1
|
Sturaro C, Ruzza C, Ferrari F, Pola P, Argentieri M, Frezza A, Marzola E, Bettegazzi B, Cattaneo S, Pietra C, Malfacini D, Calò G. In vitro pharmacological characterization of growth hormone secretagogue receptor ligands using the dynamic mass redistribution and calcium mobilization assays. Eur J Pharmacol 2024; 981:176880. [PMID: 39128804 DOI: 10.1016/j.ejphar.2024.176880] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 07/25/2024] [Accepted: 08/08/2024] [Indexed: 08/13/2024]
Abstract
Ghrelin modulates several biological functions via selective activation of the growth hormone secretagogue receptor (GHSR). GHSR agonists may be useful for the treatment of anorexia and cachexia, while antagonists and inverse agonists may represent new drugs for the treatment of metabolic and substance use disorders. Thus, the identification and pharmacodynamic characterization of new GHSR ligands is of high interest. In the present work the label-free dynamic mass redistribution (DMR) assay has been used to evaluate the pharmacological activity of a panel of GHSR ligands. This includes the endogenous peptides ghrelin, desacyl-ghrelin and LEAP2(1-14). Among synthetic compounds, the agonists anamorelin and HM01, the antagonists HM04 and YIL-781, and the inverse agonist PF-05190457 have been tested, together with HM03, R011, and H1498 from patent literature. The DMR results have been compared to those obtained in parallel experiments with the calcium mobilization assay. Ghrelin, anamorelin, HM01, and HM03 behaved as potent full GHSR agonists. YIL-781 behaved as a partial GHSR agonist and R011 as antagonist in both the assays. LEAP2(1-14) resulted a GHSR inverse agonist in DMR but not in calcium mobilization assay. PF-05190457, HM04, and H1498 behaved as GHSR inverse agonists in DMR experiments, while they acted as antagonists in calcium mobilization studies. In conclusion, this study provided a systematic pharmacodynamic characterization of several GHSR ligands in two different pharmacological assays. It demonstrated that the DMR assay can be successfully used particularly to discriminate between antagonists and inverse agonists. This study may be useful for the selection of the most appropriate compounds to be used in future studies.
Collapse
Affiliation(s)
- Chiara Sturaro
- U.O. Neurological Clinic of the University Hospital of Ferrara, Italy
| | - Chiara Ruzza
- Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy; LTTA Laboratory for Advanced Therapies, Technopole of Ferrara, Ferrara, Italy.
| | - Federica Ferrari
- Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
| | - Pietro Pola
- Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
| | - Michela Argentieri
- Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
| | - Alessia Frezza
- U.O. Neurological Clinic of the University Hospital of Ferrara, Italy
| | - Erika Marzola
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Ferrara, Italy
| | | | | | | | - Davide Malfacini
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Italy
| | - Girolamo Calò
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Italy
| |
Collapse
|
2
|
Rubinić I, Kurtov M, Likić R. Novel Pharmaceuticals in Appetite Regulation: Exploring emerging gut peptides and their pharmacological prospects. Pharmacol Res Perspect 2024; 12:e1243. [PMID: 39016695 PMCID: PMC11253306 DOI: 10.1002/prp2.1243] [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: 12/12/2023] [Revised: 03/23/2024] [Accepted: 06/22/2024] [Indexed: 07/18/2024] Open
Abstract
Obesity, a global health challenge, necessitates innovative approaches for effective management. Targeting gut peptides in the development of anti-obesity pharmaceuticals has already demonstrated significant efficacy. Ghrelin, peptide YY (PYY), cholecystokinin (CCK), and amylin are crucial in appetite regulation offering promising targets for pharmacological interventions in obesity treatment using both peptide-based and small molecule-based pharmaceuticals. Ghrelin, a sole orexigenic gut peptide, has a potential for anti-obesity therapies through various approaches, including endogenous ghrelin neutralization, ghrelin receptor antagonists, ghrelin O-acyltransferase, and functional inhibitors. Anorexigenic gut peptides, peptide YY, cholecystokinin, and amylin, have exhibited appetite-reducing effects in animal models and humans. Overcoming substantial obstacles is imperative for translating these findings into clinically effective pharmaceuticals. Peptide YY and cholecystokinin analogues, characterized by prolonged half-life and resistance to proteolytic enzymes, present viable options. Positive allosteric modulators emerge as a novel approach for modulating the cholecystokinin pathway. Amylin is currently the most promising, with both amylin analogues and dual amylin and calcitonin receptor agonists (DACRAs) progressing to advanced stages of clinical trials. Despite persistent challenges, innovative pharmaceutical strategies provide a glimpse into the future of anti-obesity therapies.
Collapse
Affiliation(s)
- Igor Rubinić
- Department of Basic and Clinical Pharmacology and Toxicology, Faculty of MedicineUniversity of RijekaRijekaCroatia
- Clinical Pharmacology unitClinical Hospital Center RijekaRijekaCroatia
| | - Marija Kurtov
- Division of Clinical Pharmacology and Toxicology, Department of Internal MedicineUniversity Hospital “Sveti Duh”ZagrebCroatia
| | - Robert Likić
- Department of Internal MedicineSchool of Medicine University of ZagrebZagrebCroatia
| |
Collapse
|
3
|
Shankar K, Metzger NP, Lawrence C, Gupta D, Osborne-Lawrence S, Varshney S, Singh O, Richard CP, Zaykov AN, Rolfts R, DuBois BN, Perez-Tilve D, Mani BK, Hammer STG, Zigman JM. A long-acting LEAP2 analog reduces hepatic steatosis and inflammation and causes marked weight loss in mice. Mol Metab 2024; 84:101950. [PMID: 38697291 PMCID: PMC11103953 DOI: 10.1016/j.molmet.2024.101950] [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: 01/18/2024] [Revised: 04/01/2024] [Accepted: 04/26/2024] [Indexed: 05/04/2024] Open
Abstract
OBJECTIVE The number of individuals affected by metabolic dysfunction associated fatty liver disease [1] is on the rise, yet hormonal contributors to the condition remain incompletely described and only a single FDA-approved treatment is available. Some studies suggest that the hormones ghrelin and LEAP2, which act as agonist and antagonist/inverse agonist, respectively, for the G protein coupled receptor GHSR, may influence the development of MAFLD. For instance, ghrelin increases hepatic fat whereas synthetic GHSR antagonists do the opposite. Also, hepatic steatosis is less prominent in standard chow-fed ghrelin-KO mice but more prominent in 42% high-fat diet-fed female LEAP2-KO mice. METHODS Here, we sought to determine the therapeutic potential of a long-acting LEAP2 analog (LA-LEAP2) to treat MAFLD in mice. LEAP2-KO and wild-type littermate mice were fed a Gubra-Amylin-NASH (GAN) diet for 10 or 40 wks, with some randomized to an additional 28 or 10 days of GAN diet, respectively, while treated with LA-LEAP2 vs Vehicle. Various metabolic parameters were followed and biochemical and histological assessments of MAFLD were made. RESULTS Among the most notable metabolic effects, daily LA-LEAP2 administration to both LEAP2-KO and wild-type littermates during the final 4 wks of a 14 wk-long GAN diet challenge markedly reduced liver weight, hepatic triglycerides, plasma ALT, hepatic microvesicular steatosis, hepatic lobular inflammation, NASH activity scores, and prevalence of higher-grade fibrosis. These changes were accompanied by prominent reductions in body weight, without effects on food intake, and reduced plasma total cholesterol. Daily LA-LEAP2 administration during the final 10 d of a 41.5 wk-long GAN diet challenge also reduced body weight, plasma ALT, and plasma total cholesterol in LEAP2-KO and wild-type littermates and prevalence of higher grade fibrosis in LEAP2-KO mice. CONCLUSIONS Administration of LA-LEAP2 to mice fed a MAFLD-prone diet markedly improves several facets of MAFLD, including hepatic steatosis, hepatic lobular inflammation, higher-grade hepatic fibrosis, and transaminitis. These changes are accompanied by prominent reductions in body weight and lowered plasma total cholesterol. Taken together, these data suggest that LEAP2 analogs such as LA-LEAP2 hold promise for the treatment of MAFLD and obesity.
Collapse
Affiliation(s)
- Kripa Shankar
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-9077, USA
| | - Nathan P Metzger
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-9077, USA
| | - Connor Lawrence
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-9077, USA
| | - Deepali Gupta
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-9077, USA
| | - Sherri Osborne-Lawrence
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-9077, USA
| | - Salil Varshney
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-9077, USA
| | - Omprakash Singh
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-9077, USA
| | - Corine P Richard
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-9077, USA
| | | | - Rebecca Rolfts
- Novo Nordisk Lexington, 33 Hayden Ave, Lexington, MA 02421, USA
| | - Barent N DuBois
- Novo Nordisk Lexington, 33 Hayden Ave, Lexington, MA 02421, USA
| | - Diego Perez-Tilve
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Bharath K Mani
- Novo Nordisk Lexington, 33 Hayden Ave, Lexington, MA 02421, USA
| | - Suntrea T G Hammer
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jeffrey M Zigman
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-9077, USA; Division of Endocrinology, Department of Internal Medicine, UT Southwestern Medical Center, USA; Department of Psychiatry, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390, USA.
| |
Collapse
|
4
|
Kulkarni SS, Singh O, Zigman JM. The intersection between ghrelin, metabolism and circadian rhythms. Nat Rev Endocrinol 2024; 20:228-238. [PMID: 38123819 PMCID: PMC11760189 DOI: 10.1038/s41574-023-00927-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/16/2023] [Indexed: 12/23/2023]
Abstract
Despite the growing popular interest in sleep and diet, many gaps exist in our scientific understanding of the interaction between circadian rhythms and metabolism. In this Review, we explore a promising, bidirectional role for ghrelin in mediating this interaction. Ghrelin both influences and is influenced by central and peripheral circadian systems. Specifically, we focus on how ghrelin impacts outputs of circadian rhythm, including neuronal activity, circulating growth hormone levels, locomotor activity and eating behaviour. We also consider the effects of circadian rhythms on ghrelin expression and the consequences of disrupted circadian patterns, such as shift work and jet lag, on ghrelin secretion. Our Review is aimed at both the casual reader interested in gaining more insight into the scientific context surrounding the trending topics of sleep and metabolism, as well as experienced scientists in the fields of ghrelin and circadian biology seeking inspiration and a comprehensive overview of how these fields are related.
Collapse
Affiliation(s)
- Soumya S Kulkarni
- Medical Scientist Training Program, UT Southwestern Medical Center, Dallas, TX, USA
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA
| | - Omprakash Singh
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA
| | - Jeffrey M Zigman
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA.
- Division of Endocrinology, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA.
- Department of Psychiatry, UT Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
5
|
Howe SL, Holdom CJ, McCombe PA, Henderson RD, Zigman JM, Ngo ST, Steyn FJ. Associations of postprandial ghrelin, liver-expressed antimicrobial peptide 2 and leptin levels with body composition, disease progression and survival in patients with amyotrophic lateral sclerosis. Eur J Neurol 2024; 31:e16052. [PMID: 37658515 PMCID: PMC10840749 DOI: 10.1111/ene.16052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 08/06/2023] [Accepted: 08/17/2023] [Indexed: 09/03/2023]
Abstract
BACKGROUND AND PURPOSE Loss of appetite contributes to weight loss and faster disease progression in amyotrophic lateral sclerosis (ALS). Impairment of appetite control in ALS may include altered production or action of orexigenic (i.e., ghrelin) and anorexigenic (i.e., liver-expressed antimicrobial peptide 2 [LEAP2] and leptin) hormones. We aimed to determine if postprandial circulating ghrelin levels, LEAP2 levels, LEAP2:ghrelin molar ratio and leptin levels differ in ALS patients compared to non-neurodegenerative disease controls, and whether they are associated with disease progression and body composition. METHODS In this prospective natural history study, we assessed postprandial plasma levels of ghrelin, LEAP2 and leptin in patients with ALS (cases; n = 46) and controls (controls; n = 43). For cases, measures were compared to changes in body weight, body composition and clinical outcomes. RESULTS Postprandial ghrelin level was decreased by 52% in cases compared to controls (p = 0.013). LEAP2:ghrelin molar ratio was increased by 249% (p = 0.009), suggesting greater ghrelin resistance. Patients with lower LEAP2:ghrelin tended to have better functional capacity at assessment, as inferred by the ALS Functional Rating Scale-Revised (τ = -0.179, p = 0.086). Furthermore, ghrelin and LEAP2:ghrelin molar ratio correlated with diagnostic delay (ghrelin, τ = 0.223, p = 0.029; LEAP2:ghrelin, τ = -0.213, p = 0.037). Baseline ghrelin level, LEAP2 level, LEAP2:ghrelin ratio and leptin level were, however, not predictive of change in functional capacity during follow-up. Also, patients with higher postprandial ghrelin levels (hazard ratio [HR] 1.375, p = 0.048), and lower LEAP2:ghelin ratios (HR 0.828, p = 0.051) had an increased risk of earlier death. CONCLUSIONS Reduced postprandial ghrelin levels, coupled with increased LEAP2:ghrelin molar ratios, suggests a loss of ghrelin action in patients with ALS. Given ghrelin's actions on appetite, metabolism and neuroprotection, reduced ghrelin and greater ghrelin resistance could contribute to impaired capacity to tolerate the physiological impact of disease. Comprehensive studies are needed to explain how ghrelin and LEAP2 contribute to body weight regulation and disease progression in ALS.
Collapse
Affiliation(s)
- Stephanie L. Howe
- Australian Institute for Bioengineering and NanotechnologyThe University of QueenslandBrisbaneQueenslandAustralia
| | - Cory J. Holdom
- Australian Institute for Bioengineering and NanotechnologyThe University of QueenslandBrisbaneQueenslandAustralia
- Centre for Clinical ResearchThe University of QueenslandBrisbaneQueenslandAustralia
| | - Pamela A. McCombe
- Centre for Clinical ResearchThe University of QueenslandBrisbaneQueenslandAustralia
- Department of NeurologyRoyal Brisbane and Women's HospitalBrisbaneQueenslandAustralia
| | - Robert D. Henderson
- Centre for Clinical ResearchThe University of QueenslandBrisbaneQueenslandAustralia
- Department of NeurologyRoyal Brisbane and Women's HospitalBrisbaneQueenslandAustralia
| | - Jeffrey M. Zigman
- Center for Hypothalamic Research, Department of Internal MedicineUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| | - Shyuan T. Ngo
- Australian Institute for Bioengineering and NanotechnologyThe University of QueenslandBrisbaneQueenslandAustralia
- Centre for Clinical ResearchThe University of QueenslandBrisbaneQueenslandAustralia
- Department of NeurologyRoyal Brisbane and Women's HospitalBrisbaneQueenslandAustralia
| | - Frederik J. Steyn
- Department of NeurologyRoyal Brisbane and Women's HospitalBrisbaneQueenslandAustralia
- School of Biomedical SciencesThe University of QueenslandBrisbaneQueenslandAustralia
| |
Collapse
|
6
|
Richardson RS, Sulima A, Rice KC, Kucharczk JA, Janda KD, Nisbett KE, Koob GF, Vendruscolo LF, Leggio L. Pharmacological GHSR (ghrelin receptor) blockade reduces alcohol binge-like drinking in male and female mice. Neuropharmacology 2023; 238:109643. [PMID: 37369277 PMCID: PMC10513123 DOI: 10.1016/j.neuropharm.2023.109643] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/20/2023] [Accepted: 06/22/2023] [Indexed: 06/29/2023]
Abstract
Ghrelin is a peptide that is produced by endocrine cells that are primarily localized in the stomach. Ghrelin receptors (GHSR) are expressed in the brain and periphery. Preclinical and clinical studies support a role for ghrelin in alcohol drinking and seeking. The GHSR has been suggested to be a potential pharmacotherapeutic target for alcohol use disorder (AUD). However, the role of the ghrelin system and its potential modulation by biological sex on binge-like drinking has not been comprehensively investigated. The present study tested six GHSR antagonists in an alcohol binge-like drinking procedure in male and female mice. Systemic administration of the GHSR antagonists JMV2959, PF-5190457, PF-6870961, and HM-04 reduced alcohol intake in both male and female mice. YIL-781 decreased intake in males, and LEAP2 (likely peripherally restricted) did not reduce intake in mice of either sex. We also administered LEAP2 and JMV2959 intracerebroventricularly to investigate whether the effects of GHSR blockade on alcohol intake are mediated by central receptors. The central administration of LEAP2 and JMV2959 decreased alcohol intake, particularly in high-drinking animals. Finally, in a preliminary experiment, an anti-ghrelin vaccine was examined for its potential effect on binge-like drinking and had no effect. In all experiments, there was a lack of meaningful sex differences. These findings suggest that central GHSR mediates binge-like alcohol intake. These data reveal novel pharmacological compounds with translational potential in the treatment of AUD and provide further evidence of the GHSR as a potential treatment target for AUD.
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, MD, USA; Neurobiology of Addiction Section, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA; Stress & 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; 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
| | - Agnieszka Sulima
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Kenner C Rice
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Jed A Kucharczk
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Kim D Janda
- Department of Chemistry, Department of Immunology and Microbial Science, The Skaggs Institute for Chemical Biology, WIRM Institute for Research and Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Khalin E Nisbett
- Neurobiology of Addiction Section, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA; Stress & 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; Graduate Program in Neuroscience, Graduate College, University of Illinois Chicago, Chicago, IL, 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 & 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, MD, USA; Department of Behavioral and Social Sciences, Center for Alcohol and Addiction Studies, Brown University, Providence, RI, 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
|
7
|
Nobari H, Azarian S, Saedmocheshi S, Valdés-Badilla P, García Calvo T. Narrative review: The role of circadian rhythm on sports performance, hormonal regulation, immune system function, and injury prevention in athletes. Heliyon 2023; 9:e19636. [PMID: 37809566 PMCID: PMC10558889 DOI: 10.1016/j.heliyon.2023.e19636] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 08/21/2023] [Accepted: 08/29/2023] [Indexed: 10/10/2023] Open
Abstract
Objectives This study was a narrative review of the importance of circadian rhythm (CR), describes the underlying mechanisms of CR in sports performance, emphasizes the reciprocal link between CR, endocrine homeostasis and sex differences, and the unique role of the circadian clock in immune system function and coordination. Method As a narrative review study, a comprehensive search was conducted in PubMed, Scopus, and Web of Science (core collection) databases using the keywords "circadian rhythm", "sports performance", "hormonal regulation", "immune system", and "injury prevention". Inclusion criteria were studies published in English and peer-reviewed journals until July 2023. Studies that examined the role of CR in sports performance, hormonal status, immune system function, and injury prevention in athletes were selected for review. Results CR is followed by almost all physiological and biochemical activities in the human body. In humans, the superchiasmatic nucleus controls many daily biorhythms under solar time, including the sleep-wake cycle. A body of literature indicates that the peak performance of essential indicators of sports performance is primarily in the afternoon hours, and the evening of actions occurs roughly at the peak of core body temperature. Recent studies have demonstrated that the time of day that exercise is performed affects the achievement of good physical performance. This review also shows various biomarkers of cellular damage in weariness and the underlying mechanisms of diurnal fluctuations. According to the clock, CR can be synchronized with photonic and non-photonic stimuli (i.e., temperature, physical activity, and food intake), and feeding patterns and diet changes can affect CR and redox markers. It also emphasizes the reciprocal links between CR and endocrine homeostasis, the specific role of the circadian clock in coordinating immune system function, and the relationship between circadian clocks and sex differences. Conclusion The interaction between insufficient sleep and time of day on performance has been established in this study because it is crucial to balance training, recovery, and sleep duration to attain optimal sports performance.
Collapse
Affiliation(s)
- Hadi Nobari
- Faculty of Sport Sciences, University of Extremadura, 10003, Cáceres, Spain
- Department of Exercise Physiology, Faculty of Educational Sciences and Psychology, University of Mohaghegh Ardabili, Ardabil, 56199-11367, Iran
| | - Somayeh Azarian
- Department of Exercise Physiology, Faculty of Educational Sciences and Psychology, University of Mohaghegh Ardabili, Ardabil, 56199-11367, Iran
| | - Saber Saedmocheshi
- Department of Physical Education and Sport Sciences, Faculty of Humanities and Social Sciences, University of Kurdistan, Sanandaj, 66177-15175, Iran
| | - Pablo Valdés-Badilla
- Department of Physical Activity Sciences, Faculty of Education Sciences, Universidad Católica del Maule, Talca, 3530000, Chile
- Sports Coach Career, School of Education, Universidad Viña del Mar, Viña del Mar, 2520000, Chile
| | - Tomás García Calvo
- Faculty of Sport Sciences, University of Extremadura, 10003, Cáceres, Spain
| |
Collapse
|
8
|
Abdel-Malek M, Yang L, Miras AD. Pharmacotherapy for chronic obesity management: a look into the future. Intern Emerg Med 2023; 18:1019-1030. [PMID: 37249754 PMCID: PMC10326094 DOI: 10.1007/s11739-023-03237-4] [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: 08/17/2022] [Accepted: 02/17/2023] [Indexed: 05/31/2023]
Abstract
Substantial leaps have been made in the drug discovery front in tackling the growing pandemic of obesity and its metabolic co-morbidities. Greater mechanistic insight and understanding of the gut-brain molecular pathways at play have enabled the pursuit of novel therapeutic agents that possess increasingly efficacious weight-lowering potential whilst remaining safe and tolerable for clinical use. In the wake of glucagon-like peptide 1 (GLP-1) based therapy, we look at recent advances in gut hormone biology that have fermented the development of next generation pharmacotherapy in diabesity that harness synergistic potential. In this paper, we review the latest data from the SURPASS and SURMOUNT clinical trials for the novel 'twincretin', known as Tirzepatide, which has demonstrated sizeable body weight reduction as well as glycaemic efficacy. We also provide an overview of amylin-based combination strategies and other emerging therapies in the pipeline that are similarly providing great promise for the future of chronic management of obesity.
Collapse
Affiliation(s)
| | - Lisa Yang
- Imperial College Healthcare NHS Trust, London, UK
| | - Alexander Dimitri Miras
- School of Medicine, Ulster University, Derry~Londonderry, UK
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| |
Collapse
|
9
|
Inceu AI, Neag MA, Craciun AE, Buzoianu AD. Gut Molecules in Cardiometabolic Diseases: The Mechanisms behind the Story. Int J Mol Sci 2023; 24:3385. [PMID: 36834796 PMCID: PMC9965280 DOI: 10.3390/ijms24043385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/03/2023] [Accepted: 02/06/2023] [Indexed: 02/10/2023] Open
Abstract
Atherosclerotic cardiovascular disease is the most common cause of morbidity and mortality worldwide. Diabetes mellitus increases cardiovascular risk. Heart failure and atrial fibrillation are associated comorbidities that share the main cardiovascular risk factors. The use of incretin-based therapies promoted the idea that activation of alternative signaling pathways is effective in reducing the risk of atherosclerosis and heart failure. Gut-derived molecules, gut hormones, and gut microbiota metabolites showed both positive and detrimental effects in cardiometabolic disorders. Although inflammation plays a key role in cardiometabolic disorders, additional intracellular signaling pathways are involved and could explain the observed effects. Revealing the involved molecular mechanisms could provide novel therapeutic strategies and a better understanding of the relationship between the gut, metabolic syndrome, and cardiovascular diseases.
Collapse
Affiliation(s)
- Andreea-Ioana Inceu
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
| | - Maria-Adriana Neag
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
| | - Anca-Elena Craciun
- Department of Diabetes, and Nutrition Diseases, Iuliu Hatieganu University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania
| | - Anca-Dana Buzoianu
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
| |
Collapse
|
10
|
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: 11] [Impact Index Per Article: 5.5] [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
|
11
|
Les approches thérapeutiques non invasives de l’obésité : hier, aujourd’hui et demain. NUTR CLIN METAB 2022. [DOI: 10.1016/j.nupar.2022.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
12
|
O'Connor RM, Kenny PJ. Utility of 'substance use disorder' as a heuristic for understanding overeating and obesity. Prog Neuropsychopharmacol Biol Psychiatry 2022; 118:110580. [PMID: 35636576 DOI: 10.1016/j.pnpbp.2022.110580] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 05/24/2022] [Accepted: 05/24/2022] [Indexed: 02/07/2023]
Abstract
Rates of obesity and obesity-associated diseases have increased dramatically in countries with developed economies. Substance use disorders (SUDs) are characterized by the persistent use of the substance despite negative consequences. It has been hypothesized that overconsumption of palatable energy dense food can elicit SUD-like maladaptive behaviors that contribute to persistent caloric intake beyond homeostatic need even in the face of negative consequences. Palatable food and drugs of abuse act on many of the same motivation-related circuits in the brain, and can induce, at least superficially, similar molecular, cellular, and physiological adaptations on these circuits. As such, applying knowledge about the neurobiological mechanisms of SUDs may serve as useful heuristic to better understand the persistent overconsumption of palatable food that contributes to obesity. However, many important differences exist between the actions of drugs of abuse and palatable food in the brain. This warrants caution when attributing weight gain and obesity to the manifestation of a putative SUD-related behavioral disorder. Here, we describe similarities and differences between compulsive drug use in SUDs and overconsumption in obesity and consider the merit of the concept of "food addiction".
Collapse
Affiliation(s)
- Richard M O'Connor
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, United States of America
| | - Paul J Kenny
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, United States of America.
| |
Collapse
|
13
|
Piper NBC, Whitfield EA, Stewart GD, Xu X, Furness SGB. Targeting appetite and satiety in diabetes and obesity, via G protein-coupled receptors. Biochem Pharmacol 2022; 202:115115. [PMID: 35671790 DOI: 10.1016/j.bcp.2022.115115] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 05/26/2022] [Accepted: 05/27/2022] [Indexed: 11/17/2022]
Abstract
Type 2 diabetes and obesity have reached pandemic proportions throughout the world, so much so that the World Health Organisation coined the term "Globesity" to help encapsulate the magnitude of the problem. G protein-coupled receptors (GPCRs) are highly tractable drug targets due to their wide involvement in all aspects of physiology and pathophysiology, indeed, GPCRs are the targets of approximately 30% of the currently approved drugs. GPCRs are also broadly involved in key physiologies that underlie type 2 diabetes and obesity including feeding reward, appetite and satiety, regulation of blood glucose levels, energy homeostasis and adipose function. Despite this, only two GPCRs are the target of approved pharmaceuticals for treatment of type 2 diabetes and obesity. In this review we discuss the role of these, and select other candidate GPCRs, involved in various facets of type 2 diabetic or obese pathophysiology, how they might be targeted and the potential reasons why pharmaceuticals against these targets have not progressed to clinical use. Finally, we provide a perspective on the current development pipeline of anti-obesity drugs that target GPCRs.
Collapse
Affiliation(s)
- Noah B C Piper
- Receptor Transducer Coupling Laboratory, School of Biomedical Sciences, Faculty of Medicine, University of Queensland, St. Lucia, QLD 4072, Australia
| | - Emily A Whitfield
- Receptor Transducer Coupling Laboratory, School of Biomedical Sciences, Faculty of Medicine, University of Queensland, St. Lucia, QLD 4072, Australia
| | - Gregory D Stewart
- Drug Discovery Biology Laboratory, Monash Institute of Pharmaceutical Sciences & Department of Pharmacology Monash University, Parkville, VIC 3052, Australia
| | - Xiaomeng Xu
- Drug Discovery Biology Laboratory, Monash Institute of Pharmaceutical Sciences & Department of Pharmacology Monash University, Parkville, VIC 3052, Australia
| | - Sebastian G B Furness
- Receptor Transducer Coupling Laboratory, School of Biomedical Sciences, Faculty of Medicine, University of Queensland, St. Lucia, QLD 4072, Australia; Drug Discovery Biology Laboratory, Monash Institute of Pharmaceutical Sciences & Department of Pharmacology Monash University, Parkville, VIC 3052, Australia.
| |
Collapse
|
14
|
Péraldi-Roux S, Bayle M, M'Kadmi C, Damian M, Vaillé J, Fernandez G, Paula Cornejo M, Marie J, Banères JL, Ben Haj Salah K, Fehrentz JA, Cantel S, Perello M, Denoyelle S, Oiry C, Neasta J. Design and Characterization of a Triazole-Based Growth Hormone Secretagogue Receptor Modulator Inhibiting the Glucoregulatory and Feeding Actions of Ghrelin. Biochem Pharmacol 2022; 202:115114. [DOI: 10.1016/j.bcp.2022.115114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/26/2022] [Accepted: 05/26/2022] [Indexed: 11/02/2022]
|
15
|
Giorgioni G, Del Bello F, Quaglia W, Botticelli L, Cifani C, Micioni Di Bonaventura E, Micioni Di Bonaventura MV, Piergentili A. Advances in the Development of Nonpeptide Small Molecules Targeting Ghrelin Receptor. J Med Chem 2022; 65:3098-3118. [PMID: 35157454 PMCID: PMC8883476 DOI: 10.1021/acs.jmedchem.1c02191] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Ghrelin is an octanoylated peptide acting by the activation of the growth hormone secretagogue receptor, namely, GHS-R1a. The involvement of ghrelin in several physiological processes, including stimulation of food intake, gastric emptying, body energy balance, glucose homeostasis, reduction of insulin secretion, and lipogenesis validates the considerable interest in GHS-R1a as a promising target for the treatment of numerous disorders. Over the years, several GHS-R1a ligands have been identified and some of them have been extensively studied in clinical trials. The recently resolved structures of GHS-R1a bound to ghrelin or potent ligands have provided useful information for the design of new GHS-R1a drugs. This perspective is focused on the development of recent nonpeptide small molecules acting as GHS-R1a agonists, antagonists, and inverse agonists, bearing classical or new molecular scaffolds, as well as on radiolabeled GHS-R1a ligands developed for imaging. Moreover, the pharmacological effects of the most studied ligands have been discussed.
Collapse
Affiliation(s)
- Gianfabio Giorgioni
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, Via Madonna delle Carceri, 62032 Camerino, Italy
| | - Fabio Del Bello
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, Via Madonna delle Carceri, 62032 Camerino, Italy
| | - Wilma Quaglia
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, Via Madonna delle Carceri, 62032 Camerino, Italy
| | - Luca Botticelli
- School of Pharmacy, Pharmacology Unit, University of Camerino, Via Madonna delle Carceri 9, 62032 Camerino, Italy
| | - Carlo Cifani
- School of Pharmacy, Pharmacology Unit, University of Camerino, Via Madonna delle Carceri 9, 62032 Camerino, Italy
| | - E Micioni Di Bonaventura
- School of Pharmacy, Pharmacology Unit, University of Camerino, Via Madonna delle Carceri 9, 62032 Camerino, Italy
| | - M V Micioni Di Bonaventura
- School of Pharmacy, Pharmacology Unit, University of Camerino, Via Madonna delle Carceri 9, 62032 Camerino, Italy
| | - Alessandro Piergentili
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, Via Madonna delle Carceri, 62032 Camerino, Italy
| |
Collapse
|
16
|
Vohra MS, Benchoula K, Serpell CJ, Hwa WE. AgRP/NPY and POMC neurons in the arcuate nucleus and their potential role in treatment of obesity. Eur J Pharmacol 2022; 915:174611. [PMID: 34798121 DOI: 10.1016/j.ejphar.2021.174611] [Citation(s) in RCA: 87] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 10/27/2021] [Accepted: 10/27/2021] [Indexed: 02/08/2023]
Abstract
Obesity is a major health crisis affecting over a third of the global population. This multifactorial disease is regulated via interoceptive neural circuits in the brain, whose alteration results in excessive body weight. Certain central neuronal populations in the brain are recognised as crucial nodes in energy homeostasis; in particular, the hypothalamic arcuate nucleus (ARC) region contains two peptide microcircuits that control energy balance with antagonistic functions: agouti-related peptide/neuropeptide-Y (AgRP/NPY) signals hunger and stimulates food intake; and pro-opiomelanocortin (POMC) signals satiety and reduces food intake. These neuronal peptides levels react to energy status and integrate signals from peripheral ghrelin, leptin, and insulin to regulate feeding and energy expenditure. To manage obesity comprehensively, it is crucial to understand cellular and molecular mechanisms of information processing in ARC neurons, since these regulate energy homeostasis. Importantly, a specific strategy focusing on ARC circuits needs to be devised to assist in treating obese patients and maintaining weight loss with minimal or no side effects. The aim of this review is to elucidate the recent developments in the study of AgRP-, NPY- and POMC-producing neurons, specific to their role in controlling metabolism. The impact of ghrelin, leptin, and insulin signalling via action of these neurons is also surveyed, since they also impact energy balance through this route. Lastly, we present key proteins, targeted genes, compounds, drugs, and therapies that actively work via these neurons and could potentially be used as therapeutic targets for treating obesity conditions.
Collapse
Affiliation(s)
- Muhammad Sufyan Vohra
- School of Medicine, Faculty of Health and Medical Sciences, Taylor's University Lakeside Campus, 47500, Subang Jaya, Selangor Darul Ehsan, Malaysia
| | - Khaled Benchoula
- School of Medicine, Faculty of Health and Medical Sciences, Taylor's University Lakeside Campus, 47500, Subang Jaya, Selangor Darul Ehsan, Malaysia
| | - Christopher J Serpell
- School of Physical Sciences, Ingram Building, University of Kent, Canterbury, Kent, CT2 7NH, United Kingdom
| | - Wong Eng Hwa
- School of Medicine, Faculty of Health and Medical Sciences, Taylor's University Lakeside Campus, 47500, Subang Jaya, Selangor Darul Ehsan, Malaysia.
| |
Collapse
|
17
|
Gjermeni E, Kirstein AS, Kolbig F, Kirchhof M, Bundalian L, Katzmann JL, Laufs U, Blüher M, Garten A, Le Duc D. Obesity-An Update on the Basic Pathophysiology and Review of Recent Therapeutic Advances. Biomolecules 2021; 11:1426. [PMID: 34680059 PMCID: PMC8533625 DOI: 10.3390/biom11101426] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/16/2021] [Accepted: 09/22/2021] [Indexed: 12/13/2022] Open
Abstract
Obesity represents a major public health problem with a prevalence increasing at an alarming rate worldwide. Continuous intensive efforts to elucidate the complex pathophysiology and improve clinical management have led to a better understanding of biomolecules like gut hormones, antagonists of orexigenic signals, stimulants of fat utilization, and/or inhibitors of fat absorption. In this article, we will review the pathophysiology and pharmacotherapy of obesity including intersection points to the new generation of antidiabetic drugs. We provide insight into the effectiveness of currently approved anti-obesity drugs and other therapeutic avenues that can be explored.
Collapse
Affiliation(s)
- Erind Gjermeni
- Department of Electrophysiology, Heart Center Leipzig at University of Leipzig, 04289 Leipzig, Germany;
- Department of Cardiology, Median Centre for Rehabilitation Schmannewitz, 04774 Dahlen, Germany;
| | - Anna S. Kirstein
- Pediatric Research Center, University Hospital for Children and Adolescents, Leipzig University, 04103 Leipzig, Germany; (A.S.K.); (F.K.); (A.G.)
| | - Florentien Kolbig
- Pediatric Research Center, University Hospital for Children and Adolescents, Leipzig University, 04103 Leipzig, Germany; (A.S.K.); (F.K.); (A.G.)
| | - Michael Kirchhof
- Department of Cardiology, Median Centre for Rehabilitation Schmannewitz, 04774 Dahlen, Germany;
| | - Linnaeus Bundalian
- Institute of Human Genetics, University Medical Center Leipzig, 04103 Leipzig, Germany;
| | - Julius L. Katzmann
- Klinik und Poliklinik für Kardiologie, University Clinic Leipzig, 04103 Leipzig, Germany; (J.L.K.); (U.L.)
| | - Ulrich Laufs
- Klinik und Poliklinik für Kardiologie, University Clinic Leipzig, 04103 Leipzig, Germany; (J.L.K.); (U.L.)
| | - Matthias Blüher
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, 04103 Leipzig, Germany;
| | - Antje Garten
- Pediatric Research Center, University Hospital for Children and Adolescents, Leipzig University, 04103 Leipzig, Germany; (A.S.K.); (F.K.); (A.G.)
| | - Diana Le Duc
- Institute of Human Genetics, University Medical Center Leipzig, 04103 Leipzig, Germany;
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, 04103 Leipzig, Germany;
- Department of Evolutionary Genetics, Max Planck Institute for Evolutionary Anthropology, 04103 Leipzig, Germany
| |
Collapse
|
18
|
Shankar K, Takemi S, Gupta D, Varshney S, Mani BK, Osborne-Lawrence S, Metzger NP, Richard CP, Berglund ED, Zigman JM. Ghrelin cell-expressed insulin receptors mediate meal- and obesity-induced declines in plasma ghrelin. JCI Insight 2021; 6:e146983. [PMID: 34473648 PMCID: PMC8492315 DOI: 10.1172/jci.insight.146983] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 08/04/2021] [Indexed: 01/20/2023] Open
Abstract
Mechanisms underlying postprandial and obesity-associated plasma ghrelin reductions are incompletely understood. Here, using ghrelin cell-selective insulin receptor-KO (GhIRKO) mice, we tested the impact of insulin, acting via ghrelin cell-expressed insulin receptors (IRs), to suppress ghrelin secretion. Insulin reduced ghrelin secretion from cultured gastric mucosal cells of control mice but not from those of GhIRKO mice. Acute insulin challenge and insulin infusion during both hyperinsulinemic-hypoglycemic clamps and hyperinsulinemic-euglycemic clamps lowered plasma ghrelin in control mice but not GhIRKO mice. Thus, ghrelin cell-expressed IRs are required for insulin-mediated reductions in plasma ghrelin. Furthermore, interventions that naturally raise insulin (glucose gavage, refeeding following fasting, and chronic high-fat diet) also lowered plasma ghrelin only in control mice - not GhIRKO mice. Thus, meal- and obesity-associated increases in insulin, acting via ghrelin cell-expressed IRs, represent a major, direct negative modulator of ghrelin secretion in vivo, as opposed to ingested or metabolized macronutrients. Refed GhIRKO mice exhibited reduced plasma insulin, highlighting ghrelin's actions to inhibit insulin release via a feedback loop. Moreover, GhIRKO mice required reduced glucose infusion rates during hyperinsulinemic-hypoglycemic clamps, suggesting that suppressed ghrelin release resulting from direct insulin action on ghrelin cells usually limits ghrelin's full potential to protect against insulin-induced hypoglycemia.
Collapse
Affiliation(s)
- Kripa Shankar
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Shota Takemi
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
- Area of Regulatory Biology, Division of Life Science, Graduate School of Science and Engineering, Saitama University, Sakuraku, Saitama, Japan
| | - Deepali Gupta
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Salil Varshney
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Bharath K. Mani
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Sherri Osborne-Lawrence
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Nathan P. Metzger
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Corine P. Richard
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Eric D. Berglund
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Jeffrey M. Zigman
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
- Division of Endocrinology, Department of Internal Medicine, and
- Department of Psychiatry, UT Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
19
|
Ouerghi N, Feki M, Bragazzi NL, Knechtle B, Hill L, Nikolaidis PT, Bouassida A. Ghrelin Response to Acute and Chronic Exercise: Insights and Implications from a Systematic Review of the Literature. Sports Med 2021; 51:2389-2410. [PMID: 34374968 PMCID: PMC8514378 DOI: 10.1007/s40279-021-01518-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/05/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND Ghrelin is a peptide hormone predominantly produced by the stomach. It exerts a wide range of functions including stimulating growth hormone release and regulating appetite, food intake, and glucose and lipid metabolism. Since physical exercise affects all these aspects, a particular interest is accorded to the relationship between ghrelin and exercise. This systematic review aimed to summarize the current available data on the topic for a better understanding of the relationship. METHODS An extensive computerized search was performed in the PubMed and SPORTDiscus databases for retrieving relevant articles. The search contained the following keywords: ghrelin, appetite-related peptides, gastrointestinal peptides, gastrointestinal hormones, exercise, acute exercise, chronic exercise, training, and physical activity. Studies investigating the effects of acute/chronic exercise on circulating forms of ghrelin were included. RESULTS The initial search identified 840 articles. After screening, 80 articles were included. Despite a heterogeneity of studies and a variability of the findings, the review suggests that acute exercise suppresses acyl ghrelin production regardless of the participants and the exercise characteristics. Long- and very long-term exercise training programs mostly resulted in increased total and des-acyl ghrelin production. The increase is more noticeable in overweight/obese individuals, and is most likely due to weight loss resulting from the training program. CONCLUSION The review suggests that exercise may impact ghrelin production. While the precise mechanisms are unclear, the effects are likely due to blood flow redistribution and weight loss for acute and chronic exercise, respectively. These changes are expected to be metabolically beneficial. Further research is needed for a better understanding of the relationship between ghrelin and exercise.
Collapse
Affiliation(s)
- Nejmeddine Ouerghi
- High Institute of Sport and Physical Education of Kef, UR13JS01, University of Jendouba, 7100, Kef, Tunisia.,Faculty of Medicine of Tunis, Rabta Hospital, LR99ES11, University of Tunis El Manar, 1007, Tunis, Tunisia
| | - Moncef Feki
- Faculty of Medicine of Tunis, Rabta Hospital, LR99ES11, University of Tunis El Manar, 1007, Tunis, Tunisia
| | - Nicola Luigi Bragazzi
- Postgraduate School of Public Health, Department of Health Sciences (DISSAL), University of Genoa, 16132, Genoa, Italy
| | - Beat Knechtle
- Medbase St. Gallen Am Vadianplatz, Vadianstrasse 26, 9001, St. Gallen, Switzerland. .,Institute of Primary Care, University of Zurich, Zurich, Switzerland.
| | - Lee Hill
- Division of Gastroenterology and Nutrition, Department of Pediatrics, McMaster University, Hamilton, L8S 4L8, Canada
| | | | - Anissa Bouassida
- High Institute of Sport and Physical Education of Kef, UR13JS01, University of Jendouba, 7100, Kef, Tunisia
| |
Collapse
|
20
|
Gupta D, Patterson AM, Osborne-Lawrence S, Bookout AL, Varshney S, Shankar K, Singh O, Metzger NP, Richard CP, Wyler SC, Elmquist JK, Zigman JM. Disrupting the ghrelin-growth hormone axis limits ghrelin's orexigenic but not glucoregulatory actions. Mol Metab 2021; 53:101258. [PMID: 34023483 PMCID: PMC8203846 DOI: 10.1016/j.molmet.2021.101258] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/26/2021] [Accepted: 05/14/2021] [Indexed: 12/19/2022] Open
Abstract
Objective Acyl-ghrelin regulates eating, body weight, blood glucose, and GH secretion upon binding to its receptor GHSR (growth hormone secretagogue receptor; ghrelin receptor). GHSR is distributed in several brain regions and some peripheral cell-types including pituitary somatotrophs. The objective of the current study was to determine the functional significance of acyl-ghrelin's action on GHSR-expressing somatotrophs in mediating GH secretion and several of acyl-ghrelin's metabolic actions. Methods GH-IRES-Cre mice and loxP-flanked (floxed) GHSR mice were newly developed and then crossed to one another to generate mice that lacked GHSR selectively from somatotrophs. Following validation of mice with somatotroph-selective GHSR deletion, metabolic responses of these mice and control littermates were assessed following both acute and chronic acyl-ghrelin administration, a 24-h fast, and a prolonged 60% chronic caloric restriction protocol modeling starvation. Results In mice with somatotroph-selective GHSR deletion, a single peripheral injection of acyl-ghrelin failed to induce GH secretion or increase food intake, unlike wild-type and other littermate control groups. However, the usual acute blood glucose increase in response to the acyl-ghrelin bolus was preserved. Similarly, chronic s.c. acyl-ghrelin administration to mice with somatotroph-selective GHSR deletion failed to increase plasma GH, food intake, or body weight. Physiologically elevating plasma acyl-ghrelin via a 24-h fast also failed to raise plasma GH and resulted in a limited hyperphagic response upon food reintroduction in mice with somatotroph-selective GHSR deletion, although those mice nonetheless did not exhibit an exaggerated reduction in blood glucose. Physiologically elevating plasma acyl-ghrelin via a 15-day caloric restriction protocol which provided only 40% of usual daily calories failed to raise plasma GH in mice with somatotroph-selective GHSR deletion, although those mice did not exhibit life-threatening hypoglycemia. Conclusions These results reveal that direct engagement of GHSR-expressing somatotrophs is required for a peripheral ghrelin bolus to acutely stimulate GH secretion and the actions of chronic acyl-ghrelin delivery and physiological plasma acyl-ghrelin elevations to increase plasma GH. These results also suggest that actions of acyl-ghrelin to increase food intake and body weight are reliant on direct activation of GHSRs expressed on somatotrophs. Furthermore, these results suggest that the glucoregulatory actions of acyl-ghrelin – in particular, its actions to raise blood glucose when acutely administered, prevent small blood glucose drops following a 24-h fast, and avert life-threatening hypoglycemia during an acute-on-chronic caloric restriction protocol – do not depend on GHSR expression by somatotrophs. Mice with pituitary somatotroph-selective GHSR deletion were generated. Somatotroph-expressed GHSRs mediate GH secretion and food intake after acute ghrelin. Body weight effects of chronic ghrelin infusion require somatotroph-expressed GHSRs. Somatotroph-expressed GHSRs enable GH to increase upon chronic caloric restriction. Mice lacking somatotroph GHSRs maintain euglycemia upon chronic caloric restriction.
Collapse
Affiliation(s)
- Deepali Gupta
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA
| | - Anna M Patterson
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA
| | - Sherri Osborne-Lawrence
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA
| | - Angie L Bookout
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA
| | - Salil Varshney
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA
| | - Kripa Shankar
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA
| | - Omprakash Singh
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA
| | - Nathan P Metzger
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA
| | - Corine P Richard
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA
| | - Steven C Wyler
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA
| | - Joel K Elmquist
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA; Division of Endocrinology, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA; Department of Psychiatry, UT Southwestern Medical Center, Dallas, TX, USA
| | - Jeffrey M Zigman
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA; Division of Endocrinology, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA; Department of Psychiatry, UT Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
21
|
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
|
22
|
Nunez‐Salces M, Li H, Feinle‐Bisset C, Young RL, Page AJ. The regulation of gastric ghrelin secretion. Acta Physiol (Oxf) 2021; 231:e13588. [PMID: 33249751 DOI: 10.1111/apha.13588] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 11/19/2020] [Accepted: 11/23/2020] [Indexed: 12/13/2022]
Abstract
Ghrelin is a gastric hormone with multiple physiological functions, including the stimulation of food intake and adiposity. It is well established that circulating ghrelin levels are closely associated with feeding patterns, rising strongly before a meal and lowering upon food intake. However, the mechanisms underlying the modulation of ghrelin secretion are not fully understood. The purpose of this review is to discuss current knowledge on the circadian oscillation of circulating ghrelin levels, the neural mechanisms stimulating fasting ghrelin levels and peripheral mechanisms modulating postprandial ghrelin levels. Furthermore, the therapeutic potential of targeting the ghrelin pathway is discussed in the context of the treatment of various metabolic disorders, including obesity, type 2 diabetes, diabetic gastroparesis and Prader-Willi syndrome. Moreover, eating disorders including anorexia nervosa, bulimia nervosa and binge-eating disorder are also discussed.
Collapse
Affiliation(s)
- Maria Nunez‐Salces
- Vagal Afferent Research Group Adelaide Medical School The University of Adelaide Adelaide SA Australia
- Centre of Research Excellence in Translating Nutritional Science to Good Health Adelaide Medical School The University of Adelaide Adelaide SA Australia
- Nutrition, Diabetes & Gut Health, Lifelong Health Theme South Australian Health & Medical Research Institute (SAHMRI) Adelaide SA Australia
| | - Hui Li
- Vagal Afferent Research Group Adelaide Medical School The University of Adelaide Adelaide SA Australia
- Centre of Research Excellence in Translating Nutritional Science to Good Health Adelaide Medical School The University of Adelaide Adelaide SA Australia
- Nutrition, Diabetes & Gut Health, Lifelong Health Theme South Australian Health & Medical Research Institute (SAHMRI) Adelaide SA Australia
| | - Christine Feinle‐Bisset
- Centre of Research Excellence in Translating Nutritional Science to Good Health Adelaide Medical School The University of Adelaide Adelaide SA Australia
| | - Richard L. Young
- Centre of Research Excellence in Translating Nutritional Science to Good Health Adelaide Medical School The University of Adelaide Adelaide SA Australia
- Nutrition, Diabetes & Gut Health, Lifelong Health Theme South Australian Health & Medical Research Institute (SAHMRI) Adelaide SA Australia
- Intestinal Nutrient Sensing Group Adelaide Medical School The University of Adelaide Adelaide SA Australia
| | - Amanda J. Page
- Vagal Afferent Research Group Adelaide Medical School The University of Adelaide Adelaide SA Australia
- Centre of Research Excellence in Translating Nutritional Science to Good Health Adelaide Medical School The University of Adelaide Adelaide SA Australia
- Nutrition, Diabetes & Gut Health, Lifelong Health Theme South Australian Health & Medical Research Institute (SAHMRI) Adelaide SA Australia
| |
Collapse
|
23
|
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
|
24
|
Kaiser J, Krippeit-Drews P, Drews G. Acyl-Ghrelin Influences Pancreatic β-Cell Function by Interference with K ATP Channels. Diabetes 2021; 70:423-435. [PMID: 33154069 DOI: 10.2337/db20-0231] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 11/03/2020] [Indexed: 11/13/2022]
Abstract
The aim for this study was to elucidate how the hypothalamic hunger-inducing hormone acyl-ghrelin (AG), which is also produced in the pancreas, affects β-cell function, with particular attention to the role of ATP-sensitive K+ (KATP) channels and the exact site of action of the hormone. AG hyperpolarized the membrane potential and decreased cytoplasmic calcium concentration [Ca2+]c and glucose-stimulated insulin secretion (GSIS). These effects were abolished in β-cells from SUR1-knockout (KO) mice. AG increased KATP current but only in a configuration with intact metabolism. Unacylated ghrelin counteracted the effects of AG. The influence of AG on membrane potential and GSIS could only be averted in the combined presence of a ghrelin receptor (GHSR1a) antagonist and an inverse agonist. The inhibition of GSIS by AG could be prevented by dibutyryl cyclic-cAMP or 3-isobutyl-1-methylxanthine and the somatostatin (SST) receptor 2-5 antagonist H6056. These data indicate that AG indirectly opens KATP channels probably by interference with the cAMP/cAMP-dependent protein kinase pathway, resulting in a decrease of [Ca2+]c and GSIS. The experiments with SUR1-KO β-cells point to a direct effect of AG on β-cells and not, as earlier suggested, to an exclusive effect by AG-induced SST release from δ-cells. Nevertheless, SST receptors may be involved in the effect of AG, possibly by heteromerization of AG and SST receptors.
Collapse
Affiliation(s)
- Julia Kaiser
- Department of Pharmacology, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Peter Krippeit-Drews
- Department of Pharmacology, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Gisela Drews
- Department of Pharmacology, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| |
Collapse
|
25
|
Liang Y, Yin W, Yin Y, Zhang W. Ghrelin Based Therapy of Metabolic Diseases. Curr Med Chem 2021; 28:2565-2576. [PMID: 32538716 PMCID: PMC11213490 DOI: 10.2174/0929867327666200615152804] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 05/04/2020] [Accepted: 05/18/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Ghrelin, a unique 28 amino acid peptide hormone secreted by the gastric X/A like cells, is an endogenous ligand of the growth hormone secretagogue receptor (GHSR). Ghrelin-GHSR signaling has been found to exert various physiological functions, including stimulation of appetite, regulation of body weight, lipid and glucose metabolism, and increase of gut motility and secretion. This system is thus critical for energy homeostasis. OBJECTIVE The objective of this review is to highlight the strategies of ghrelin-GHSR based intervention for therapy of obesity and its related metabolic diseases. RESULTS Therapeutic strategies of metabolic disorders targeting the ghrelin-GHSR pathway involve neutralization of circulating ghrelin by antibodies and RNA spiegelmers, antagonism of ghrelin receptor by its antagonists and inverse agonists, inhibition of ghrelin O-acyltransferase (GOAT), as well as potential pharmacological approach to decrease ghrelin synthesis and secretion. CONCLUSION Various compounds targeting the ghrelin-GHSR system have shown promising efficacy for the intervention of obesity and relevant metabolic disorders in animals and in vitro. Further clinical trials to validate their efficacy in human beings are urgently needed.
Collapse
Affiliation(s)
- Yuan Liang
- Key Laboratory of Molecular Cardiovascular Science, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Wenzhen Yin
- Key Laboratory of Molecular Cardiovascular Science, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Yue Yin
- Key Laboratory of Molecular Cardiovascular Science, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Weizhen Zhang
- Key Laboratory of Molecular Cardiovascular Science, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
- Department of Surgery, University of Michigan Medical Center, Ann Arbor, MI 48109-0346, USA
| |
Collapse
|
26
|
Reich N, Hölscher C. Acylated Ghrelin as a Multi-Targeted Therapy for Alzheimer's and Parkinson's Disease. Front Neurosci 2020; 14:614828. [PMID: 33381011 PMCID: PMC7767977 DOI: 10.3389/fnins.2020.614828] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 11/27/2020] [Indexed: 12/13/2022] Open
Abstract
Much thought has been given to the impact of Amyloid Beta, Tau and Alpha-Synuclein in the development of Alzheimer's disease (AD) and Parkinson's disease (PD), yet the clinical failures of the recent decades indicate that there are further pathological mechanisms at work. Indeed, besides amyloids, AD and PD are characterized by the culminative interplay of oxidative stress, mitochondrial dysfunction and hyperfission, defective autophagy and mitophagy, systemic inflammation, BBB and vascular damage, demyelination, cerebral insulin resistance, the loss of dopamine production in PD, impaired neurogenesis and, of course, widespread axonal, synaptic and neuronal degeneration that leads to cognitive and motor impediments. Interestingly, the acylated form of the hormone ghrelin has shown the potential to ameliorate the latter pathologic changes, although some studies indicate a few complications that need to be considered in the long-term administration of the hormone. As such, this review will illustrate the wide-ranging neuroprotective properties of acylated ghrelin and critically evaluate the hormone's therapeutic benefits for the treatment of AD and PD.
Collapse
Affiliation(s)
- Niklas Reich
- Biomedical & Life Sciences Division, Lancaster University, Lancaster, United Kingdom
| | - Christian Hölscher
- Neurology Department, A Second Hospital, Shanxi Medical University, Taiyuan, China.,Research and Experimental Center, Henan University of Chinese Medicine, Zhengzhou, China
| |
Collapse
|
27
|
"A LEAP 2 conclusions? Targeting the ghrelin system to treat obesity and diabetes". Mol Metab 2020; 46:101128. [PMID: 33246141 PMCID: PMC8085568 DOI: 10.1016/j.molmet.2020.101128] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 11/15/2020] [Accepted: 11/20/2020] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND The hormone ghrelin stimulates food intake, promotes adiposity, increases body weight, and elevates blood glucose. Consequently, alterations in plasma ghrelin levels and the functioning of other components of the broader ghrelin system have been proposed as potential contributors to obesity and diabetes. Furthermore, targeting the ghrelin system has been proposed as a novel therapeutic strategy for obesity and diabetes. SCOPE OF REVIEW The current review focuses on the potential for targeting ghrelin and other proteins comprising the ghrelin system as a treatment for obesity and diabetes. The main components of the ghrelin system are introduced. Data supporting a role for the endogenous ghrelin system in the development of obesity and diabetes along with data that seemingly refute such a role are outlined. An argument for further research into the development of ghrelin system-targeted therapeutic agents is delineated. Also, an evidence-based discussion of potential factors and contexts that might influence the efficacy of this class of therapeutics is provided. MAJOR CONCLUSIONS It would not be a "leap to" conclusions to suggest that agents which target the ghrelin system - including those that lower acyl-ghrelin levels, raise LEAP2 levels, block GHSR activity, and/or raise desacyl-ghrelin signaling - could represent efficacious novel treatments for obesity and diabetes.
Collapse
|
28
|
Sustkova‐Fiserova M, Puskina N, Havlickova T, Lapka M, Syslova K, Pohorala V, Charalambous C. Ghrelin receptor antagonism of fentanyl-induced conditioned place preference, intravenous self-administration, and dopamine release in the nucleus accumbens in rats. Addict Biol 2020; 25:e12845. [PMID: 31696597 DOI: 10.1111/adb.12845] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 09/26/2019] [Accepted: 10/02/2019] [Indexed: 12/22/2022]
Abstract
The extended occurrence of fentanils abuse associated with the dramatic increase in opioid fatal overdoses and dependence strongly emphasizes insufficiencies in opioid addiction treatment. Recently, the growth hormone secretagogue receptor (GHS-R1A) antagonism was proposed as a promising mechanism for drug addiction therapy. However, the role of GHS-R1A and its endogenous ligand ghrelin in opioid abuse is still unclear. Therefore, the aim of our study was to clarify whether the GHS-R1A antagonist JMV2959 could reduce the fentanyl-induced conditioned place preference (CPP), the fentanyl intravenous self-administration (IVSA), and the tendency to relapse, but also whether JMV2959 could significantly influence the fentanyl-induced dopamine efflux in the nucleus accumbens (NAC) in rats, that importantly participates in opioids' reinforcing effects. Following an ongoing fentanyl self-administration, JMV2959 3 mg/kg was administered intraperitoneally 20 minutes before three consequent daily 360-minute IVSA sessions under a fixed ratio FR1, which significantly reduced the number of active lever-pressing, the number of infusions, and the fentanyl intake. Pretreatment with JMV2959 also reduced the fentanyl-seeking/relapse-like behaviour tested in rats on the 12th day of the forced abstinence period. Pretreatment with JMV2959 significantly and dose-dependently reduced the manifestation of fentanyl-CPP. The fentanyl-CPP development was reduced after the simultaneous administration of JMV2959 with fentanyl during conditioning. The JMV2959 significantly reduced the accumbens dopamine release induced by subcutaneous and intravenous fentanyl. Simultaneously, it affected the concentration of byproducts associated with dopamine metabolism in the NAC. Our findings suggest that GHS-R1A importantly participates in the rewarding/reinforcing effects of fentanyl.
Collapse
Affiliation(s)
| | - Nina Puskina
- Department of Addictology First Faculty of Medicine Charles University Czech Republic
| | - Tereza Havlickova
- Department of Pharmacology, Third Faculty of Medicine Charles University Czech Republic
| | - Marek Lapka
- Department of Pharmacology, Third Faculty of Medicine Charles University Czech Republic
| | - Kamila Syslova
- Laboratory of Medicinal Diagnostics Department of Organic Technology ICT Czech Republic
| | - Veronika Pohorala
- Department of Pharmacology, Third Faculty of Medicine Charles University Czech Republic
| | | |
Collapse
|
29
|
Childs MD, Luyt LG. A Decade's Progress in the Development of Molecular Imaging Agents Targeting the Growth Hormone Secretagogue Receptor. Mol Imaging 2020; 19:1536012120952623. [PMID: 33104445 PMCID: PMC8865914 DOI: 10.1177/1536012120952623] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
The growth hormone secretagogue receptor 1a (GHSR), also called the ghrelin receptor, is a G protein-coupled receptor known to play an important metabolic role in the regulation of various physiological processes, including energy expenditure, growth hormone secretion, and cell proliferation. This receptor has been implicated in numerous health issues including obesity, gastrointestinal disorders, type II diabetes, and regulation of body weight in patients with Prader-Willi syndrome, and there has been growing interest in studying its mechanism of behavior to unlock further applications of GHSR-targeted therapeutics. In addition, the GHSR is expressed in various types of cancer including prostate, breast, and testicular cancers, while aberrant expression has been reported in cardiac disease. Targeted molecular imaging of the GHSR could provide insights into its role in biological processes related to these disease states. Over the past decade, imaging probes targeting this receptor have been discovered for the imaging modalities PET, SPECT, and optical imaging. High-affinity analogues of ghrelin, the endogenous ligand for the GHSR, as well as small molecule inhibitors have been developed and evaluated both in vitro and in pre-clinical models. This review provides a comprehensive overview of the molecular imaging agents targeting the GHSR reported to the end of 2019.
Collapse
Affiliation(s)
- Marina D Childs
- Department of Chemistry, University of Western Ontario, London, Ontario, Canada
| | - Leonard G Luyt
- Department of Chemistry, University of Western Ontario, London, Ontario, Canada.,Lawson Health Research Institute, London, Ontario, Canada.,Department of Oncology, University of Western Ontario, London, Ontario, Canada.,Department of Medical Imaging, University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
30
|
Lindqvist A, Shcherbina L, Prasad RB, Miskelly MG, Abels M, Martínez-Lopéz JA, Fred RG, Nergård BJ, Hedenbro J, Groop L, Hjerling-Leffler J, Wierup N. Ghrelin suppresses insulin secretion in human islets and type 2 diabetes patients have diminished islet ghrelin cell number and lower plasma ghrelin levels. Mol Cell Endocrinol 2020; 511:110835. [PMID: 32371087 DOI: 10.1016/j.mce.2020.110835] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 04/07/2020] [Accepted: 04/21/2020] [Indexed: 01/22/2023]
Abstract
It is not known how ghrelin affects insulin secretion in human islets from patients with type 2 diabetes (T2D) or whether islet ghrelin expression or circulating ghrelin levels are altered in T2D. Here we sought out to identify the effect of ghrelin on insulin secretion in human islets and the impact of T2D on circulating ghrelin levels and on islet ghrelin cells. The effect of ghrelin on insulin secretion was assessed in human T2D and non-T2D islets. Ghrelin expression was assessed with RNA-sequencing (n = 191) and immunohistochemistry (n = 21). Plasma ghrelin was measured with ELISA in 40 T2D and 40 non-T2D subjects. Ghrelin exerted a glucose-dependent insulin-suppressing effect in islets from both T2D and non-T2D donors. Compared with non-T2D donors, T2D donors had reduced ghrelin mRNA expression and 75% less islet ghrelin cells, and ghrelin mRNA expression correlated negatively with HbA1c. T2D subjects had 25% lower fasting plasma ghrelin levels than matched controls. Thus, ghrelin has direct insulin-suppressing effects in human islets and T2D patients have lower fasting ghrelin levels, likely as a result of reduced number of islet ghrelin cells. These findings support inhibition of ghrelin signaling as a potential therapeutic avenue for stimulation of insulin secretion in T2D patients.
Collapse
Affiliation(s)
- A Lindqvist
- Lund University Diabetes Centre, Lund University, Malmö, Sweden
| | - L Shcherbina
- Lund University Diabetes Centre, Lund University, Malmö, Sweden
| | - R B Prasad
- Lund University Diabetes Centre, Lund University, Malmö, Sweden
| | - M G Miskelly
- Lund University Diabetes Centre, Lund University, Malmö, Sweden
| | - M Abels
- Lund University Diabetes Centre, Lund University, Malmö, Sweden
| | - J A Martínez-Lopéz
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - R G Fred
- Lund University Diabetes Centre, Lund University, Malmö, Sweden
| | | | - J Hedenbro
- Lund University Diabetes Centre, Lund University, Malmö, Sweden; Aleris Obesitas, Lund, Sweden
| | - L Groop
- Lund University Diabetes Centre, Lund University, Malmö, Sweden; Finnish Institute of Molecular Medicine, Helsinki, Finland
| | - J Hjerling-Leffler
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - N Wierup
- Lund University Diabetes Centre, Lund University, Malmö, Sweden.
| |
Collapse
|
31
|
Williams DM, Nawaz A, Evans M. Drug Therapy in Obesity: A Review of Current and Emerging Treatments. Diabetes Ther 2020; 11:1199-1216. [PMID: 32297119 PMCID: PMC7261312 DOI: 10.1007/s13300-020-00816-y] [Citation(s) in RCA: 119] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Indexed: 12/25/2022] Open
Abstract
Whilst the prevalence of obesity continues to increase at an alarming rate worldwide, the personal and economic burden of obesity-related complications becomes ever more important. Whilst dietary and lifestyle measures remain the fundamental focus of the patient to counter obesity, more frequently pharmacological and/or surgical interventions are required. Nevertheless, these therapies are often limited by weight loss efficacy, side effects, surgical risks and frequently obesity relapse. Currently, only five drug therapies are approved for the specific treatment of obesity. However, our understanding of the pathophysiology of obesity and of gut hormones has developed precipitously over the last 20-30 years. As a result, there has been a recent movement to create and use analogues that manipulate these gut hormones to support weight loss. In this article we review the efficacy of the currently approved drug therapies and discuss future potential drug mechanisms and early clinical trial results exploring these budding avenues. We discuss the use of glucagon-like peptide-1 (GLP-1) analogues as monotherapy and unimolecular dual or triple agonists that exploit the GLP-1 receptor and/or the gastric inhibitory peptide (GIP) receptor and/or the glucagon receptor. We also explore the use of sodium-glucose co-transporter-2 (SGLT-2) inhibitors, amylin mimetics, leptin analogues, ghrelin antagonists and centrally acting agents to suppress appetite [neuropeptide Y (NPY) antagonists, melanocortin-4 receptor (MC4R) agonists and cannabinoid-1 receptor antagonists]. Whilst further evidence is required to support their clinical use, preclinical and early clinical trial results are encouraging.
Collapse
Affiliation(s)
- David M Williams
- Department of Diabetes and Endocrinology, University Hospital Llandough, Cardiff, UK.
| | - Asif Nawaz
- Department of Diabetes and Endocrinology, University Hospital Llandough, Cardiff, UK
| | - Marc Evans
- Department of Diabetes and Endocrinology, University Hospital Llandough, Cardiff, UK
| |
Collapse
|
32
|
Gewehr MCF, Silverio R, Rosa-Neto JC, Lira FS, Reckziegel P, Ferro ES. Peptides from Natural or Rationally Designed Sources Can Be Used in Overweight, Obesity, and Type 2 Diabetes Therapies. Molecules 2020; 25:E1093. [PMID: 32121443 PMCID: PMC7179135 DOI: 10.3390/molecules25051093] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 02/20/2020] [Accepted: 02/25/2020] [Indexed: 12/18/2022] Open
Abstract
Overweight and obesity are among the most prominent health problems in the modern world, mostly because they are either associated with or increase the risk of other diseases such as type 2 diabetes, hypertension, and/or cancer. Most professional organizations define overweight and obesity according to individual body-mass index (BMI, weight in kilograms divided by height squared in meters). Overweight is defined as individuals with BMI from 25 to 29, and obesity as individuals with BMI ≥30. Obesity is the result of genetic, behavioral, environmental, physiological, social, and cultural factors that result in energy imbalance and promote excessive fat deposition. Despite all the knowledge concerning the pathophysiology of obesity, which is considered a disease, none of the existing treatments alone or in combination can normalize blood glucose concentration and prevent debilitating complications from obesity. This review discusses some new perspectives for overweight and obesity treatments, including the use of the new orally active cannabinoid peptide Pep19, the advantage of which is the absence of undesired central nervous system effects usually experienced with other cannabinoids.
Collapse
Affiliation(s)
- Mayara C. F. Gewehr
- Department of Pharmacology, Biomedical Sciences Institute, University of São Paulo (USP), São Paulo 05508-000, Brazil;
| | - Renata Silverio
- Department of Pharmacology, Center of Biological Sciences, Federal University of Santa Catarina (UFSC), Florianópolis 88040-900, Brazil;
| | - José Cesar Rosa-Neto
- Department of Cell and Developmental Biology, Biomedical Sciences Institute, University of São Paulo (USP), São Paulo 05508-000, Brazil;
| | - Fabio S. Lira
- Department of Physical Education, São Paulo State University (UNESP), Presidente Prudente 19060-900, Brazil;
| | - Patrícia Reckziegel
- Department of Pharmacology, National Institute of Pharmacology and Molecular Biology (INFAR), Federal University of São Paulo (UNIFESP), São Paulo 05508-000, Brazil;
| | - Emer S. Ferro
- Department of Pharmacology, Biomedical Sciences Institute, University of São Paulo (USP), São Paulo 05508-000, Brazil;
| |
Collapse
|
33
|
Abstract
PURPOSE OF REVIEW Obesity is affecting over 600 million adults worldwide and has numerous negative effects on health. Since ghrelin positively regulates food intake and body weight, targeting its signaling to induce weight loss under conditions of obesity seems promising. Thus, the present work reviews and discusses different possibilities to alter ghrelin signaling. RECENT FINDINGS Ghrelin signaling can be altered by RNA Spiegelmers, GHSR/Fc, ghrelin-O-acyltransferase inhibitors as well as antagonists, and inverse agonists of the ghrelin receptor. PF-05190457 is the first inverse agonist of the ghrelin receptor tested in humans shown to inhibit growth hormone secretion, gastric emptying, and reduce postprandial glucose levels. Effects on body weight were not examined. Although various highly promising agents targeting ghrelin signaling exist, so far, they were mostly only tested in vitro or in animal models. Further research in humans is thus needed to further assess the effects of ghrelin antagonism on body weight especially under conditions of obesity.
Collapse
Affiliation(s)
- Martha A Schalla
- Charité Center for Internal Medicine and Dermatology, Charité Center for Internal Medicine and Dermatology, Department for Psychosomatic Medicine; Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Andreas Stengel
- Charité Center for Internal Medicine and Dermatology, Charité Center for Internal Medicine and Dermatology, Department for Psychosomatic Medicine; Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.
- Department of Psychosomatic Medicine and Psychotherapy, Medical University Hospital Tübingen, Tübingen, Germany.
| |
Collapse
|
34
|
Rose F, Bloom S, Tan T. Novel approaches to anti-obesity drug discovery with gut hormones over the past 10 years. Expert Opin Drug Discov 2019; 14:1151-1159. [DOI: 10.1080/17460441.2019.1646243] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Frances Rose
- Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, UK
| | - Stephen Bloom
- Department of Investigative Medicine, Imperial College London, London, UK
| | - Tricia Tan
- Department of Investigative Medicine, Imperial College London, London, UK
| |
Collapse
|
35
|
Gray SM, Page LC, Tong J. Ghrelin regulation of glucose metabolism. J Neuroendocrinol 2019; 31:e12705. [PMID: 30849212 PMCID: PMC6688917 DOI: 10.1111/jne.12705] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 03/04/2019] [Accepted: 03/05/2019] [Indexed: 12/14/2022]
Abstract
Ghrelin and its receptor, the growth hormone secretagogue receptor 1a (GHSR1a), are implicated in the regulation of glucose metabolism via direct actions in the pancreatic islet, as well as peripheral insulin-sensitive tissues and the brain. Although many studies have explored the role of ghrelin in glucose tolerance and insulin secretion, a complete mechanistic understanding remains to be clarified. This review highlights the local expression and function of ghrelin and GHSR1a in pancreatic islets and how this axis may modulate insulin secretion from pancreatic β-cells. Additionally, we discuss the effect of ghrelin on in vivo glucose metabolism in rodents and humans, as well as the metabolic circumstances under which the action of ghrelin may predominate.
Collapse
Affiliation(s)
- Sarah. M. Gray
- Duke Molecular Physiology Institute, Duke University, Durham, NC 27701
| | - Laura C. Page
- Division of Endocrinology, Department of Pediatrics, Duke University, Durham, NC 27701
| | - Jenny Tong
- Duke Molecular Physiology Institute, Duke University, Durham, NC 27701
- Division of Endocrinology, Department of Pediatrics, Duke University, Durham, NC 27701
- Division of Endocrinology, Metabolism, and Nutrition, Department of Medicine, Duke University, Durham, NC 27701
| |
Collapse
|
36
|
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: 45] [Impact Index Per Article: 7.5] [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
|
37
|
Buckinx A, Van Den Herrewegen Y, Pierre A, Cottone E, Ben Haj Salah K, Fehrentz JA, Kooijman R, De Bundel D, Smolders I. Differential Effects of a Full and Biased Ghrelin Receptor Agonist in a Mouse Kindling Model. Int J Mol Sci 2019; 20:ijms20102480. [PMID: 31137460 PMCID: PMC6567032 DOI: 10.3390/ijms20102480] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 05/15/2019] [Accepted: 05/15/2019] [Indexed: 12/23/2022] Open
Abstract
The ghrelin system has received substantial recognition as a potential target for novel anti-seizure drugs. Ghrelin receptor (ghrelin-R) signaling is complex, involving Gαq/11, Gαi/o, Gα12/13, and β-arrestin pathways. In this study, we aimed to deepen our understanding regarding signaling pathways downstream the ghrelin-R responsible for mediating anticonvulsive effects in a kindling model. Mice were administered the proconvulsive dopamine 1 receptor-agonist, SKF81297, to gradually induce a kindled state. Prior to every SKF81297 injection, mice were treated with a ghrelin-R full agonist (JMV-1843), a Gαq and Gα12 biased ligand unable to recruit β-arrestin (YIL781), a ghrelin-R antagonist (JMV-2959), or saline. Mice treated with JMV-1843 had fewer and less severe seizures compared to saline-treated controls, while mice treated with YIL781 experienced longer and more severe seizures. JMV-2959 treatment did not lead to differences in seizure severity and number. Altogether, these results indicate that the Gαq or Gα12 signaling pathways are not responsible for mediating JMV-1843′s anticonvulsive effects and suggest a possible involvement of β-arrestin signaling in the anticonvulsive effects mediated by ghrelin-R modulation.
Collapse
Affiliation(s)
- An Buckinx
- Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information, Research Group Experimental Pharmacology, Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussels, Belgium.
| | - Yana Van Den Herrewegen
- Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information, Research Group Experimental Pharmacology, Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussels, Belgium.
| | - Anouk Pierre
- Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information, Research Group Experimental Pharmacology, Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussels, Belgium.
| | - Eleonora Cottone
- Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information, Research Group Experimental Pharmacology, Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussels, Belgium.
| | - Khoubaib Ben Haj Salah
- Max Mousseron Institute of Biomolecules UMR524, CNRS, University of Montpellier, Ecole Nationale Supérieure de Chimie de Montpellier, 34090 Montpellier, France.
| | - Jean-Alain Fehrentz
- Max Mousseron Institute of Biomolecules UMR524, CNRS, University of Montpellier, Ecole Nationale Supérieure de Chimie de Montpellier, 34090 Montpellier, France.
| | - Ron Kooijman
- Research Group Experimental Pharmacology, Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), 1050 Brussels, Belgium.
| | - Dimitri De Bundel
- Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information, Research Group Experimental Pharmacology, Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussels, Belgium.
| | - Ilse Smolders
- Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information, Research Group Experimental Pharmacology, Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussels, Belgium.
| |
Collapse
|
38
|
Mani BK, Shankar K, Zigman JM. Ghrelin's Relationship to Blood Glucose. Endocrinology 2019; 160:1247-1261. [PMID: 30874792 PMCID: PMC6482034 DOI: 10.1210/en.2019-00074] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 03/09/2019] [Indexed: 12/16/2022]
Abstract
Much effort has been directed at studying the orexigenic actions of administered ghrelin and the potential effects of the endogenous ghrelin system on food intake, food reward, body weight, adiposity, and energy expenditure. Although endogenous ghrelin's actions on some of these processes remain ambiguous, its glucoregulatory actions have emerged as well-recognized features during extreme metabolic conditions. The blood glucose-raising actions of ghrelin are beneficial during starvation-like conditions, defending against life-threatening falls in blood glucose, but they are seemingly detrimental in obese states and in certain monogenic forms of diabetes, contributing to hyperglycemia. Also of interest, blood glucose negatively regulates ghrelin secretion. This article reviews the literature suggesting the existence of a blood glucose-ghrelin axis and highlights the factors that mediate the glucoregulatory actions of ghrelin, especially during metabolic extremes such as starvation and diabetes.
Collapse
Affiliation(s)
- Bharath K Mani
- Division of Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
- Division of Endocrinology and Metabolism, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Kripa Shankar
- Division of Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
- Division of Endocrinology and Metabolism, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Jeffrey M Zigman
- Division of Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
- Division of Endocrinology and Metabolism, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, Texas
- Correspondence: Jeffrey M. Zigman, MD, PhD, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, Texas 75390. E-mail:
| |
Collapse
|
39
|
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
|
40
|
Translating biased signaling in the ghrelin receptor system into differential in vivo functions. Proc Natl Acad Sci U S A 2018; 115:E10255-E10264. [PMID: 30301804 DOI: 10.1073/pnas.1804003115] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Biased signaling has been suggested as a means of selectively modulating a limited fraction of the signaling pathways for G-protein-coupled receptor family members. Hence, biased ligands may allow modulation of only the desired physiological functions and not elicit undesired effects associated with pharmacological treatments. The ghrelin receptor is a highly sought antiobesity target, since the gut hormone ghrelin in humans has been shown to increase both food intake and fat accumulation. However, it also modulates mood, behavior, growth hormone secretion, and gastric motility. Thus, blocking all pathways of this receptor may give rise to potential side effects. In the present study, we describe a highly promiscuous signaling capacity for the ghrelin receptor. We tested selected ligands for their ability to regulate the various pathways engaged by the receptor. Among those, a biased ligand, YIL781, was found to activate the Gαq/11 and Gα12 pathways selectively without affecting the engagement of β-arrestin or other G proteins. YIL781 was further characterized for its in vivo physiological functions. In combination with the use of mice in which Gαq/11 was selectively deleted in the appetite-regulating AgRP neurons, this biased ligand allowed us to demonstrate that selective blockade of Gαq/11, without antagonism at β-arrestin or other G-protein coupling is sufficient to decrease food intake.
Collapse
|
41
|
Havlickova T, Charalambous C, Lapka M, Puskina N, Jerabek P, Sustkova-Fiserova M. Ghrelin Receptor Antagonism of Methamphetamine-Induced Conditioned Place Preference and Intravenous Self-Administration in Rats. Int J Mol Sci 2018; 19:ijms19102925. [PMID: 30261633 PMCID: PMC6213741 DOI: 10.3390/ijms19102925] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 09/15/2018] [Accepted: 09/24/2018] [Indexed: 02/06/2023] Open
Abstract
Methamphetamine abuse imposes a significant burden on individuals and society worldwide, and an effective therapy of methamphetamine addiction would provide distinguished social benefits. Ghrelin significantly participates in reinforcing neurobiological mechanisms of stimulants, including amphetamines; thus, ghrelin antagonism is proposed as a promising addiction treatment. The aim of our study was to elucidate whether the pretreatment with growth hormone secretagogue receptor (GHS-R1A) antagonist, substance JMV2959, could reduce the methamphetamine intravenous self-administration (IVSA) and the tendency to relapse, and whether JMV2959 could reduce or prevent methamphetamine-induced conditioned place preference (CPP) in rats. Following an adequate maintenance period, JMV2959 3 mg/kg was administered intraperitoneally 20 min before three consequent daily 180 min sessions of methamphetamine IVSA under a fixed ratio FR1, which significantly reduced the number of active lever-pressings, the number of infusions, and the amount of the consumed methamphetamine dose. Pretreatment with JMV2959 also reduced or prevented relapse-like behavior tested in rats on the 12th day of the abstinence period. Pretreatment with JMV2959 significantly reduced the expression of methamphetamine-induced CPP. Simultaneous administration of JMV2959 with methamphetamine during the conditioning period significantly reduced the methamphetamine-CPP. Our results encourage further research of the ghrelin antagonism as a potential new pharmacological tool for methamphetamine addiction treatment.
Collapse
Affiliation(s)
- Tereza Havlickova
- Department of Pharmacology, Third Faculty of Medicine, Charles University, Ruska 87, 1010034 Prague, Czech Republic.
| | - Chrysostomos Charalambous
- Department of Pharmacology, Third Faculty of Medicine, Charles University, Ruska 87, 1010034 Prague, Czech Republic.
| | - Marek Lapka
- Department of Pharmacology, Third Faculty of Medicine, Charles University, Ruska 87, 1010034 Prague, Czech Republic.
| | - Nina Puskina
- Department of Addictology, First Faculty of Medicine, Charles University, Apolinarska 4, 212800 Prague, Czech Republic.
| | - Pavel Jerabek
- Department of Pharmacology, Third Faculty of Medicine, Charles University, Ruska 87, 1010034 Prague, Czech Republic.
| | - Magdalena Sustkova-Fiserova
- Department of Pharmacology, Third Faculty of Medicine, Charles University, Ruska 87, 1010034 Prague, Czech Republic.
| |
Collapse
|
42
|
Napolitano T, Silvano S, Vieira A, Balaji S, Garrido-Utrilla A, Friano ME, Atlija J, Collombat P. Role of ghrelin in pancreatic development and function. Diabetes Obes Metab 2018; 20 Suppl 2:3-10. [PMID: 30230184 DOI: 10.1111/dom.13385] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 05/23/2018] [Accepted: 05/25/2018] [Indexed: 11/28/2022]
Abstract
Ghrelin is a gastric peptide with anabolic functions. It acutely stimulates growth hormone (GH) secretion from the anterior pituitary glands and modulates hypothalamic circuits that control food intake and energy expenditure. Besides its central activity, ghrelin is also involved in the regulation of pancreatic development and physiology. Particularly, several studies highlighted the ability of ghrelin to sustain β-cell viability and proliferation. Furthermore, ghrelin seems to exert inhibitory effects on pancreatic acinar and endocrine secretory functions. Due to its pleiotropic activity on energy metabolism, ghrelin has become a topic of great interest for experimental research focused on type II diabetes and obesity. The aim of this review is to illustrate the complex and not fully understood interplay between ghrelin, pancreas and glucose homeostasis.
Collapse
Affiliation(s)
- Tiziana Napolitano
- Inserm, CNRS, iBV, University of Nice Sophia Antipolis, Nice, France
- iBV, Institut de Biologie Valrose, Centre de Biochimie, University of Nice Sophia Antipolis, Nice Cedex 2, France
| | - Serena Silvano
- Inserm, CNRS, iBV, University of Nice Sophia Antipolis, Nice, France
- iBV, Institut de Biologie Valrose, Centre de Biochimie, University of Nice Sophia Antipolis, Nice Cedex 2, France
| | - Andhira Vieira
- Inserm, CNRS, iBV, University of Nice Sophia Antipolis, Nice, France
- iBV, Institut de Biologie Valrose, Centre de Biochimie, University of Nice Sophia Antipolis, Nice Cedex 2, France
| | - Shruti Balaji
- Inserm, CNRS, iBV, University of Nice Sophia Antipolis, Nice, France
- iBV, Institut de Biologie Valrose, Centre de Biochimie, University of Nice Sophia Antipolis, Nice Cedex 2, France
| | - Anna Garrido-Utrilla
- Inserm, CNRS, iBV, University of Nice Sophia Antipolis, Nice, France
- iBV, Institut de Biologie Valrose, Centre de Biochimie, University of Nice Sophia Antipolis, Nice Cedex 2, France
| | - Marika E Friano
- Inserm, CNRS, iBV, University of Nice Sophia Antipolis, Nice, France
- iBV, Institut de Biologie Valrose, Centre de Biochimie, University of Nice Sophia Antipolis, Nice Cedex 2, France
| | - Josipa Atlija
- Inserm, CNRS, iBV, University of Nice Sophia Antipolis, Nice, France
- iBV, Institut de Biologie Valrose, Centre de Biochimie, University of Nice Sophia Antipolis, Nice Cedex 2, France
| | - Patrick Collombat
- Inserm, CNRS, iBV, University of Nice Sophia Antipolis, Nice, France
- iBV, Institut de Biologie Valrose, Centre de Biochimie, University of Nice Sophia Antipolis, Nice Cedex 2, France
| |
Collapse
|
43
|
Hope DCD, Tan TMM, Bloom SR. No Guts, No Loss: Toward the Ideal Treatment for Obesity in the Twenty-First Century. Front Endocrinol (Lausanne) 2018; 9:442. [PMID: 30158899 PMCID: PMC6104129 DOI: 10.3389/fendo.2018.00442] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 07/17/2018] [Indexed: 12/25/2022] Open
Abstract
Over the last century, our knowledge of the processes which control appetite and weight regulation has developed significantly. The understanding of where gut hormones fit into the control of energy homeostasis in addition to the rapid advancement of pharmacotherapeutics has paved the way for the development of novel gut hormone analogs to target weight loss. Currently, bariatric surgery remains the most efficacious treatment for obesity. The emergence of gut hormone analogs may provide a useful non-surgical addition to the armamentarium in treating obesity. Simply targeting single gut hormone pathways may be insufficiently efficacious, and combination/multiple-agonist approaches may be necessary to obtain the results required for clear clinical impact.
Collapse
Affiliation(s)
- David C D Hope
- Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, United Kingdom
| | - Tricia M M Tan
- Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, United Kingdom
| | - Stephen R Bloom
- Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, United Kingdom
| |
Collapse
|
44
|
Li Q, Yu Q, Lin L, Zhang H, Peng M, Jing C, Xu G. Hypothalamic peroxisome proliferator-activated receptor gamma regulates ghrelin production and food intake. Neuropeptides 2018; 69:39-45. [PMID: 29655655 DOI: 10.1016/j.npep.2018.04.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 04/06/2018] [Accepted: 04/06/2018] [Indexed: 02/06/2023]
Abstract
Peroxisome proliferator-activated receptor-γ (PPARγ) regulates fatty acid storage, glucose metabolism, and food intake. Ghrelin, a gastric hormone, provides a hunger signal to the central nervous system to stimulate appetite. However, the effects of PPARγ on ghrelin production are still unclear. In the present study, the effects of PPARγ on ghrelin production were examined in lean- or high-fat diet-induced obese (DIO) C57BL/6J mice and mHypoE-42 cells, a hypothalamic cell line. 3rd intracerebroventricular injection of adenoviral-directed overexpression of PPARγ (Ad-PPARγ) reduced hypothalamic and plasma ghrelin, food intake in both lean C57BL/6J mice and diet-induced obese mice. These changes were associated with a significant increase in mechanistic target of rapamycin complex 1 (mTORC1) activity. Overexpression of PPARγ enhanced mTORC1 signaling and suppressed ghrelin production in cultured mHypoE-42 cells. Our results suggest that hypothalamic PPARγ plays a vital role in ghrelin production and food intake in mice.
Collapse
Affiliation(s)
- Qingjie Li
- Department of Physiology, School of Medicine, Jinan University, 601 Huangpu Avenue West, Tianhe District, Guangzhou, Guangdong 510632, China
| | - Quan Yu
- Central Laboratory, School of Medicine, Jinan University, Guangzhou, China
| | - Li Lin
- Department of Physiology, School of Medicine, Jinan University, 601 Huangpu Avenue West, Tianhe District, Guangzhou, Guangdong 510632, China
| | - Heng Zhang
- Department of Physiology, School of Medicine, Jinan University, 601 Huangpu Avenue West, Tianhe District, Guangzhou, Guangdong 510632, China
| | - Miao Peng
- Department of Physiology, School of Medicine, Jinan University, 601 Huangpu Avenue West, Tianhe District, Guangzhou, Guangdong 510632, China
| | - Chunxia Jing
- Department of Epidemiology, School of Medicine, Jinan University, Guangzhou, China
| | - Geyang Xu
- Department of Physiology, School of Medicine, Jinan University, 601 Huangpu Avenue West, Tianhe District, Guangzhou, Guangdong 510632, China.
| |
Collapse
|
45
|
Pustovit RV, Callaghan B, Ringuet MT, Kerr NF, Hunne B, Smyth IM, Pietra C, Furness JB. Evidence that central pathways that mediate defecation utilize ghrelin receptors but do not require endogenous ghrelin. Physiol Rep 2018; 5:5/15/e13385. [PMID: 28801520 PMCID: PMC5555902 DOI: 10.14814/phy2.13385] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 07/17/2017] [Accepted: 07/20/2017] [Indexed: 01/24/2023] Open
Abstract
In laboratory animals and in human, centrally penetrant ghrelin receptor agonists, given systemically or orally, cause defecation. Animal studies show that the effect is due to activation of ghrelin receptors in the spinal lumbosacral defecation centers. However, it is not known whether there is a physiological role of ghrelin or the ghrelin receptor in the control of defecation. Using immunohistochemistry and immunoassay, we detected and measured ghrelin in the stomach, but were unable to detect ghrelin by either method in the lumbosacral spinal cord, or other regions of the CNS. In rats in which the thoracic spinal cord was transected 5 weeks before, the effects of a ghrelin agonist on colorectal propulsion were significantly enhanced, but defecation caused by water avoidance stress (WAS) was reduced. In knockout rats that expressed no ghrelin and in wild‐type rats, WAS‐induced defecation was reduced by a ghrelin receptor antagonist, to similar extents. We conclude that the ghrelin receptors of the lumbosacral defecation centers have a physiological role in the control of defecation, but that their role is not dependent on ghrelin. This implies that a transmitter other than ghrelin engages the ghrelin receptor or a ghrelin receptor complex.
Collapse
Affiliation(s)
- Ruslan V Pustovit
- Department of Anatomy & Neuroscience, University of Melbourne, Parkville, Victoria, Australia.,Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia
| | - Brid Callaghan
- Department of Anatomy & Neuroscience, University of Melbourne, Parkville, Victoria, Australia
| | - Mitchell T Ringuet
- Department of Anatomy & Neuroscience, University of Melbourne, Parkville, Victoria, Australia.,Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia
| | - Nicole F Kerr
- Department of Anatomy & Neuroscience, University of Melbourne, Parkville, Victoria, Australia
| | - Billie Hunne
- Department of Anatomy & Neuroscience, University of Melbourne, Parkville, Victoria, Australia
| | - Ian M Smyth
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Claudio Pietra
- Helsinn Research and Preclinical Department, Lugano, Switzerland
| | - John B Furness
- Department of Anatomy & Neuroscience, University of Melbourne, Parkville, Victoria, Australia .,Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia
| |
Collapse
|
46
|
Abstract
Ghrelin, a gastric-derived acylated peptide, regulates energy homeostasis by transmitting information about peripheral nutritional status to the brain, and is essential for protecting organisms against famine. Ghrelin operates brain circuits to regulate homeostatic and hedonic feeding. Recent research advances have shed new light on ghrelin's multifaceted roles in cellular homeostasis, which could maintain the internal environment and overcome metaflammation in metabolic organs. Here, we highlight our current understanding of the regulatory mechanisms of the ghrelin system in energy metabolism and cellular homeostasis and its clinical trials. Future studies of ghrelin will further elucidate how the stomach regulates systemic homeostasis.
Collapse
Affiliation(s)
- Shigehisa Yanagi
- Divisions of Neurology, Respirology, Endocrinology and Metabolism, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Kiyotake, Miyazaki 889-1692, Japan
| | - Takahiro Sato
- Molecular Genetics, Institute of Life Science, Kurume University, Kurume 839-0864, Japan
| | - Kenji Kangawa
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka 565-8565, Japan
| | - Masamitsu Nakazato
- Divisions of Neurology, Respirology, Endocrinology and Metabolism, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Kiyotake, Miyazaki 889-1692, Japan; AMED-CREST, Japan Agency for Medical Research and Development, Chiyoda-ku, Tokyo 100-0004, Japan.
| |
Collapse
|
47
|
Liu S, Chen S, Ren J, Li B, Qin B. Ghrelin protects retinal ganglion cells against rotenone via inhibiting apoptosis, restoring mitochondrial function, and activating AKT-mTOR signaling. Neuropeptides 2018; 67:63-70. [PMID: 29174113 DOI: 10.1016/j.npep.2017.11.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2017] [Revised: 10/25/2017] [Accepted: 11/17/2017] [Indexed: 02/06/2023]
Abstract
Ghrelin, a 28-amino acid peptide hormone, has protective effects on neuronal cells. The present study aimed to examine the neuroprotective effects of ghrelin on the rat retinal ganglion cells in the rotenone-induced in vitro model of Parkinson's disease (PD). Cell viability and cell apoptosis were determined by MTT assay and flow cytometry, respectively. Mitochondrial functions were detected by mitochondrial complex I activity assay and mitochondrial membrane potential (MMP) assay. The mRNA and protein expression levels were determined by qRT-PCR and western blot, respectively. Rotenone significantly suppressed cell viability and increased cell apoptosis, also decreased the mitochondrial complex I activity as well as MMP in rat retinal ganglion cell line (RGC-5). Growth hormone secretagogue receptor (Ghsr) siRNA transfection significantly suppressed the expression of Ghsr in RGC-5 cells. Ghrelin treatment attenuated the effects of rotenone-induced changes in cell viability, cell apoptosis and mitochondrial functions in RGC-5 cells. Post-transcriptional suppression by Ghsr siRNA transfection and treatment with GHS-R antagonist, YIL781, both significantly attenuated the effects of ghrelin in RGC-5 cells. Rotenone decreased the protein levels of Bcl-2 and increased the protein levels of Bax, cleaved caspase-3 and cleaved caspase-9, and this effect was reversed by ghrelin treatment. Ghrelin also prevented the inhibitory effects of rotenone on the AKT-mTOR signaling. The effects of ghrelin on the rotenone-induced changes in apoptosis-related protein levels and AKT-mTOR signaling were attenuated by Ghsr siRNA transfection and treatment with YIL781 in the RGC-5 cells. In addition, both rapamycin and AKT inhibitor IV pre-treatment significantly attenuated the effects of ghrelin on rotenone-induced changes in cell viability and cell apoptosis. In conclusion, ghrelin by acting on the GSH-R to protect rat retinal ganglion cells against rotenone via inhibiting apoptosis and restore mitochondrial functions in RGC-5 cells, and this effect was partially associated with the AKT-mTOR signaling pathway in RGC-5 cells.
Collapse
Affiliation(s)
- Shenwen Liu
- Shenzhen Eye Hospital, Affiliated Shenzhen Eye Hospital of Jinan University, Joint College of Optometry, Shenzhen University, Shenzhen Key Laboratory of Ophthalmology, Shenzhen City, Guangdong Province, China
| | - Sheng Chen
- Shenzhen Eye Hospital, Affiliated Shenzhen Eye Hospital of Jinan University, Joint College of Optometry, Shenzhen University, Shenzhen Key Laboratory of Ophthalmology, Shenzhen City, Guangdong Province, China
| | - Jing Ren
- Shenzhen Eye Hospital, Affiliated Shenzhen Eye Hospital of Jinan University, Joint College of Optometry, Shenzhen University, Shenzhen Key Laboratory of Ophthalmology, Shenzhen City, Guangdong Province, China
| | - Baijun Li
- Shenzhen Eye Hospital, Affiliated Shenzhen Eye Hospital of Jinan University, Joint College of Optometry, Shenzhen University, Shenzhen Key Laboratory of Ophthalmology, Shenzhen City, Guangdong Province, China
| | - Bo Qin
- Shenzhen Eye Hospital, Affiliated Shenzhen Eye Hospital of Jinan University, Joint College of Optometry, Shenzhen University, Shenzhen Key Laboratory of Ophthalmology, Shenzhen City, Guangdong Province, China.
| |
Collapse
|
48
|
Poher AL, Tschöp MH, Müller TD. Ghrelin regulation of glucose metabolism. Peptides 2018; 100:236-242. [PMID: 29412824 PMCID: PMC5805851 DOI: 10.1016/j.peptides.2017.12.015] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 12/15/2017] [Accepted: 12/16/2017] [Indexed: 02/07/2023]
Abstract
The a 28-amino acid peptide ghrelin was discovered in 1999 as a growth hormone (GH) releasing peptide. Soon after its discovery, ghrelin was found to increase body weight and adiposity by acting on the hypothalamic melanocortinergic system. Subsequently, ghrelin was found to exert a series of metabolic effects, overall testifying ghrelin a pleiotropic nature of broad pharmacological interest. Ghrelin acts through the growth hormone secretagogue-receptor (GHS-R), a seven transmembrane G protein-coupled receptor with high expression in the anterior pituitary, pancreatic islets, thyroid gland, heart and various regions of the brain. Among ghrelins numerous metabolic effects are the most prominent the stimulation of appetite via activation of orexigenic hypothalamic neurocircuits and the food-intake independent stimulation of lipogenesis, which both together lead to an increase in body weight and adiposity. Ghrelin effects beyond the regulation of appetite and GH secretion include the regulation of gut motility, sleep-wake rhythm, taste sensation, reward seeking behaviour, and the regulation of glucose metabolism. The latter received recently increasing recognition because pharmacological inhibition of ghrelin signaling might be of therapeutic value to improve insuin resistance and type 2 diabetes. In this review we highlight the multifaceted nature of ghrelin and summarize its glucoregulatory action and discuss the pharmacological value of ghrelin pathway inhibition for the treatment of glucose intolerance and type 2 diabetes.
Collapse
Affiliation(s)
- Anne-Laure Poher
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center (HDC), Helmholtz Zentrum München and German National Diabetes Center (DZD), 85764, Neuherberg, Germany
| | - Matthias H Tschöp
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center (HDC), Helmholtz Zentrum München and German National Diabetes Center (DZD), 85764, Neuherberg, Germany; Division of Metabolic Diseases, Department of Medicine, Technische Universität München, 80333, Munich, Germany
| | - Timo D Müller
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center (HDC), Helmholtz Zentrum München and German National Diabetes Center (DZD), 85764, Neuherberg, Germany.
| |
Collapse
|
49
|
Sun EWL, Martin AM, Young RL, Keating DJ. The Regulation of Peripheral Metabolism by Gut-Derived Hormones. Front Endocrinol (Lausanne) 2018; 9:754. [PMID: 30662430 PMCID: PMC6328484 DOI: 10.3389/fendo.2018.00754] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 11/27/2018] [Indexed: 12/13/2022] Open
Abstract
Enteroendocrine cells lining the gut epithelium constitute the largest endocrine organ in the body and secrete over 20 different hormones in response to cues from ingested foods and changes in nutritional status. Not only do these hormones convey signals from the gut to the brain via the gut-brain axis, they also act directly on metabolically important peripheral targets in a highly concerted fashion to maintain energy balance and glucose homeostasis. Gut-derived hormones released during fasting tend to be orexigenic and have hyperglycaemic potential. Conversely, gut hormones secreted postprandially generally promote satiety and facilitate glucose clearance. Although some of the metabolic benefits conferred by bariatric surgeries have been ascribed to changes in the secretory profiles of various gut hormones, the therapeutic potential of the enteroendocrine system as a viable target against metabolic diseases remain largely underexploited, except for incretin-mimetics. This review provides a brief overview of the physiological importance and highlights the therapeutic potential of the following gut hormones: serotonin, glucose-dependent insulinotropic peptide, glucagon-like peptide 1, oxyntomodulin, peptide YY, insulin-like peptide 5, and ghrelin.
Collapse
Affiliation(s)
- Emily W. L. Sun
- College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Alyce M. Martin
- College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Richard L. Young
- Nutrition and Metabolism, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
| | - Damien J. Keating
- College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
- Nutrition and Metabolism, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
- *Correspondence: Damien J. Keating
| |
Collapse
|
50
|
Mani BK, Zigman JM. Ghrelin as a Survival Hormone. Trends Endocrinol Metab 2017; 28:843-854. [PMID: 29097101 PMCID: PMC5777178 DOI: 10.1016/j.tem.2017.10.001] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 10/03/2017] [Accepted: 10/05/2017] [Indexed: 12/22/2022]
Abstract
Ghrelin administration induces food intake and body weight gain. Based on these actions, the ghrelin system was initially proposed as an antiobesity target. Subsequent studies using genetic mouse models have raised doubts about the role of the endogenous ghrelin system in mediating body weight homeostasis or obesity. However, this is not to say that the endogenous ghrelin system is not important metabolically or otherwise. Here we review an emerging concept in which the endogenous ghrelin system serves an essential function during extreme nutritional and psychological challenges to defend blood glucose, protect body weight, avoid exaggerated depression, and ultimately allow survival.
Collapse
Affiliation(s)
- Bharath K Mani
- Divisions of Hypothalamic Research and Endocrinology, Department of Internal Medicine, and Department of Psychiatry, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390-9077, USA
| | - Jeffrey M Zigman
- Divisions of Hypothalamic Research and Endocrinology, Department of Internal Medicine, and Department of Psychiatry, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390-9077, USA.
| |
Collapse
|