1
|
Richardson RS, Kryszak LA, Vendruscolo JCM, Koob GF, Vendruscolo LF, Leggio L. GHSR blockade, but not reduction of peripherally circulating ghrelin via β 1-adrenergic receptor antagonism, decreases binge-like alcohol drinking in mice. Mol Psychiatry 2025; 30:1047-1056. [PMID: 39232198 PMCID: PMC11835741 DOI: 10.1038/s41380-024-02713-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 08/17/2024] [Accepted: 08/21/2024] [Indexed: 09/06/2024]
Abstract
Alcohol use disorder (AUD) and binge drinking are highly prevalent public health issues. The stomach-derived peptide ghrelin, and its receptor, the growth hormone secretagogue receptor (GHSR), both of which are expressed in the brain and periphery, are implicated in alcohol-related outcomes. We previously found that systemic and central administration of GHSR antagonists reduced binge-like alcohol drinking, whereas a ghrelin vaccine did not. Thus, we hypothesized that central GHSR drives binge-like alcohol drinking independently of peripheral ghrelin. To investigate this hypothesis, we antagonized β1-adrenergic receptors (β1ARs), which are required for peripheral ghrelin release, and combined them with GHSR blockers. We found that both systemic β1AR antagonism with atenolol (peripherally restricted) and metoprolol (brain permeable) robustly decreased plasma ghrelin levels. Also, ICV administration of atenolol had no effect on peripheral endogenous ghrelin levels. However, only metoprolol, but not atenolol, decreased binge-like alcohol drinking. The β1AR antagonism also did not prevent the effects of the GHSR blockers JMV2959 and PF-5190457 in decreasing binge-like alcohol drinking. These results suggest that the GHSR rather than peripheral endogenous ghrelin is involved in binge-like alcohol drinking. Thus, GHSRs and β1ARs represent possible targets for therapeutic intervention for AUD, including the potential combination of drugs that target these two systems.
Collapse
Affiliation(s)
- Rani S Richardson
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore and Bethesda, MD, USA
- Neurobiology of Addiction Section, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
- University of North Carolina School of Medicine MD/PhD Program, University of North Carolina, Chapel Hill, NC, USA
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, USA
- Stress and Addiction Neuroscience Unit, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore, MD, USA
| | - Lindsay A Kryszak
- National Institute on Drug Abuse Intramural Research Program Translational Analytical Core, National Institutes of Health, Baltimore, MD, USA
| | - Janaina C M Vendruscolo
- Neurobiology of Addiction Section, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
- Stress and Addiction Neuroscience Unit, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore, MD, USA
| | - George F Koob
- Neurobiology of Addiction Section, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Leandro F Vendruscolo
- Stress and Addiction Neuroscience Unit, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore, MD, USA.
| | - Lorenzo Leggio
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore and Bethesda, MD, USA.
- National Institute on Drug Abuse Intramural Research Program Translational Analytical Core, National Institutes of Health, Baltimore, MD, USA.
- Department of Behavioral and Social Sciences, Center for Alcohol and Addiction Studies, Brown University, Providence, RI, USA.
- Medication Development Program, Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD, USA.
- Division of Addiction Medicine, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA.
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC, USA.
| |
Collapse
|
2
|
Yanik T, Durhan ST. Pro-Opiomelanocortin and Melanocortin Receptor 3 and 4 Mutations in Genetic Obesity. Biomolecules 2025; 15:209. [PMID: 40001512 PMCID: PMC11853658 DOI: 10.3390/biom15020209] [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: 12/11/2024] [Revised: 01/21/2025] [Accepted: 01/27/2025] [Indexed: 02/27/2025] Open
Abstract
Genetic obesity results from loss-of-function mutations, including those affecting the leptin-melanocortin system, which regulates body weight. Pro-opiomelanocortin (POMC)-derived neurohormones act as ligands for melanocortin receptors (MCRs), regulating the leptin-melanocortin pathway through protein-protein interactions. Loss-of-function mutations in the genes encoding POMC, MC3R, and MC4R can lead to the dysregulation of energy expenditure and feeding balance, early-onset obesity, and developmental dysregulation. Recent studies have identified new genetic regulatory mechanisms and potential biomarker regions for the POMC gene and MC4R secondary messenger pathway associated with obesity. Recent advances in crystal structure studies have enhanced our understanding of the protein interactions in this pathway. This narrative review focuses on recent developments in two key areas related to POMC regulation and the leptin-melanocortin pathway: (1) genetic variations in and functions of POMC, and (2) MC3R and MC4R variants that lead to genetic obesity in humans. Understanding these novel mutations in POMC and MC4R/MC3R, as well as their structural and intracellular mechanisms, may help identify strategies for the treatment and diagnosis of obesity, particularly childhood obesity.
Collapse
Affiliation(s)
- Tulin Yanik
- Department of Biological Sciences, Middle East Technical University, Ankara 06800, Türkiye
| | - Seyda Tugce Durhan
- Department of Biochemistry, Middle East Technical University, Ankara 06800, Türkiye;
| |
Collapse
|
3
|
Rosendo-Silva D, Lopes E, Monteiro-Alfredo T, Falcão-Pires I, Eickhoff H, Viana S, Reis F, Pires AS, Abrantes AM, Botelho MF, Seiça R, Matafome P. The adipose tissue melanocortin 3 receptor is targeted by ghrelin and leptin and may be a therapeutic target in obesity. Mol Cell Endocrinol 2024; 594:112367. [PMID: 39293775 DOI: 10.1016/j.mce.2024.112367] [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: 07/24/2024] [Revised: 09/03/2024] [Accepted: 09/07/2024] [Indexed: 09/20/2024]
Abstract
OBJECTIVE Obesity is linked to perturbations in energy balance mechanisms, including ghrelin and leptin actions at the hypothalamic circuitry of neuropeptide Y (NPY) and melanocortin. However, information about the regulation of this system in the periphery is still scarce. Our objective was to study the regulation of the NPY/melanocortin system in the adipose tissue (AT) and evaluate its therapeutic potential for obesity and type 2 diabetes. METHODS The expression of the NPY/melanocortin receptors' levels was assessed in the visceral AT of individuals with obesity and altered metabolism. Protein levels of these receptors were evaluated in cultured adipocytes incubated with ghrelin (30 and 100 ng/mL) and leptin (1 and 10 nM) and in the AT of an animal model with a mutation in the leptin receptor (ZSF1 rat), to understand their regulation by leptin and ghrelin. The vertical sleeve gastrectomy animal model was used to evaluate the putative therapeutic potential of the NPY/melanocortin system. RESULTS In this study, we unravelled that leptin (1 nM and 10 nM) selectively reduced the levels of NPY5R and MC3R but no other NPYR/MCRs in cultured adipocytes. In turn, acylated ghrelin (100 ng/mL) significantly increased NPY1R, but the inhibition of its receptor also abrogates MC3R levels. However, in the Lepr-deficient ZSF1 rat, both NPY5R and MC3R levels were reduced, along with other NPYRs and MCRs, suggesting that leptin resistance negatively affects NPY and melanocortin signalling. In human adipose tissue, we found a downregulation of genes encoding the NPY and melanocortin receptors in the visceral AT of individuals with obesity and insulin resistance, being correlated with genes regulating metabolic activity. Additionally, diabetic obese rats submitted to vertical sleeve gastrectomy showed increased levels of NPY, melanocortin, ghrelin, and leptin receptors in the AT, including MC3R, suggesting it may constitute a therapeutic target in obesity. CONCLUSIONS Our results suggest that the AT NPY/melanocortin system, particularly the MC3R, may be involved in the neuroendocrine regulation of adipocyte metabolism. Altogether, our work shows MC3R is under the control of the ghrelin/leptin duo, is reduced in patients with obesity and prediabetes, and may constitute a therapeutic target in obesity.
Collapse
Affiliation(s)
- Daniela Rosendo-Silva
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR) and Institute of Physiology, Faculty of Medicine, Coimbra, Portugal; University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal; Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal.
| | - Eduardo Lopes
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR) and Institute of Physiology, Faculty of Medicine, Coimbra, Portugal; University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
| | - Tamaeh Monteiro-Alfredo
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR) and Institute of Physiology, Faculty of Medicine, Coimbra, Portugal; University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
| | - Inês Falcão-Pires
- UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine of the University of Porto, Porto, Portugal
| | - Hans Eickhoff
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR) and Institute of Physiology, Faculty of Medicine, Coimbra, Portugal
| | - Sofia Viana
- University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal; Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal; University of Coimbra, iCBR and Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, Coimbra, Portugal; Polytechnic University of Coimbra, Coimbra Health School (ESTeSC), Coimbra, Portugal
| | - Flávio Reis
- University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal; Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal; University of Coimbra, iCBR and Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, Coimbra, Portugal
| | - Ana Salomé Pires
- University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal; Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal; University of Coimbra, iCBR Area of Environment Genetics and Oncobiology (CIMAGO), Institute of Biophysics, Faculty of Medicine, Coimbra, Portugal
| | - Ana Margarida Abrantes
- University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal; Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal; University of Coimbra, iCBR Area of Environment Genetics and Oncobiology (CIMAGO), Institute of Biophysics, Faculty of Medicine, Coimbra, Portugal
| | - Maria Filomena Botelho
- University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal; Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal; University of Coimbra, iCBR Area of Environment Genetics and Oncobiology (CIMAGO), Institute of Biophysics, Faculty of Medicine, Coimbra, Portugal
| | - Raquel Seiça
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR) and Institute of Physiology, Faculty of Medicine, Coimbra, Portugal
| | - Paulo Matafome
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR) and Institute of Physiology, Faculty of Medicine, Coimbra, Portugal; University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal; Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal; Polytechnic University of Coimbra, Coimbra Health School (ESTeSC), Coimbra, Portugal.
| |
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
|
Hölscher C. Glucagon-like peptide-1 class drugs show clear protective effects in Parkinson's and Alzheimer's disease clinical trials: A revolution in the making? Neuropharmacology 2024; 253:109952. [PMID: 38677445 DOI: 10.1016/j.neuropharm.2024.109952] [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: 04/05/2024] [Accepted: 04/11/2024] [Indexed: 04/29/2024]
Abstract
Parkinson's disease (PD) is a complex syndrome for which there is no disease-modifying treatment on the market. However, a group of drugs from the Glucagon-like peptide-1 (GLP-1) class have shown impressive improvements in clinical phase II trials. Exendin-4 (Bydureon), Liraglutide (Victoza, Saxenda) and Lixisenatide (Adlyxin), drugs that are on the market as treatments for diabetes, have shown clear effects in improving motor activity in patients with PD in phase II clinical trials. In addition, Liraglutide has shown improvement in cognition and brain shrinkage in a phase II trial in patients with Alzheimer disease (AD). Two phase III trials testing the GLP-1 drug semaglutide (Wegovy, Ozempic, Rybelsus) are ongoing. This perspective article will summarize the clinical results obtained so far in this novel research area. We are at a crossroads where GLP-1 class drugs are emerging as a new treatment strategy for PD and for AD. Newer drugs that have been designed to enter the brain easier are being developed already show improved effects in preclinical studies compared with the older GLP-1 class drugs that had been developed to treat diabetes. The future looks bright for new treatments for AD and PD.
Collapse
Affiliation(s)
- Christian Hölscher
- Henan Academy of Innovations in Medical Science, Neurodegeneration Research Group, 451100 Xinzheng, Henan province, China.
| |
Collapse
|
6
|
Smith A, Rodrigues T, Wallace C, Mezher K, MacAulay B, Prowse R, Hyland L, Abizaid A. Growth Hormone Secretagogue Receptor (GHSR) Signaling in the Ventral Tegmental Area (VTA) Mediates Feeding Produced by Chronic Social Defeat Stress in Male Mice. Neuroscience 2024; 547:17-27. [PMID: 38583506 DOI: 10.1016/j.neuroscience.2024.03.022] [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: 12/20/2023] [Revised: 03/04/2024] [Accepted: 03/18/2024] [Indexed: 04/09/2024]
Abstract
Ghrelin, a hormone secreted by the stomach, binds to the growth hormone secretagogue receptor (GHSR) in various brain regions to produce a number of behavioral effects that include increased feeding motivation. During social defeat stress, ghrelin levels rise in correlation with increased feeding and potentially play a role in attenuating the anxiogenic effects of social defeat. One region implicated in the feeding effects of ghrelin is the ventral tegmental area (VTA), a region implicated in reward seeking behaviors, and linked to social defeat in mice. Here we examined the role of GHSR signaling in the VTA in feeding behavior in mice exposed to social defeat stress. Male C57BL/J6 mice that were socially defeated once daily for 3 weeks ate more, had higher plasma ghrelin level and increased GHSR expression in the VTA compared to non-stressed mice. Socially defeated GHSR KO mice failed to increase their caloric intake in response to this stressor but rescue of GHSR expression in the VTA restored feeding responses. Finally, we pharmacologically blocked VTA GHSR signalling with JMV2959 infused via an indwelling VTA cannula connected to a minipump. Vehicle-treated mice increased their caloric intake during social defeat, but JMV2959-infusions attenuated feeding responses and increased anxiety-like behaviors. The data suggest that GHSR signalling in the VTA is critical for the increases in appetite observed during chronic social defeat stress. Furthermore, these data support the idea that GHSR signaling in the VTA may also have anxiolytic effects, and blocking GHSR in this region may result in an anxiety-like phenotype.
Collapse
Affiliation(s)
- Andrea Smith
- Carleton University, Neuroscience Department, Ottawa, ON, Canada
| | - Trevor Rodrigues
- Carleton University, Neuroscience Department, Ottawa, ON, Canada
| | - Caroline Wallace
- Carleton University, Neuroscience Department, Ottawa, ON, Canada
| | - Karen Mezher
- Carleton University, Neuroscience Department, Ottawa, ON, Canada
| | - Brenna MacAulay
- Carleton University, Neuroscience Department, Ottawa, ON, Canada
| | - Rebecca Prowse
- Carleton University, Neuroscience Department, Ottawa, ON, Canada
| | - Lindsay Hyland
- Carleton University, Neuroscience Department, Ottawa, ON, Canada
| | - Alfonso Abizaid
- Carleton University, Neuroscience Department, Ottawa, ON, Canada.
| |
Collapse
|
7
|
Wang Y, Dong Z, An Z, Jin W. Cancer cachexia: Focus on cachexia factors and inter-organ communication. Chin Med J (Engl) 2024; 137:44-62. [PMID: 37968131 PMCID: PMC10766315 DOI: 10.1097/cm9.0000000000002846] [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: 05/25/2023] [Indexed: 11/17/2023] Open
Abstract
ABSTRACT Cancer cachexia is a multi-organ syndrome and closely related to changes in signal communication between organs, which is mediated by cancer cachexia factors. Cancer cachexia factors, being the general name of inflammatory factors, circulating proteins, metabolites, and microRNA secreted by tumor or host cells, play a role in secretory or other organs and mediate complex signal communication between organs during cancer cachexia. Cancer cachexia factors are also a potential target for the diagnosis and treatment. The pathogenesis of cachexia is unclear and no clear effective treatment is available. Thus, the treatment of cancer cachexia from the perspective of the tumor ecosystem rather than from the perspective of a single molecule and a single organ is urgently needed. From the point of signal communication between organs mediated by cancer cachexia factors, finding a deeper understanding of the pathogenesis, diagnosis, and treatment of cancer cachexia is of great significance to improve the level of diagnosis and treatment. This review begins with cancer cachexia factors released during the interaction between tumor and host cells, and provides a comprehensive summary of the pathogenesis, diagnosis, and treatment for cancer cachexia, along with a particular sight on multi-organ signal communication mediated by cancer cachexia factors. This summary aims to deepen medical community's understanding of cancer cachexia and may conduce to the discovery of new diagnostic and therapeutic targets for cancer cachexia.
Collapse
Affiliation(s)
- Yongfei Wang
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu 730000, China
- Institute of Cancer Neuroscience, Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu 730000, China
| | - Zikai Dong
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu 730000, China
- Institute of Cancer Neuroscience, Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu 730000, China
| | - Ziyi An
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu 730000, China
- Institute of Cancer Neuroscience, Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu 730000, China
| | - Weilin Jin
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu 730000, China
- Institute of Cancer Neuroscience, Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu 730000, China
| |
Collapse
|
8
|
Sadee W. Ligand-Free Signaling of G-Protein-Coupled Receptors: Physiology, Pharmacology, and Genetics. Molecules 2023; 28:6375. [PMID: 37687205 PMCID: PMC10489045 DOI: 10.3390/molecules28176375] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 08/28/2023] [Accepted: 08/30/2023] [Indexed: 09/10/2023] Open
Abstract
G-protein-coupled receptors (GPCRs) are ubiquitous sensors and regulators of cellular functions. Each GPCR exists in complex aggregates with multiple resting and active conformations. Designed to detect weak stimuli, GPCRs can also activate spontaneously, resulting in basal ligand-free signaling. Agonists trigger a cascade of events leading to an activated agonist-receptor G-protein complex with high agonist affinity. However, the ensuing signaling process can further remodel the receptor complex to reduce agonist affinity, causing rapid ligand dissociation. The acutely activated ligand-free receptor can continue signaling, as proposed for rhodopsin and μ opioid receptors, resulting in robust receptor activation at low agonist occupancy with enhanced agonist potency. Continued receptor stimulation can further modify the receptor complex, regulating sustained ligand-free signaling-proposed to play a role in opioid dependence. Basal, acutely agonist-triggered, and sustained elevated ligand-free signaling could each have distinct functions, reflecting multi-state conformations of GPCRs. This review addresses basal and stimulus-activated ligand-free signaling, its regulation, genetic factors, and pharmacological implications, focusing on opioid and serotonin receptors, and the growth hormone secretagogue receptor (GHSR). The hypothesis is proposed that ligand-free signaling of 5-HT2A receptors mediate therapeutic effects of psychedelic drugs. Research avenues are suggested to close the gaps in our knowledge of ligand-free GPCR signaling.
Collapse
Affiliation(s)
- Wolfgang Sadee
- Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA;
- Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA 94158, USA
- Aether Therapeutics Inc., Austin, TX 78756, USA
| |
Collapse
|
9
|
Ghrelin/GHS-R1A antagonism in memory test and its effects on central molecular signaling involved in addiction in rats. Pharmacol Biochem Behav 2023; 224:173528. [PMID: 36870422 DOI: 10.1016/j.pbb.2023.173528] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 12/23/2022] [Accepted: 02/12/2023] [Indexed: 03/06/2023]
Abstract
Central ghrelin signaling seems to play important role in addiction as well as memory processing. Antagonism of the growth hormone secretagogue receptor (GHS-R1A) has been recently proposed as a promising tool for the unsatisfactory drug addiction therapy. However, molecular aspects of GHS-R1A involvement in specific brain regions remain unclear. The present study demonstrated for the first time that acute as well as subchronic (4 days) administration of the experimental GHS-R1A antagonist JMV2959 in usual intraperitoneal doses including 3 mg/kg, had no influence on memory functions tested in the Morris Water Maze in rats as well as no significant effects on the molecular markers linked with memory processing in selected brain areas in rats, specifically on the β-actin, c-Fos, two forms of the calcium/calmodulin-dependent protein kinase II (CaMKII, p-CaMKII) and the cAMP-response element binding protein (CREB, p-CREB), within the medial prefrontal cortex (mPFC), nucleus accumbens (NAc), dorsal striatum, and hippocampus (HIPP). Furthermore, following the methamphetamine intravenous self-administration in rats, the 3 mg/kg JMV2959 pretreatment significantly reduced or prevented the methamphetamine-induced significant decrease of hippocampal β-actin and c-Fos as well as it prevented the significant decrease of CREB in the NAC and mPFC. These results imply, that the GHS-R1A antagonist/JMV2959 might reduce/prevent some of the memory-linked molecular changes elicited by methamphetamine addiction within brain structures associated with memory (HIPP), reward (NAc), and motivation (mPFC), which may contribute to the previously observed significant JMV2959-induced reduction of the methamphetamine self-administration and drug-seeking behavior in the same animals. Further research is necessary to corroborate these results.
Collapse
|
10
|
Yanik T, Durhan ST. Specific Functions of Melanocortin 3 Receptor (MC3R). J Clin Res Pediatr Endocrinol 2023; 15:1-6. [PMID: 36053086 PMCID: PMC9976164 DOI: 10.4274/jcrpe.galenos.2022.2022-5-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Melanocortin 3 receptor (MC3R) is a G-protein coupled receptor which has been defined mostly as a regulator of the appetite/hunger balance mechanisms to date. In addition to its function regarding the weight gain and appetite control mechanisms of MC3R, recent studies have shown that MC3R controls growth, puberty, and circadian rhythms as well. Despite the drastic effects of MC3R deficiency in humans and other mammals, its cellular mechanisms are still under investigation. In this review paper, we aimed to point out the importance of MC3R regulations in three main areas: 1) its impact on weight and appetite control, 2) its role in the control of growth, puberty, and the circadian rhythm, and, 3) its protein-protein interactions and cellular mechanisms.
Collapse
Affiliation(s)
- Tulin Yanik
- Middle East Technical University, Department of Biological Sciences, Ankara, Turkey,* Address for Correspondence: Middle East Technical University, Department of Biological Sciences, Ankara, Turkey Phone: +90 312 210 64 65 E-mail:
| | - Seyda Tugce Durhan
- Middle East Technical University, Department of Biochemistry, Ankara, Turkey
| |
Collapse
|
11
|
Gross JD, Zhou Y, Barak LS, Caron MG. Ghrelin receptor signaling in health and disease: a biased view. Trends Endocrinol Metab 2023; 34:106-118. [PMID: 36567228 PMCID: PMC9852078 DOI: 10.1016/j.tem.2022.12.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 11/23/2022] [Accepted: 12/06/2022] [Indexed: 12/25/2022]
Abstract
As allosteric complexes, G-protein-coupled receptors (GPCRs) respond to extracellular stimuli and pleiotropically couple to intracellular transducers to elicit signaling pathway-dependent effects in a process known as biased signaling or functional selectivity. One such GPCR, the ghrelin receptor (GHSR1a), has a crucial role in restoring and maintaining metabolic homeostasis during disrupted energy balance. Thus, pharmacological modulation of GHSR1a bias could offer a promising strategy to treat several metabolism-based disorders. Here, we summarize current evidence supporting GHSR1a functional selectivity in vivo and highlight recent structural data. We propose that precise determinations of GHSR1a molecular pharmacology and pathway-specific physiological effects will enable discovery of GHSR1a drugs with tailored signaling profiles, thereby providing safer and more effective treatments for metabolic diseases.
Collapse
Affiliation(s)
- Joshua D Gross
- Department of Cell Biology, Duke University, Durham, NC 27710, USA
| | - Yang Zhou
- Department of Cell Biology, Duke University, Durham, NC 27710, USA
| | - Lawrence S Barak
- Department of Cell Biology, Duke University, Durham, NC 27710, USA.
| | - Marc G Caron
- Department of Cell Biology, Duke University, Durham, NC 27710, USA
| |
Collapse
|
12
|
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
|
13
|
O’Reilly C, Lin L, Wang H, Fluckey J, Sun Y. Ablation of Ghrelin Receptor Mitigates the Metabolic Decline of Aging Skeletal Muscle. Genes (Basel) 2022; 13:1368. [PMID: 36011279 PMCID: PMC9407208 DOI: 10.3390/genes13081368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/23/2022] [Accepted: 07/26/2022] [Indexed: 02/04/2023] Open
Abstract
The orexigenic hormone ghrelin has multifaceted roles in health and disease. We have reported that ablation of the ghrelin receptor, growth hormone secretagogue receptor (GHS-R), protects against metabolic dysfunction of adipose tissues in aging. Our further observation interestingly revealed that GHS-R deficiency phenocopies the effects of myokine irisin. In this study, we aim to determine whether GHS-R affects the metabolic functions of aging skeletal muscle and whether GHS-R regulates the muscular functions via irisin. We first studied the expression of metabolic signature genes in gastrocnemius muscle of young, middle-aged and old mice. Then, old GHS-R knockout (Ghsr-/-) mice and their wild type counterparts were used to assess the impact of GHS-R ablation on the metabolic characteristics of gastrocnemius and soleus muscle. There was an increase of GHS-R expression in skeletal muscle during aging, inversely correlated with the decline of metabolic functions. Remarkedly the muscle of old GHS-R knockout (Ghsr-/-) mice exhibited a youthful metabolic profile and better maintenance of oxidative type 2 muscle fibers. Furthermore, old Ghsr-/- mice showed improved treadmill performance, supporting better functionality. Also intriguing to note was the fact that old GHS-R-ablated mice showed increased expression of the irisin precursor FNDC5 in the muscle and elevated plasma irisin levels in circulation, which supports a potential interrelationship between GHS-R and irisin. Overall, our work suggests that GHS-R has deleterious effects on the metabolism of aging muscle, which may be at least partially mediated by myokine irisin.
Collapse
Affiliation(s)
- Colleen O’Reilly
- Department of Health and Kinesiology, Texas A & M University, College Station, TX 77843, USA;
| | - Ligen Lin
- USDA/ARS Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA;
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, China
| | - Hongying Wang
- Department of Nutrition, Texas A & M University, College Station, TX 77843, USA;
| | - James Fluckey
- Department of Health and Kinesiology, Texas A & M University, College Station, TX 77843, USA;
| | - Yuxiang Sun
- USDA/ARS Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA;
- Department of Nutrition, Texas A & M University, College Station, TX 77843, USA;
| |
Collapse
|
14
|
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
|
15
|
Wellman M, Budin R, Woodside B, Abizaid A. Energetic demands of lactation produce an increase in the expression of growth hormone secretagogue receptor in the hypothalamus and ventral tegmental area of the rat despite a reduction in circulating ghrelin. J Neuroendocrinol 2022; 34:e13126. [PMID: 35365872 DOI: 10.1111/jne.13126] [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] [Received: 11/05/2021] [Revised: 01/07/2022] [Accepted: 02/08/2022] [Indexed: 12/26/2022]
Abstract
Lactating rats show changes in the secretion of hormones and brain signals that promote hyperphagia and facilitate the production of milk. Little is known, however, about the role of ghrelin in the mechanisms sustaining lactational hyperphagia. Here, we used Wistar female rats that underwent surgery to sever the galactophores to prevent milk delivery (GC rats) and decrease the energetic drain of milk delivery. We compared plasma acyl-ghrelin concentrations and growth hormone secretagogue receptor (GHSR) mRNA expression in different brain regions of GC rats with those of sham operated lactating and nonlactating rats. Additional lactating and nonlactating rats were implanted with cannulae aimed at the lateral ventricles and were used to compare feeding responses to central ghrelin or GHSR antagonist infusions to those of nonlactating rats receiving similar infusions on day 14-16 postpartum (pp). Results show lower plasma acyl-ghrelin concentrations on day 15 pp sham operated lactating rats compared to GC or nonlactating rats. These changes occur in association with increased GHSR mRNA expression in the hypothalamic arcuate nucleus (ARC) and ventral tegmental area (VTA) of sham operated lactating rats. Despite lactational hyperphagia, infusions of ghrelin (0.25 or 1 μg) resulted in similar increases in food intake in lactating and nonlactating rats. In addition, infusions of the GHSR antagonist JMV3002 (4 μg in 1 μl of vehicle) produced greater suppression of food intake in lactating rats than in nonlactating rats. These data suggest that, despite lower plasma ghrelin, the energetic drain of lactation increases sensitivity to the orexigenic effects of ghrelin in brain regions important for food intake and energy balance, and these events are associated with lactational hyperphagia.
Collapse
Affiliation(s)
- Martin Wellman
- Neuroscience Department, Carleton University, Ottawa, Ontario, Canada
| | - Radek Budin
- Centre for Studies in Behavioural Neurobiology, Psychology Department, Concordia University, Montreal, Quebec, Canada
| | - Barbara Woodside
- Centre for Studies in Behavioural Neurobiology, Psychology Department, Concordia University, Montreal, Quebec, Canada
| | - Alfonso Abizaid
- Neuroscience Department, Carleton University, Ottawa, Ontario, Canada
| |
Collapse
|
16
|
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
|
17
|
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
|
18
|
Hölscher C. Protective properties of GLP-1 and associated peptide hormones in neurodegenerative disorders. Br J Pharmacol 2022; 179:695-714. [PMID: 33900631 PMCID: PMC8820183 DOI: 10.1111/bph.15508] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 04/21/2021] [Accepted: 04/22/2021] [Indexed: 12/11/2022] Open
Abstract
Type 2 diabetes mellitus and the associated desensitisation of insulin signalling has been identified as a risk factor for progressive neurodegenerative disorders such as Alzheimer's disease, Parkinson's disease and others. Glucagon-like peptide 1 (GLP-1) is a hormone that has growth factor-like and neuroprotective properties. Several clinical trials have been conducted, testing GLP-1 receptor agonists in patients with Alzheimer's disease, Parkinson's disease or diabetes-induced memory impairments. The trials showed clear improvements in Alzheimer's disease, Parkinson's disease and diabetic patients. Glucose-dependent insulinotropic polypeptide/gastric inhibitory peptide (GIP) is the 'sister' incretin hormone of GLP-1. GIP analogues have shown neuroprotective effects in animal models of disease and can improve on the effects of GLP-1. Novel dual GLP-1/GIP receptor agonists have been developed that can enter the brain at an enhanced rate. The improved neuroprotective effects of these drugs suggest that they are superior to single GLP-1 receptor agonists and could provide disease-modifying care for Alzheimer's disease and Parkinson's disease patients. LINKED ARTICLES: This article is part of a themed issue on GLP1 receptor ligands (BJP 75th Anniversary). To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v179.4/issuetoc.
Collapse
Affiliation(s)
- Christian Hölscher
- The Second Associated Hospital, Neurology DepartmentShanxi Medical UniversityTaiyuanChina
- Academy of Chinese Medical ScienceHenan University of Chinese MedicineZhengzhouChina
| |
Collapse
|
19
|
Sustkova-Fiserova M, Charalambous C, Khryakova A, Certilina A, Lapka M, Šlamberová R. The Role of Ghrelin/GHS-R1A Signaling in Nonalcohol Drug Addictions. Int J Mol Sci 2022; 23:761. [PMID: 35054944 PMCID: PMC8776007 DOI: 10.3390/ijms23020761] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 01/05/2022] [Accepted: 01/06/2022] [Indexed: 01/27/2023] Open
Abstract
Drug addiction causes constant serious health, social, and economic burden within the human society. The current drug dependence pharmacotherapies, particularly relapse prevention, remain limited, unsatisfactory, unreliable for opioids and tobacco, and even symptomatic for stimulants and cannabinoids, thus, new more effective treatment strategies are researched. The antagonism of the growth hormone secretagogue receptor type A (GHS-R1A) has been recently proposed as a novel alcohol addiction treatment strategy, and it has been intensively studied in experimental models of other addictive drugs, such as nicotine, stimulants, opioids and cannabinoids. The role of ghrelin signaling in these drugs effects has also been investigated. The present review aims to provide a comprehensive overview of preclinical and clinical studies focused on ghrelin's/GHS-R1A possible involvement in these nonalcohol addictive drugs reinforcing effects and addiction. Although the investigation is still in its early stage, majority of the existing reviewed experimental results from rodents with the addition of few human studies, that searched correlations between the genetic variations of the ghrelin signaling or the ghrelin blood content with the addictive drugs effects, have indicated the importance of the ghrelin's/GHS-R1As involvement in the nonalcohol abused drugs pro-addictive effects. Further research is necessary to elucidate the exact involved mechanisms and to verify the future potential utilization and safety of the GHS-R1A antagonism use for these drug addiction therapies, particularly for reducing the risk of relapse.
Collapse
Affiliation(s)
- Magdalena Sustkova-Fiserova
- Department of Pharmacology, Third Faculty of Medicine, Charles University, Ruska 87, 100 00 Prague, Czech Republic; (C.C.); (A.K.); (A.C.); (M.L.)
| | - Chrysostomos Charalambous
- Department of Pharmacology, Third Faculty of Medicine, Charles University, Ruska 87, 100 00 Prague, Czech Republic; (C.C.); (A.K.); (A.C.); (M.L.)
| | - Anna Khryakova
- Department of Pharmacology, Third Faculty of Medicine, Charles University, Ruska 87, 100 00 Prague, Czech Republic; (C.C.); (A.K.); (A.C.); (M.L.)
| | - Alina Certilina
- Department of Pharmacology, Third Faculty of Medicine, Charles University, Ruska 87, 100 00 Prague, Czech Republic; (C.C.); (A.K.); (A.C.); (M.L.)
| | - Marek Lapka
- Department of Pharmacology, Third Faculty of Medicine, Charles University, Ruska 87, 100 00 Prague, Czech Republic; (C.C.); (A.K.); (A.C.); (M.L.)
| | - Romana Šlamberová
- Department of Physiology, Third Faculty of Medicine, Charles University, Ke Karlovu 4, 120 00 Prague, Czech Republic;
| |
Collapse
|
20
|
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
|
21
|
Nakanishi Y, Higuchi J, Honda N, Komura N. [Pharmacological profile and clinical efficacy of anamorelin HCl (ADLUMIZ ®Tablets), the first orally available drug for cancer cachexia with ghrelin-like action in Japan]. Nihon Yakurigaku Zasshi 2021; 156:370-381. [PMID: 34719572 DOI: 10.1254/fpj.21046] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Anamorelin hydrochloride (hereinafter referred to as anamorelin) is an orally active, small-molecule drug with a similar pharmacological action to ghrelin, an endogenous ligand of growth hormone secretagogue receptor type 1a (GHS-R1a). It was first approved in Japan for the treatment of cancer cachexia, characterized by weight loss and anorexia. Anamorelin stimulated the secretion of growth hormone (GH) from cultured rat pituitary cells and increased plasma GH levels by oral administration to rats, pigs and humans. When anamorelin was orally administered once daily for 6 days to rats, larger body weight gain associated with increased food consumption compared to the control group was observed from after the first dose. Anamorelin is a selective agonist for GHS-R1a and enhanced GHS-R1a-mediated pituitary GH secretion and increased food consumption, resulting in body weight gain. In the two Japanese phase II studies in patients with cancer cachexia associated with non-small cell lung cancer (NSCLC), improvement of lean body mass (LBM) and body weight losses and anorexia were demonstrated. The tumor types of target patients in the Japanese phase III study were colorectal, gastric, and pancreatic cancer. As a result, maintenance and increase of LBM and body weight as well as improvement of anorexia were observed, and the efficacy against cancer cachexia associated with colorectal, gastric, and pancreatic cancer was confirmed. There were no observed events considered to be significant safety risks. In conclusion, anamorelin is expected to provide a new therapeutic option for cancer cachexia for which no effective treatment has been available.
Collapse
Affiliation(s)
- Yasutomo Nakanishi
- Discovery & Research, Research Center of Specialty, Ono Pharmaceutical Co., Ltd
| | - Junya Higuchi
- Discovery & Research, Research promotion, Ono Pharmaceutical Co., Ltd
| | | | - Naoyuki Komura
- Clinical Development, Clinical Development Planning II, Ono Pharmaceutical Co., Ltd
| |
Collapse
|
22
|
Airapetov MI, Eresko SO, Lebedev AA, Bychkov ER, Shabanov PD. Expression of the growth hormone secretagogue receptor 1a (GHS-R1a) in the brain. Physiol Rep 2021; 9:e15113. [PMID: 34755494 PMCID: PMC8578894 DOI: 10.14814/phy2.15113] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 10/23/2021] [Accepted: 10/26/2021] [Indexed: 12/17/2022] Open
Abstract
The review presents data on the expression of growth hormone secretagogue receptor 1a (GHS-R1a) in the brain regions in model animals (zebrafish, rodents, primates), and in the human brain. Studies show widespread distribution of the receptor in the brain, which evidences the involvement of the receptor in many physiological processes. Using various organisms, data have been obtained regarding the participation of the GHS-R1a in the regulation of the anti- and pro-inflammatory response, proliferation, and apoptosis. It is known that the receptor plays an important role in eating behavior and is also involved in the pathogenetic mechanisms of drug addiction, obesity, and chronic alcohol consumption. Based on this, research is underway with the use of various therapeutic agents that can be used for the pharmacological correction of these conditions. This review also presents hypothetical pathways of intracellular signaling, in which GHS-R1a may participate. A complete understanding of these mechanisms has not yet been reached. The ghrelin intracellular signaling seem to be specific to brain region and, probably, also depend on the metabolic or stress status of the organism.
Collapse
Affiliation(s)
- Marat I. Airapetov
- Department of NeuropharmacologyInstitute of Experimental MedicineSt. PetersburgRussia
- Department of PharmacologySt. Petersburg State Pediatric Medical UniversitySt. PetersburgRussia
| | - Sergei O. Eresko
- Department of NeuropharmacologyInstitute of Experimental MedicineSt. PetersburgRussia
- Research and Training Center of Molecular and Cellular TechnologiesSt. Petersburg State Chemical Pharmaceutical UniversitySt PetersburgRussia
- Department of BiologySaint‐Petersburg State UniversitySt PetersburgRussia
| | - Andrei A. Lebedev
- Department of NeuropharmacologyInstitute of Experimental MedicineSt. PetersburgRussia
| | - Evgenii R. Bychkov
- Department of NeuropharmacologyInstitute of Experimental MedicineSt. PetersburgRussia
- Department of PharmacologyKirov Military Medical AcademySt. PetersburgRussia
| | - Petr D. Shabanov
- Department of NeuropharmacologyInstitute of Experimental MedicineSt. PetersburgRussia
- Department of PharmacologyKirov Military Medical AcademySt. PetersburgRussia
| |
Collapse
|
23
|
de Melo Reis RA, Isaac AR, Freitas HR, de Almeida MM, Schuck PF, Ferreira GC, Andrade-da-Costa BLDS, Trevenzoli IH. Quality of Life and a Surveillant Endocannabinoid System. Front Neurosci 2021; 15:747229. [PMID: 34776851 PMCID: PMC8581450 DOI: 10.3389/fnins.2021.747229] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 10/01/2021] [Indexed: 12/11/2022] Open
Abstract
The endocannabinoid system (ECS) is an important brain modulatory network. ECS regulates brain homeostasis throughout development, from progenitor fate decision to neuro- and gliogenesis, synaptogenesis, brain plasticity and circuit repair, up to learning, memory, fear, protection, and death. It is a major player in the hypothalamic-peripheral system-adipose tissue in the regulation of food intake, energy storage, nutritional status, and adipose tissue mass, consequently affecting obesity. Loss of ECS control might affect mood disorders (anxiety, hyperactivity, psychosis, and depression), lead to drug abuse, and impact neurodegenerative (Alzheimer's, Parkinson, Huntington, Multiple, and Amyotrophic Lateral Sclerosis) and neurodevelopmental (autism spectrum) disorders. Practice of regular physical and/or mind-body mindfulness and meditative activities have been shown to modulate endocannabinoid (eCB) levels, in addition to other players as brain-derived neurotrophic factor (BDNF). ECS is involved in pain, inflammation, metabolic and cardiovascular dysfunctions, general immune responses (asthma, allergy, and arthritis) and tumor expansion, both/either in the brain and/or in the periphery. The reason for such a vast impact is the fact that arachidonic acid, a precursor of eCBs, is present in every membrane cell of the body and on demand eCBs synthesis is regulated by electrical activity and calcium shifts. Novel lipid (lipoxins and resolvins) or peptide (hemopressin) players of the ECS also operate as regulators of physiological allostasis. Indeed, the presence of cannabinoid receptors in intracellular organelles as mitochondria or lysosomes, or in nuclear targets as PPARγ might impact energy consumption, metabolism and cell death. To live a better life implies in a vigilant ECS, through healthy diet selection (based on a balanced omega-3 and -6 polyunsaturated fatty acids), weekly exercises and meditation therapy, all of which regulating eCBs levels, surrounded by a constructive social network. Cannabidiol, a diet supplement has been a major player with anti-inflammatory, anxiolytic, antidepressant, and antioxidant activities. Cognitive challenges and emotional intelligence might strengthen the ECS, which is built on a variety of synapses that modify human behavior. As therapeutically concerned, the ECS is essential for maintaining homeostasis and cannabinoids are promising tools to control innumerous targets.
Collapse
Affiliation(s)
- Ricardo Augusto de Melo Reis
- Laboratory of Neurochemistry, Institute of Biophysics Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Alinny Rosendo Isaac
- Laboratory of Neurochemistry, Institute of Biophysics Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Hércules Rezende Freitas
- Laboratory of Neuroenergetics and Inborn Errors of Metabolism, Institute of Medical Biochemistry Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Mariana Macedo de Almeida
- Laboratory of Molecular Endocrinology, Institute of Biophysics Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Patricia Fernanda Schuck
- Laboratory of Neuroenergetics and Inborn Errors of Metabolism, Institute of Medical Biochemistry Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gustavo Costa Ferreira
- Laboratory of Neuroenergetics and Inborn Errors of Metabolism, Institute of Medical Biochemistry Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Isis Hara Trevenzoli
- Laboratory of Molecular Endocrinology, Institute of Biophysics Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
24
|
Airapetov MI, Eresko SO, Lebedev AA, Bychkov ER, Shabanov PD. Expression of Ghrelin Receptor GHS-R1a in the Brain (Mini Review). Mol Biol 2021. [DOI: 10.1134/s002689332103002x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
25
|
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
|
26
|
Contribution of growth hormone secretagogue receptor (GHSR) signaling in the ventral tegmental area (VTA) to the regulation of social motivation in male mice. Transl Psychiatry 2021; 11:230. [PMID: 33879778 PMCID: PMC8058340 DOI: 10.1038/s41398-021-01350-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 03/19/2021] [Accepted: 03/30/2021] [Indexed: 01/22/2023] Open
Abstract
Most psychiatric disorders are characterized by deficits in the ability to interact socially with others. Ghrelin, a hormone normally associated with the regulation of glucose utilization and appetite, is also implicated in the modulation of motivated behaviors including those associated with food and sex rewards. Here we hypothesized that deficits in ghrelin receptor (growth hormone secretagogue receptor; GHSR) signaling are also associated with deficits in social motivation in male mice. To test this hypothesis, we compared social motivation in male mice lacking GHSR or mice treated with the GHSR antagonist JMV2959 with that of WT or vehicle-treated mice. GHSR signaling in dopamine cells of the ventral tegmental area (VTA) has been implicated in the control of sexual behavior, thus we further hypothesized that GHSR signaling in the VTA is important for social motivation. Thus, we conducted studies where we delivered JMV2959 to block GHSR in the VTA of mice, and studies where we rescued the expression of GHSR in the VTA of GHSR knockout (KO) mice. Mice lacking GHSR or injected with JMV2959 peripherally for 3 consecutive days displayed lower social motivation as reflected by a longer latency to approach a novel conspecific and shorter interaction time compared to WT or vehicle-treated controls. Furthermore, intra-VTA infusion of JMV2959 resulted in longer latencies to approach a novel conspecific, whereas GHSR KO mice with partial rescue of the GHSR showed decreased latencies to begin a novel social interaction. Together, these data suggest that GHSR in the VTA facilitate social approach in male mice, and GHSR-signaling deficits within the VTA result in reduced motivation to interact socially.
Collapse
|
27
|
Schalla MA, Taché Y, Stengel A. Neuroendocrine Peptides of the Gut and Their Role in the Regulation of Food Intake. Compr Physiol 2021; 11:1679-1730. [PMID: 33792904 DOI: 10.1002/cphy.c200007] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The regulation of food intake encompasses complex interplays between the gut and the brain. Among them, the gastrointestinal tract releases different peptides that communicate the metabolic state to specific nuclei in the hindbrain and the hypothalamus. The present overview gives emphasis on seven peptides that are produced by and secreted from specialized enteroendocrine cells along the gastrointestinal tract in relation with the nutritional status. These established modulators of feeding are ghrelin and nesfatin-1 secreted from gastric X/A-like cells, cholecystokinin (CCK) secreted from duodenal I-cells, glucagon-like peptide 1 (GLP-1), oxyntomodulin, and peptide YY (PYY) secreted from intestinal L-cells and uroguanylin (UGN) released from enterochromaffin (EC) cells. © 2021 American Physiological Society. Compr Physiol 11:1679-1730, 2021.
Collapse
Affiliation(s)
- Martha A Schalla
- Charité Center for Internal Medicine and Dermatology, Department for Psychosomatic Medicine, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Yvette Taché
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, CURE: Digestive Diseases Research Center, David Geffen School of Medicine, UCLA, Los Angeles, California, USA.,VA Greater Los Angeles Healthcare System, Los Angeles, California, USA
| | - Andreas Stengel
- Charité Center for Internal Medicine and Dermatology, Department for Psychosomatic Medicine, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany.,Department of Psychosomatic Medicine and Psychotherapy, Medical University Hospital Tübingen, Tübingen, Germany
| |
Collapse
|
28
|
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
|
29
|
Cannabinoid-Induced Conditioned Place Preference, Intravenous Self-Administration, and Behavioral Stimulation Influenced by Ghrelin Receptor Antagonism in Rats. Int J Mol Sci 2021; 22:ijms22052397. [PMID: 33673659 PMCID: PMC7957642 DOI: 10.3390/ijms22052397] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 02/13/2021] [Accepted: 02/21/2021] [Indexed: 11/24/2022] Open
Abstract
Cannabis/cannabinoids are widely used for recreational and therapy purposes, but their risks are largely disregarded. However, cannabinoid-associated use disorders and dependence are alarmingly increasing and an effective treatment is lacking. Recently, the growth hormone secretagogue receptor (GHSR1A) antagonism was proposed as a promising mechanism for drug addiction therapy. However, the role of GHS-R1A and its endogenous ligand ghrelin in cannabinoid abuse remains unclear. Therefore, the aim of our study was to investigate whether the GHS-R1A antagonist JMV2959 could reduce the tetrahydrocannabinol (THC)-induced conditioned place preference (CPP) and behavioral stimulation, the WIN55,212-2 intravenous self-administration (IVSA), and the tendency to relapse. Following an ongoing WIN55,212-2 self-administration, JMV2959 3 mg/kg was administered intraperitoneally 20 min before three consequent daily 120-min IVSA sessions under a fixed ratio FR1, which significantly reduced the number of the active lever-pressing, the number of infusions, and the cannabinoid intake. Pretreatment with JMV2959 suggested reduction of the WIN55,212-2-seeking/relapse-like behavior tested in rats on the twelfth day of the forced abstinence period. On the contrary, pretreatment with ghrelin significantly increased the cannabinoid IVSA as well as enhanced the relapse-like behavior. Co-administration of ghrelin with JMV2959 abolished/reduced the significant efficacy of the GHS-R1A antagonist in the cannabinoid IVSA. Pretreatment with JMV2959 significantly and dose-dependently reduced the manifestation of THC-induced CPP. The THC-CPP development was reduced after the simultaneous administration of JMV2959 with THC during conditioning. JMV2959 also significantly reduced the THC-induced behavioral stimulation in the LABORAS cage. Our findings suggest that GHS-R1A importantly participates in the rewarding/reinforcing effects of cannabinoids.
Collapse
|
30
|
Charalambous C, Lapka M, Havlickova T, Syslova K, Sustkova-Fiserova M. Alterations in Rat Accumbens Dopamine, Endocannabinoids and GABA Content During WIN55,212-2 Treatment: The Role of Ghrelin. Int J Mol Sci 2020; 22:ijms22010210. [PMID: 33379212 PMCID: PMC7795825 DOI: 10.3390/ijms22010210] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 12/21/2020] [Accepted: 12/24/2020] [Indexed: 01/22/2023] Open
Abstract
The endocannabinoid/CB1R system as well as the central ghrelin signalling with its growth hormone secretagogoue receptors (GHS-R1A) are importantly involved in food intake and reward/reinforcement processing and show distinct overlaps in distribution within the relevant brain regions including the hypothalamus (food intake), the ventral tegmental area (VTA) and the nucleus accumbens (NAC) (reward/reinforcement). The significant mutual interaction between these systems in food intake has been documented; however, the possible role of ghrelin/GHS-R1A in the cannabinoid reinforcement effects and addiction remain unclear. Therefore, the principal aim of the present study was to investigate whether pretreatment with GHS-R1A antagonist/JMV2959 could reduce the CB1R agonist/WIN55,212-2–induced dopamine efflux in the nucleus accumbens shell (NACSh), which is considered a crucial trigger impulse of the addiction process. The synthetic aminoalklylindol cannabinoid WIN55,212-2 administration into the posterior VTA induced significant accumbens dopamine release, which was significantly reduced by the 3 mg/kg i.p. JMV2959 pretreatment. Simultaneously, the cannabinoid-increased accumbens dopamine metabolic turnover was significantly augmented by the JMV2959 pretreament. The intracerebral WIN55,212-2 administration also increased the endocannabinoid arachidonoylethanolamide/anandamide and the 2-arachidonoylglycerol/2-AG extracellular levels in the NACSh, which was moderately but significantly attenuated by the JMV2959 pretreatment. Moreover, the cannabinoid-induced decrease in accumbens γ-aminobutyric acid/gamma-aminobutyric acid levels was reversed by the JMV2959 pretreatment. The behavioural study in the LABORAS cage showed that 3 mg/kg JMV2959 pretreatment also significantly reduced the systemic WIN55,212-2-induced behavioural stimulation. Our results demonstrate that the ghrelin/GHS-R1A system significantly participates in the rewarding/reinforcing effects of the cannabinoid/CB1 agonist that are involved in cannabinoid addiction processing.
Collapse
Affiliation(s)
- Chrysostomos Charalambous
- Department of Addictology, First Faculty of Medicine, Charles University, Apolinarska 4, 128 00 Prague 2, Czech Republic;
| | - Marek Lapka
- Department of Pharmacology, Third Faculty of Medicine, Charles University, Ruska 87, 100 34 Prague 10, Czech Republic; (M.L.); (T.H.)
| | - Tereza Havlickova
- Department of Pharmacology, Third Faculty of Medicine, Charles University, Ruska 87, 100 34 Prague 10, Czech Republic; (M.L.); (T.H.)
| | - Kamila Syslova
- Laboratory of Medicinal Diagnostics, Department of Organic Technology, University of Chemistry and Technology Prague, Technicka 5, 166 28 Prague 6, Czech Republic;
| | - Magdalena Sustkova-Fiserova
- Department of Pharmacology, Third Faculty of Medicine, Charles University, Ruska 87, 100 34 Prague 10, Czech Republic; (M.L.); (T.H.)
- Correspondence: ; Tel.: +420-267-102-450; Fax: +420-267-102-461
| |
Collapse
|
31
|
Xiao X, Bi M, Jiao Q, Chen X, Du X, Jiang H. A new understanding of GHSR1a--independent of ghrelin activation. Ageing Res Rev 2020; 64:101187. [PMID: 33007437 DOI: 10.1016/j.arr.2020.101187] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/13/2020] [Accepted: 09/21/2020] [Indexed: 12/13/2022]
Abstract
Growth hormone secretagogue receptor 1a (GHSR1a), a member of the G protein-coupled receptor (GPCR) family, is a functional receptor of ghrelin. The expression levels and activities of GHSR1a are affected by various factors. In past years, it has been found that the ghrelin-GHSR1a system can perform biological functions such as anti-inflammation, anti-apoptosis, and anti-oxidative stress. In addition to mediating the effect of ghrelin, GHSR1a also has abnormally high constitutive activity; that is, it can still transmit intracellular signals without activation of the ghrelin ligand. This constitutive activity affects brain functions, growth and development of the body; therefore, it has profound impacts on neurodegenerative diseases and some other age-related diseases. In addition, GHSR1a can also form homodimers or heterodimers with other GPCRs, affecting the release of neurotransmitters, appetite regulation, cell proliferation and insulin release. Therefore, further understanding of the constitutive activities and dimerization of GHSR1a will enable us to better clarify the characteristics of GHSR1a and provide more therapeutic targets for drug development. Here, we focus on the roles of GHSR1a in various biological functions and provide a comprehensive summary of the current research on GHSR1a to provide broader therapeutic prospects for age-related disease treatment.
Collapse
Affiliation(s)
- Xue Xiao
- 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, 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, 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, China
| | - 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, 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, 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, China.
| |
Collapse
|
32
|
Stone LA, Harmatz ES, Goosens KA. Ghrelin as a Stress Hormone: Implications for Psychiatric Illness. Biol Psychiatry 2020; 88:531-540. [PMID: 32912426 DOI: 10.1016/j.biopsych.2020.05.013] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 05/11/2020] [Accepted: 05/11/2020] [Indexed: 12/28/2022]
Abstract
The stress response is an adaptive means of maintaining physiological homeostasis in the face of changing environmental conditions. However, protracted recruitment of stress systems can precipitate wear and tear on the body and may lead to many forms of disease. The mechanisms underlying the connection between chronic stress and disease are not fully understood and are likely multifactorial. In this review, we evaluate the possibility that the hormone ghrelin may contribute to the pathophysiology that follows chronic stress. Since ghrelin was discovered as a pro-hunger hormone, many additional roles for it have been identified, including in learning, memory, reward, and stress. We describe the beneficial effects that ghrelin exerts in healthy mammals and discuss that prolonged exposure to ghrelin has been linked to maladaptive responses and behaviors in the realm of psychiatric disease. In addition, we consider whether chronic stress-associated altered ghrelin signaling may enhance susceptibility to posttraumatic stress disorder and comorbid conditions such as major depressive disorder and alcohol use disorder. Finally, we explore the possibility that ghrelin-based therapeutics could eventually form the basis of a treatment strategy for illnesses that are linked to chronic stress and potentially also ghrelin dysregulation, and we identify critical avenues for future research in this regard.
Collapse
Affiliation(s)
| | | | - Ki A Goosens
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York.
| |
Collapse
|
33
|
Pietraszko W, Furgala A, Gorecka-Mazur A, Kwinta B, Kaszuba-Zwoinska J, Polak J, Fiszer U, Gil K, Krygowska-Wajs A. Assessments of plasma acyl-ghrelin levels in Parkinson's disease patients treated with deep brain stimulation. Peptides 2020; 128:170299. [PMID: 32305796 DOI: 10.1016/j.peptides.2020.170299] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 02/26/2020] [Accepted: 03/12/2020] [Indexed: 12/26/2022]
Abstract
Gastrointestinal dysfunction is the most common non-motor symptom in Parkinson's disease (PD) with rates rising as the disease progresses. Deep brain stimulation of subthalamic nucleus (STN DBS) improves motor functions in advanced PD. However, the effect of STN DBS on ghrelin concentration and consequently on motility disturbances as well as body weight is unclear. The objective of this study was to assess acyl-ghrelin levels in comparison to weight in advanced PD patients treated with STN DBS. Plasma concentrations of acyl-ghrelin was measured in 29 PD patients in the fasting state and at 30, 60, 120, and 180 min after a standard meal preoperatively and 3 months after surgery. The level of acyl-ghrelin in PD patients were compared with 30 age and sex-matched healthy controls. We reported that mean plasma acyl-ghrelin levels were decreased in PD patients before STN DBS in fasting (p = 0.0003) and in 30 min postprandial phase (p = 0.04) compared with healthy controls. The plasma acyl-ghrelin levels after STN DBS increased in pre-prandial and postprandial phase in PD patients at the investigated time points. Body weight gained on average 2.33 kg during the first 3 months after surgery. There was no correlation between the acyl-ghrelin plasma levels and BMI. After STN DBS in fasting and postprandial phase plasma acyl-ghrelin levels were increased. The results showed that STN DBS therapy elicited a modification of ghrelin levels, increasing its concentration in pre- and postprandial state. In addition, body weight was increased during 3 months after surgery.
Collapse
Affiliation(s)
- Wojciech Pietraszko
- Department of Neurosurgery, Jagiellonian University, Medical College, Krakow, Botaniczna 3, Poland
| | - Agata Furgala
- Department of Pathophysiology, Jagiellonian University, Medical College, Krakow, Czysta 18, Poland
| | - Agnieszka Gorecka-Mazur
- Department of Pathophysiology, Jagiellonian University, Medical College, Krakow, Czysta 18, Poland
| | - Borys Kwinta
- Department of Neurosurgery, Jagiellonian University, Medical College, Krakow, Botaniczna 3, Poland
| | - Jolanta Kaszuba-Zwoinska
- Department of Pathophysiology, Jagiellonian University, Medical College, Krakow, Czysta 18, Poland
| | - Jaroslaw Polak
- Department of Neurosurgery, Jagiellonian University, Medical College, Krakow, Botaniczna 3, Poland
| | - Urszula Fiszer
- Department of Neurology and Epileptology, Centre of Postgraduate Medical Education, Warsaw, Czerniakowska 231, Poland
| | - Krzysztof Gil
- Department of Pathophysiology, Jagiellonian University, Medical College, Krakow, Czysta 18, Poland
| | - Anna Krygowska-Wajs
- Department of Neurology, Jagiellonian University, Medical College, Krakow, Botaniczna 3, Poland.
| |
Collapse
|
34
|
Chen X, Yu Y, Zheng P, Jin T, He M, Zheng M, Song X, Jones A, Huang XF. Olanzapine increases AMPK-NPY orexigenic signaling by disrupting H1R-GHSR1a interaction in the hypothalamic neurons of mice. Psychoneuroendocrinology 2020; 114:104594. [PMID: 32007669 DOI: 10.1016/j.psyneuen.2020.104594] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 12/22/2019] [Accepted: 01/17/2020] [Indexed: 12/11/2022]
Abstract
Second generation antipsychotics, particularly olanzapine, induce severe obesity, which is associated with their antagonistic effect on the histamine H1 receptor (H1R). We have previously demonstrated that oral administration of olanzapine increases the concentration of neuropeptide Y (NPY) in the hypothalamus of rats, accompanied by hyperphagia and weight gain. However, it is unclear if the increased NPY after olanzapine administration is due to its direct effect on hypothalamic neurons and its H1R antagonistic property. In the present study, we showed that with an inverted U-shape dose-response curve, olanzapine increased NPY expression in the NPY-GFP hypothalamic neurons; however, this was not the case in the hypothalamic neurons of H1R knockout mice. Olanzapine inhibited the interaction of H1R and GHSR1a (ghrelin receptor) in the primary mouse hypothalamic neurons and NPY-GFP neurons examined by confocal fluorescence resonance energy transfer (FRET) technology. Furthermore, an H1R agonist, FMPH inhibited olanzapine activation of GHSR1a downstream signaling pAMPK and transcription factors of NPY (pFOXO1 and pCREB) in the hypothalamic NPY-GFP cell. However, an olanzapine analogue (E-Olan) with lower affinity to H1R presented negligible enhancement of pCREB within the nucleus of NPY neurons. These findings suggest that the H1R antagonist property of olanzapine inhibits the interaction of H1R and GHSR1a, activates GHSR1a downstream signaling pAMPK-FOXO1/pCREB and increases hypothalamic NPY: this could be one of the important molecular mechanisms of H1R antagonism of olanzapine-induced obesity in antipsychotic management of psychiatric disorders.
Collapse
Affiliation(s)
- Xiaoqi Chen
- Department of Endocrinology and Rheumatology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Illawarra Health and Medical Research Institute and Molecular Horizons, School of Medicine, University of Wollongong, NSW, 2522, Australia
| | - Yinghua Yu
- Department of Pathogen Biology and Immunology, Xuzhou Medical University and Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou, Jiangsu 221004, China.
| | - Peng Zheng
- Illawarra Health and Medical Research Institute and Molecular Horizons, School of Medicine, University of Wollongong, NSW, 2522, Australia
| | - Tiantian Jin
- Illawarra Health and Medical Research Institute and Molecular Horizons, School of Medicine, University of Wollongong, NSW, 2522, Australia
| | - Meng He
- School of Chemistry, Wuhan University of Technology, Wuhan, China
| | - Mingxuan Zheng
- Department of Pathogen Biology and Immunology, Xuzhou Medical University and Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou, Jiangsu 221004, China
| | - Xueqin Song
- School of Psychiatry, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Alison Jones
- Illawarra Health and Medical Research Institute and Molecular Horizons, School of Medicine, University of Wollongong, NSW, 2522, Australia
| | - Xu-Feng Huang
- Illawarra Health and Medical Research Institute and Molecular Horizons, School of Medicine, University of Wollongong, NSW, 2522, Australia.
| |
Collapse
|
35
|
Hölscher C. Brain insulin resistance: role in neurodegenerative disease and potential for targeting. Expert Opin Investig Drugs 2020; 29:333-348. [PMID: 32175781 DOI: 10.1080/13543784.2020.1738383] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Introduction: This review evaluates the novel strategy of treating Alzheimer's and Parkinson's disease (AD and PD) withdrugs that initially have been developed to treat type 2 diabetes. As insulin signalling has been found to be de-sensitized in the brains of patients, drugs that can re-sensitize insulin signalling have been tested to evaluate if this strategy can alter disease progression.Areas covered: The review will give an overview of preclinical and clinical tests in AD and PD of drugs activating insulin receptors, glucagon-like peptide -1 (GLP-1) receptors, and glucose-dependent insulinotropic polypeptide (GIP) receptors.Expert opinion: Insulin, GLP-1 and GIP receptor agonists have shown good effects in preclinical studies. First clinical trials in MCI/AD patients have shown that insulin can improve on key pathological symptoms of AD such as memory impairment, brain activity, neuronal energy utilization, and inflammation markers. A GLP-1 receptor agonist has shown disease-modifying effects in PD patients, and first pilot studies have shown encouraging effects of a GLP-1 receptor agonist in AD patients. Novel dual GLP-1/GIP receptor agonists that cross the blood brain barrier show superior neuroprotective effects compared to single GLP-1 or GIP receptor agonists, and show great promise as novel treatments of AD and PD.
Collapse
Affiliation(s)
- Christian Hölscher
- Second Hospital, Neurology Department, Shanxi Medical University, Taiyuan, Shanxi, PR China.,Research and Experimental Center, Henan University of Chinese Medicine, Zhengzhou, Henan, PR China
| |
Collapse
|
36
|
Abizaid A, Hougland JL. Ghrelin Signaling: GOAT and GHS-R1a Take a LEAP in Complexity. Trends Endocrinol Metab 2020; 31:107-117. [PMID: 31636018 PMCID: PMC7299083 DOI: 10.1016/j.tem.2019.09.006] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 09/25/2019] [Accepted: 09/27/2019] [Indexed: 12/30/2022]
Abstract
Ghrelin and the growth hormone secretagogue receptor 1a (GHS-R1a) are important targets for disorders related to energy balance and metabolic regulation. Pharmacological control of ghrelin signaling is a promising avenue to address health issues involving appetite, weight gain, obesity, and related metabolic disorders, and may be an option for patients suffering from wasting conditions like cachexia. In this review, we summarize recent developments in the biochemistry of ghrelin and GHS-R1a signaling. These include unravelling the enzymatic transformations that generate active ghrelin and the discovery of multiple proteins that interact with ghrelin and GHS-R1a to regulate signaling. Furthermore, we propose that harnessing these processes will lead to highly selective treatments to address obesity, diabetes, and other metabolism-linked disorders.
Collapse
Affiliation(s)
- Alfonso Abizaid
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada
| | - James L Hougland
- Department of Chemistry, Syracuse University, Syracuse, NY, USA.
| |
Collapse
|
37
|
Cornejo MP, Castrogiovanni D, Schiöth HB, Reynaldo M, Marie J, Fehrentz JA, Perello M. Growth hormone secretagogue receptor signalling affects high-fat intake independently of plasma levels of ghrelin and LEAP2, in a 4-day binge eating model. J Neuroendocrinol 2019; 31:e12785. [PMID: 31469195 DOI: 10.1111/jne.12785] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 08/16/2019] [Accepted: 08/27/2019] [Indexed: 12/25/2022]
Abstract
The growth hormone secretagogue receptor (GHSR) is a G protein-coupled receptor that is highly expressed in the central nervous system. GHSR acts as a receptor for ghrelin and for liver-expressed antimicrobial peptide 2 (LEAP2), which blocks ghrelin-evoked activity. GHSR also displays ligand-independent activity, including a high constitutive activity that signals in the absence of ghrelin and is reduced by LEAP2. GHSR activity modulates a variety of food intake-related behaviours, including binge eating. Previously, we reported that GHSR-deficient mice daily and time-limited exposed to a high-fat (HF) diet display an attenuated binge-like HF intake compared to wild-type mice. In the present study, we aimed to determine whether ligand-independent GHSR activity affects binge-like HF intake in a 4-day binge-like eating protocol. We found that plasma levels of ghrelin and LEAP2 were not modified in mice exposed to this binge-like eating protocol. Moreover, systemic administration of ghrelin or LEAP2 did not alter HF intake in our experimental conditions. Interestingly, we found that central administration of LEAP2 or K-(D-1-Nal)-FwLL-NH2 , which are both blockers of constitutive GHSR activity, reduced binge-like HF intake, whereas central administration of ghrelin or the ghrelin-evoked GHSR activity blockers [D-Lys3]-GHRP-6 and JMV2959 did not modify binge-like HF intake. Taken together, current data indicate that GHSR activity in the brain affects binge-like HF intake in mice independently of plasma levels of ghrelin and LEAP2.
Collapse
Affiliation(s)
- María Paula Cornejo
- 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, Argentina
| | - Daniel Castrogiovanni
- 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, Argentina
| | - Helgi B Schiöth
- Department of Neuroscience, Functional Pharmacology, Uppsala University, Uppsala, Sweden
- Institute for Translational Medicine and Biotechnology, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Mirta Reynaldo
- 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, Argentina
| | - Jacky Marie
- Faculté de Pharmacie, Institut des Biomolécules Max Mousseron, UMR 5247 CNRS-Université Montpellier-ENSCM, Montpellier, France
| | - Jean-Alain Fehrentz
- Faculté de Pharmacie, Institut des Biomolécules Max Mousseron, UMR 5247 CNRS-Université Montpellier-ENSCM, Montpellier, 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, Argentina
| |
Collapse
|
38
|
Dunn DP, Bastacky JM, Gray CC, Abtahi S, Currie PJ. Role of mesolimbic ghrelin in the acquisition of cocaine reward. Neurosci Lett 2019; 709:134367. [DOI: 10.1016/j.neulet.2019.134367] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 07/01/2019] [Accepted: 07/02/2019] [Indexed: 12/27/2022]
|
39
|
A ghrelin receptor and oxytocin receptor heterocomplex impairs oxytocin mediated signalling. Neuropharmacology 2019; 152:90-101. [DOI: 10.1016/j.neuropharm.2018.12.022] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 11/21/2018] [Accepted: 12/18/2018] [Indexed: 12/31/2022]
|
40
|
Ratcliff M, Rees D, McGrady S, Buntwal L, Hornsby AKE, Bayliss J, Kent BA, Bussey T, Saksida L, Beynon AL, Howell OW, Morgan AH, Sun Y, Andrews ZB, Wells T, Davies JS. Calorie restriction activates new adult born olfactory-bulb neurones in a ghrelin-dependent manner but acyl-ghrelin does not enhance subventricular zone neurogenesis. J Neuroendocrinol 2019; 31:e12755. [PMID: 31179562 DOI: 10.1111/jne.12755] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 06/06/2019] [Accepted: 06/06/2019] [Indexed: 12/25/2022]
Abstract
The ageing and degenerating brain show deficits in neural stem/progenitor cell (NSPC) plasticity that are accompanied by impairments in olfactory discrimination. Emerging evidence suggests that the gut hormone ghrelin plays an important role in protecting neurones, promoting synaptic plasticity and increasing hippocampal neurogenesis in the adult brain. In the present study, we investigated the role of ghrelin with respect to modulating adult subventricular zone (SVZ) NSPCs that give rise to new olfactory bulb (OB) neurones. We characterised the expression of the ghrelin receptor, growth hormone secretagogue receptor (GHSR), using an immunohistochemical approach in GHSR-eGFP reporter mice to show that GHSR is expressed in several regions, including the OB but not in the SVZ of the lateral ventricle. These data suggest that acyl-ghrelin does not mediate a direct effect on NSPC in the SVZ. Consistent with these findings, treatment with acyl-ghrelin or genetic silencing of GHSR did not alter NSPC proliferation within the SVZ. Similarly, using a bromodeoxyuridine pulse-chase approach, we show that peripheral treatment of adult rats with acyl-ghrelin did not increase the number of new adult-born neurones in the granule cell layer of the OB. These data demonstrate that acyl-ghrelin does not increase adult OB neurogenesis. Finally, we investigated whether elevating ghrelin indirectly, via calorie restriction (CR), regulated the activity of new adult-born cells in the OB. Overnight CR induced c-Fos expression in new adult-born OB cells but not in developmentally born cells, whereas neuronal activity was absent following re-feeding. These effects were not present in ghrelin-/- mice, suggesting that adult-born cells are uniquely sensitive to changes in ghrelin mediated by fasting and re-feeding. In summary, ghrelin does not promote neurogenesis in the SVZ and OB; however, new adult-born OB cells are activated by CR in a ghrelin-dependent manner.
Collapse
Affiliation(s)
- Michael Ratcliff
- Molecular Neurobiology, Institute of Life Sciences, School of Medicine, Swansea University, Swansea, UK
| | - Daniel Rees
- Molecular Neurobiology, Institute of Life Sciences, School of Medicine, Swansea University, Swansea, UK
| | - Scott McGrady
- Molecular Neurobiology, Institute of Life Sciences, School of Medicine, Swansea University, Swansea, UK
| | - Luke Buntwal
- Molecular Neurobiology, Institute of Life Sciences, School of Medicine, Swansea University, Swansea, UK
| | - Amanda K E Hornsby
- Molecular Neurobiology, Institute of Life Sciences, School of Medicine, Swansea University, Swansea, UK
| | - Jaqueline Bayliss
- Biomedical Discovery Unit, Department of Physiology, Monash University, Melbourne, Victoria, Australia
| | - Brianne A Kent
- Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | | | | | - Amy L Beynon
- Molecular Neurobiology, Institute of Life Sciences, School of Medicine, Swansea University, Swansea, UK
| | - Owain W Howell
- Molecular Neurobiology, Institute of Life Sciences, School of Medicine, Swansea University, Swansea, UK
| | - Alwena H Morgan
- Molecular Neurobiology, Institute of Life Sciences, School of Medicine, Swansea University, Swansea, UK
| | - Yuxiang Sun
- Department of Nutrition and Food Science, Texas A&M University, College Station, Texas, USA
| | - Zane B Andrews
- Biomedical Discovery Unit, Department of Physiology, Monash University, Melbourne, Victoria, Australia
| | - Timothy Wells
- School of Biosciences, Cardiff University, Cardiff, UK
| | - Jeffrey S Davies
- Molecular Neurobiology, Institute of Life Sciences, School of Medicine, Swansea University, Swansea, UK
| |
Collapse
|
41
|
Serrenho D, Santos SD, Carvalho AL. The Role of Ghrelin in Regulating Synaptic Function and Plasticity of Feeding-Associated Circuits. Front Cell Neurosci 2019; 13:205. [PMID: 31191250 PMCID: PMC6546032 DOI: 10.3389/fncel.2019.00205] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Accepted: 04/24/2019] [Indexed: 12/21/2022] Open
Abstract
Synaptic plasticity of the neuronal circuits associated with feeding behavior is regulated by peripheral signals as a response to changes in the energy status of the body. These signals include glucose, free fatty acids, leptin and ghrelin and are released into circulation, being able to reach the brain. Ghrelin, a small peptide released from the stomach, is an orexigenic hormone produced in peripheral organs, and its action regulates food intake, body weight and glucose homeostasis. Behavioral studies show that ghrelin is implicated in the regulation of both hedonic and homeostatic feeding and of cognition. Ghrelin-induced synaptic plasticity has been described in neuronal circuits associated with these behaviors. In this review, we discuss the neuromodulatory mechanisms induced by ghrelin in regulating synaptic plasticity in three main neuronal circuits previously associated with feeding behaviors, namely hypothalamic (homeostatic feeding), ventral tegmental (hedonic and motivational feeding) and hippocampal (cognitive) circuits. Given the central role of ghrelin in regulating feeding behaviors, and the altered ghrelin levels associated with metabolic disorders such as obesity and anorexia, it is of paramount relevance to understand the effects of ghrelin on synaptic plasticity of neuronal circuits associated with feeding behaviors.
Collapse
Affiliation(s)
- Débora Serrenho
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal.,Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal.,PhD Program in Experimental Biology and Biomedicine (PDBEB), University of Coimbra, Coimbra, Portugal
| | - Sandra D Santos
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal.,Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal
| | - Ana Luísa Carvalho
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal.,Department of Life Sciences, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
42
|
Wenthur CJ, Gautam R, Zhou B, Vendruscolo LF, Leggio L, Janda KD. Ghrelin Receptor Influence on Cocaine Reward is Not Directly Dependent on Peripheral Acyl-Ghrelin. Sci Rep 2019; 9:1841. [PMID: 30755699 PMCID: PMC6372697 DOI: 10.1038/s41598-019-38549-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 12/31/2018] [Indexed: 12/30/2022] Open
Abstract
The peptide hormone acyl-ghrelin and its receptor, GHSR1a, represent intriguing therapeutic targets due to their actions in metabolic homeostasis and reward activity. However, this pleotropic activity makes it difficult to intervene in this system without inducing unwanted effects. Thus, it is desirable to identify passive and active regulatory mechanisms that allow differentiation between functional domains. Anatomical restriction by the blood brain barrier represents one major passive regulatory mechanism. However, it is likely that the ghrelin system is subject to additional passive mechanisms that promote independent regulation of orexigenic behavior and reward processing. By applying acyl-ghrelin sequestering antibodies, it was determined that peripheral sequestration of acyl-ghrelin is sufficient to blunt weight gain, but not cocaine rewarding effects. However, both weight gain and reward-associated behaviors were shown to be blocked by direct antagonism of GHSR1a. Overall, these data indicate that GHSR1a effects on reward are independent from peripheral acyl-ghrelin binding, whereas centrally-mediated alteration of energy storage requires peripheral acyl-ghrelin binding. This demonstration of variable ligand-dependence amongst functionally-distinct GHSR1a populations is used to generate a regulatory model for functional manipulation of specific effects when attempting to therapeutically target the ghrelin system.
Collapse
Affiliation(s)
- Cody J Wenthur
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
- Department of Pharmacy, University of Wisconsin - Madison, Madison, WI, USA
| | - Ritika Gautam
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | - Bin Zhou
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | - Leandro F Vendruscolo
- Neurobiology of Addiction Section, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Lorenzo Leggio
- Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology, National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research and National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Bethesda, MD, USA
- Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, Brown University, Providence, RI, USA
| | - Kim D Janda
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA.
- Department of Immunology and Microbial Science, The Skaggs Institute for Chemical Biology, and The Worm Institute for Research and Medicine (WIRM), The Scripps Research Institute, La Jolla, CA, USA.
| |
Collapse
|
43
|
Martínez Damonte V, Rodríguez SS, Raingo J. Growth hormone secretagogue receptor constitutive activity impairs voltage-gated calcium channel-dependent inhibitory neurotransmission in hippocampal neurons. J Physiol 2018; 596:5415-5428. [PMID: 30199095 DOI: 10.1113/jp276256] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 09/06/2018] [Indexed: 12/14/2022] Open
Abstract
KEY POINTS Presynaptic CaV 2 voltage-gated calcium channels link action potentials arriving at the presynaptic terminal to neurotransmitter release. Hence, their regulation is essential to fine tune brain circuitry. CaV 2 channels are highly sensitive to G protein-coupled receptor (GPCR) modulation. Our previous data indicated that growth hormone secretagogue receptor (GHSR) constitutive activity impairs CaV 2 channels by decreasing their surface density. We present compelling support for the impact of CaV 2.2 channel inhibition by agonist-independent GHSR activity exclusively on GABA release in hippocampal cultures. We found that this selectivity arises from a high reliance of GABA release on CaV 2.2 rather than on CaV 2.1 channels. Our data provide new information on the effects of the ghrelin-GHSR system on synaptic transmission, suggesting a putative physiological role of the constitutive signalling of a GPCR that is expressed at high levels in brain areas with restricted access to its natural agonist. ABSTRACT Growth hormone secretagogue receptor (GHSR) displays high constitutive activity, independent of its endogenous ligand, ghrelin. Unlike ghrelin-induced GHSR activity, the physiological role of GHSR constitutive activity and the mechanisms that underlie GHSR neuronal modulation remain elusive. We previously demonstrated that GHSR constitutive activity modulates presynaptic CaV 2 voltage-gated calcium channels. Here we postulate that GHSR constitutive activity-mediated modulation of CaV 2 channels could be relevant in the hippocampus since this brain area has high GHSR expression but restricted access to ghrelin. We performed whole-cell patch-clamp in hippocampal primary cultures from E16- to E18-day-old C57BL6 wild-type and GHSR-deficient mice after manipulating GHSR expression with lentiviral transduction. We found that GHSR constitutive activity impairs CaV 2.1 and CaV 2.2 native calcium currents and that CaV 2.2 basal impairment leads to a decrease in GABA but not glutamate release. We postulated that this selective effect is related to a higher CaV 2.2 over CaV 2.1 contribution to GABA release (∼40% for CaV 2.2 in wild-type vs. ∼20% in wild-type GHSR-overexpressing cultures). This effect of GHSR constitutive activity is conserved in hippocampal brain slices, where GHSR constitutive activity reduces local GABAergic transmission of the granule cell layer (intra-granule cell inhibitory postsynaptic current (IPSC) size ∼-67 pA in wild-type vs. ∼-100 pA in GHSR-deficient mice), whereas the glutamatergic output from the dentate gyrus to CA3 remains unchanged. In summary, we found that GHSR constitutive activity impairs IPSCs both in hippocampal primary cultures and in brain slices through a CaV 2-dependent mechanism without affecting glutamatergic transmission.
Collapse
Affiliation(s)
- Valentina Martínez Damonte
- Multidisciplinary Institute of Cell Biology (IMBICE), National Council of Science and Technology (CONICET), Buenos Aires Comision of Science (CIC) and La Plata University (UNLP), La Plata, Argentina
| | - Silvia Susana Rodríguez
- Multidisciplinary Institute of Cell Biology (IMBICE), National Council of Science and Technology (CONICET), Buenos Aires Comision of Science (CIC) and La Plata University (UNLP), La Plata, Argentina
| | - Jesica Raingo
- Multidisciplinary Institute of Cell Biology (IMBICE), National Council of Science and Technology (CONICET), Buenos Aires Comision of Science (CIC) and La Plata University (UNLP), La Plata, Argentina
| |
Collapse
|
44
|
Xue Q, Bai B, Ji B, Chen X, Wang C, Wang P, Yang C, Zhang R, Jiang Y, Pan Y, Cheng B, Chen J. Ghrelin Through GHSR1a and OX1R Heterodimers Reveals a Gαs-cAMP-cAMP Response Element Binding Protein Signaling Pathway in Vitro. Front Mol Neurosci 2018; 11:245. [PMID: 30065627 PMCID: PMC6056640 DOI: 10.3389/fnmol.2018.00245] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Accepted: 06/25/2018] [Indexed: 01/19/2023] Open
Abstract
Growth hormone secretagogue receptor 1α (GHSR1a) and Orexin 1 receptor (OX1R) are involved in various important physiological processes, and have many similar characteristics in function and distribution in peripheral tissues and the central nervous system. We explored the possibility of heterodimerization between GHSR1a and OX1R and revealed a signal transduction pathway mechanism. In this study, bioluminescence and fluorescence resonance energy transfer and co-immunoprecipitation (Co-IP) analyses were performed to demonstrate the formation of functional GHSR1a/OX1R heterodimers. This showed that a peptide corresponding to the 5-transmembrane domain of OX1R impaired heterodimer construction. We found that ghrelin stimulated GHSR1a/OX1R heterodimer cells to increase the activation of Gαs protein, compared to the cells that express GHSR1a. Stimulation of GHSR1a/OX1R heterodimers with orexin-A did not alter GPCR interactions with Gα protein subunits. GHSR1a/OX1R heterodimers induced Gαs and downstream signaling pathway activity, including increase of cAMP-response element luciferase reporter activity and cAMP levels. In addition, ghrelin induced a higher proliferation rate in SH-SY5Y cells than in controls. This suggests that ghrelin GHSR1a/OX1R heterodimers promotes an upregulation of a Gαs-cAMP-cAMP-responsive element signaling pathway in vitro and an increase in neuroblastoma cell proliferation.
Collapse
Affiliation(s)
- Qingjie Xue
- Neurobiology Institute, Jining Medical University, Jining, China.,Department of Pathogenic Biology, Jining Medical University, Jining, China
| | - Bo Bai
- Neurobiology Institute, Jining Medical University, Jining, China
| | - Bingyuan Ji
- Neurobiology Institute, Jining Medical University, Jining, China
| | - Xiaoyu Chen
- Department of Physiology, Taishan Medical University, Taian, China
| | - Chunmei Wang
- Neurobiology Institute, Jining Medical University, Jining, China
| | - Peixiang Wang
- Neurobiology Institute, Jining Medical University, Jining, China
| | - Chunqing Yang
- Neurobiology Institute, Jining Medical University, Jining, China
| | - Rumin Zhang
- Neurobiology Institute, Jining Medical University, Jining, China
| | - Yunlu Jiang
- Neurobiology Institute, Jining Medical University, Jining, China
| | - Yanyou Pan
- Neurobiology Institute, Jining Medical University, Jining, China
| | - Baohua Cheng
- Neurobiology Institute, Jining Medical University, Jining, China
| | - Jing Chen
- Neurobiology Institute, Jining Medical University, Jining, China.,Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, United Kingdom
| |
Collapse
|
45
|
Gorzkiewicz A, Szemraj J. Brain endocannabinoid signaling exhibits remarkable complexity. Brain Res Bull 2018; 142:33-46. [PMID: 29953913 DOI: 10.1016/j.brainresbull.2018.06.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 06/06/2018] [Accepted: 06/21/2018] [Indexed: 01/04/2023]
Abstract
The endocannabinoid (eCB) signaling system is one of the most extensive of the mammalian brain. Despite the involvement of only few specific ligands and receptors, the system encompasses a vast diversity of triggered mechanisms and driven effects. It mediates a wide range of phenomena, including the regulation of transmitter release, neural excitability, synaptic plasticity, impulse spread, long-term neuronal potentiation, neurogenesis, cell death, lineage segregation, cell migration, inflammation, oxidative stress, nociception and the sleep cycle. It is also known to be involved in the processes of learning and memory formation. This extensive scope of action is attained by combining numerous variables. In a properly functioning brain, the correlations of these variables are kept in a strictly controlled balance; however, this balance is disrupted in many pathological conditions. However, while this balance is known to be disrupted by drugs in the case of addicts, the stimuli and mechanisms influencing the neurodegenerating brain remain elusive. This review examines the multiple factors and phenomena affecting the eCB signaling system in the brain. It evaluates techniques of controlling the eCB system to identify the obstacles in their applications and highlights the crucial interdependent variables that may influence biomedical research outcomes.
Collapse
Affiliation(s)
- Anna Gorzkiewicz
- Medical University of Lodz, ul.Mazowiecka 6/8, 92-215, Lodz, Poland.
| | - Janusz Szemraj
- Medical University of Lodz, ul.Mazowiecka 6/8, 92-215, Lodz, Poland
| |
Collapse
|
46
|
Kirsch M, Mertens W. On the Drive Specificity of Freudian Drives for the Generation of SEEKING Activities: The Importance of the Underestimated Imperative Motor Factor. Front Psychol 2018; 9:616. [PMID: 29774002 PMCID: PMC5943553 DOI: 10.3389/fpsyg.2018.00616] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2017] [Accepted: 04/12/2018] [Indexed: 12/19/2022] Open
Abstract
Doubters of Freud's theory of drives frequently mentioned that his approach is outdated and therefore cannot be useful for solving current problems in patients with mental disorders. At present, many scientists believe that affects rather than drives are of utmost importance for the emotional life and the theoretical framework of affective neuroscience, developed by Panksepp, strongly underpinned this view. Panksepp evaluated seven so-called command systems and the SEEKING system is therein of central importance. Panksepp used Pankseppian drives as inputs for the SEEKING system but noted the missing explanation of drive-specific generation of SEEKING activities in his description. Drive specificity requires dual action of the drive: the activation of a drive-specific brain area and the release of the neurotransmitter dopamine. Noticeably, as Freud claimed drive specificity too, it was here analyzed whether a Freudian drive can evoke the generation of drive-specific SEEKING activities. Special importance was addressed to the imperative motor factor in Freud's drive theory because Panksepp's formulations focused on neural pathways without specifying underlying neurotransmitter/endocrine factors impelling motor activity. As Panksepp claimed sleep as a Pankseppian drive, we firstly had to classified sleep as a Freudian drive by using three evaluated criteria for a Freudian drive. After that it was possible to identify the imperative motor factors of hunger, thirst, sex, and sleep. Most importantly, all of these imperative motor factors can both activate a drive-specific brain area and release dopamine from dopaminergic neurons, i.e., they can achieve the so-called drive specificity. Surprisingly, an impaired Freudian drive can alter via endocrinological pathways the concentration of the imperative motor factor of a second Freudian drive, obviously in some independence to the level of the metabolic deficit, thereby offering the possibility to modulate the generation of SEEKING activities of this second Freudian drive. This novel possibility might help to refine the general understanding of the action of Freudian drives. As only imperative motor factors of Freudian drives can guarantee drive specificity for the generation of SEEKING activities, the impact of Freud's construct Eros (with its constituents hunger, thirst, sex, and sleep) should be revisited.
Collapse
Affiliation(s)
- Michael Kirsch
- Institute of Physiological Chemistry, University Hospital Essen, Essen, Germany
| | - Wolfgang Mertens
- Division of Clinical Psychology and Psychotherapy, Department of Psychology, Faculty of Psychology and Educational Sciences, Ludwig Maximilian University of Munich, Munich, Germany
| |
Collapse
|
47
|
Bocchio-Chiavetto L, Zanardini R, Tosato S, Ventriglia M, Ferrari C, Bonetto C, Lasalvia A, Giubilini F, Fioritti A, Pileggi F, Pratelli M, Pavanati M, Favaro A, De Girolamo G, Frisoni GB, Ruggeri M, Gennarelli M. Immune and metabolic alterations in first episode psychosis (FEP) patients. Brain Behav Immun 2018; 70:315-324. [PMID: 29548996 DOI: 10.1016/j.bbi.2018.03.013] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Revised: 03/09/2018] [Accepted: 03/11/2018] [Indexed: 12/14/2022] Open
Abstract
The molecular underpinnings associated to first episode psychosis (FEP) remains to be elucidated, but compelling evidence supported an association of FEP with blood alterations in biomarkers related to immune system, growth factors and metabolism regulators. Many of these studies have not been already confirmed in larger samples or have not considered the FEP diagnostic subgroups. In order to identify biochemical signatures of FEP, the serum levels of the growth factors BDNF and VEGF, the immune regulators IL-1RA, IL-6, IL-10 and IL-17, RANTES/CCL5, MIP-1b/CCL4, IL-8 and the metabolic regulators C-peptide, ghrelin, GIP, GLP-1, glucagon, insulin, leptin, PAI-1, resistin and visfatin were analysed in 260 subjects collected in the GET UP project. The results indicated an increase of MIP-1b/CCL4, VEGF, IL-6 and PAI-1, while IL-17, ghrelin, glucagon and GLP-1 were decreased in the whole sample of FEP patients (p < 0.01 for all markers except for PAI-1 p < 0.05). No differences were evidenced for these markers among the diagnostic groups that constitute the FEP sample, whereas IL-8 is increased only in patients with a diagnosis of affective psychosis. The principal component analysis (PCA) and variable importance analysis (VIA) indicated that MIP-1b/CCL4, ghrelin, glucagon, VEGF and GLP-1 were the variables mostly altered in FEP patients. On the contrary, none of the analysed markers nor a combination of them can discriminate between FEP diagnostic subgroups. These data evidence a profile of immune and metabolic alterations in FEP patients, providing new information on the molecular mechanism associated to the psychosis onset for the development of preventive strategies and innovative treatment targets.
Collapse
Affiliation(s)
- Luisella Bocchio-Chiavetto
- IRCCS Centro S. Giovanni di Dio, Fatebenefratelli, Brescia, Italy; Faculty of Psychology, eCampus University, Novedrate (Como), Italy.
| | | | - Sarah Tosato
- Section of Psychiatry, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Mariacarla Ventriglia
- Fatebenefratelli Foundation, AFaR Division, Fatebenefratelli Hospital, Isola Tiberina, Rome, Italy
| | - Clarissa Ferrari
- IRCCS Centro S. Giovanni di Dio, Fatebenefratelli, Brescia, Italy
| | - Chiara Bonetto
- Section of Psychiatry, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Antonio Lasalvia
- Unit of Psychiatry, Azienda Ospedaliera Universitaria Integrata (AOUI), Verona, Italy
| | | | | | | | | | - Michele Pavanati
- Department of Medical Sciences of Communication and Behavior, Section of Psychiatry, The Consultation-Liaison Psychiatric Service and Psychiatric Unit, University of Ferrara, Italy
| | - Angela Favaro
- Department of Neurosciences, University of Padua and Azienda Ospedaliera, Padua, Italy
| | | | - Giovanni Battista Frisoni
- IRCCS Centro S. Giovanni di Dio, Fatebenefratelli, Brescia, Italy; Geneva University Hospital and University of Geneva, Switzerland
| | - Mirella Ruggeri
- Section of Psychiatry, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Massimo Gennarelli
- IRCCS Centro S. Giovanni di Dio, Fatebenefratelli, Brescia, Italy; Dept. of Molecular and Translational Medicine, Division of Biology and Genetics, University of Brescia, Italy
| |
Collapse
|
48
|
GHSR-D2R heteromerization modulates dopamine signaling through an effect on G protein conformation. Proc Natl Acad Sci U S A 2018; 115:4501-4506. [PMID: 29632174 DOI: 10.1073/pnas.1712725115] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The growth hormone secretagogue receptor (GHSR) and dopamine receptor (D2R) have been shown to oligomerize in hypothalamic neurons with a significant effect on dopamine signaling, but the molecular processes underlying this effect are still obscure. We used here the purified GHSR and D2R to establish that these two receptors assemble in a lipid environment as a tetrameric complex composed of two each of the receptors. This complex further recruits G proteins to give rise to an assembly with only two G protein trimers bound to a receptor tetramer. We further demonstrate that receptor heteromerization directly impacts on dopamine-mediated Gi protein activation by modulating the conformation of its α-subunit. Indeed, association to the purified GHSR:D2R heteromer triggers a different active conformation of Gαi that is linked to a higher rate of GTP binding and a faster dissociation from the heteromeric receptor. This is an additional mechanism to expand the repertoire of GPCR signaling modulation that could have implications for the control of dopamine signaling in normal and physiopathological conditions.
Collapse
|
49
|
More SV, Choi DK. Emerging preclinical pharmacological targets for Parkinson's disease. Oncotarget 2018; 7:29835-63. [PMID: 26988916 PMCID: PMC5045437 DOI: 10.18632/oncotarget.8104] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 02/08/2016] [Indexed: 12/14/2022] Open
Abstract
Parkinson's disease (PD) is a progressive neurological condition caused by the degeneration of dopaminergic neurons in the basal ganglia. It is the most prevalent form of Parkinsonism, categorized by cardinal features such as bradykinesia, rigidity, tremors, and postural instability. Due to the multicentric pathology of PD involving inflammation, oxidative stress, excitotoxicity, apoptosis, and protein aggregation, it has become difficult to pin-point a single therapeutic target and evaluate its potential application. Currently available drugs for treating PD provide only symptomatic relief and do not decrease or avert disease progression resulting in poor patient satisfaction and compliance. Significant amount of understanding concerning the pathophysiology of PD has offered a range of potential targets for PD. Several emerging targets including AAV-hAADC gene therapy, phosphodiesterase-4, potassium channels, myeloperoxidase, acetylcholinesterase, MAO-B, dopamine, A2A, mGlu5, and 5-HT-1A/1B receptors are in different stages of clinical development. Additionally, alternative interventions such as deep brain stimulation, thalamotomy, transcranial magnetic stimulation, and gamma knife surgery, are also being developed for patients with advanced PD. As much as these therapeutic targets hold potential to delay the onset and reverse the disease, more targets and alternative interventions need to be examined in different stages of PD. In this review, we discuss various emerging preclinical pharmacological targets that may serve as a new promising neuroprotective strategy that could actually help alleviate PD and its symptoms.
Collapse
Affiliation(s)
- Sandeep Vasant More
- Department of Biotechnology, College of Biomedical and Health Science, Konkuk University, Chungju, South Korea
| | - Dong-Kug Choi
- Department of Biotechnology, College of Biomedical and Health Science, Konkuk University, Chungju, South Korea
| |
Collapse
|
50
|
Mahbod P, Smith EP, Fitzgerald ME, Morano RL, Packard BA, Ghosal S, Scheimann JR, Perez-Tilve D, Herman JP, Tong J. Desacyl Ghrelin Decreases Anxiety-like Behavior in Male Mice. Endocrinology 2018; 159:388-399. [PMID: 29155981 PMCID: PMC5761608 DOI: 10.1210/en.2017-00540] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 11/10/2017] [Indexed: 11/19/2022]
Abstract
Ghrelin is a 28-amino acid polypeptide that regulates feeding, glucose metabolism, and emotionality (stress, anxiety, and depression). Plasma ghrelin circulates as desacyl ghrelin (DAG) or, in an acylated form, acyl ghrelin (AG), through the actions of ghrelin O-acyltransferase (GOAT), exhibiting low or high affinity, respectively, for the growth hormone secretagogue receptor (GHSR) 1a. We investigated the role of endogenous AG, DAG, and GHSR1a signaling on anxiety and stress responses using ghrelin knockout (Ghr KO), GOAT KO, and Ghsr stop-floxed (Ghsr null) mice. Behavioral and hormonal responses were tested in the elevated plus maze and light/dark (LD) box. Mice lacking both AG and DAG (Ghr KO) increased anxiety-like behaviors across tests, whereas anxiety reactions were attenuated in DAG-treated Ghr KO mice and in mice lacking AG (GOAT KO). Notably, loss of GHSR1a (Ghsr null) did not affect anxiety-like behavior in any test. Administration of AG and DAG to Ghr KO mice with lifelong ghrelin deficiency reduced anxiety-like behavior and decreased phospho-extracellular signal-regulated kinase phosphorylation in the Edinger-Westphal nucleus in wild-type mice, a site normally expressing GHSR1a and involved in stress- and anxiety-related behavior. Collectively, our data demonstrate distinct roles for endogenous AG and DAG in regulation of anxiety responses and suggest that the behavioral impact of ghrelin may be context dependent.
Collapse
Affiliation(s)
- Parinaz Mahbod
- Department of Psychiatry and Behavioral Neuroscience,
University of Cincinnati, Cincinnati, Ohio 45267
| | - Eric P. Smith
- Department of Medicine, University of Cincinnati,
Cincinnati, Ohio 45267
| | - Maureen E. Fitzgerald
- Department of Psychiatry and Behavioral Neuroscience,
University of Cincinnati, Cincinnati, Ohio 45267
| | - Rachel L. Morano
- Department of Psychiatry and Behavioral Neuroscience,
University of Cincinnati, Cincinnati, Ohio 45267
| | - Benjamin A. Packard
- Department of Psychiatry and Behavioral Neuroscience,
University of Cincinnati, Cincinnati, Ohio 45267
| | - Sriparna Ghosal
- Department of Psychiatry and Behavioral Neuroscience,
University of Cincinnati, Cincinnati, Ohio 45267
| | - Jessie R. Scheimann
- Department of Psychiatry and Behavioral Neuroscience,
University of Cincinnati, Cincinnati, Ohio 45267
| | - Diego Perez-Tilve
- Department of Medicine, University of Cincinnati,
Cincinnati, Ohio 45267
| | - James P. Herman
- Department of Psychiatry and Behavioral Neuroscience,
University of Cincinnati, Cincinnati, Ohio 45267
| | - Jenny Tong
- Division of Endocrinology, Metabolism and Nutrition,
Department of Medicine, Duke University, Durham, North Carolina 27708
| |
Collapse
|