance in obese, dysglycemic mice in vivo. In contrast, metabolic improvements were attenuated when access to bromocriptine was restricted either to the CNS through intracerebroventricular administration or delayed access to the CNS via BrMeI. Our findings demonstrate that the coordinated actions of both CNS and peripheral D2-like receptors are required for correcting dysglycemia. Ultimately, the development of a first-generation of drugs designed to selectively target the periphery provides a blueprint for dissecting mechanisms of central versus peripheral DA signaling and paves the way for novel strategies to treat dysglycemia.
Collapse
Affiliation(s)
- Alessandro Bonifazi
- Medicinal Chemistry Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Michael Ellenberger
- Medicinal Chemistry Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Zachary J. Farino
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Rana Rais
- Department of Neurology, Johns Hopkins Drug Discovery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sandra Pereira
- Centre for Addiction and Mental Health, Toronto, ON, Canada
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | | | - Comfort A. Boateng
- Medicinal Chemistry Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Amy J. Eshleman
- Research Service, VA Portland Health Care System, Portland, Oregon, USA
- Departments of Behavioral Neuroscience and Psychiatry, Oregon Health & Science University, Portland, OR, USA
| | - Aaron Janowsky
- Research Service, VA Portland Health Care System, Portland, Oregon, USA
- Departments of Behavioral Neuroscience and Psychiatry, Oregon Health & Science University, Portland, OR, USA
- Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Margaret K. Hahn
- Centre for Addiction and Mental Health, Toronto, ON, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
- Department of Pharmacology, University of Toronto, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Banting & Best Diabetes Centre, Toronto, ON, Canada
| | - Gary J. Schwartz
- The Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, NY, USA
- Division of Endocrinology, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Barbara S. Slusher
- Department of Neurology, Johns Hopkins Drug Discovery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Amy Hauck Newman
- Medicinal Chemistry Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Zachary Freyberg
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA, USA
- Lead Contact
| |
Collapse
|
6
|
Browning KN. Colokinetic co-operation to control colorectal motility. J Physiol 2023; 601:5139-5140. [PMID: 37942673 PMCID: PMC11492826 DOI: 10.1113/jp285688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 10/27/2023] [Indexed: 11/10/2023] Open
Affiliation(s)
- Kirsteen N Browning
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, PA, USA
| |
Collapse
|
7
|
Ringuet MT, Koo A, Furness SGB, McDougall SJ, Furness JB. Sites and mechanisms of action of colokinetics at dopamine, ghrelin and serotonin receptors in the rodent lumbosacral defecation centre. J Physiol 2023; 601:5195-5211. [PMID: 37772438 PMCID: PMC10952827 DOI: 10.1113/jp285217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 09/13/2023] [Indexed: 09/30/2023] Open
Abstract
Agonists of dopamine D2 receptors (D2R), 5-hydroxytryptamine (5-HT, serotonin) receptors (5-HTR) and ghrelin receptors (GHSR) activate neurons in the lumbosacral defecation centre, and act as 'colokinetics', leading to increased propulsive colonic motility, in vivo. In the present study, we investigated which neurons in the lumbosacral defecation centre express the receptors and whether dopamine, serotonin and ghrelin receptor agonists act on the same lumbosacral preganglionic neurons (PGNs). We used whole cell electrophysiology to record responses from neurons in the lumbosacral defecation centre, following colokinetic application, and investigated their expression profiles and the chemistries of their neural inputs. Fluorescence in situ hybridisation revealed Drd2, Ghsr and Htr2C transcripts were colocalised in lumbosacral PGNs of mice, and immunohistochemistry showed that these neurons have closely associated tyrosine hydroxylase and 5-HT boutons. Previous studies showed that they do not receive ghrelin inputs. Whole cell electrophysiology in adult mice spinal cord revealed that dopamine, serotonin, α-methylserotonin and capromorelin each caused inward, excitatory currents in overlapping populations of lumbosacral PGNs. Furthermore, dopamine caused increased frequency of both IPSCs and EPSCs in a cohort of D2R neurons. Tetrodotoxin blocked the IPSCs and EPSCs, revealing a post-synaptic excitatory action of dopamine. In lumbosacral PGNs of postnatal day 7-14 rats, only dopamine's postsynaptic effects were observed. Furthermore, inward, excitatory currents evoked by dopamine were reduced by the GHSR antagonist, YIL781. We conclude that lumbosacral PGNs are the site where the action of endogenous ligands of D2R and 5-HT2R converge, and that GHSR act as a cis-modulator of D2R expressed by the same neurons. KEY POINTS: Dopamine, 5-hydroxytryptamine (5-HT, serotonin) and ghrelin (GHSR) receptor agonists increase colorectal motility and have been postulated to act at receptors on parasympathetic preganglionic neurons (PGNs) in the lumbosacral spinal cord. We aimed to determine which neurons in the lumbosacral spinal cord express dopamine, serotonin and GHSR receptors, their neural inputs, and whether agonists at these receptors excite them. We show that dopamine, serotonin and ghrelin receptor transcripts are contained in the same PGNs and that these neurons have closely associated tyrosine hydroxylase and serotonin boutons. Whole cell electrophysiology revealed that dopamine, serotonin and GHSR receptor agonists induce an inward excitatory current in overlapping populations of lumbosacral PGNs. Dopamine-induced excitation was reversed by GHSR antagonism. The present study demonstrates that lumbosacral PGNs are the site at which actions of endogenous ligands of dopamine D2 receptors and 5-HT type 2 receptors converge. Ghrelin receptors are functional, but their role appears to be as modulators of dopamine effects at D2 receptors.
Collapse
Affiliation(s)
- Mitchell T. Ringuet
- Department of Anatomy & PhysiologyUniversity of MelbourneMelbourneVICAustralia
| | - Ada Koo
- Department of Anatomy & PhysiologyUniversity of MelbourneMelbourneVICAustralia
| | - Sebastian G. B. Furness
- School of Biomedical SciencesUniversity of QueenslandBrisbaneQLDAustralia
- Monash Institute of Pharmaceutical SciencesMelbourneVICAustralia
| | - Stuart J. McDougall
- Florey Institute of Neuroscience and Mental HealthUniversity of MelbourneMelbourneVICAustralia
| | - John B. Furness
- Department of Anatomy & PhysiologyUniversity of MelbourneMelbourneVICAustralia
- Florey Institute of Neuroscience and Mental HealthUniversity of MelbourneMelbourneVICAustralia
| |
Collapse
|
8
|
Guidolin D, Tortorella C, Marcoli M, Cervetto C, De Caro R, Maura G, Agnati LF. Modulation of Neuron and Astrocyte Dopamine Receptors via Receptor-Receptor Interactions. Pharmaceuticals (Basel) 2023; 16:1427. [PMID: 37895898 PMCID: PMC10610355 DOI: 10.3390/ph16101427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 09/29/2023] [Accepted: 10/04/2023] [Indexed: 10/29/2023] Open
Abstract
Dopamine neurotransmission plays critical roles in regulating complex cognitive and behavioral processes including reward, motivation, reinforcement learning, and movement. Dopamine receptors are classified into five subtypes, widely distributed across the brain, including regions responsible for motor functions and specific areas related to cognitive and emotional functions. Dopamine also acts on astrocytes, which express dopamine receptors as well. The discovery of direct receptor-receptor interactions, leading to the formation of multimeric receptor complexes at the cell membrane and providing the cell decoding apparatus with flexible dynamics in terms of recognition and signal transduction, has expanded the knowledge of the G-protein-coupled receptor-mediated signaling processes. The purpose of this review article is to provide an overview of currently identified receptor complexes containing dopamine receptors and of their modulatory action on dopamine-mediated signaling between neurons and between neurons and astrocytes. Pharmacological possibilities offered by targeting receptor complexes in terms of addressing neuropsychiatric disorders associated with altered dopamine signaling will also be briefly discussed.
Collapse
Affiliation(s)
- Diego Guidolin
- Department of Neuroscience, University of Padova, 35122 Padova, Italy; (C.T.); (R.D.C.)
| | - Cinzia Tortorella
- Department of Neuroscience, University of Padova, 35122 Padova, Italy; (C.T.); (R.D.C.)
| | - Manuela Marcoli
- Department of Pharmacy, University of Genova, 16126 Genova, Italy; (M.M.); (C.C.); (G.M.)
| | - Chiara Cervetto
- Department of Pharmacy, University of Genova, 16126 Genova, Italy; (M.M.); (C.C.); (G.M.)
| | - Raffaele De Caro
- Department of Neuroscience, University of Padova, 35122 Padova, Italy; (C.T.); (R.D.C.)
| | - Guido Maura
- Department of Pharmacy, University of Genova, 16126 Genova, Italy; (M.M.); (C.C.); (G.M.)
| | - Luigi F. Agnati
- Department of Biomedical, Metabolic Sciences and Neuroscience, University of Modena and Reggio Emilia, 41121 Modena, Italy;
| |
Collapse
|
9
|
Geisler CE, Hayes MR. Metabolic hormone action in the VTA: Reward-directed behavior and mechanistic insights. Physiol Behav 2023; 268:114236. [PMID: 37178855 PMCID: PMC10330780 DOI: 10.1016/j.physbeh.2023.114236] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 04/10/2023] [Accepted: 05/10/2023] [Indexed: 05/15/2023]
Abstract
Dysfunctional signaling in midbrain reward circuits perpetuates diseases characterized by compulsive overconsumption of rewarding substances such as substance abuse, binge eating disorder, and obesity. Ventral tegmental area (VTA) dopaminergic activity serves as an index for how rewarding stimuli are perceived and triggers behaviors necessary to obtain future rewards. The evolutionary linking of reward with seeking and consuming palatable foods ensured an organism's survival, and hormone systems that regulate appetite concomitantly developed to regulate motivated behaviors. Today, these same mechanisms serve to regulate reward-directed behavior around food, drugs, alcohol, and social interactions. Understanding how hormonal regulation of VTA dopaminergic output alters motivated behaviors is essential to leveraging therapeutics that target these hormone systems to treat addiction and disordered eating. This review will outline our current understanding of the mechanisms underlying VTA action of the metabolic hormones ghrelin, glucagon-like peptide-1, amylin, leptin, and insulin to regulate behavior around food and drugs of abuse, highlighting commonalities and differences in how these five hormones ultimately modulate VTA dopamine signaling.
Collapse
Affiliation(s)
- Caroline E Geisler
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Matthew R Hayes
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
10
|
Wang L, Gao J, Cao X, Du J, Cao L, Nie Z, Xu G, Dong Z. Integrated Analysis of Transcriptomics and Metabolomics Unveil the Novel Insight of One-Year-Old Precocious Mechanism in the Chinese Mitten Crab, Eriocheir sinensis. Int J Mol Sci 2023; 24:11171. [PMID: 37446357 DOI: 10.3390/ijms241311171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 06/29/2023] [Accepted: 07/03/2023] [Indexed: 07/15/2023] Open
Abstract
Eriocheir sinensis is traditionally a native high-value crab that is widely distributed in eastern Asia, and the precocity is considered the bottleneck problem affecting the development of the industry. The precocious E. sinensis is defined as a crab that reaches complete sexual maturation during the first year of its lifespan rather than as normally in the second year. However, the exact regulatory mechanisms underlying the precocity are still unclear to date. This study is the first to explore the mechanism of precocity with transcriptome-metabolome association analysis between the precocious and normal sexually mature E. sinensis. Our results indicated that the phenylalanine metabolism (map00360) and neuroactive ligand-receptor interaction (map04080) pathways play an important role in the precocity in the ovary of E. sinensis. In map00360, the predicted aromatic-L-amino-acid decarboxylase and 4-hydroxyphenylpyruvate dioxygenase isoform X1 genes and the phenethylamine, phenylethyl alcohol, trans-2-hydroxycinnamate, and L-tyrosine metabolites were all down-regulated in the ovary of the precocious E. sinensis. The map04080 was the common KEGG pathway in the ovary and hepatopancreas between the precocious and normal crab. In the ovary, the predicted growth hormone secretagogue receptor type 1 gene was up-regulated, and the L-glutamate metabolite was down-regulated in the precocious E. sinensis. In the hepatopancreas, the predicted forkhead box protein I2 gene and taurine metabolite were up-regulated and the the L-glutamate metabolite was down-regulated in the precocious crab. There was no common pathway in the testis. Numerous common pathways in the hepatopancreas between male precocious and normal crab were identified. The specific amino acids, fatty acids and flavorful nucleotide (inosine monophosphate (MP), cytidine MP, adenosine MP, uridine MP, and guanosine MP) contents in the hepatopancreas and gonads further confirmed the above omics results. Our results suggest that the phenylalanine metabolism may affect the ovarian development by changing the contents of the neurotransmitter and tyrosine. The neuroactive ligand-receptor interaction pathway may affect the growth by changing the expressions of related genes and affect the umami taste of the gonads and hepatopancreas through the differences of L-glutamate metabolite in the precocious E. sinensis. The results provided valuable and novel insights on the precocious mechanism and may have a significant impact on the development of the E. sinensis aquaculture industry.
Collapse
Affiliation(s)
- Lanmei Wang
- Key Laboratory of Freshwater, Fisheries and Germplasm Resources Utilization, Freshwater Fisheries Research Centre of Chinese Academy of Fishery Sciences, Ministry of Agriculture and Rural Affairs, Wuxi 214081, China
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi 214081, China
| | - Jiancao Gao
- Key Laboratory of Freshwater, Fisheries and Germplasm Resources Utilization, Freshwater Fisheries Research Centre of Chinese Academy of Fishery Sciences, Ministry of Agriculture and Rural Affairs, Wuxi 214081, China
| | - Xi Cao
- College of Fisheries and Life Science, Shanghai Ocean University, Shanghai 201306, China
| | - Jinliang Du
- Key Laboratory of Freshwater, Fisheries and Germplasm Resources Utilization, Freshwater Fisheries Research Centre of Chinese Academy of Fishery Sciences, Ministry of Agriculture and Rural Affairs, Wuxi 214081, China
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi 214081, China
| | - Liping Cao
- Key Laboratory of Freshwater, Fisheries and Germplasm Resources Utilization, Freshwater Fisheries Research Centre of Chinese Academy of Fishery Sciences, Ministry of Agriculture and Rural Affairs, Wuxi 214081, China
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi 214081, China
| | - Zhijuan Nie
- Key Laboratory of Freshwater, Fisheries and Germplasm Resources Utilization, Freshwater Fisheries Research Centre of Chinese Academy of Fishery Sciences, Ministry of Agriculture and Rural Affairs, Wuxi 214081, China
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi 214081, China
| | - Gangchun Xu
- Key Laboratory of Freshwater, Fisheries and Germplasm Resources Utilization, Freshwater Fisheries Research Centre of Chinese Academy of Fishery Sciences, Ministry of Agriculture and Rural Affairs, Wuxi 214081, China
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi 214081, China
- College of Fisheries and Life Science, Shanghai Ocean University, Shanghai 201306, China
| | - Zaijie Dong
- Key Laboratory of Freshwater, Fisheries and Germplasm Resources Utilization, Freshwater Fisheries Research Centre of Chinese Academy of Fishery Sciences, Ministry of Agriculture and Rural Affairs, Wuxi 214081, China
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi 214081, China
- College of Fisheries and Life Science, Shanghai Ocean University, Shanghai 201306, China
| |
Collapse
|
11
|
Chen J, Wang Z, Zhang R, Yin H, Wang P, Wang C, Jiang Y. Heterodimerization of apelin and opioid receptor-like 1 receptors mediates apelin-13-induced G protein biased signaling. Life Sci 2023:121892. [PMID: 37364634 DOI: 10.1016/j.lfs.2023.121892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 06/17/2023] [Accepted: 06/23/2023] [Indexed: 06/28/2023]
Abstract
The apelin receptor (APJ) and the opioid-related nociceptin receptor 1 (ORL1) are family A G protein-coupled receptors that participate in a variety of physiological processes. The distribution and function of APJ and ORL1 in the nervous system and peripheral tissues are similar; however, the detailed mechanism of how these two receptors modulate signaling and physiological effects remains unclear. Here, we examined whether APJ and ORL1 form dimers, and investigated signal transduction pathways. The endogenous co-expression of APJ and ORL1 in SH-SY5Y cells was confirmed by western blotting and RT-PCR. Bioluminescence and fluorescence resonance energy transfer assays, as well as a proximity ligation assay and co-immunoprecipitation experiments, demonstrated that APJ and ORL1 heterodimerize in HEK293 cells. We found that the APJ-ORL1 heterodimer is selectively activated by apelin-13, which causes the dimer to couple to Gαi proteins and reduce the recruitment of GRKs and β-arrestins to the dimer. We showed that the APJ-ORL1 dimer exhibits biased signaling, in which G protein-dependent signaling pathways override β-arrestin-dependent signaling pathways. Our results demonstrate that the structural interface of the APJ-ORL1 dimer switches from transmembrane domain TM1/TM2 in the inactive state to TM5 in the active state. We used mutational analysis and BRET assays to identify key residues in TM5 (APJ L2185.55, APJ I2245.61, and ORL1 L2295.52) responsible for the receptor-receptor interaction. These results provide important information on the APJ-ORL1 heterodimer and may assist the design of new drugs targeting biased signaling pathways for treatment of pain and cardiovascular and metabolic diseases.
Collapse
Affiliation(s)
- Jing Chen
- Neurobiology Institute, Jining Medical University, Jining, China; Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, United Kingdom.
| | - Zhengwen Wang
- Neurobiology Institute, Jining Medical University, Jining, China
| | - Rumin Zhang
- Neurobiology Institute, Jining Medical University, Jining, China
| | - Haiyan Yin
- Neurobiology Institute, Jining Medical University, Jining, China
| | - Peixiang Wang
- Neurobiology Institute, Jining Medical University, Jining, China
| | - Chunmei Wang
- Neurobiology Institute, Jining Medical University, Jining, China
| | - Yunlu Jiang
- Neurobiology Institute, Jining Medical University, Jining, China.
| |
Collapse
|
12
|
Jin R, Sun S, Hu Y, Zhang H, Sun X. Neuropeptides Modulate Feeding via the Dopamine Reward Pathway. Neurochem Res 2023:10.1007/s11064-023-03954-4. [PMID: 37233918 DOI: 10.1007/s11064-023-03954-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 05/13/2023] [Accepted: 05/16/2023] [Indexed: 05/27/2023]
Abstract
Dopamine (DA) is a catecholamine neurotransmitter widely distributed in the central nervous system. It participates in various physiological functions, such as feeding, anxiety, fear, sleeping and arousal. The regulation of feeding is exceptionally complex, involving energy homeostasis and reward motivation. The reward system comprises the ventral tegmental area (VTA), nucleus accumbens (NAc), hypothalamus, and limbic system. This paper illustrates the detailed mechanisms of eight typical orexigenic and anorexic neuropeptides that regulate food intake through the reward system. According to recent literature, neuropeptides released from the hypothalamus and other brain regions regulate reward feeding predominantly through dopaminergic neurons projecting from the VTA to the NAc. In addition, their effect on the dopaminergic system is mediated by the prefrontal cortex, paraventricular thalamus, laterodorsal tegmental area, amygdala, and complex neural circuits. Research on neuropeptides involved in reward feeding can help identify more targets to treat diseases with metabolic disorders, such as obesity.
Collapse
Affiliation(s)
- Ruijie Jin
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
- Department of Clinical Medicine, Medical College, Qingdao University, Qingdao, China
| | - Shanbin Sun
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
- Department of Clinical Medicine, Medical College, Qingdao University, Qingdao, China
| | - Yang Hu
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
- Department of Clinical Medicine, Medical College, Qingdao University, Qingdao, China
| | - Hongfei Zhang
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
- Department of Clinical Medicine, Medical College, Qingdao University, Qingdao, China
| | - Xiangrong Sun
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China.
| |
Collapse
|
13
|
Guidolin D, Tortorella C, Marcoli M, Cervetto C, Maura G, Agnati LF. Receptor-receptor interactions and microvesicle exchange as mechanisms modulating signaling between neurons and astrocytes. Neuropharmacology 2023; 231:109509. [PMID: 36935005 DOI: 10.1016/j.neuropharm.2023.109509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 02/21/2023] [Accepted: 03/16/2023] [Indexed: 03/19/2023]
Abstract
It is well known that astrocytes play a significant metabolic role in the nervous tissue, maintaining the homeostasis of the extracellular space and of the blood-brain barrier, and providing trophic support to neurons. In addition, however, evidence exists indicating astrocytes as important elements for brain activity through signaling exchange with neurons. Astrocytes, indeed, can sense synaptic activity and their molecular machinery responds to neurotransmitters released by neurons with cytoplasmic Ca2+ elevations that, in turn, stimulate the release of neuroactive substances (gliotransmitters) influencing nearby neurons. In both cell types the recognition and transduction of this complex pattern of signals is mediated by specific receptors that are also involved in mechanisms tuning the intercellular cross-talk between astrocytes and neurons. Two of these mechanisms are the focus of the present discussion. The first concerns direct receptor-receptor interactions leading to the formation at the cell membrane of multimeric receptor complexes. The cooperativity that emerges in the actions of orthosteric and allosteric ligands of the monomers forming the assembly provides the cell decoding apparatus with sophisticated and flexible dynamics in terms of recognition and signal transduction pathways. A further mechanism of plasticity involving receptors is based on the transfer of elements of the cellular signaling apparatus via extracellular microvesicles acting as protective containers, which can lead to transient changes in the transmitting/decoding capabilities of the target cell.
Collapse
Affiliation(s)
- Diego Guidolin
- Department of Neuroscience, Section of Anatomy, University of Padova, 35121, Padova, Italy.
| | - Cinzia Tortorella
- Department of Neuroscience, Section of Anatomy, University of Padova, 35121, Padova, Italy
| | - Manuela Marcoli
- Department of Pharmacy, Center of Excellence for Biomedical Research, University of Genova, 16126, Genova, Italy
| | - Chiara Cervetto
- Department of Pharmacy, Center of Excellence for Biomedical Research, University of Genova, 16126, Genova, Italy
| | - Guido Maura
- Department of Pharmacy, Center of Excellence for Biomedical Research, University of Genova, 16126, Genova, Italy
| | - Luigi F Agnati
- Department of Biomedical Sciences, University of Modena and Reggio Emilia, 41125, Modena, Italy
| |
Collapse
|
14
|
Quinpirole ameliorates nigral dopaminergic neuron damage in Parkinson's disease mouse model through activating GHS-R1a/D 2R heterodimers. Acta Pharmacol Sin 2023:10.1038/s41401-023-01063-0. [PMID: 36899113 PMCID: PMC10374575 DOI: 10.1038/s41401-023-01063-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 02/12/2023] [Indexed: 03/12/2023] Open
Abstract
Growth hormone secretagogue receptor 1a (GHS-R1a) is an important G protein-coupled receptor (GPCR) that regulates a variety of functions by binding to ghrelin. It has been shown that the dimerization of GHS-R1a with other receptors also affects ingestion, energy metabolism, learning and memory. Dopamine type 2 receptor (D2R) is a GPCR mainly distributed in the ventral tegmental area (VTA), substantia nigra (SN), striatum and other brain regions. In this study we investigated the existence and function of GHS-R1a/D2R heterodimers in nigral dopaminergic neurons in Parkinson's disease (PD) models in vitro and in vivo. By conducting immunofluorescence staining, FRET and BRET analyses, we confirmed that GHS-R1a and D2R could form heterodimers in PC-12 cells and in the nigral dopaminergic neurons of wild-type mice. This process was inhibited by MPP+ or MPTP treatment. Application of QNP (10 μM) alone significantly increased the viability of MPP+-treated PC-12 cells, and administration of quinpirole (QNP, 1 mg/kg, i.p. once before and twice after MPTP injection) significantly alleviated motor deficits in MPTP-induced PD mice model; the beneficial effects of QNP were abolished by GHS-R1a knockdown. We revealed that the GHS-R1a/D2R heterodimers could increase the protein levels of tyrosine hydroxylase in the SN of MPTP-induced PD mice model through the cAMP response element binding protein (CREB) signaling pathway, ultimately promoting dopamine synthesis and release. These results demonstrate a protective role for GHS-R1a/D2R heterodimers in dopaminergic neurons, providing evidence for the involvement of GHS-R1a in PD pathogenesis independent of ghrelin.
Collapse
|
15
|
Bono F, Tomasoni Z, Mutti V, Sbrini G, Kumar R, Longhena F, Fiorentini C, Missale C. G Protein-Dependent Activation of the PKA-Erk1/2 Pathway by the Striatal Dopamine D1/D3 Receptor Heteromer Involves Beta-Arrestin and the Tyrosine Phosphatase Shp-2. Biomolecules 2023; 13:473. [PMID: 36979407 PMCID: PMC10046256 DOI: 10.3390/biom13030473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 02/23/2023] [Accepted: 03/01/2023] [Indexed: 03/08/2023] Open
Abstract
The heteromer composed of dopamine D1 and D3 receptors (D1R-D3R) has been defined as a structure able to trigger Erk1/2 and Akt signaling in a G protein-independent, beta-arrestin 1-dependent way that is physiologically expressed in the ventral striatum and is likely involved in the control of locomotor activity. Indeed, abnormal levels of D1R-D3R heteromer in the dorsal striatum have been correlated with the development of L-DOPA-induced dyskinesia (LID) in Parkinson's disease patients, a motor complication associated with striatal D1R signaling, thus requiring Gs protein and PKA activity to activate Erk1/2. Therefore, to clarify the role of the D1R/D3R heteromer in LID, we investigated the signaling pathway induced by the heteromer using transfected cells and primary mouse striatal neurons. Collectively, we found that in both the cell models, D1R/D3R heteromer-induced activation of Erk1/2 exclusively required the D1R molecular effectors, such as Gs protein and PKA, with the contribution of the phosphatase Shp-2 and beta-arrestins, indicating that heterodimerization with the D3R abolishes the specific D3R-mediated signaling but strongly allows D1R signals. Therefore, while in physiological conditions the D1R/D3R heteromer could represent a mechanism that strengthens the D1R activity, its pathological expression may contribute to the abnormal PKA-Shp-2-Erk1/2 pathway connected with LID.
Collapse
Affiliation(s)
- Federica Bono
- Section of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123 Brescia, Italy
| | - Zaira Tomasoni
- Section of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123 Brescia, Italy
| | - Veronica Mutti
- Section of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123 Brescia, Italy
| | - Giulia Sbrini
- Section of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123 Brescia, Italy
| | - Rajesh Kumar
- Seattle Children’s Research Institute, 1920 Terry Ave., Seattle, WA 98101, USA
| | - Francesca Longhena
- Section of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123 Brescia, Italy
| | - Chiara Fiorentini
- Section of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123 Brescia, Italy
| | - Cristina Missale
- Section of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123 Brescia, Italy
| |
Collapse
|
16
|
Zhu R, Tian P, Zhang H, Wang G, Chen W. Gut microbiome-brain interactions in anorexia nervosa: Potential mechanisms and regulatory strategies. Neuropharmacology 2023; 224:109315. [PMID: 36356938 DOI: 10.1016/j.neuropharm.2022.109315] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 09/29/2022] [Accepted: 10/30/2022] [Indexed: 11/09/2022]
Abstract
Anorexia nervosa (AN) is a psychiatric disorder characterised by malnutrition, fear of weight gain, and body image disturbances. The aetiology of AN is complex, and may involve environmental factors, genetic factors, and biochemical factors, with the latter meaning that AN may be closely associated with neurons, neurotransmitters, and hormones related to appetite and emotional regulation. In addition, an increasing number of studies have shown there is a link between the intestinal microbiota and psychiatric disorders, such as depression. However, few studies and reviews have focused on AN and gut microbes. Accordingly, in this review, we examine the potential pathogenesis of AN in terms of changes in the gut microbiota and its metabolites, and their effects on AN. The neurobiological function of the nervous system in relation to AN are also been mentioned. Furthermore, we suggest future research directions for this field, and note that probiotics may be developed for use as dietary supplements to help alleviate AN in patients.
Collapse
Affiliation(s)
- Ran Zhu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, PR China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Peijun Tian
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, PR China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, PR China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China; National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, 214122, China; (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou, 225004, China; Wuxi Translational Medicine Research Center and Jiangsu Translational Medicine Research Institute Wuxi Branch, Wuxi, Jiangsu, 214122, China
| | - Gang Wang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, PR China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China; National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, 214122, China; (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou, 225004, China.
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, PR China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China; Wuxi Translational Medicine Research Center and Jiangsu Translational Medicine Research Institute Wuxi Branch, Wuxi, Jiangsu, 214122, China
| |
Collapse
|
17
|
Ghrelin system in Alzheimer's disease. Curr Opin Neurobiol 2023; 78:102655. [PMID: 36527939 PMCID: PMC10395051 DOI: 10.1016/j.conb.2022.102655] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 11/16/2022] [Indexed: 12/23/2022]
Abstract
Alzheimer's disease (AD) is the most common type of dementia in seniors. Current efforts to understand the etiopathogenesis of this neurodegenerative disorder have brought forth questions about systemic factors in the development of AD. Ghrelin is a brain-gut peptide that is activated by ghrelin O-acyltransferase (GOAT) and signals via its receptor, growth hormone secretagogue receptor (GHSR). With increasing recognition of the neurotropic effects of ghrelin, the role of ghrelin system deregulation in the development of AD has been accentuated in recent years. In this review, we summarized recent research progress regarding the mechanisms of ghrelin signaling dysregulation and its contribution to AD brain pathology. In addition, we also discussed the therapeutic potential of strategies targeting ghrelin signaling for the treatment of this neurological disease.
Collapse
|
18
|
Tezenas-du-Montcel C, Tolle V. La régulation de la prise alimentaire au travers des actions antagonistes de la ghréline et du LEAP-2. CAHIERS DE NUTRITION ET DE DIÉTÉTIQUE 2023. [DOI: 10.1016/j.cnd.2022.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
|
19
|
Jerlhag E. Animal studies reveal that the ghrelin pathway regulates alcohol-mediated responses. Front Psychiatry 2023; 14:1050973. [PMID: 36970276 PMCID: PMC10030715 DOI: 10.3389/fpsyt.2023.1050973] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 02/21/2023] [Indexed: 03/29/2023] Open
Abstract
Alcohol use disorder (AUD) is often described as repeated phases of binge drinking, compulsive alcohol-taking, craving for alcohol during withdrawal, and drinking with an aim to a reduce the negative consequences. Although multifaceted, alcohol-induced reward is one aspect influencing the former three of these. The neurobiological mechanisms regulating AUD processes are complex and one of these systems is the gut-brain peptide ghrelin. The vast physiological properties of ghrelin are mediated via growth hormone secretagogue receptor (GHSR, ghrelin receptor). Ghrelin is well known for its ability to control feeding, hunger, and metabolism. Moreover, ghrelin signaling appears central for alcohol-mediated responses; findings reviewed herein. In male rodents GHSR antagonism reduces alcohol consumption, prevents relapse drinking, and attenuates the motivation to consume alcohol. On the other hand, ghrelin increases the consumption of alcohol. This ghrelin-alcohol interaction is also verified to some extent in humans with high alcohol consumption. In addition, either pharmacological or genetic suppression of GHSR decreases several alcohol-related effects (behavioral or neurochemical). Indeed, this suppression blocks the alcohol-induced hyperlocomotion and dopamine release in nucleus accumbens as well as ablates the alcohol reward in the conditioned place preference model. Although not fully elucidated, this interaction appears to involve areas central for reward, such as the ventral tegmental area (VTA) and brain nodes targeted by VTA projections. As reviewed briefly, the ghrelin pathway does not only modulate alcohol-mediated effects, it regulates reward-related behaviors induced by addictive drugs. Although personality traits like impulsivity and risk-taking behaviors are common in patients with AUD, the role of the ghrelin pathway thereof is unknown and remains to be studied. In summary, the ghrelin pathway regulates addiction processes like AUD and therefore the possibility that GHSR antagonism reduces alcohol or drug-taking should be explored in randomized clinical trials.
Collapse
|
20
|
Freyberg Z, Gittes GK. Roles of Pancreatic Islet Catecholamine Neurotransmitters in Glycemic Control and in Antipsychotic Drug-Induced Dysglycemia. Diabetes 2023; 72:3-15. [PMID: 36538602 PMCID: PMC9797319 DOI: 10.2337/db22-0522] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 09/24/2022] [Indexed: 12/24/2022]
Abstract
Catecholamine neurotransmitters dopamine (DA) and norepinephrine (NE) are essential for a myriad of functions throughout the central nervous system, including metabolic regulation. These molecules are also present in the pancreas, and their study may shed light on the effects of peripheral neurotransmission on glycemic control. Though sympathetic innervation to islets provides NE that signals at local α-cell and β-cell adrenergic receptors to modify hormone secretion, α-cells and β-cells also synthesize catecholamines locally. We propose a model where α-cells and β-cells take up catecholamine precursors in response to postprandial availability, preferentially synthesizing DA. The newly synthesized DA signals in an autocrine/paracrine manner to regulate insulin and glucagon secretion and maintain glycemic control. This enables islets to couple local catecholamine signaling to changes in nutritional state. We also contend that the DA receptors expressed by α-cells and β-cells are targeted by antipsychotic drugs (APDs)-some of the most widely prescribed medications today. Blockade of local DA signaling contributes significantly to APD-induced dysglycemia, a major contributor to treatment discontinuation and development of diabetes. Thus, elucidating the peripheral actions of catecholamines will provide new insights into the regulation of metabolic pathways and may lead to novel, more effective strategies to tune metabolism and treat diabetes.
Collapse
Affiliation(s)
- Zachary Freyberg
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA
| | - George K. Gittes
- Division of Pediatric Surgery, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
21
|
Borroto-Escuela DO, Cuesta-Marti C, Lopez-Salas A, Chruścicka-Smaga B, Crespo-Ramírez M, Tesoro-Cruz E, Palacios-Lagunas DA, Perez de la Mora M, Schellekens H, Fuxe K. The oxytocin receptor represents a key hub in the GPCR heteroreceptor network: potential relevance for brain and behavior. Front Mol Neurosci 2022; 15:1055344. [PMID: 36618821 PMCID: PMC9812438 DOI: 10.3389/fnmol.2022.1055344] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 11/11/2022] [Indexed: 12/24/2022] Open
Abstract
In the last 10 years, it has become increasingly clear that large numbers of axon collaterals extend from the oxytocin (OXT) hypothalamic axons, especially the parvocellular components, to other brain regions. Consequently, the OXT signaling system forms, like other monoamine axons, a rich functional network across several brain regions. In this manuscript, we review the recently indicated higher order G-protein coupled heteroreceptor complexes of the oxytocin receptor (OXTR), and how these, via allosteric receptor-receptor interactions modulate the recognition, signaling, and trafficking of the participating receptor protomers and their potential impact for brain and behavior. The major focus will be on complexes of the OXTR protomer with the dopamine D2 receptor (D2R) protomer and the serotonin 2A (5-HT2AR) and 2C (5-HT2CR) receptor protomers. Specifically, the existence of D2R-OXTR heterocomplexes in the nucleus accumbens and the caudate putamen of rats has led to a postulated function for this heteromer in social behavior. Next, a physical interaction between OXTRs and the growth hormone secretagogue or ghrelin receptor (GHS-R1a) was demonstrated, which consequently was able to attenuate OXTR-mediated Gαq signaling. This highlights the potential of ghrelin-targeted therapies to modulate oxytocinergic signaling with relevance for appetite regulation, anxiety, depression, and schizophrenia. Similarly, evidence for 5-HT2AR-OXTR heteromerization in the pyramidal cell layer of CA2 and CA3 in the dorsal hippocampus and in the nucleus accumbens shell was demonstrated. This complex may offer new strategies for the treatment of both mental disease and social behavior. Finally, the 5-HT2CR-OXTR heterocomplexes were demonstrated in the CA1, CA2, and CA3 regions of the dorsal hippocampus. Future work should be done to investigate the precise functional consequence of region-specific OXTR heteromerization in the brain, as well across the periphery, and whether the integration of neuronal signals in the brain may also involve higher order OXTR-GHS-R1a heteroreceptor complexes including the dopamine (DA), noradrenaline (NA) or serotonin (5-HT) receptor protomers or other types of G-protein coupled receptors (GPCRs).
Collapse
Affiliation(s)
- Dasiel O. Borroto-Escuela
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden,Receptomics and Brain Disorders Lab, Department of Human Physiology, Faculty of Medicine, University of Malaga, Málaga, Spain,Department of Biomolecular Science, Section of Morphology, Physiology and Environmental Biology, University of Urbino, Urbino, Italy,*Correspondence: Dasiel O. Borroto-Escuela Harriët Schellekens
| | - Cristina Cuesta-Marti
- APC Microbiome Ireland, University College CorkCork, Ireland,Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Alexander Lopez-Salas
- Receptomics and Brain Disorders Lab, Department of Human Physiology, Faculty of Medicine, University of Malaga, Málaga, Spain
| | | | - Minerva Crespo-Ramírez
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Emiliano Tesoro-Cruz
- Unidad de Investigación Biomédica en Inmunología e Infectología, Hospital de Infectología, Centro Médico Nacional La Raza, IMSS, Ciudad de México, Mexico
| | | | - Miguel Perez de la Mora
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Harriët Schellekens
- APC Microbiome Ireland, University College CorkCork, Ireland,Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland,*Correspondence: Dasiel O. Borroto-Escuela Harriët Schellekens
| | - Kjell Fuxe
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
22
|
Reich N, Hölscher C. Beyond Appetite: Acylated Ghrelin As A Learning, Memory and Fear Behavior-modulating Hormone. Neurosci Biobehav Rev 2022; 143:104952. [DOI: 10.1016/j.neubiorev.2022.104952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 04/27/2022] [Accepted: 11/05/2022] [Indexed: 11/10/2022]
|
23
|
Caniceiro AB, Bueschbell B, Schiedel AC, Moreira IS. Class A and C GPCR Dimers in Neurodegenerative Diseases. Curr Neuropharmacol 2022; 20:2081-2141. [PMID: 35339177 PMCID: PMC9886835 DOI: 10.2174/1570159x20666220327221830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 02/21/2022] [Accepted: 03/23/2022] [Indexed: 11/22/2022] Open
Abstract
Neurodegenerative diseases affect over 30 million people worldwide with an ascending trend. Most individuals suffering from these irreversible brain damages belong to the elderly population, with onset between 50 and 60 years. Although the pathophysiology of such diseases is partially known, it remains unclear upon which point a disease turns degenerative. Moreover, current therapeutics can treat some of the symptoms but often have severe side effects and become less effective in long-term treatment. For many neurodegenerative diseases, the involvement of G proteincoupled receptors (GPCRs), which are key players of neuronal transmission and plasticity, has become clearer and holds great promise in elucidating their biological mechanism. With this review, we introduce and summarize class A and class C GPCRs, known to form heterodimers or oligomers to increase their signalling repertoire. Additionally, the examples discussed here were shown to display relevant alterations in brain signalling and had already been associated with the pathophysiology of certain neurodegenerative diseases. Lastly, we classified the heterodimers into two categories of crosstalk, positive or negative, for which there is known evidence.
Collapse
Affiliation(s)
- Ana B. Caniceiro
- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; ,These authors contributed equally to this work.
| | - Beatriz Bueschbell
- PhD Programme in Experimental Biology and Biomedicine, Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Casa Costa Alemão, 3030-789 Coimbra, Portugal; ,These authors contributed equally to this work.
| | - Anke C. Schiedel
- Department of Pharmaceutical & Medicinal Chemistry, Pharmaceutical Institute, University of Bonn, D-53121 Bonn, Germany;
| | - Irina S. Moreira
- University of Coimbra, Department of Life Sciences, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal; ,Center for Neuroscience and Cell Biology, Center for Innovative Biomedicine and Biotechnology, 3004-504 Coimbra, Portugal,Address correspondence to this author at the Center for Neuroscience and Cell Biology, Center for Innovative Biomedicine and Biotechnology, 3004-504 Coimbra, Portugal; E-mail:
| |
Collapse
|
24
|
Ringuet MT, Furness JB, Furness SGB. G protein-coupled receptor interactions and modification of signalling involving the ghrelin receptor, GHSR1a. J Neuroendocrinol 2022; 34:e13077. [PMID: 34931385 DOI: 10.1111/jne.13077] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 12/05/2021] [Indexed: 12/28/2022]
Abstract
The growth hormone secretagogue receptor 1a (GHSR1a) is intriguing because of its potential as a therapeutic target and its diverse molecular interactions. Initial studies of the receptor focused on the potential therapeutic ability for growth hormone (GH) release to reduce wasting in aging individuals, as well as food intake regulation for treatment of cachexia. Known roles of GHSR1a now extend to regulation of neurogenesis, learning and memory, gastrointestinal motility, glucose/lipid metabolism, the cardiovascular system, neuronal protection, motivational salience, and hedonic feeding. Ghrelin, the endogenous agonist of GHSR1a, is primarily located in the stomach and is absent from the central nervous system (CNS), including the spinal cord. However, ghrelin in the circulation does have access to a small number of CNS sites, including the arcuate nucleus, which is important in feeding control. At some sites, such as at somatotrophs, GHSR1a has high constitutive activity. Typically, ghrelin-dependent and constitutive GHSR1a activation occurs via Gαq/11 pathways. In vitro and in vivo data suggest that GHSR1a heterodimerises with multiple G protein-coupled receptors (GPCRs), including dopamine D1 and D2, serotonin 2C, orexin, oxytocin and melanocortin 3 receptors (MCR3), as well as the MCR3 accessory protein, MRAP2, providing possible mechanisms for its many physiological effects. In all cases, the receptor interaction changes downstream signalling and the responses to receptor agonists. This review discusses the signalling mechanisms of GHSR1a alone and in combination with other GPCRs, and explores the physiological consequences of GHSR1a coupling with other GPCRs.
Collapse
Affiliation(s)
- Mitchell Ty Ringuet
- Department of Anatomy and Physiology, University of Melbourne, Parkville, VIC, Australia
| | - John Barton Furness
- Department of Anatomy and Physiology, University of Melbourne, Parkville, VIC, Australia
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
| | | |
Collapse
|
25
|
Involvement of the ghrelin system in the maintenance and reinstatement of cocaine-motivated behaviors: a role of adrenergic action at peripheral β1 receptors. Neuropsychopharmacology 2022; 47:1449-1460. [PMID: 34923576 PMCID: PMC9206024 DOI: 10.1038/s41386-021-01249-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 11/22/2021] [Accepted: 11/28/2021] [Indexed: 12/30/2022]
Abstract
Cocaine addiction is a significant medical and public concern. Despite decades of research effort, development of pharmacotherapy for cocaine use disorder remains largely unsuccessful. This may be partially due to insufficient understanding of the complex biological mechanisms involved in the pathophysiology of this disorder. In the present study, we show that: (1) elevation of ghrelin by cocaine plays a critical role in maintenance of cocaine self-administration and cocaine-seeking motivated by cocaine-conditioned stimuli; (2) acquisition of cocaine-taking behavior is associated with the acquisition of stimulatory effects of cocaine by cocaine-conditioned stimuli on ghrelin secretion, and with an upregulation of ghrelin receptor mRNA levels in the ventral tegmental area (VTA); (3) blockade of ghrelin signaling by pretreatment with JMV2959, a selective ghrelin receptor antagonist, dose-dependently inhibits reinstatement of cocaine-seeking triggered by either cocaine or yohimbine in behaviorally extinguished animals with a history of cocaine self-administration; (4) JMV2959 pretreatment also inhibits brain stimulation reward (BSR) and cocaine-potentiated BSR maintained by optogenetic stimulation of VTA dopamine neurons in DAT-Cre mice; (5) blockade of peripheral adrenergic β1 receptors by atenolol potently attenuates the elevation in circulating ghrelin induced by cocaine and inhibits cocaine self-administration and cocaine reinstatement triggered by cocaine. These findings demonstrate that the endogenous ghrelin system plays an important role in cocaine-related addictive behaviors and suggest that manipulating and targeting this system may be viable for mitigating cocaine use disorder.
Collapse
|
26
|
Piper NBC, Whitfield EA, Stewart GD, Xu X, Furness SGB. Targeting appetite and satiety in diabetes and obesity, via G protein-coupled receptors. Biochem Pharmacol 2022; 202:115115. [PMID: 35671790 DOI: 10.1016/j.bcp.2022.115115] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 05/26/2022] [Accepted: 05/27/2022] [Indexed: 11/17/2022]
Abstract
Type 2 diabetes and obesity have reached pandemic proportions throughout the world, so much so that the World Health Organisation coined the term "Globesity" to help encapsulate the magnitude of the problem. G protein-coupled receptors (GPCRs) are highly tractable drug targets due to their wide involvement in all aspects of physiology and pathophysiology, indeed, GPCRs are the targets of approximately 30% of the currently approved drugs. GPCRs are also broadly involved in key physiologies that underlie type 2 diabetes and obesity including feeding reward, appetite and satiety, regulation of blood glucose levels, energy homeostasis and adipose function. Despite this, only two GPCRs are the target of approved pharmaceuticals for treatment of type 2 diabetes and obesity. In this review we discuss the role of these, and select other candidate GPCRs, involved in various facets of type 2 diabetic or obese pathophysiology, how they might be targeted and the potential reasons why pharmaceuticals against these targets have not progressed to clinical use. Finally, we provide a perspective on the current development pipeline of anti-obesity drugs that target GPCRs.
Collapse
Affiliation(s)
- Noah B C Piper
- Receptor Transducer Coupling Laboratory, School of Biomedical Sciences, Faculty of Medicine, University of Queensland, St. Lucia, QLD 4072, Australia
| | - Emily A Whitfield
- Receptor Transducer Coupling Laboratory, School of Biomedical Sciences, Faculty of Medicine, University of Queensland, St. Lucia, QLD 4072, Australia
| | - Gregory D Stewart
- Drug Discovery Biology Laboratory, Monash Institute of Pharmaceutical Sciences & Department of Pharmacology Monash University, Parkville, VIC 3052, Australia
| | - Xiaomeng Xu
- Drug Discovery Biology Laboratory, Monash Institute of Pharmaceutical Sciences & Department of Pharmacology Monash University, Parkville, VIC 3052, Australia
| | - Sebastian G B Furness
- Receptor Transducer Coupling Laboratory, School of Biomedical Sciences, Faculty of Medicine, University of Queensland, St. Lucia, QLD 4072, Australia; Drug Discovery Biology Laboratory, Monash Institute of Pharmaceutical Sciences & Department of Pharmacology Monash University, Parkville, VIC 3052, Australia.
| |
Collapse
|
27
|
Prins K, Huisman M, McLuskey A, Mies R, Karels B, Delhanty PJD, Visser JA. Ghrelin deficiency sex-dependently affects food intake, locomotor activity, and adipose and hepatic gene expression in a binge-eating mouse model. Am J Physiol Endocrinol Metab 2022; 322:E494-E507. [PMID: 35403437 DOI: 10.1152/ajpendo.00432.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Binge-eating disorder is the most prevalent eating disorder diagnosed, affecting three times more women than men. Ghrelin stimulates appetite and reward signaling, and loss of its receptor reduces binge-eating behavior in male mice. Here, we examined the influence of ghrelin itself on binge-eating behavior in both male and female mice. Five-wk-old wild-type (WT) and ghrelin-deficient (Ghrl-/-) mice were housed individually in indirect calorimetry cages for 9 wks. Binge-like eating was induced by giving mice ad libitum chow, but time-restricted access to a Western-style diet (WD; 2 h access, 3 days/wk) in the light phase (BE); control groups received ad libitum chow (CO), or ad libitum access to both diets (CW). All groups of BE mice showed binge-eating behavior, eating up to 60% of their 24-h intake during the WD access period. Subsequent dark phase chow intake was decreased in Ghrl-/- mice and remained decreased in Ghrl-/- females on nonbinge days. Also, nonbinge day locomotor activity was lower in Ghrl-/- than in WT BE females. Upon euthanasia, Ghrl-/- BE mice weighed less and had a lower lean body mass percentage than WT BE mice. In BE and CW groups, ghrelin and sex altered the expression of genes involved in lipid processing, thermogenesis, and aging in white adipose tissue and livers. We conclude that, although ghrelin deficiency does not hamper the development of binge-like eating, it sex-dependently alters food intake timing, locomotor activity, and metabolism. These results add to the growing body of evidence that ghrelin signaling is sexually dimorphic.NEW & NOTEWORTHY Ghrelin, a peptide hormone secreted from the gut, is involved in hunger and reward signaling, which are altered in binge-eating disorder. Although sex differences have been described in both binge-eating and ghrelin signaling, this interaction has not been fully elucidated. Here, we show that ghrelin deficiency affects the behavior and metabolism of mice in a binge-like eating paradigm, and that the sex of the mice impacts the magnitude and direction of these effects.
Collapse
Affiliation(s)
- Karina Prins
- Department of Internal Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Martin Huisman
- Department of Internal Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Anke McLuskey
- Department of Internal Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Rosinda Mies
- Department of Internal Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Bas Karels
- Department of Internal Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Patric J D Delhanty
- Department of Internal Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Jenny A Visser
- Department of Internal Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
28
|
Guidolin D, Tortorella C, Marcoli M, Maura G, Agnati LF. Intercellular Communication in the Central Nervous System as Deduced by Chemical Neuroanatomy and Quantitative Analysis of Images: Impact on Neuropharmacology. Int J Mol Sci 2022; 23:5805. [PMID: 35628615 PMCID: PMC9145073 DOI: 10.3390/ijms23105805] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/19/2022] [Accepted: 05/20/2022] [Indexed: 01/25/2023] Open
Abstract
In the last decades, new evidence on brain structure and function has been acquired by morphological investigations based on synergic interactions between biochemical anatomy approaches, new techniques in microscopy and brain imaging, and quantitative analysis of the obtained images. This effort produced an expanded view on brain architecture, illustrating the central nervous system as a huge network of cells and regions in which intercellular communication processes, involving not only neurons but also other cell populations, virtually determine all aspects of the integrative function performed by the system. The main features of these processes are described. They include the two basic modes of intercellular communication identified (i.e., wiring and volume transmission) and mechanisms modulating the intercellular signaling, such as cotransmission and allosteric receptor-receptor interactions. These features may also open new possibilities for the development of novel pharmacological approaches to address central nervous system diseases. This aspect, with a potential major impact on molecular medicine, will be also briefly discussed.
Collapse
Affiliation(s)
- Diego Guidolin
- Department of Neuroscience, Section of Anatomy, University of Padova, 35121 Padova, Italy;
| | - Cinzia Tortorella
- Department of Neuroscience, Section of Anatomy, University of Padova, 35121 Padova, Italy;
| | - Manuela Marcoli
- Department of Pharmacy, Center of Excellence for Biomedical Research, University of Genova, 16126 Genova, Italy; (M.M.); (G.M.)
| | - Guido Maura
- Department of Pharmacy, Center of Excellence for Biomedical Research, University of Genova, 16126 Genova, Italy; (M.M.); (G.M.)
| | - Luigi F. Agnati
- Department of Biomedical Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy;
| |
Collapse
|
29
|
Péraldi-Roux S, Bayle M, M'Kadmi C, Damian M, Vaillé J, Fernandez G, Paula Cornejo M, Marie J, Banères JL, Ben Haj Salah K, Fehrentz JA, Cantel S, Perello M, Denoyelle S, Oiry C, Neasta J. Design and Characterization of a Triazole-Based Growth Hormone Secretagogue Receptor Modulator Inhibiting the Glucoregulatory and Feeding Actions of Ghrelin. Biochem Pharmacol 2022; 202:115114. [DOI: 10.1016/j.bcp.2022.115114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/26/2022] [Accepted: 05/26/2022] [Indexed: 11/02/2022]
|
30
|
Chen X, Dong J, Jiao Q, Du X, Bi M, Jiang H. "Sibling" battle or harmony: crosstalk between nesfatin-1 and ghrelin. Cell Mol Life Sci 2022; 79:169. [PMID: 35239020 PMCID: PMC11072372 DOI: 10.1007/s00018-022-04193-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 02/02/2022] [Accepted: 02/04/2022] [Indexed: 12/17/2022]
Abstract
Ghrelin was first identified as an endogenous ligand of the growth hormone secretagogue receptor (GHSR) in 1999, with the function of stimulating the release of growth hormone (GH), while nesfatin-1 was identified in 2006. Both peptides are secreted by the same kind of endocrine cells, X/A-like cells in the stomach. Compared with ghrelin, nesfatin-1 exerts opposite effects on energy metabolism, glucose metabolism, gastrointestinal functions and regulation of blood pressure, but exerts similar effects on anti-inflammation and neuroprotection. Up to now, nesfatin-1 remains as an orphan ligand because its receptor has not been identified. Several studies have shown the effects of nesfatin-1 are dependent on the receptor of ghrelin. We herein compare the effects of nesfatin-1 and ghrelin in several aspects and explore the possibility of their interactions.
Collapse
Affiliation(s)
- Xi Chen
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, People's Republic of China
| | - Jing Dong
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, People's Republic of China
| | - Qian Jiao
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, People's Republic of China
| | - Xixun Du
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, People's Republic of China
| | - Mingxia Bi
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, People's Republic of China
| | - Hong Jiang
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, People's Republic of China.
| |
Collapse
|
31
|
Gupta S, Mukhopadhyay S, Mitra A. Therapeutic potential of GHSR-1A antagonism in alcohol dependence, a review. Life Sci 2022; 291:120316. [PMID: 35016882 DOI: 10.1016/j.lfs.2022.120316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 01/05/2022] [Accepted: 01/05/2022] [Indexed: 11/28/2022]
Abstract
Growth hormone secretagogue receptor type 1A (GHSR-1A) is a functional receptor of orexigenic peptide ghrelin and is highly expressed in mesolimbic dopaminergic systems that regulate incentive value of artificial reward in substance abuse. Interestingly, GHSR-1A has also shown ligand-independent constitutive activity. Alcohol use disorder (AUD) is one of the growing concerns worldwide as it involves complex neuro-psycho-endocrinological interactions. Positive correlation of acylated ghrelin and alcohol-induced human brain response in the right and left ventral striatum are evident. In the last decade, the beneficial effects of ghrelin receptor (GHSR-1A) antagonism to suppress artificial reward circuitries and induce self-control for alcohol consumption have drawn significant attention from researchers. In this updated review, we summarize the available recent preclinical, clinical, and experimental data to discuss functional, molecular actions of central ghrelin-GHSR-1A signaling in different craving levels for alcohol as well as to promote "GHSR-1A antagonism" as one of the potential therapies in early abstinence.
Collapse
Affiliation(s)
- Shreyasi Gupta
- Department of Zoology, Triveni Devi Bhalotia College, Raniganj, Paschim Bardhaman 713 347, West Bengal, India
| | - Sanchari Mukhopadhyay
- Department of Psychiatry, National Institute of Mental Health and Neurosciences, Hombegowda Nagar, Bengaluru 560029, India
| | - Arkadeep Mitra
- Department of Zoology, City College, 102/1, Raja Rammohan Sarani, Kolkata 700 009, West Bengal, India.
| |
Collapse
|
32
|
Dezaki K, Yada T. Status of ghrelin as an islet hormone and paracrine/autocrine regulator of insulin secretion. Peptides 2022; 148:170681. [PMID: 34728253 DOI: 10.1016/j.peptides.2021.170681] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 10/06/2021] [Accepted: 10/28/2021] [Indexed: 12/20/2022]
Abstract
Ghrelin is expressed in the pancreatic islet cells as well as the stomach. In the perfused pancreas and isolated islets, GHS-R antagonism, ghrelin immunoneutralization and ghrelin-knockout (Ghr-KO) all increase glucose-induced insulin release. Thus, pharmacological, immunological and genetic blockades of ghrelin in the pancreatic islets all markedly augment glucose-induced insulin release, showing that islet-derived ghrelin physiologically restricts insulin release in rodents. In this review, we focus on the current understanding of the following key questions: 1) from which islet cells ghrelin is released, 2) on which islet cells ghrelin acts, and 3) mechanisms by which the islet-derived ghrelin inhibits insulin secretion.
Collapse
Affiliation(s)
- Katsuya Dezaki
- Iryo Sosei University, Faculty of Pharmacy, 5-5-1, Chuodai Iino, Iwaki, Fukushima, 970-8551, Japan.
| | - Toshihiko Yada
- Kansai Electric Power Medical Research Institute, Center for Integrative Physiology, Division of Integrative Physiology, 1-5-6 Minatojimaminamimachi, Chuo-ku, Kobe, 650-0047, Japan; Division of Diabetes, Metabolism and Endocrinology, Kobe University Graduate School of Medicine, 7-5-2 Kusunoki-Cho, Chuo-Ku, Kobe, 650-0017, Japan.
| |
Collapse
|
33
|
Tufvesson-Alm M, Shevchouk OT, Jerlhag E. Insight into the role of the gut-brain axis in alcohol-related responses: Emphasis on GLP-1, amylin, and ghrelin. Front Psychiatry 2022; 13:1092828. [PMID: 36699502 PMCID: PMC9868418 DOI: 10.3389/fpsyt.2022.1092828] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 12/15/2022] [Indexed: 01/11/2023] Open
Abstract
Alcohol use disorder (AUD) contributes substantially to global morbidity and mortality. Given the heterogenicity of this brain disease, available pharmacological treatments only display efficacy in sub-set of individuals. The need for additional treatment options is thus substantial and is the goal of preclinical studies unraveling neurobiological mechanisms underlying AUD. Although these neurobiological processes are complex and numerous, one system gaining recent attention is the gut-brain axis. Peptides of the gut-brain axis include anorexigenic peptide like glucagon-like peptide-1 (GLP-1) and amylin as well as the orexigenic peptide ghrelin. In animal models, agonists of the GLP-1 or amylin receptor and ghrelin receptor (GHSR) antagonists reduce alcohol drinking, relapse drinking, and alcohol-seeking. Moreover, these three gut-brain peptides modulate alcohol-related responses (behavioral and neurochemical) in rodents, suggesting that the alcohol reduction may involve a suppression of alcohol's rewarding properties. Brain areas participating in the ability of these gut-brain peptides to reduce alcohol-mediated behaviors/neurochemistry involve those important for reward. Human studies support these preclinical studies as polymorphisms of the genes encoding for GLP-1 receptor or the ghrelin pathway are associated with AUD. Moreover, a GLP-1 receptor agonist decreases alcohol drinking in overweight patients with AUD and an inverse GHSR agonist reduces alcohol craving. Although preclinical and clinical studies reveal an interaction between the gut-brain axis and AUD, additional studies should explore this in more detail.
Collapse
Affiliation(s)
- Maximilian Tufvesson-Alm
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Olesya T Shevchouk
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Elisabet Jerlhag
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
34
|
Labarthe A, Zizzari P, Fiquet O, Lebrun N, Veldhuis JD, Roelfsema F, Chauveau C, Bohlooly-Y M, Epelbaum J, Tolle V. Effect of Growth Hormone Secretagogue Receptor Deletion on Growth, Pulsatile Growth Hormone Secretion, and Meal Pattern in Male and Female Mice. Neuroendocrinology 2022; 112:215-234. [PMID: 33774644 DOI: 10.1159/000516147] [Citation(s) in RCA: 6] [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] [Received: 11/03/2020] [Accepted: 03/25/2021] [Indexed: 11/19/2022]
Abstract
INTRODUCTION While the vast majority of research investigating the role of ghrelin or its receptor, GHS-R1a, in growth, feeding, and metabolism has been conducted in male rodents, very little is known about sex differences in this system. Furthermore, the role of GHS-R1a signaling in the control of pulsatile GH secretion and its link with growth or metabolic parameters has never been characterized. METHODS We assessed the sex-specific contribution of GHS-R1a signaling in the activity of the GH/IGF-1 axis, metabolic parameters, and feeding behavior in adolescent (5-6 weeks old) or adult (10-19 weeks old) GHS-R KO (Ghsr-/-) and WT (Ghsr+/+) male and female mice. RESULTS Adult Ghsr-/- male and female mice displayed deficits in weight and linear growth that were correlated with reduced GH pituitary contents in males only. GHS-R1a deletion was associated with reduced meal frequency and increased meal intervals, as well as reduced hypothalamic GHRH and NPY mRNA in males, not females. In adult, GH release from Ghsr-/- mice pituitary explants ex vivo was reduced independently of the sex. However, in vivo pulsatile GH secretion decreased in adult but not adolescent Ghsr-/- females, while in males, GHS-R1a deletion was associated with reduction in pulsatile GH secretion during adolescence exclusively. In males, linear growth did not correlate with pulsatile GH secretion, but rather with ApEn, a measure that reflects irregularity of the rhythmic secretion. Fat mass, plasma leptin concentrations, or ambulatory activity did not predict differences in GH secretion. DISCUSSION/CONCLUSION These results point to a sex-dependent dimorphic effect of GHS-R1a signaling to modulate pulsatile GH secretion and meal pattern in mice with different compensatory mechanisms occurring in the hypothalamus of adult males and females after GHS-R1a deletion. Altogether, we show that GHS-R1a signaling plays a more critical role in the regulation of pulsatile GH secretion during adolescence in males and adulthood in females.
Collapse
Affiliation(s)
- Alexandra Labarthe
- Université de Paris, UMRS_1266 INSERM, Institute of Psychiatry and Neuroscience of Paris, Paris, France
| | - Philippe Zizzari
- Université de Paris, UMRS_1266 INSERM, Institute of Psychiatry and Neuroscience of Paris, Paris, France
| | - Oriane Fiquet
- Université de Paris, UMRS_1266 INSERM, Institute of Psychiatry and Neuroscience of Paris, Paris, France
| | - Nicolas Lebrun
- Université de Paris, UMRS_1266 INSERM, Institute of Psychiatry and Neuroscience of Paris, Paris, France
| | - Johannes D Veldhuis
- Endocrine Research Unit, Department of Medicine, Mayo School of Graduate Medical Education, Clinical Translational Science Center, Mayo Clinic, Rochester, New York, USA
| | - Ferdinand Roelfsema
- Department of Internal Medicine, Section of Endocrinology and Metabolism, Leiden University Medical Center, Leiden, The Netherlands
| | - Christophe Chauveau
- Marrow Adiposity and Bone Lab - MABLab ULR 4490, University Littoral Côte d'Opale, Boulogne-sur-Mer, France
- University Lille, CHU Lille, Lille, France
| | - Mohammad Bohlooly-Y
- Translational Genomics, Discovery Sciences, Biopharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Jacques Epelbaum
- Université de Paris, UMRS_1266 INSERM, Institute of Psychiatry and Neuroscience of Paris, Paris, France
- UMR CNRS/MNHN 7179, Mécanismes Adaptatifs et Evolution, Brunoy, France
| | - Virginie Tolle
- Université de Paris, UMRS_1266 INSERM, Institute of Psychiatry and Neuroscience of Paris, Paris, France
| |
Collapse
|
35
|
Deschaine SL, Leggio L. From "Hunger Hormone" to "It's Complicated": Ghrelin Beyond Feeding Control. Physiology (Bethesda) 2022; 37:5-15. [PMID: 34964687 PMCID: PMC8742734 DOI: 10.1152/physiol.00024.2021] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Discovered as a peptide involved in releasing growth hormone, ghrelin was initially characterized as the "hunger hormone." However, emerging research indicates that ghrelin appears to play an important part in relaying information regarding nutrient availability and value and adjusting physiological and motivational processes accordingly. These functions make ghrelin an interesting therapeutic candidate for metabolic and neuropsychiatric diseases involving disrupted nutrition that can further potentiate the rewarding effect of maladaptive behaviors.
Collapse
Affiliation(s)
- Sara L. Deschaine
- 1Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Baltimore and Bethesda, Maryland
| | - Lorenzo Leggio
- 1Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Baltimore and Bethesda, Maryland,2Medication Development Program, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, Maryland,3Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, School of Public Health, Brown University, Providence, Rhode Island,4Division of Addiction Medicine, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland,5Department of Neuroscience, Georgetown University Medical Center, Washington, District of Columbia
| |
Collapse
|
36
|
Shevchouk OT, Tufvesson-Alm M, Jerlhag E. An Overview of Appetite-Regulatory Peptides in Addiction Processes; From Bench to Bed Side. Front Neurosci 2021; 15:774050. [PMID: 34955726 PMCID: PMC8695496 DOI: 10.3389/fnins.2021.774050] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 11/08/2021] [Indexed: 12/17/2022] Open
Abstract
There is a substantial need for new pharmacological treatments of addiction, and appetite-regulatory peptides are implied as possible candidates. Appetite regulation is complex and involves anorexigenic hormones such as glucagon-like peptide-1 (GLP-1) and amylin, and orexigenic peptides like ghrelin and all are well-known for their effects on feeding behaviors. This overview will summarize more recent physiological aspects of these peptides, demonstrating that they modulate various aspects of addiction processes. Findings from preclinical, genetic, and experimental clinical studies exploring the association between appetite-regulatory peptides and the acute or chronic effects of addictive drugs will be introduced. Short or long-acting GLP-1 receptor agonists independently attenuate the acute rewarding properties of addictive drugs or reduce the chronic aspects of drugs. Genetic variation of the GLP-1 system is associated with alcohol use disorder. Also, the amylin pathway modulates the acute and chronic behavioral responses to addictive drugs. Ghrelin has been shown to activate reward-related behaviors. Moreover, ghrelin enhances, whereas pharmacological or genetic suppression of the ghrelin receptor attenuates the responses to various addictive drugs. Genetic studies and experimental clinical studies further support the associations between ghrelin and addiction processes. Further studies should explore the mechanisms modulating the ability of appetite-regulatory peptides to reduce addiction, and the effects of combination therapies or different diets on substance use are warranted. In summary, these studies provide evidence that appetite-regulatory peptides modulate reward and addiction processes, and deserve to be investigated as potential treatment target for addiction.
Collapse
Affiliation(s)
- Olesya T Shevchouk
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Maximilian Tufvesson-Alm
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Elisabet Jerlhag
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
37
|
Differential Influence of Pueraria lobata Root Extract and Its Main Isoflavones on Ghrelin Levels in Alcohol-Treated Rats. Pharmaceuticals (Basel) 2021; 15:ph15010025. [PMID: 35056082 PMCID: PMC8777655 DOI: 10.3390/ph15010025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/22/2021] [Accepted: 12/22/2021] [Indexed: 12/19/2022] Open
Abstract
The study was carried out on alcohol-preferring male Wistar rats. The following drugs were repeatedly (28×) administered: acamprosate (500 mg/kg, p.o.), naltrexone (0.1 mg/kg, i.p), and Pueraria lobata (kudzu) root extract (KU) (500 mg/kg, p.o.) and its isoflavones: daidzin (40 mg/kg, p.o.) and puerarin (150 mg/kg, p.o.). Their effects on a voluntary alcohol intake were assessed. KU and alcohol were also given for 9 days in an experiment on alcohol tolerance development. Finally, total and active ghrelin levels in peripheral blood serum were measured by ELISA method. Acamprosate, naltrexone, daidzin, and puerarin, reducing the alcohol intake, caused an increase in both forms of ghrelin levels. On the contrary, though KU inhibited the alcohol intake and alcohol tolerance development, it reduced ghrelin levels in alcohol-preferring rats. The changes of ghrelin concentration could play a role as an indicator of the currently used drugs. The other effect on the KU-induced shift in ghrelin levels in the presence of alcohol requires further detailed study.
Collapse
|
38
|
Colvin KJ, Killen HS, Kanter MJ, Halperin MC, Engel L, Dickinson MB, Fimmel AI, Holland JG, Currie PJ. Differential effects of intra-ventral tegmental area ghrelin and glucagon-like peptide-1 on the stimulatory action of D-amphetamine and cocaine-induced ethanol intake in male Sprague Dawley rats. Behav Brain Res 2021; 421:113726. [PMID: 34954300 DOI: 10.1016/j.bbr.2021.113726] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 12/15/2021] [Accepted: 12/17/2021] [Indexed: 12/24/2022]
Abstract
In order to further elucidate the role of mesolimbic peptides in the expression of ethanol reward, the present study investigated the effects of ghrelin and glucagon-like peptide-1 (GLP-1) on ethanol intake, in addition to ethanol intake stimulated by systemic d-amphetamine or cocaine treatment. While a number of studies suggest that ghrelin plays an important role in mesolimbic reward, emerging data now indicate that GLP-1 receptor mechanisms inhibit reward signaling, possibly by directly or indirectly inhibiting ghrelinergic activity within the mesolimbic system. In the present study all rats were initially habituated to a 6% ethanol solution. We then demonstrated that intraperitoneal injections of d-amphetamine and cocaine increased ethanol intake compared to the vehicle condition. In subsequent testing we examined the effects of ventral tegmental area (VTA) ghrelin or vehicle paired with a fixed dose of d-amphetamine or vehicle. In separate rats we then investigated the impact of the GLP-1 agonist exendin-4 (Ex-4), injected into the VTA, on ethanol intake alone, or when Ex-4 was co-administered with d-amphetamine or cocaine. Our results indicated that VTA ghrelin significantly increased ethanol intake, and most importantly, potentiated the effect of d-amphetamine and cocaine on ethanol consumption. Conversely, VTA Ex-4 inhibited ethanol intake and antagonized the stimulatory effect of d-amphetamine and cocaine on ethanol consumption. In a final study we further demonstrated that VTA Ex-4 treatment significantly inhibited the combined stimulatory effects of ghrelin paired with d-amphetamine or ghrelin paired with cocaine. Overall our findings are consistent with a critical role for both ghrelin and GLP-1 receptor mechanisms in mesolimbic ethanol reward circuitry. Moreover, our results further suggest that ghrelin and GLP-1 modulate the stimulatory effect of psychostimulants on ethanol intake.
Collapse
Affiliation(s)
- Kayla J Colvin
- Department of Psychology, Reed College, 3203 SE Woodstock Blvd., Portland, OR 97202, USA
| | - Henry S Killen
- Department of Psychology, Reed College, 3203 SE Woodstock Blvd., Portland, OR 97202, USA
| | - Maxwell J Kanter
- Department of Psychology, Reed College, 3203 SE Woodstock Blvd., Portland, OR 97202, USA
| | - Maximilian C Halperin
- Department of Psychology, Reed College, 3203 SE Woodstock Blvd., Portland, OR 97202, USA
| | - Liv Engel
- Department of Psychology, Reed College, 3203 SE Woodstock Blvd., Portland, OR 97202, USA
| | - Matthew B Dickinson
- Department of Psychology, Reed College, 3203 SE Woodstock Blvd., Portland, OR 97202, USA
| | - Anna I Fimmel
- Department of Psychology, Reed College, 3203 SE Woodstock Blvd., Portland, OR 97202, USA
| | - James G Holland
- Department of Psychology, Reed College, 3203 SE Woodstock Blvd., Portland, OR 97202, USA
| | - Paul J Currie
- Department of Psychology, Reed College, 3203 SE Woodstock Blvd., Portland, OR 97202, USA.
| |
Collapse
|
39
|
Price ML, Ley CD, Gorvin CM. The emerging role of heterodimerisation and interacting proteins in ghrelin receptor function. J Endocrinol 2021; 252:R23-R39. [PMID: 34663757 PMCID: PMC8630777 DOI: 10.1530/joe-21-0206] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Accepted: 10/18/2021] [Indexed: 11/14/2022]
Abstract
Ghrelin is a peptide hormone secreted primarily by the stomach that acts upon the growth hormone secretagogue receptor (GHSR1), a G protein-coupled receptor whose functions include growth hormone secretion, appetite regulation, energy expenditure, regulation of adiposity, and insulin release. Following the discovery that GHSR1a stimulates food intake, receptor antagonists were developed as potential therapies to regulate appetite. However, despite reductions in signalling, the desired effects on appetite were absent. Studies in the past 15 years have demonstrated GHSR1a can interact with other transmembrane proteins, either by direct binding (i.e. heteromerisation) or via signalling cross-talk. These interactions have various effects on GHSR1a signalling including preferential coupling to one pathway (i.e. biased signalling), coupling to a unique G protein (G protein switching), suppression of GHSR1a signalling, and enhancement of signalling by both receptors. While many of these interactions have been shown in cells overexpressing the proteins of interest and remain to be verified in tissues, substantial evidence exists showing that GHSR1a and the dopamine receptor D1 (DRD1) form heteromers, which promote synaptic plasticity and formation of hippocampal memory. Additionally, a reduction in GHSR1a-DRD1 complexes in favour of establishment of GHSR1a-Aβ complexes correlates with Alzheimer's disease, indicating that GHSR1a heteromers may have pathological functions. Herein, we summarise the evidence published to date describing interactions between GHSR1a and transmembrane proteins, discuss the experimental strengths and limitations of these studies, describe the physiological evidence for each interaction, and address their potential as novel drug targets for appetite regulation, Alzheimer's disease, insulin secretion, and inflammation.
Collapse
Affiliation(s)
- Maria L Price
- Institute of Metabolism and Systems Research and Centre for Endocrinology, Diabetes and Metabolism, University of Birmingham, Birmingham, UK
- Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, Birmingham, UK
| | - Cameron D Ley
- Institute of Metabolism and Systems Research and Centre for Endocrinology, Diabetes and Metabolism, University of Birmingham, Birmingham, UK
- Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, Birmingham, UK
| | - Caroline M Gorvin
- Institute of Metabolism and Systems Research and Centre for Endocrinology, Diabetes and Metabolism, University of Birmingham, Birmingham, UK
- Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, Birmingham, UK
- Correspondence should be addressed to C M Gorvin:
| |
Collapse
|
40
|
Farrokhi R, Babapour V, Zendehdel M, Asghari A, Gilanpour H. Role of Dopaminergic and Cannabinoidergic Receptors on Ghrelin-Induced Hypophagia in 5-Day-Old Broiler Chicken. ARCHIVES OF RAZI INSTITUTE 2021; 76:935-948. [PMID: 35096329 PMCID: PMC8790998 DOI: 10.22092/ari.2020.351261.1514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 08/24/2020] [Indexed: 01/24/2023]
Abstract
The present study aimed to identify the role of dopaminergic and cannabinoidergic systems in the ghrelin-induced hypophagia among meat-type chickens. In the first experiment, intracerebroventricular (ICV) injection was applied to birds with control solution, D1 receptor antagonist (5 nmol), ghrelin (6 nmol), and D1 receptor antagonist plus ghrelin. The second to sixth experiments were similar to the first one, with the difference that D2 receptor antagonist (5 nmol), D3 receptor antagonist (6.4 nmol), D4 receptor antagonist (6 nmol), the precursor of dopamine (125 nmol), and 6-hydroxy dopamine (150 nmol) instead of D1 antagonist were injected into the broiler chickens. In experiment 7, control solution and different levels of ghrelin antagonists (5, 10, and 20 nmol) were injected. In experiment 8, the chickens were ICV injected with control solution, ghrelin antagonist (10 nmol), dopamine (40 nmol), and ghrelin antagonist plus dopamine. In experiments 9 and 10, CB1 and CB2 receptors antagonist (6.25µg and 5µg) were co-injected with ghrelin (6 nmol), respectively, measuring the food intake for 120 min after the injection. It was observed that ghrelin ICV injection considerably reduced food intake, whereas ghrelin antagonist increased food intake, depending on the dose (P<0.05). In addition, ghrelin-induced hypophagia was significantly attenuated by D1 receptor antagonist and 6-hydroxy dopamine (P<0.05), while the dopamine precursor considerably elevated the ghrelin-induced food intake (P<0.05). The dopamine-induced feeding behavior was diminished by the co-administration of [D-Lys-3]-GHRP-6 (10 nmol)+dopamine (40 nmol) (P<0.05). In addition, CB1 receptor antagonists enhanced the ghrelin influence on food intake (P<0.05). The results implied that the hypophagic impact of ghrelin was probably mediated by D1 and CB1 receptors within neonatal broilers.
Collapse
Affiliation(s)
- R Farrokhi
- Department of Basic Sciences, Faculty of Veterinary Medicine, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - V Babapour
- Department of Basic Sciences, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - M Zendehdel
- Department of Basic Sciences, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - A Asghari
- Department of Clinical Science, Faculty of Veterinary Medicine, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - H Gilanpour
- Department of Basic Sciences, Faculty of Veterinary Medicine, Science and Research Branch, Islamic Azad University, Tehran, Iran
| |
Collapse
|
41
|
Hassouna R, Fernandez G, Lebrun N, Fiquet O, Roelfsema F, Labarthe A, Zizzari P, Tomasetto C, Epelbaum J, Viltart O, Chauveau C, Perello M, Tolle V. Ghrelin Gene Deletion Alters Pulsatile Growth Hormone Secretion in Adult Female Mice. Front Endocrinol (Lausanne) 2021; 12:754522. [PMID: 34721302 PMCID: PMC8549963 DOI: 10.3389/fendo.2021.754522] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 09/16/2021] [Indexed: 11/13/2022] Open
Abstract
Using preproghrelin-deficient mice (Ghrl-/-), we previously observed that preproghrelin modulates pulsatile growth hormone (GH) secretion in post-pubertal male mice. However, the role of ghrelin and its derived peptides in the regulation of growth parameters or feeding in females is unknown. We measured pulsatile GH secretion, growth, metabolic parameters and feeding behavior in adult Ghrl-/- and Ghrl+/+ male and female mice. We also assessed GH release from pituitary explants and hypothalamic growth hormone-releasing hormone (GHRH) expression and immunoreactivity. Body weight and body fat mass, linear growth, spontaneous food intake and food intake following a 48-h fast, GH pituitary contents and GH release from pituitary explants ex vivo, fasting glucose and glucose tolerance were not different among adult Ghrl-/- and Ghrl+/+ male or female mice. In vivo, pulsatile GH secretion was decreased, while approximate entropy, that quantified orderliness of secretion, was increased in adult Ghrl-/- females only, defining more irregular GH pattern. The number of neurons immunoreactive for GHRH visualized in the hypothalamic arcuate nucleus was increased in adult Ghrl-/- females, as compared to Ghrl+/+ females, whereas the expression of GHRH was not different amongst groups. Thus, these results point to sex-specific effects of preproghrelin gene deletion on pulsatile GH secretion, but not feeding, growth or metabolic parameters, in adult mice.
Collapse
Affiliation(s)
- Rim Hassouna
- Université de Paris, UMR-S 1266 INSERM, Institute of Psychiatry and Neuroscience of Paris, Paris, France
| | - Gimena Fernandez
- Laboratory of Neurophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA). National University of La Plata], La Plata, Buenos Aires, Argentina
| | - Nicolas Lebrun
- Université de Paris, UMR-S 1266 INSERM, Institute of Psychiatry and Neuroscience of Paris, Paris, France
| | - Oriane Fiquet
- Université de Paris, UMR-S 1266 INSERM, Institute of Psychiatry and Neuroscience of Paris, Paris, France
| | - Ferdinand Roelfsema
- Department of Internal Medicine, Section of Endocrinology and Metabolism, Leiden University Medical Center, Leiden, Netherlands
| | - Alexandra Labarthe
- Université de Paris, UMR-S 1266 INSERM, Institute of Psychiatry and Neuroscience of Paris, Paris, France
| | - Philippe Zizzari
- Université de Paris, UMR-S 1266 INSERM, Institute of Psychiatry and Neuroscience of Paris, Paris, France
| | - Catherine Tomasetto
- Institut de génétique et de biologie moléculaire et cellulaire (IGBMC), UMR7104 CNRS/U1258 INSERM, Université de Strasbourg, Illkirch, France
| | - Jacques Epelbaum
- Université de Paris, UMR-S 1266 INSERM, Institute of Psychiatry and Neuroscience of Paris, Paris, France
- UMR CNRS/MNHN 7179, Mécanismes Adaptatifs et Evolution, Brunoy, France
| | - Odile Viltart
- Université de Paris, UMR-S 1266 INSERM, Institute of Psychiatry and Neuroscience of Paris, Paris, France
- Université de Lille, Faculté des Sciences et Technologies, Villeneuve d’Ascq, France
| | - Christophe Chauveau
- Marrow Adiposity and Bone Lab - MABLab ULR 4490, Univ. Littoral Côte d’Opale, Boulogne-sur-Mer, Univ. Lille and CHU Lille, Lille, France
| | - Mario Perello
- Laboratory of Neurophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA). National University of La Plata], La Plata, Buenos Aires, Argentina
| | - Virginie Tolle
- Université de Paris, UMR-S 1266 INSERM, Institute of Psychiatry and Neuroscience of Paris, Paris, France
| |
Collapse
|
42
|
Mustafá ER, Cordisco González S, Damian M, Cantel S, Denoyelle S, Wagner R, Schiöth HB, Fehrentz JA, Banères JL, Perelló M, Raingo J. LEAP2 Impairs the Capability of the Growth Hormone Secretagogue Receptor to Regulate the Dopamine 2 Receptor Signaling. Front Pharmacol 2021; 12:712437. [PMID: 34447311 PMCID: PMC8383165 DOI: 10.3389/fphar.2021.712437] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 07/26/2021] [Indexed: 12/12/2022] Open
Abstract
The growth hormone secretagogue receptor (GHSR) signals in response to ghrelin, but also acts via ligand-independent mechanisms that include either constitutive activation or interaction with other G protein-coupled receptors, such as the dopamine 2 receptor (D2R). A key target of GHSR in neurons is voltage-gated calcium channels type 2.2 (CaV2.2). Recently, the liver-expressed antimicrobial peptide 2 (LEAP2) was recognized as a novel GHSR ligand, but the mechanism of action of LEAP2 on GHSR is not well understood. Here, we investigated the role of LEAP2 on the canonical and non-canonical modes of action of GHSR on CaV2.2 function. Using a heterologous expression system and patch-clamp recordings, we found that LEAP2 impairs the reduction of CaV2.2 currents induced by ghrelin-evoked and constitutive GHSR activities, acting as a GHSR antagonist and inverse agonist, respectively. We also found that LEAP2 prevents GHSR from modulating the effects of D2R signaling on CaV2.2 currents, and that the GHSR-binding N-terminal region LEAP2 underlies these effects. Using purified labeled receptors assembled into lipid nanodiscs and Forster Resonance Energy Transfer (FRET) assessments, we found that the N-terminal region of LEAP2 stabilizes an inactive conformation of GHSR that is dissociated from Gq protein and, consequently, reverses the effect of GHSR on D2R-dependent Gi activation. Thus, our results provide critical molecular insights into the mechanism mediating LEAP2 modulation of GHSR.
Collapse
Affiliation(s)
- Emilio R Mustafá
- Laboratory of Electrophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata (UNLP)], La Plata, Argentina
| | - Santiago Cordisco González
- Laboratory of Electrophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata (UNLP)], La Plata, Argentina
| | - Marjorie Damian
- Institut des Biomolécules Max Mousseron (IBMM), Université Montpellier, CNRS, Montpellier, France
| | - Sonia Cantel
- Institut des Biomolécules Max Mousseron (IBMM), Université Montpellier, CNRS, Montpellier, France
| | - Severine Denoyelle
- Institut des Biomolécules Max Mousseron (IBMM), Université Montpellier, CNRS, Montpellier, France
| | - Renaud Wagner
- Plateforme IMPReSs, CNRS UMR7242, Biotechnologie et Signalisation Cellulaire, École Supérieure de Biotechnologie de Strasbourg, Strasbourg, France
| | - Helgi B Schiöth
- Department of Neuroscience, Uppsala University, Uppsala, Sweden.,Institute for Translational Medicine and Biothechnology, I. M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Jean-Alain Fehrentz
- Institut des Biomolécules Max Mousseron (IBMM), Université Montpellier, CNRS, Montpellier, France
| | - Jean-Louis Banères
- Institut des Biomolécules Max Mousseron (IBMM), Université Montpellier, CNRS, Montpellier, France
| | - Mario Perelló
- Laboratory of Neurophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata (UNLP)], La Plata, Argentina
| | - Jesica Raingo
- Laboratory of Electrophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata (UNLP)], La Plata, Argentina
| |
Collapse
|
43
|
Beheshti S, Dehestani H. Differential expression levels of the hippocampal ghrelin and its receptor mRNA during memory consolidation. Behav Brain Res 2021; 408:113270. [PMID: 33811951 DOI: 10.1016/j.bbr.2021.113270] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 03/23/2021] [Accepted: 03/28/2021] [Indexed: 12/30/2022]
Abstract
Ghrelin is a peptide, secreted mainly from the stomach. But, it is also produced in the brain. Studies have confirmed the positive impact of ghrelin on memory formation. However, the expression levels of ghrelin or its receptors were not measured in the brain during the process of memory formation. The probable alteration in the expression levels of ghrelin or its receptors in the brain during memory formation can be a reason for the contribution of its signaling in this process. We quantified the gene expression levels of ghrelin and its receptors in the hippocampus during fear and spatial memory consolidation. Thirty- nine adult male Wistar rats weighing 180-220 g were utilized. Memory consolidation was evaluated using the inhibitory avoidance task and Morris water maze. Rats were euthanized at different times (1, 3, and 24 h) post-training and their hippocampi were removed and freezed directly in liquid nitrogen. Quantitative real-time polymerize chain reaction (PCR) was used to quantify the messenger ribonucleic acid (mRNA) expression levels of the hippocampal ghrelin and its receptors. The mRNA levels of ghrelin exhibited a significant increase, 24 h post-training in the inhibitory avoidance task, while its receptor levels were down-regulated. Also, the mRNA expression levels of the hippocampal ghrelin were not changed significantly during memory consolidation in the Morris water maze, while its receptor showed a significant increase, 24 h post-training. The results show a differential profile of the expression levels of the hippocampal ghrelin or its receptor mRNA during fear or spatial memory consolidation. This proposes that a local increase in the hippocampal ghrelin or its receptor levels might be crucial for fear, and spatial memory consolidation. However, due to the small sample sizes, it is worth noting the preliminary nature of the conclusions in the present study.
Collapse
Affiliation(s)
- Siamak Beheshti
- Department of Plant and Animal Biology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran.
| | - Hadi Dehestani
- Department of Plant and Animal Biology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| |
Collapse
|
44
|
Fujita W. [MOPr-DOPr heteromer: the meaning and possibility as novel therapeutic target for pain control]. Nihon Yakurigaku Zasshi 2021; 156:134-138. [PMID: 33952839 DOI: 10.1254/fpj.20103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Many studies suggest opioid receptor (OPr) dimerization modulates the pharmacological properties of opiates. Specifically, heteromerization between OPr types has been reported to lead to changes in intracellular signaling. Thus, ligands targeting heteromers are expected to be novel therapeutic targets with reduced side effects. The heteromers of mu (MOPr) and delta (DOPr) are detected in brain regions involved in pain processing. The bivalent ligand or small molecule were identified as a MOPr-DOPr targeting ligand. These ligands exhibit antinociceptive properties similar to that of morphine with lesser antinociceptive tolerance as compared to morphine. Studies exploring the in vivo regulation of MOPr-DOPr heteromers, showed chronic morphine administration leads to an upregulation of these heteromers in select brain regions. Exploration of mechanisms underlying this phenomenon led us to the G protein-coupled receptor chaperone, RTP4, that is induced by chronic morphine and facilitates the heteromerization of MOPr and DOPr. In this review, I will introduce the simulated structure or property of MOPr-DOPr heteromer, its targeting ligands, and its intracellular regulatory mechanism that include a key molecule like RTP4 that could serve as a scaffold for the development of novel therapeutic drugs with reduced adverse effects, and hence may take place of the conventional clinical opioids.
Collapse
Affiliation(s)
- Wakako Fujita
- Department of Medical Pharmacology, Nagasaki University Graduate School of Biomedical Sciences
| |
Collapse
|
45
|
Furness JB, Pustovit RV, Syder AJ, Ringuet MT, Yoo EJ, Fanjul A, Wykosky J, Fothergill LJ, Whitfield EA, Furness SGB. Dopamine and ghrelin receptor co-expression and interaction in the spinal defecation centers. Neurogastroenterol Motil 2021; 33:e14051. [PMID: 33264473 DOI: 10.1111/nmo.14051] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 10/20/2020] [Accepted: 11/12/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND Dopamine receptor 2 (DRD2) and ghrelin receptor (GHSR1a) agonists both stimulate defecation by actions at the lumbosacral defecation center. Dopamine is in nerve terminals surrounding autonomic neurons of the defecation center, whereas ghrelin is not present in the spinal cord. Dopamine at D2 receptors generally inhibits neurons, but at the defecation center, its effect is excitatory. METHODS In vivo recording of defecation and colorectal propulsion was used to investigate interaction between DRD2 and GHSR1a. Localization studies were used to determine sites of receptor expression in rat and human spinal cord. KEY RESULTS Dopamine, and the DRD2 agonist, quinpirole, directly applied to the lumbosacral cord, caused defecation. The effect of intrathecal dopamine was inhibited by the GHSR1a antagonist, YIL781, given systemically, but YIL781 was not an antagonist at DRD2. The DRD2 agonist, pramipexole, administered systemically caused colorectal propulsion that was prevented when the pelvic nerves were cut. Drd2 and Ghsr were expressed together in autonomic preganglionic neurons at the level of the defecation centers in rat and human. Behaviorally induced defecation (caused by water avoidance stress) was reduced by the DRD2 antagonist, sulpiride. We had previously shown it is reduced by YIL781. CONCLUSIONS AND INFERENCES Our observations imply that dopamine is a transmitter of the defecation pathways whose actions are exerted through interacting dopamine (D2) and ghrelin receptors on lumbosacral autonomic neurons that project to the colorectum. The results explain the excitation by dopamine agonists and the conservation of GHSR1a in the absence of ghrelin.
Collapse
Affiliation(s)
- John B Furness
- Florey Institute of Neuroscience and Mental Health, Parkville, Vic., Australia.,Department of Anatomy & Neuroscience, University of Melbourne, Parkville, Vic., Australia
| | - Ruslan V Pustovit
- Florey Institute of Neuroscience and Mental Health, Parkville, Vic., Australia.,Department of Anatomy & Neuroscience, University of Melbourne, Parkville, Vic., Australia
| | - Andrew J Syder
- Gastroenterology Drug Discovery Unit, Takeda Pharmaceutical Company Limited, San Diego, CA, USA
| | - Mitchell T Ringuet
- Florey Institute of Neuroscience and Mental Health, Parkville, Vic., Australia.,Department of Anatomy & Neuroscience, University of Melbourne, Parkville, Vic., Australia
| | - Eun Ji Yoo
- Gastroenterology Drug Discovery Unit, Takeda Pharmaceutical Company Limited, San Diego, CA, USA
| | - Andrea Fanjul
- Gastroenterology Drug Discovery Unit, Takeda Pharmaceutical Company Limited, San Diego, CA, USA
| | - Jill Wykosky
- Gastroenterology Drug Discovery Unit, Takeda Pharmaceutical Company Limited, San Diego, CA, USA
| | - Linda J Fothergill
- Florey Institute of Neuroscience and Mental Health, Parkville, Vic., Australia.,Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Vic., Australia
| | - Emily A Whitfield
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Vic., Australia
| | - Sebastian G B Furness
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Vic., Australia
| |
Collapse
|
46
|
Misganaw D. Heteromerization of dopaminergic receptors in the brain: Pharmacological implications. Pharmacol Res 2021; 170:105600. [PMID: 33836279 DOI: 10.1016/j.phrs.2021.105600] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/17/2021] [Accepted: 04/02/2021] [Indexed: 12/15/2022]
Abstract
Dopamine exerts its physiological effects through two subtypes of receptors, i.e. the receptors of the D1 family (D1R and D5R) and the D2 family (D2R, D3R, and D4R), which differ in their pattern of distribution, affinity, and signaling. The D1-like subfamily (D1R and D5R) are coupled to Gαs/olf proteins to activate adenylyl cyclase whereas the D2-like receptors are coupled to Gαi/o subunits and suppress the activity of adenylyl cyclase. Dopamine receptors are capable of forming homodimers, heterodimers, and higher-order oligomeric complexes, resulting in a change in the individual protomers' recognition, signaling, and pharmacology. Heteromerization has the potential to modify the canonical pharmacological features of individual monomeric units such as ligand affinity, activation, signaling, and cellular trafficking through allosteric interactions, reviving the field and introducing a new pharmacological target. Since heteromers are expressed and formed in a tissue-specific manner, they could provide the framework to design selective and effective drug candidates, such as brain-penetrant heterobivalent drugs and interfering peptides, with limited side effects. Therefore, heteromerization could be a promising area of pharmacology research, as it could contribute to the development of novel pharmacological interventions for dopamine dysregulated brain disorders such as addiction, schizophrenia, cognition, Parkinson's disease, and other motor-related disorders. This review is articulated based on the three criteria established by the International Union of Basic and Clinical Pharmacology for GPCR heterodimers (IUPHAR): evidence of co-localization and physical interactions in native or primary tissue, presence of a new physiological and functional property than the individual protomers, and loss of interaction and functional fingerprints upon heterodimer disruption.
Collapse
Affiliation(s)
- Desye Misganaw
- Pharmacology and Toxicology Unit, Department of Pharmacy, College of Medicine and Health Science, Wollo University, P.O. Box 1145, Dessie, Ethiopia.
| |
Collapse
|
47
|
Biased signaling: A viable strategy to drug ghrelin receptors for the treatment of obesity. Cell Signal 2021; 83:109976. [PMID: 33713808 DOI: 10.1016/j.cellsig.2021.109976] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 02/23/2021] [Accepted: 03/08/2021] [Indexed: 02/07/2023]
Abstract
Obesity is a global burden and a chronic ailment with damaging overall health effects. Ghrelin, an octanoylated 28 amino acid peptide hormone, is secreted from the oxyntic mucosa of the stomach. Ghrelin acts on regions of the hypothalamus to regulate feeding behavior and glucose homeostasis through its G protein-coupled receptor. Recently, several central pathways modulating the metabolic actions of ghrelin have been reported. While these signaling pathways can be inhibited or activated by antagonists or agonists, they can also be discriminatingly activated in a "biased" response to impart different degrees of activation in distinct pathways downstream of the receptor. Here, we review recent ghrelin biased signaling findings as well as characteristics of ghrelin hormone and its receptors pertinent for biased signaling. We then evaluate the feasibility for ghrelin receptor biased signaling as a strategy for the development of effective pharmacotherapy in obesity treatment.
Collapse
|
48
|
Ribeiro LF, Catarino T, Carvalho M, Cortes L, Santos SD, Opazo PO, Ribeiro LR, Oliveiros B, Choquet D, Esteban JA, Peça J, Carvalho AL. Ligand-independent activity of the ghrelin receptor modulates AMPA receptor trafficking and supports memory formation. Sci Signal 2021; 14:14/670/eabb1953. [PMID: 33593997 DOI: 10.1126/scisignal.abb1953] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The biological signals of hunger, satiety, and memory are interconnected. The role of the hormone ghrelin in regulating feeding and memory makes ghrelin receptors attractive targets for associated disorders. We investigated the effects of the high ligand-independent activity of the ghrelin receptor GHS-R1a on the physiology of excitatory synapses in the hippocampus. Blocking this activity produced a decrease in the synaptic content of AMPA receptors in hippocampal neurons and a reduction in GluA1 phosphorylation at Ser845 Reducing the ligand-independent activity of GHS-R1a increased the surface diffusion of AMPA receptors and impaired AMPA receptor-dependent synaptic delivery induced by chemical long-term potentiation. Accordingly, we found that blocking this GHS-R1a activity impaired spatial and recognition memory in mice. These observations support a role for the ligand-independent activity of GHS-R1a in regulating AMPA receptor trafficking under basal conditions and in the context of synaptic plasticity that underlies learning.
Collapse
Affiliation(s)
- Luís F Ribeiro
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal.
| | - Tatiana Catarino
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal.,University of Coimbra, IIIUC-Institute for Interdisciplinary Research, 3030-789 Coimbra, Portugal
| | - Mário Carvalho
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal.,MIT-Portugal Bioengineering Systems Doctoral Program, NOVA University of Lisbon, 1099-85, Lisboa, Portugal
| | - Luísa Cortes
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal.,University of Coimbra, IIIUC-Institute for Interdisciplinary Research, 3030-789 Coimbra, Portugal
| | - Sandra D Santos
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal.,University of Coimbra, IIIUC-Institute for Interdisciplinary Research, 3030-789 Coimbra, Portugal
| | - Patricio O Opazo
- University of Bordeaux, Interdisciplinary Institute for Neuroscience, UMR 5297, 33000 Bordeaux, France.,CNRS, UMR 5297, 33000 Bordeaux, France
| | - Lyn Rosenbrier Ribeiro
- Functional and Mechanistic Safety, Clinical Pharmacology and Safety Sciences, R&D AstraZeneca, Cambridge CB2 0SL, UK
| | - Bárbara Oliveiros
- Laboratory of Biostatistics and Medical Informatics (LBIM), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal.,Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Daniel Choquet
- University of Bordeaux, Interdisciplinary Institute for Neuroscience, UMR 5297, 33000 Bordeaux, France.,CNRS, UMR 5297, 33000 Bordeaux, France.,Bordeaux Imaging Center, UMS 3420, CNRS-Bordeaux University, US4 INSERM, 33000 Bordeaux, France
| | - José A Esteban
- Department of Molecular Neurobiology, Centro de Biología Molecular "Severo Ochoa," Consejo Superior de Investigaciones Científicas (CSIC)/Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - João Peça
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal.,University of Coimbra, Department of Life Sciences, 3000-456 Coimbra, Portugal
| | - Ana Luísa Carvalho
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal. .,University of Coimbra, Department of Life Sciences, 3000-456 Coimbra, Portugal
| |
Collapse
|
49
|
Cornejo MP, Mustafá ER, Cassano D, Banères JL, Raingo J, Perello M. The ups and downs of growth hormone secretagogue receptor signaling. FEBS J 2021; 288:7213-7229. [PMID: 33460513 DOI: 10.1111/febs.15718] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 01/05/2021] [Accepted: 01/14/2021] [Indexed: 12/14/2022]
Abstract
The growth hormone secretagogue receptor (GHSR) has emerged as one of the most fascinating molecules from the perspective of neuroendocrine control. GHSR is mainly expressed in the pituitary and the brain, and plays key roles regulating not only growth hormone secretion but also food intake, adiposity, body weight, glucose homeostasis and other complex functions. Quite atypically, GHSR signaling displays a basal constitutive activity that can be up- or downregulated by two digestive system-derived hormones: the octanoylated-peptide ghrelin and the liver-expressed antimicrobial peptide 2 (LEAP2), which was recently recognized as an endogenous GHSR ligand. The existence of two ligands with contrary actions indicates that GHSR activity can be tightly regulated and that the receptor displays the capability to integrate such opposing inputs in order to provide a balanced intracellular signal. This article provides a summary of the current understanding of the biology of ghrelin, LEAP2 and GHSR and discusses the reconceptualization of the cellular and physiological implications of the ligand-regulated GHSR signaling, based on the latest findings.
Collapse
Affiliation(s)
- María P Cornejo
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET), Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], Buenos Aires, Argentina
| | - Emilio R Mustafá
- Laboratory of Electrophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET), Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], Buenos Aires, Argentina
| | - Daniela Cassano
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET), Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], Buenos Aires, Argentina
| | - Jean-Louis Banères
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, CNRS, Université de Montpellier, Ecole Nationale Supérieure de Chimie de Montpellier, Faculté de Pharmacie, Montpellier cedex 5, France
| | - Jesica Raingo
- Laboratory of Electrophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET), Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], Buenos Aires, Argentina
| | - Mario Perello
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET), Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], Buenos Aires, Argentina
| |
Collapse
|
50
|
Khelifa MS, Skov LJ, Holst B. Biased Ghrelin Receptor Signaling and the Dopaminergic System as Potential Targets for Metabolic and Psychological Symptoms of Anorexia Nervosa. Front Endocrinol (Lausanne) 2021; 12:734547. [PMID: 34646236 PMCID: PMC8503187 DOI: 10.3389/fendo.2021.734547] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 08/16/2021] [Indexed: 12/15/2022] Open
Abstract
Anorexia Nervosa (AN) is a complex disease that impairs the metabolic, mental and physiological health of affected individuals in a severe and sometimes lethal way. Many of the common symptoms in AN patients, such as reduced food intake, anxiety, impaired gut motility or overexercising are connected to both the orexigenic gut hormone ghrelin and the dopaminergic system. Targeting the ghrelin receptor (GhrR) to treat AN seems a promising possibility in current research. However, GhrR signaling is highly complex. First, the GhrR can activate four known intracellular pathways Gαq, Gαi/o, Gα12/13 and the recruitment of β-arrestin. Biased signaling provides the possibility to activate or inhibit only one or a subset of the intracellular pathways of a pleiotropic receptor. This allows specific targeting of physiological functions without adverse effects. Currently little is known on how biased signaling could specifically modulate GhrR effects. Second, GhrR signaling has been shown to be interconnected with the dopaminergic system, particularly in the context of AN symptoms. This review highlights that a biased agonist for the GhrR may be a promising target for the treatment of AN, however extensive and systematic translational studies are still needed and the connection to the dopaminergic system has to be taken into account.
Collapse
|