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: 0] [Impact Index Per Article: 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
|
Nestor LJ, Ersche KD. Gut Hormones: Possible Mediators of Addictive Disorders? Eur Addict Res 2024:1-8. [PMID: 39389039 DOI: 10.1159/000540743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 08/02/2024] [Indexed: 10/12/2024]
Abstract
BACKGROUND Alcohol and drug dependence are major health and economic burdens to society. One of the major challenges to reducing this burden will be to develop more effective and better tolerated medications that target alternative mechanisms in the brain. While the dopamine system has been well characterized for mediating the reward value of drugs, there is evidence that the endocrine system also conveys signals to the same neural systems using gut hormones. SUMMARY These gut hormones, produced in the stomach and intestine and that regulate food intake, have also been shown to control the use of other substances, such as alcohol and drugs of abuse. Examples of such hormones are ghrelin and glucagon-like peptide-1, which exert their effects on dopamine transmission in parts of the brain known to be involved in some of the core features of addiction, such as reward sensitivity. KEY MESSAGES This raises the possibility that gut hormone systems may play a pivotal role in addictive disorders. This review will briefly outline emerging evidence that the ghrelin and glucagon-like peptide-1 hormones are contrasting mediators of alcohol and drug use and may present a promising alternative target for treatment intervention in addictive disorders.
Collapse
Affiliation(s)
- Liam J Nestor
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - Karen D Ersche
- Department of Psychiatry, University of Cambridge, Cambridge, UK
- Department of Systems Neuroscience, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
- Department of Addictive Behaviour and Addiction Medicine, Central Institute of Mental Health, University of Heidelberg, Mannheim, Germany
| |
Collapse
|
3
|
Witley S, Edvardsson CE, Aranäs C, Tufvesson-Alm M, Stalberga D, Green H, Vestlund J, Jerlhag E. Des-acyl ghrelin reduces alcohol intake and alcohol-induced reward in rodents. Transl Psychiatry 2024; 14:277. [PMID: 38965230 PMCID: PMC11224403 DOI: 10.1038/s41398-024-02996-8] [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: 03/01/2024] [Revised: 06/23/2024] [Accepted: 06/27/2024] [Indexed: 07/06/2024] Open
Abstract
The mechanisms contributing to alcohol use disorder (AUD) are complex and the orexigenic peptide ghrelin, which enhances alcohol reward, is implied as a crucial modulator. The major proportion of circulating ghrelin is however the non-octanoylated form of ghrelin, des-acyl ghrelin (DAG), whose role in reward processes is unknown. As recent studies show that DAG decreases food intake, we hypothesize that DAG attenuates alcohol-related responses in animal models. Acute and repeated DAG treatment dose-dependently decreased alcohol drinking in male and female rats. In these alcohol-consuming male rats, repeated DAG treatment causes higher levels of dopamine metabolites in the ventral tegmental area, an area central to reward processing. The role of DAG in reward processing is further supported as DAG prevents alcohol-induced locomotor stimulation, reward in the conditioned place preference paradigm, and dopamine release in the nucleus accumbens in male rodents. On the contrary, DAG does not alter the memory of alcohol reward or affect neurotransmission in the hippocampus, an area central to memory. Further, circulating DAG levels are positively correlated with alcohol drinking in female but not male rats. Studies were conducted in attempts to identify tentative targets of DAG, which currently are unknown. Data from these recombinant cell system revealed that DAG does not bind to either of the monoamine transporters, 5HT2A, CB1, or µ-opioid receptors. Collectively, our data show that DAG attenuates alcohol-related responses in rodents, an effect opposite to that of ghrelin, and contributes towards a deeper insight into behaviors regulated by the ghrelinergic signaling pathway.
Collapse
Affiliation(s)
- Sarah Witley
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Christian E Edvardsson
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Cajsa Aranäs
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Maximilian Tufvesson-Alm
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Darta Stalberga
- Division of Clinical Chemistry and Pharmacology, Department of Biomedical and Clinical Sciences, Faculty of Medicine, Linköping University, Linköping, Sweden
| | - Henrik Green
- Division of Clinical Chemistry and Pharmacology, Department of Biomedical and Clinical Sciences, Faculty of Medicine, Linköping University, Linköping, Sweden
- Department of Forensic Genetics and Forensic Toxicology, National Board of Forensic Medicine, Linköping, Sweden
| | - Jesper Vestlund
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Elisabet Jerlhag
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
4
|
Lékó AH, Gregory-Flores A, Marchette RCN, Gomez JL, Vendruscolo JCM, Repunte-Canonigo V, Choung V, Deschaine SL, Whiting KE, Jackson SN, Cornejo MP, Perello M, You ZB, Eckhaus M, Rasineni K, Janda KD, Zorman B, Sumazin P, Koob GF, Michaelides M, Sanna PP, Vendruscolo LF, Leggio L. Genetic or pharmacological GHSR blockade has sexually dimorphic effects in rodents on a high-fat diet. Commun Biol 2024; 7:632. [PMID: 38796563 PMCID: PMC11127961 DOI: 10.1038/s42003-024-06303-5] [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: 09/29/2023] [Accepted: 05/08/2024] [Indexed: 05/28/2024] Open
Abstract
The stomach-derived hormone ghrelin regulates essential physiological functions. The ghrelin receptor (GHSR) has ligand-independent actions; therefore, GHSR gene deletion may be a reasonable approach to investigate the role of this system in feeding behaviors and diet-induced obesity (DIO). Here, we investigate the effects of a long-term (12-month) high-fat (HFD) versus regular diet on obesity-related measures in global GHSR-KO and wild-type (WT) Wistar male and female rats. Our main findings are that the GHSR gene deletion protects against DIO and decreases food intake during HFD in male but not in female rats. GHSR gene deletion increases thermogenesis and brain glucose uptake in male rats and modifies the effects of HFD on brain glucose metabolism in a sex-specific manner, as assessed with small animal positron emission tomography. We use RNA-sequencing to show that GHSR-KO rats have upregulated expression of genes responsible for fat oxidation in brown adipose tissue. Central administration of a novel GHSR inverse agonist, PF-5190457, attenuates ghrelin-induced food intake, but only in male, not in female mice. HFD-induced binge-like eating is reduced by inverse agonism in both sexes. Our results support GHSR as a promising target for new pharmacotherapies for obesity.
Collapse
Affiliation(s)
- András H Lékó
- 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
- Center on Compulsive Behaviors, Intramural Research Program, National Institutes of Health, Bethesda, MD, USA
| | - Adriana Gregory-Flores
- 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
| | - Renata C N Marchette
- Neurobiology of Addiction Section, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Juan L Gomez
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Unit, Neuroimaging Research Branch, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Janaina C M Vendruscolo
- Neurobiology of Addiction Section, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Vez Repunte-Canonigo
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Vicky Choung
- 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
| | - Sara L Deschaine
- 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
| | - Kimberly E Whiting
- 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
| | - Shelley N Jackson
- Translational Analytical Core, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Maria Paula Cornejo
- Grupo de Neurofisiología, Instituto Multidisciplinario de Biología Celular (IMBICE), Universidad Nacional La Plata (UNLP), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) y Comisión de Investigaciones Científicas de la Provincia de Buenos Aires (CIC-PBA), La Plata, Argentina
| | - Mario Perello
- Grupo de Neurofisiología, Instituto Multidisciplinario de Biología Celular (IMBICE), Universidad Nacional La Plata (UNLP), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) y Comisión de Investigaciones Científicas de la Provincia de Buenos Aires (CIC-PBA), La Plata, Argentina
| | - Zhi-Bing You
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Michael Eckhaus
- Pathology Service, Division of Veterinary Resources, Office of Research Services, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Karuna Rasineni
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, USA
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Kim D Janda
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | - Barry Zorman
- Department of Pediatrics, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Pavel Sumazin
- Department of Pediatrics, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - George F Koob
- Neurobiology of Addiction Section, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Michael Michaelides
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Unit, Neuroimaging Research Branch, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Pietro P Sanna
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Leandro F Vendruscolo
- Stress and Addiction Neuroscience Unit, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore, MD, USA.
| | - Lorenzo Leggio
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore, MD, USA.
- Translational Analytical Core, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA.
- Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, School of Public Health, 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
|
5
|
Kulkarni SS, Singh O, Zigman JM. The intersection between ghrelin, metabolism and circadian rhythms. Nat Rev Endocrinol 2024; 20:228-238. [PMID: 38123819 DOI: 10.1038/s41574-023-00927-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [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
|
6
|
Izat N, Bolleddula J, Abbasi A, Cheruzel L, Jones RS, Moss D, Ortega-Muro F, Parmentier Y, Peterkin VC, Tian DD, Venkatakrishnan K, Zientek MA, Barber J, Houston JB, Galetin A, Scotcher D. Challenges and Opportunities for In Vitro-In Vivo Extrapolation of Aldehyde Oxidase-Mediated Clearance: Toward a Roadmap for Quantitative Translation. Drug Metab Dispos 2023; 51:1591-1606. [PMID: 37751998 DOI: 10.1124/dmd.123.001436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 08/25/2023] [Accepted: 08/28/2023] [Indexed: 09/28/2023] Open
Abstract
Underestimation of aldehyde oxidase (AO)-mediated clearance by current in vitro assays leads to uncertainty in human dose projections, thereby reducing the likelihood of success in drug development. In the present study we first evaluated the current drug development practices for AO substrates. Next, the overall predictive performance of in vitro-in vivo extrapolation of unbound hepatic intrinsic clearance (CLint,u) and unbound hepatic intrinsic clearance by AO (CLint,u,AO) was assessed using a comprehensive literature database of in vitro (human cytosol/S9/hepatocytes) and in vivo (intravenous/oral) data collated for 22 AO substrates (total of 100 datapoints from multiple studies). Correction for unbound fraction in the incubation was done by experimental data or in silico predictions. The fraction metabolized by AO (fmAO) determined via in vitro/in vivo approaches was found to be highly variable. The geometric mean fold errors (gmfe) for scaled CLint,u (mL/min/kg) were 10.4 for human hepatocytes, 5.6 for human liver cytosols, and 5.0 for human liver S9, respectively. Application of these gmfe's as empirical scaling factors improved predictions (45%-57% within twofold of observed) compared with no correction (11%-27% within twofold), with the scaling factors qualified by leave-one-out cross-validation. A road map for quantitative translation was then proposed following a critical evaluation on the in vitro and clinical methodology to estimate in vivo fmAO In conclusion, the study provides the most robust system-specific empirical scaling factors to date as a pragmatic approach for the prediction of in vivo CLint,u,AO in the early stages of drug development. SIGNIFICANCE STATEMENT: Confidence remains low when predicting in vivo clearance of AO substrates using in vitro systems, leading to de-prioritization of AO substrates from the drug development pipeline to mitigate risk of unexpected and costly in vivo impact. The current study establishes a set of empirical scaling factors as a pragmatic tool to improve predictability of in vivo AO clearance. Developing clinical pharmacology strategies for AO substrates by utilizing mass balance/clinical drug-drug interaction data will help build confidence in fmAO.
Collapse
Affiliation(s)
- Nihan Izat
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Jayaprakasam Bolleddula
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Armina Abbasi
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Lionel Cheruzel
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Robert S Jones
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Darren Moss
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Fatima Ortega-Muro
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Yannick Parmentier
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Vincent C Peterkin
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Dan-Dan Tian
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Karthik Venkatakrishnan
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Michael A Zientek
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Jill Barber
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - J Brian Houston
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Aleksandra Galetin
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Daniel Scotcher
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| |
Collapse
|
7
|
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: 11] [Impact Index Per Article: 11.0] [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
|
8
|
Leggio L, Leko A, Gregory-Flores A, Marchette R, Gomez J, Vendruscolo J, Repunte-Canonigo V, Chuong V, Deschaine S, Whiting K, Jackson S, Cornejo M, Perello M, You ZB, Eckhaus M, Janda K, Zorman B, Sumazin P, Koob G, Michaelides M, Sanna PP, Vendruscolo L. Genetic or pharmacological GHSR blockade has sexually dimorphic effects in rodents on a high-fat diet. RESEARCH SQUARE 2023:rs.3.rs-3236045. [PMID: 37886546 PMCID: PMC10602167 DOI: 10.21203/rs.3.rs-3236045/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Abstract
The stomach-derived hormone ghrelin regulates essential physiological functions. The ghrelin receptor (GHSR) has ligand-independent actions, therefore, GHSR gene deletion may be a reasonable approach to investigate the role of this system in feeding behaviors and diet-induced obesity (DIO). Here we investigated the effects of a long-term (12 month) high-fat (HFD) versus regular diet on obesity-related measures in global GHSR-KO and wild type (WT) Wistar male and female rats. Our main findings were that the GHSR gene deletion protects against DIO and decreases food intake during HFD in male but not in female rats. GHSR gene deletion increased thermogenesis and brain glucose uptake in male rats and modified the effects of HFD on brain glucose metabolism in a sex-specific manner, as assessed with small animal positron emission tomography. RNA-sequencing was also used to show that GHSR-KO rats had upregulated expression of genes responsible for fat oxidation in brown adipose tissue. Central administration of a novel GHSR inverse agonist, PF-5190457, attenuated ghrelin-induced food intake, but only in male, not in female mice. HFD-induced binge-like eating was reduced by inverse agonism in both sexes. Our results support GHSR as a promising target for new pharmacotherapies for obesity.
Collapse
|
9
|
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
|
10
|
Fischler PV, Soyka M, Seifritz E, Mutschler J. Off-label and investigational drugs in the treatment of alcohol use disorder: A critical review. Front Pharmacol 2022; 13:927703. [PMID: 36263121 PMCID: PMC9574013 DOI: 10.3389/fphar.2022.927703] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 06/30/2022] [Indexed: 11/13/2022] Open
Abstract
Compounds known to be successful in the treatment of alcohol use disorder include the aversive agent, Disulfiram, the glutamatergic NMDA receptor antagonist, Acamprosate, and the opioid receptor antagonists, Naltrexone and Nalmefene. Although all four are effective in maintaining abstinence or reduction of alcohol consumption, only a small percentage of patients receive pharmacological treatment. In addition, many other medications have been investigated for their therapeutic potential in the treatment of alcohol use disorder. In this review we summarize and compare Baclofen, Gabapentin, Topiramate, Ondansetron, Varenicline, Aripiprazole, Quetiapine, Clozapine, Antidepressants, Lithium, Neuropeptide Y, Neuropeptide S, Corticotropin-releasing factor antagonists, Oxytocin, PF-05190457, Memantine, Ifenprodil, Samidorphan, Ondelopran, ABT-436, SSR149415, Mifepristone, Ibudilast, Citicoline, Rimonabant, Surinabant, AM4113 and Gamma-hydroxybutyrate While some have shown promising results in the treatment of alcohol use disorder, others have disappointed and should be excluded from further investigation. Here we discuss the most promising results and highlight medications that deserve further preclinical or clinical study. Effective, patient-tailored treatment will require greater understanding provided by many more preclinical and clinical studies.
Collapse
Affiliation(s)
- Pascal Valentin Fischler
- Department for Gynecology and Obstetrics, Women’s Clinic Lucerne, Cantonal Hospital of Lucerne, Lucerne, Switzerland
- *Correspondence: Pascal Valentin Fischler,
| | - Michael Soyka
- Psychiatric Hospital University of Munich, Munich, Germany
| | - Erich Seifritz
- Director of the Clinic for Psychiatry, Psychotherapy and Psychosomatics, Psychiatric University Clinic Zürich, Zürich, Switzerland
| | | |
Collapse
|
11
|
Toselli F, Golding M, Nicolaï J, Gillent E, Chanteux H. Drug clearance by aldehyde oxidase: can we avoid clinical failure? Xenobiotica 2022; 52:890-903. [PMID: 36170034 DOI: 10.1080/00498254.2022.2129519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Despite increased awareness of aldehyde oxidase (AO) as a major drug-metabolising enzyme, predicting the pharmacokinetics of its substrates remains challenging. Several drug candidates have been terminated due to high clearance, which were subsequently discovered to be AO substrates. Even retrospective extrapolation of human clearance, from models more sensitive to AO activity, often resulted in underprediction.The questions of the current work thus were: Is there an acceptable degree of in vitro AO metabolism that does not result in high in vivo human clearance? And, if so, how can this be predicted?We built an in vitro/in vivo correlation using known AO substrates, combining multiple in vitro parameters to calculate the blood metabolic clearance mediated by AO (CLbAO). This value was compared with observed blood clearance (CLb-obs), establishing cut-off CLbAO values, to discriminate between low and high CLb-obs. The model was validated using additional literature compounds, and CLb-obs was predicted in the correct category.This simple, categorical, semi-quantitative yet multi-factorial model is readily applicable in drug discovery. Further, it is valuable for high-clearance compounds, as it predicts the CLb group, rather than an exact CLb value, for the substrates of this poorly-characterised enzyme.
Collapse
Affiliation(s)
| | | | - Johan Nicolaï
- Development Science, UCB Biopharma, Braine-l'Alleud, Belgium
| | - Eric Gillent
- Development Science, UCB Biopharma, Braine-l'Alleud, Belgium
| | - Hugues Chanteux
- Development Science, UCB Biopharma, Braine-l'Alleud, Belgium
| |
Collapse
|
12
|
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: 0.5] [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
|
13
|
Abstract
Alcohol use disorder (AUD) is a highly prevalent but severely under-treated disorder, with only three widely-approved pharmacotherapies. Given that AUD is a very heterogeneous disorder, it is unlikely that one single medication will be effective for all individuals with an AUD. As such, there is a need to develop new, more effective, and diverse pharmacological treatment options for AUD with the hopes of increasing utilization and improving care. In this qualitative literature review, we discuss the efficacy, mechanism of action, and tolerability of approved, repurposed, and novel pharmacotherapies for the treatment of AUD with a clinical perspective. Pharmacotherapies discussed include: disulfiram, acamprosate, naltrexone, nalmefene, topiramate, gabapentin, varenicline, baclofen, sodium oxybate, aripiprazole, ondansetron, mifepristone, ibudilast, suvorexant, prazosin, doxazosin, N-acetylcysteine, GET73, ASP8062, ABT-436, PF-5190457, and cannabidiol. Overall, many repurposed and novel agents discussed in this review demonstrate clinical effectiveness and promise for the future of AUD treatment. Importantly, these medications also offer potential improvements towards the advancement of precision medicine and personalized treatment for the heterogeneous AUD population. However, there remains a great need to improve access to treatment, increase the menu of approved pharmacological treatments, and de-stigmatize and increase treatment-seeking for AUD.
Collapse
|
14
|
Andersen DB, Holst JJ. Peptides in the regulation of glucagon secretion. Peptides 2022; 148:170683. [PMID: 34748791 DOI: 10.1016/j.peptides.2021.170683] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 10/21/2021] [Accepted: 11/02/2021] [Indexed: 02/06/2023]
Abstract
Glucose homeostasis is maintained by the glucoregulatory hormones, glucagon, insulin and somatostatin, secreted from the islets of Langerhans. Glucagon is the body's most important anti-hypoglycemic hormone, mobilizing glucose from glycogen stores in the liver in response to fasting, thus maintaining plasma glucose levels within healthy limits. Glucagon secretion is regulated by both circulating nutrients, hormones and neuronal inputs. Hormones that may regulate glucagon secretion include locally produced insulin and somatostatin, but also urocortin-3, amylin and pancreatic polypeptide, and from outside the pancreas glucagon-like peptide-1 and 2, peptide tyrosine tyrosine and oxyntomodulin, glucose-dependent insulinotropic polypeptide, neurotensin and ghrelin, as well as the hypothalamic hormones arginine-vasopressin and oxytocin, and calcitonin from the thyroid. Each of these hormones have distinct effects, ranging from regulating blood glucose, to regulating appetite, stomach emptying rate and intestinal motility, which makes them interesting targets for treating metabolic diseases. Awareness regarding the potential effects of the hormones on glucagon secretion is important since secretory abnormalities could manifest as hyperglycemia or even lethal hypoglycemia. Here, we review the effects of each individual hormone on glucagon secretion, their interplay, and how treatments aimed at modulating the plasma levels of these hormones may also influence glucagon secretion and glycemic control.
Collapse
Affiliation(s)
- Daniel B Andersen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Panum Institute, Blegdamsvej 3B, 2200, Copenhagen N, Denmark; Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Jens J Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Panum Institute, Blegdamsvej 3B, 2200, Copenhagen N, Denmark; Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark.
| |
Collapse
|
15
|
Molecular mechanism of agonism and inverse agonism in ghrelin receptor. Nat Commun 2022; 13:300. [PMID: 35027551 PMCID: PMC8758724 DOI: 10.1038/s41467-022-27975-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 12/29/2021] [Indexed: 02/05/2023] Open
Abstract
Much effort has been invested in the investigation of the structural basis of G protein-coupled receptors (GPCRs) activation. Inverse agonists, which can inhibit GPCRs with constitutive activity, are considered useful therapeutic agents, but the molecular mechanism of such ligands remains insufficiently understood. Here, we report a crystal structure of the ghrelin receptor bound to the inverse agonist PF-05190457 and a cryo-electron microscopy structure of the active ghrelin receptor-Go complex bound to the endogenous agonist ghrelin. Our structures reveal a distinct binding mode of the inverse agonist PF-05190457 in the ghrelin receptor, different from the binding mode of agonists and neutral antagonists. Combining the structural comparisons and cellular function assays, we find that a polar network and a notable hydrophobic cluster are required for receptor activation and constitutive activity. Together, our study provides insights into the detailed mechanism of ghrelin receptor binding to agonists and inverse agonists, and paves the way to design specific ligands targeting ghrelin receptors. Ghrelin receptor regulates energy homeostasis through constitutive activity or by the ghrelin. Here the authors report two structures of ghrelin receptor bound to agonist and inverse agonist, providing insights into the mechanism of inverse agonism, which is of interest for specific ligand design.
Collapse
|
16
|
Sustkova-Fiserova M, Charalambous C, Khryakova A, Certilina A, Lapka M, Šlamberová R. The Role of Ghrelin/GHS-R1A Signaling in Nonalcohol Drug Addictions. Int J Mol Sci 2022; 23:761. [PMID: 35054944 PMCID: PMC8776007 DOI: 10.3390/ijms23020761] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 01/05/2022] [Accepted: 01/06/2022] [Indexed: 01/27/2023] Open
Abstract
Drug addiction causes constant serious health, social, and economic burden within the human society. The current drug dependence pharmacotherapies, particularly relapse prevention, remain limited, unsatisfactory, unreliable for opioids and tobacco, and even symptomatic for stimulants and cannabinoids, thus, new more effective treatment strategies are researched. The antagonism of the growth hormone secretagogue receptor type A (GHS-R1A) has been recently proposed as a novel alcohol addiction treatment strategy, and it has been intensively studied in experimental models of other addictive drugs, such as nicotine, stimulants, opioids and cannabinoids. The role of ghrelin signaling in these drugs effects has also been investigated. The present review aims to provide a comprehensive overview of preclinical and clinical studies focused on ghrelin's/GHS-R1A possible involvement in these nonalcohol addictive drugs reinforcing effects and addiction. Although the investigation is still in its early stage, majority of the existing reviewed experimental results from rodents with the addition of few human studies, that searched correlations between the genetic variations of the ghrelin signaling or the ghrelin blood content with the addictive drugs effects, have indicated the importance of the ghrelin's/GHS-R1As involvement in the nonalcohol abused drugs pro-addictive effects. Further research is necessary to elucidate the exact involved mechanisms and to verify the future potential utilization and safety of the GHS-R1A antagonism use for these drug addiction therapies, particularly for reducing the risk of relapse.
Collapse
Affiliation(s)
- Magdalena Sustkova-Fiserova
- Department of Pharmacology, Third Faculty of Medicine, Charles University, Ruska 87, 100 00 Prague, Czech Republic; (C.C.); (A.K.); (A.C.); (M.L.)
| | - Chrysostomos Charalambous
- Department of Pharmacology, Third Faculty of Medicine, Charles University, Ruska 87, 100 00 Prague, Czech Republic; (C.C.); (A.K.); (A.C.); (M.L.)
| | - Anna Khryakova
- Department of Pharmacology, Third Faculty of Medicine, Charles University, Ruska 87, 100 00 Prague, Czech Republic; (C.C.); (A.K.); (A.C.); (M.L.)
| | - Alina Certilina
- Department of Pharmacology, Third Faculty of Medicine, Charles University, Ruska 87, 100 00 Prague, Czech Republic; (C.C.); (A.K.); (A.C.); (M.L.)
| | - Marek Lapka
- Department of Pharmacology, Third Faculty of Medicine, Charles University, Ruska 87, 100 00 Prague, Czech Republic; (C.C.); (A.K.); (A.C.); (M.L.)
| | - Romana Šlamberová
- Department of Physiology, Third Faculty of Medicine, Charles University, Ke Karlovu 4, 120 00 Prague, Czech Republic;
| |
Collapse
|
17
|
Shevchouk OT, Tufvesson-Alm M, Jerlhag E. An Overview of Appetite-Regulatory Peptides in Addiction Processes; From Bench to Bed Side. Front Neurosci 2021; 15:774050. [PMID: 34955726 PMCID: PMC8695496 DOI: 10.3389/fnins.2021.774050] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 11/08/2021] [Indexed: 12/17/2022] Open
Abstract
There is a substantial need for new pharmacological treatments of addiction, and appetite-regulatory peptides are implied as possible candidates. Appetite regulation is complex and involves anorexigenic hormones such as glucagon-like peptide-1 (GLP-1) and amylin, and orexigenic peptides like ghrelin and all are well-known for their effects on feeding behaviors. This overview will summarize more recent physiological aspects of these peptides, demonstrating that they modulate various aspects of addiction processes. Findings from preclinical, genetic, and experimental clinical studies exploring the association between appetite-regulatory peptides and the acute or chronic effects of addictive drugs will be introduced. Short or long-acting GLP-1 receptor agonists independently attenuate the acute rewarding properties of addictive drugs or reduce the chronic aspects of drugs. Genetic variation of the GLP-1 system is associated with alcohol use disorder. Also, the amylin pathway modulates the acute and chronic behavioral responses to addictive drugs. Ghrelin has been shown to activate reward-related behaviors. Moreover, ghrelin enhances, whereas pharmacological or genetic suppression of the ghrelin receptor attenuates the responses to various addictive drugs. Genetic studies and experimental clinical studies further support the associations between ghrelin and addiction processes. Further studies should explore the mechanisms modulating the ability of appetite-regulatory peptides to reduce addiction, and the effects of combination therapies or different diets on substance use are warranted. In summary, these studies provide evidence that appetite-regulatory peptides modulate reward and addiction processes, and deserve to be investigated as potential treatment target for addiction.
Collapse
Affiliation(s)
- Olesya T Shevchouk
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Maximilian Tufvesson-Alm
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Elisabet Jerlhag
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
18
|
Sulima A, Akhlaghi F, Leggio L, Rice KC. Synthesis of PF-6870961, a major hydroxy metabolite of the novel ghrelin receptor inverse agonist PF-5190457. Bioorg Med Chem 2021; 50:116465. [PMID: 34662828 PMCID: PMC9837661 DOI: 10.1016/j.bmc.2021.116465] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 09/30/2021] [Accepted: 10/05/2021] [Indexed: 01/17/2023]
Abstract
Preclinical and human studies have indicated involvement of the ghrelin system in alcohol-related behaviors illuminating the possibility of using ghrelin receptor blockers as a pharmacological intervention for alcohol use disorder (AUD). Preliminary data from a recently conducted phase 1b human study with a ghrelin receptor inverse agonist, PF-5190457 (2-(2-methylimidazo[2,1-b][1,3thiazol-6-yl)-1-{2-(1R)-5-(6-methylpyrimidin-4-yl)-2,3-dihydro-1H-inden-1-yl]-2,7-diazaspiro[3.5]non-7-ylethanone), provided evidence on the safety and tolerability of this compound when co-administered with alcohol. Furthermore, the study revealed important information on the biotransformation pathways for this compound and prompted the discovery and then synthesis of a newly identified major metabolite, PF-6870961 ((R)-1-(2-(5-(2-hydroxy-6-methylpyrimidin-4-yl)-2,3-dihydro-1H-inden-1-yl)-2,7-diazaspiro[3.5]nonan-7-yl)-2-(2-methylimidazo[2,1-b]thiazol-6-yl)ethan-1-one). The metabolite was synthesized and fully characterized through a design that enabled it to be prepared in useful quantities. The synthesis provided direct access to the recently discovered PF-6870961 and is allowing researchers to conduct additional and deeper evaluation of its in vitro and in vivo properties.
Collapse
Affiliation(s)
- Agnieszka Sulima
- Medication Development Program, Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Department of Health and Human Services, 9800 Medical Center Drive, Bethesda, Maryland 20892-3373, United States; Medication Development Program, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Department of Health and Human Services, 251 Bayview Boulevard, Baltimore, Maryland 21224, United States.
| | - Fatemeh Akhlaghi
- Clinical Pharmacokinetics Research Laboratory, Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island Kingston, RI, United States
| | - Lorenzo Leggio
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Department of Health and Human Services, 251 Bayview Boulevard, Baltimore, Maryland 21224, United States; Medication Development Program, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Department of Health and Human Services, 251 Bayview Boulevard, Baltimore, Maryland 21224, United States
| | - Kenner C Rice
- Medication Development Program, Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Department of Health and Human Services, 9800 Medical Center Drive, Bethesda, Maryland 20892-3373, United States; Medication Development Program, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Department of Health and Human Services, 251 Bayview Boulevard, Baltimore, Maryland 21224, United States.
| |
Collapse
|
19
|
Novozhilova M, Mishchenko T, Kondakova E, Lavrova T, Gavrish M, Aferova S, Franceschi C, Vedunova M. Features of age-related response to sleep deprivation: in vivo experimental studies. Aging (Albany NY) 2021; 13:19108-19126. [PMID: 34320466 PMCID: PMC8386558 DOI: 10.18632/aging.203372] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 07/17/2021] [Indexed: 12/23/2022]
Abstract
Insomnia is currently considered one of the potential triggers of accelerated aging. The frequency of registered sleep-wake cycle complaints increases with age and correlates with the quality of life of elderly people. Nevertheless, whether insomnia is actually an age-associated process or whether it acts as an independent stress-factor that activates pathological processes, remains controversial. In this study, we analyzed the effects of long-term sleep deprivation modeling on the locomotor and orienting-exploratory activity, spatial learning abilities and working memory of C57BL/6 female mice of different ages. We also evaluated the modeled stress influence on morphological changes in brain tissue, the functional activity of the mitochondrial apparatus of nerve cells, and the level of DNA methylation and mRNA expression levels of the transcription factor HIF-1α (Hif1) and age-associated molecular marker PLIN2. Our findings point to the age-related adaptive capacity of female mice to the long-term sleep deprivation influence. For young (1.5 months) mice, the modeled sleep deprivation acts as a stress factor leading to weight loss against the background of increased food intake, the activation of animals' locomotor and exploratory activity, their mnestic functions, and molecular and cellular adaptive processes ensuring animal resistance both to stress and risk of accelerated aging development. Sleep deprivation in adult (7-9 months) mice is accompanied by an increase in body weight against the background of active food intake, increased locomotor and exploratory activity, gross disturbances in mnestic functions, and decreased adaptive capacity of brain cells, that potentially increasing the risk of pathological reactions and neurodegenerative processes.
Collapse
Affiliation(s)
- Maria Novozhilova
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603022, Russia
| | - Tatiana Mishchenko
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603022, Russia
| | - Elena Kondakova
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603022, Russia
| | - Tatiana Lavrova
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603022, Russia
| | - Maria Gavrish
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603022, Russia
| | - Svetlana Aferova
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603022, Russia
| | - Claudio Franceschi
- Institute of Information Technologies, Mathematics and Mechanics (ITMM), National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603022, Russia
| | - Maria Vedunova
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603022, Russia
| |
Collapse
|
20
|
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: 1.7] [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
|
21
|
Cobbina E, Lee MR, Leggio L, Akhlaghi F. A Population Pharmacokinetic Analysis of PF-5190457, a Novel Ghrelin Receptor Inverse Agonist in Healthy Volunteers and in Heavy Alcohol Drinkers. Clin Pharmacokinet 2020; 60:471-484. [PMID: 33155163 DOI: 10.1007/s40262-020-00942-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/29/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND OBJECTIVES The ghrelin receptor (GHS-R1a) is a potential target for alcohol use disorders. PF-5190457 is the first inverse agonist of GHS-R1a to progress to clinical development with potential to treat alcohol use disorder. We present a population pharmacokinetic model for PF-5190457 in non-heavy (alcohol consumption status = 0) and heavy alcohol drinkers (alcohol consumption status = 1), and identify relevant factors that can influence its pharmacokinetics. METHODS Plasma concentration-time data from non-heavy (n = 35) and heavy drinkers (n = 12) were pooled for the population pharmacokinetic model development. The influence of various covariates including alcohol consumption status was evaluated. The accuracy, precision, and robustness of the model were also evaluated using bootstrapping and visual predictive checks. RESULTS A two-compartment model best described the pharmacokinetics of PF-5190457. The apparent volume of distribution of 44.5 L, apparent clearance of 72.0 L/h, apparent peripheral volume of distribution of 271 L, apparent distributional clearance of 28.7 L/h, and first-order absorption rate constant of 0.27/h were accurate and precise. The apparent volume of distribution was 3.8-fold higher (169 L) in heavy drinkers, and correlated with a lower maximum plasma concentration in heavy drinkers compared with non-heavy drinkers at the same dose; and a corresponding reduced incidence of somnolence in heavy drinkers at doses > 50 mg. CONCLUSIONS This work provides an accurate, precise, and robust two-compartment model that describes the pharmacokinetics of PF-5190457 and suggests a possible link of PF-5190457 pharmacokinetics with somnolence. TRIAL REGISTRATION ClinicalTrials.gov identifier numbers NCT01247896 and NCT02039349.
Collapse
Affiliation(s)
- Enoch Cobbina
- Clinical Pharmacokinetics Research Laboratory, Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Office 495 A, Avedisian Hall, 7 Greenhouse Road, Kingston, RI, 02881, USA
| | - Mary R Lee
- 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
| | - 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.,Medication Development Program, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA.,Department of Behavioral and Social Sciences, Center for Alcohol and Addiction Studies, Brown University, Providence, RI, USA
| | - Fatemeh Akhlaghi
- Clinical Pharmacokinetics Research Laboratory, Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Office 495 A, Avedisian Hall, 7 Greenhouse Road, Kingston, RI, 02881, USA.
| |
Collapse
|
22
|
Tuero C, Valenti V, Rotellar F, Landecho MF, Cienfuegos JA, Frühbeck G. Revisiting the Ghrelin Changes Following Bariatric and Metabolic Surgery. Obes Surg 2020; 30:2763-2780. [PMID: 32323063 DOI: 10.1007/s11695-020-04601-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Since the description of ghrelin in 1999, several studies have dug into the effects of this hormone and its relationship with bariatric surgery. While some aspects are still unresolved, a clear connection between ghrelin and the changes after metabolic surgery have been established. Besides weight loss, a significant amelioration in obesity-related comorbidities following surgery has also been reported. These changes in patients occur in the early postoperative period, before the weight loss appears, so that amelioration may be mainly due to hormonal changes. The purpose of this review is to go through the current body of knowledge of ghrelin's physiology, as well as to update and clarify the changes that take place in ghrelin concentrations following bariatric/metabolic surgery together with their potential consolidation to outcomes.
Collapse
Affiliation(s)
- Carlota Tuero
- Obesity Area, Clínica Universidad de Navarra, Avenida Pio XII 36, 31008, Pamplona, Navarra, Spain.
- Department of Surgery, Clínica Universidad de Navarra, Pamplona, Spain.
| | - Victor Valenti
- Obesity Area, Clínica Universidad de Navarra, Avenida Pio XII 36, 31008, Pamplona, Navarra, Spain
- Department of Surgery, Clínica Universidad de Navarra, Pamplona, Spain
- CIBEROBN, Instituto de Salud Carlos III, Pamplona, Navarra, Spain
- Obesity and Adipobiology Group, IdiSNA, Pamplona, Spain
| | - Fernando Rotellar
- Obesity Area, Clínica Universidad de Navarra, Avenida Pio XII 36, 31008, Pamplona, Navarra, Spain
- Department of Surgery, Clínica Universidad de Navarra, Pamplona, Spain
- CIBEROBN, Instituto de Salud Carlos III, Pamplona, Navarra, Spain
- Obesity and Adipobiology Group, IdiSNA, Pamplona, Spain
| | - Manuel F Landecho
- Obesity Area, Clínica Universidad de Navarra, Avenida Pio XII 36, 31008, Pamplona, Navarra, Spain
- Department of Internal Medicine, General Health Check-up unit, Clínica Universidad de Navarra, Pamplona, Spain
| | - Javier A Cienfuegos
- Obesity Area, Clínica Universidad de Navarra, Avenida Pio XII 36, 31008, Pamplona, Navarra, Spain
- Department of Surgery, Clínica Universidad de Navarra, Pamplona, Spain
- CIBEROBN, Instituto de Salud Carlos III, Pamplona, Navarra, Spain
- Obesity and Adipobiology Group, IdiSNA, Pamplona, Spain
| | - Gema Frühbeck
- Obesity Area, Clínica Universidad de Navarra, Avenida Pio XII 36, 31008, Pamplona, Navarra, Spain.
- CIBEROBN, Instituto de Salud Carlos III, Pamplona, Navarra, Spain.
- Obesity and Adipobiology Group, IdiSNA, Pamplona, Spain.
- Department of Endocrinology and Nutrition, Clínica Universidad de Navarra, Pamplona, Spain.
| |
Collapse
|
23
|
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: 104] [Impact Index Per Article: 26.0] [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
|
24
|
Torz LJ, Osborne-Lawrence S, Rodriguez J, He Z, Cornejo MP, Mustafá ER, Jin C, Petersen N, Hedegaard MA, Nybo M, Damonte VM, Metzger NP, Mani BK, Williams KW, Raingo J, Perello M, Holst B, Zigman JM. Metabolic insights from a GHSR-A203E mutant mouse model. Mol Metab 2020; 39:101004. [PMID: 32339772 PMCID: PMC7242877 DOI: 10.1016/j.molmet.2020.101004] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 04/13/2020] [Accepted: 04/16/2020] [Indexed: 02/02/2023] Open
Abstract
Objective Binding of ghrelin to its receptor, growth hormone secretagogue receptor (GHSR), stimulates GH release, induces eating, and increases blood glucose. These processes may also be influenced by constitutive (ghrelin-independent) GHSR activity, as suggested by findings in short people with naturally occurring GHSR-A204E mutations and reduced food intake and blood glucose in rodents administered GHSR inverse agonists, both of which impair constitutive GHSR activity. In this study, we aimed to more fully determine the physiologic relevance of constitutive GHSR activity. Methods We generated mice with a GHSR mutation that replaces alanine at position 203 with glutamate (GHSR-A203E), which corresponds to the previously described human GHSR-A204E mutation, and used them to conduct ex vivo neuronal electrophysiology and in vivo metabolic assessments. We also measured signaling within COS-7 and HEK293T cells transfected with wild-type GHSR (GHSR-WT) or GHSR-A203E constructs. Results In COS-7 cells, GHSR-A203E resulted in lower baseline IP3 accumulation than GHSR-WT; ghrelin-induced IP3 accumulation was observed in both constructs. In HEK293T cells co-transfected with voltage-gated CaV2.2 calcium channel complex, GHSR-A203E had no effect on basal CaV2.2 current density while GHSR-WT did; both GHSR-A203E and GHSR-WT inhibited CaV2.2 current in the presence of ghrelin. In cultured hypothalamic neurons from GHSR-A203E and GHSR-deficient mice, native calcium currents were greater than those in neurons from wild-type mice; ghrelin inhibited calcium currents in cultured hypothalamic neurons from both GHSR-A203E and wild-type mice. In brain slices, resting membrane potentials of arcuate NPY neurons from GHSR-A203E mice were hyperpolarized compared to those from wild-type mice; the same percentage of arcuate NPY neurons from GHSR-A203E and wild-type mice depolarized upon ghrelin exposure. The GHSR-A203E mutation did not significantly affect body weight, body length, or femur length in the first ∼6 months of life, yet these parameters were lower in GHSR-A203E mice after 1 year of age. During a 7-d 60% caloric restriction regimen, GHSR-A203E mice lacked the usual marked rise in plasma GH and demonstrated an exaggerated drop in blood glucose. Administered ghrelin also exhibited reduced orexigenic and GH secretagogue efficacies in GHSR-A203E mice. Conclusions Our data suggest that the A203E mutation ablates constitutive GHSR activity and that constitutive GHSR activity contributes to the native depolarizing conductance of GHSR-expressing arcuate NPY neurons. Although the A203E mutation does not block ghrelin-evoked signaling as assessed using in vitro and ex vivo models, GHSR-A203E mice lack the usual acute food intake response to administered ghrelin in vivo. The GHSR-A203E mutation also blunts GH release, and in aged mice leads to reduced body length and femur length, which are consistent with the short stature of human carriers of the GHSR-A204E mutation. We generated mice with a GHSR mutation replacing Ala at position 203 with Glu. The A203E mutation ablates constitutive GHSR activity & hyperpolarizes NPY neurons. GHSR-A203E mice lack the usual orexigenic response to administered ghrelin. The GHSR-A203E mutation blunts GH release and causes reduced body length. This finding is consistent with short stature in human carriers of the GHSR-A204E mutation.
Collapse
Affiliation(s)
- Lola J Torz
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark; Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Sherri Osborne-Lawrence
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA
| | - Juan Rodriguez
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA
| | - Zhenyan He
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA
| | | | - Emilio Román Mustafá
- Laboratory of Electrophysiology of the Multidisciplinary Institute of Cell Biology [Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA)], La Plata, Buenos Aires, Argentina
| | - Chunyu Jin
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Natalia Petersen
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Morten A Hedegaard
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Maja Nybo
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Valentina Martínez Damonte
- Laboratory of Electrophysiology of the Multidisciplinary Institute of Cell Biology [Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA)], La Plata, Buenos Aires, Argentina
| | - Nathan P Metzger
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA
| | - Bharath K Mani
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA
| | - Kevin W Williams
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA
| | - Jesica Raingo
- Laboratory of Electrophysiology of the Multidisciplinary Institute of Cell Biology [Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA)], La Plata, Buenos Aires, Argentina
| | - Mario Perello
- Laboratory of Neurophysiology, La Plata, Buenos Aires, Argentina
| | - Birgitte Holst
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark; Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark.
| | - Jeffrey M Zigman
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
25
|
Lee MR, Tapocik JD, Ghareeb M, Schwandt ML, Dias AA, Le AN, Cobbina E, Farinelli LA, Bouhlal S, Farokhnia M, Heilig M, Akhlaghi F, Leggio L. The novel ghrelin receptor inverse agonist PF-5190457 administered with alcohol: preclinical safety experiments and a phase 1b human laboratory study. Mol Psychiatry 2020; 25:461-475. [PMID: 29728704 PMCID: PMC6215751 DOI: 10.1038/s41380-018-0064-y] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 02/21/2018] [Accepted: 02/22/2018] [Indexed: 12/15/2022]
Abstract
Rodent studies indicate that ghrelin receptor blockade reduces alcohol consumption. However, no ghrelin receptor blockers have been administered to heavy alcohol drinking individuals. Therefore, we evaluated the safety, tolerability, pharmacokinetic (PK), pharmacodynamic (PD) and behavioral effects of a novel ghrelin receptor inverse agonist, PF-5190457, when co-administered with alcohol. We tested the effects of PF-5190457 combined with alcohol on locomotor activity, loss-of-righting reflex (a measure of alcohol sedative actions), and on blood PF-5190457 concentrations in rats. Then, we performed a single-blind, placebo-controlled, within-subject human study with PF-5190457 (placebo/0 mg b.i.d., 50 mg b.i.d., 100 mg b.i.d.). Twelve heavy drinkers during three identical visits completed an alcohol administration session, subjective assessments, and an alcohol cue-reactivity procedure, and gave blood samples for PK/PD testing. In rats, PF-5190457 did not interact with the effects of alcohol on locomotor activity or loss-of-righting reflex. Alcohol did not affect blood PF-5190457 concentrations. In humans, all adverse events were mild or moderate and did not require discontinuation or dose reductions. Drug dose did not alter alcohol concentration or elimination, alcohol-induced stimulation or sedation, or mood during alcohol administration. Potential PD markers of PF-5190457 were acyl-to-total ghrelin ratio and insulin-like growth factor-1. PF-5190457 (100 mg b.i.d.) reduced alcohol craving during the cue-reactivity procedure. This study provides the first translational evidence of safety and tolerability of the ghrelin receptor inverse agonist PF-5190457 when co-administered with alcohol. PK/PD/behavioral findings support continued research of PF-5190457 as a potential pharmacological agent to treat alcohol use disorder.
Collapse
Affiliation(s)
- Mary R Lee
- Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology, National Institute on Alcohol Abuse and Alcoholism and National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD, USA
| | - Jenica D Tapocik
- Section on Molecular Pathophysiology, Laboratory of Clinical and Translational Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Mwlod Ghareeb
- Clinical Pharmacokinetics Research Laboratory, Department of Biomedical & Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, USA
| | - Melanie L Schwandt
- Office of the Clinical Director, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Alexandra A Dias
- Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology, National Institute on Alcohol Abuse and Alcoholism and National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD, USA
| | - April N Le
- Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology, National Institute on Alcohol Abuse and Alcoholism and National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD, USA
| | - Enoch Cobbina
- Clinical Pharmacokinetics Research Laboratory, Department of Biomedical & Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, USA
| | - Lisa A Farinelli
- Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology, National Institute on Alcohol Abuse and Alcoholism and National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD, USA
| | - Sofia Bouhlal
- Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology, National Institute on Alcohol Abuse and Alcoholism and National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD, USA
| | - Mehdi Farokhnia
- Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology, National Institute on Alcohol Abuse and Alcoholism and National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD, USA
| | - Markus Heilig
- Section on Molecular Pathophysiology, Laboratory of Clinical and Translational Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
- Center for Social and Affective Neuroscience, Linköping University, Linköping, Sweden
| | - Fatemeh Akhlaghi
- Clinical Pharmacokinetics Research Laboratory, Department of Biomedical & Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, USA
| | - Lorenzo Leggio
- Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology, National Institute on Alcohol Abuse and Alcoholism and National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD, USA.
- Department of Behavioral and Social Sciences, Center for Alcohol and Addiction Studies, Brown University, Providence, RI, USA.
| |
Collapse
|
26
|
Neurochemical Evidence of Preclinical and Clinical Reports on Target-Based Therapy in Alcohol Used Disorder. Neurochem Res 2020; 45:491-507. [PMID: 31898084 DOI: 10.1007/s11064-019-02944-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 12/13/2019] [Accepted: 12/20/2019] [Indexed: 10/25/2022]
Abstract
Alcohol use disorder (AUD) is a chronic relapsing disorder, which enforces a person to compulsively seek alcohol, restricting control over alcohol intake leads to emergence of an undesired emotional state during abstinence. There are recent advances for better understanding of neurocircuitry involved in the pathophysiology of AUD. Alcohol interaction with neuronal membrane proteins results in changes in neuronal circuits. It is also linked with the potential medication and their clinical validation concerning their pharmacological targets for alcoholic abstinence. This review covers research work from the past few decades on the therapeutic advances on treatment of alcohol dependence; further detailing the fundamental neurochemical mechanisms after alcohol administration. It also covers interaction of alcohol with GABAergic, glutaminergic, dopaminergic, serotonergic and opioid systems. This review further elaborated the neurobiology of noradrenergic, cholinergic and cannabinoid systems and their interaction with AUD. Elaborative information of potential drug targets under current exploration for AUD treatment with their mechanisms are reported here along with clinical outcomes and the associated side effects.
Collapse
|
27
|
Ghrelin Aggravates Prostate Enlargement in Rats with Testosterone-Induced Benign Prostatic Hyperplasia, Stromal Cell Proliferation, and Smooth Muscle Contraction in Human Prostate Tissues. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:4748312. [PMID: 31885795 PMCID: PMC6893282 DOI: 10.1155/2019/4748312] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 10/01/2019] [Accepted: 10/30/2019] [Indexed: 12/15/2022]
Abstract
Epidemiologic studies revealed a context between lower urinary tract symptoms (LUTS) suggestive of benign prostatic hyperplasia (BPH) and metabolic syndrome. However, molecular mechanisms underlying this relationship are largely unknown. Prostate enlargement and increased prostate smooth muscle tone are important factors in the pathophysiology of LUTS suggestive of BPH. In the present study, we studied effects of the metabolic hormone ghrelin on prostate enlargement in rats with experimentally induced BPH, growth of cultured stromal cells from human prostate (WPMY-1), and smooth muscle contraction of human prostate tissues. Ghrelin (20 nmol/kg daily, p.o., 2 weeks) increased prostate size in rats with testosterone-induced BPH. Microarray identified 114 ghrelin-upregulated genes (2-fold or more) in these prostates, with possible roles in growth, smooth muscle contraction, or metabolism. 12 genes were selected for further analyses. In human prostate tissues, mRNA levels of 11 of them correlated positively with ghrelin receptor (GHSR) expression, but only two with the degree of BPH. Accordingly, no correlation was evident between GHSR expression level and BPH in human prostate tissues. In WPMY-1 cells, the GHRS agonist MK0677 upregulated 11 of the selected genes. MK0677 induced proliferation of WPMY-1 cells, shown by EdU assay, colony formation, proliferation markers, flow cytometry, and viability. In myographic measurements, GHSR agonists enhanced contractions of human prostate strips. Together, ghrelin may aggravate prostate enlargement, stromal cell growth, and prostate smooth muscle contraction in BPH. Ghrelin may deteriorate urethral obstruction independently from BPH, qualifying the ghrelin system as an attractive new target to be tested for LUTS treatment in BPH.
Collapse
|
28
|
Lee MR, Farokhnia M, Cobbina E, Saravanakumar A, Li X, Battista JT, Farinelli LA, Akhlaghi F, Leggio L. Endocrine effects of the novel ghrelin receptor inverse agonist PF-5190457: Results from a placebo-controlled human laboratory alcohol co-administration study in heavy drinkers. Neuropharmacology 2019; 170:107788. [PMID: 31557492 DOI: 10.1016/j.neuropharm.2019.107788] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 08/30/2019] [Accepted: 09/19/2019] [Indexed: 11/30/2022]
Abstract
Both animal and human work suggests that the ghrelin system may be involved in the mechanisms that regulate the development and maintenance of alcohol use disorder. Previously, in a Phase 1b study, we tested pharmacological blockade of the growth hormone secretagogue receptor 1a (GHS-R1a, also known as the ghrelin receptor), in heavy drinking individuals with PF-5190457, an orally bioavailable, potent and selective GHS-R1a inverse agonist. We report here the effects of PF-5190457 on endocrine blood concentrations of amylin, gastric inhibitory polypeptide, glucagon-like peptide 1, insulin, leptin, pancreatic polypeptide, peptide YY, thyroid stimulating hormone, free triiodothyronine (T3), thyroxine (T4), cortisol, prolactin, and glucose during PF-5190457 dosing, as compared to placebo, in absence of alcohol as well as during an alcohol challenge when PF-5190457 was on steady-state. Blood hormone levels were largely unaffected by PF-5190457, both during dosing and in the context of alcohol challenge. The safety-related relevance of these findings to further develop PF-5190547 in alcohol use disorder is discussed. CLINICALTRIALS.GOV: NCT02039349. This article is part of the special issue on 'Neuropeptides'.
Collapse
Affiliation(s)
- Mary R Lee
- 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
| | - Mehdi Farokhnia
- 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 on Compulsive Behaviors, National Institutes of Health, Bethesda, MD, USA
| | - Enoch Cobbina
- Clinical Pharmacokinetics Research Laboratory, Department of Biomedical & Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, USA
| | - Anitha Saravanakumar
- Clinical Pharmacokinetics Research Laboratory, Department of Biomedical & Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, USA
| | - Xiaobai Li
- Biostatistics and Clinical Epidemiology Service, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Jillian T Battista
- 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
| | - Lisa A Farinelli
- 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
| | - Fatemeh Akhlaghi
- Clinical Pharmacokinetics Research Laboratory, Department of Biomedical & Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, 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 on Compulsive Behaviors, National Institutes of Health, Bethesda, MD, USA; Medication Development Program, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA; Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, Brown University, Providence, RI, USA.
| |
Collapse
|
29
|
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
|
30
|
Vestlund J, Bergquist F, Eckernäs D, Licheri V, Adermark L, Jerlhag E. Ghrelin signalling within the rat nucleus accumbens and skilled reach foraging. Psychoneuroendocrinology 2019; 106:183-194. [PMID: 30999229 DOI: 10.1016/j.psyneuen.2019.04.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 03/12/2019] [Accepted: 04/06/2019] [Indexed: 01/23/2023]
Abstract
Motivation alters behaviour in a complex manner and nucleus accumbens (NAc) shell has been implied as a key structure regulating such behaviour. Recent studies show that acute ghrelin signalling enhances motivation when assessed in a simple motor task. The aim of the present study was to define the role of ghrelin signalling on motivation in a more complex motor behaviour. Rats were tested in the Montoya staircase, an animal model of skilled reach foraging assessed by the number of sucrose pellets consumed. Electrophysiological recordings were conducted to explore the neurophysiological correlates of ghrelin signalling. The initial electrophysiological results displayed that ex vivo administration of ghrelin increased NAc shell output in brain slices from drug- and training-naïve rats. In rats with an acquired skilled reach performance, acute as well as repeated treatment with a ghrelin receptor (GHSR-1 A) antagonist (JMV2959) decreased the number of sucrose pellets consumed. Moreover, infusion of JMV2959 into NAc shell reduced this consumption. Sub-chronic, during ten days, JMV2959 treatment during training on the Montoya staircase reduced the number of pellets consumed, whereas ghrelin improved this behaviour. In addition, field potential and whole cell recordings were conducted in NAc shell of rats that had been treated with ghrelin or GHSR-1 A antagonist during training on the Montoya staircase. Sub-chronic administration of ghrelin during motor-skill learning selectively increased the frequency of inhibitory transmission in the NAc shell, resulting in a net suppression of accumbal output. Collectively these data suggest that ghrelin signalling in NAc shell enhances skilled reached foraging tentatively by increasing the motivation.
Collapse
Affiliation(s)
- Jesper Vestlund
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Filip Bergquist
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Daniel Eckernäs
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Valentina Licheri
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Louise Adermark
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Elisabet Jerlhag
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
31
|
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: 1.2] [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
|
32
|
Adusumalli S, Jamwal R, Obach RS, Ryder TF, Leggio L, Akhlaghi F. Role of Molybdenum-Containing Enzymes in the Biotransformation of the Novel Ghrelin Receptor Inverse Agonist PF-5190457: A Reverse Translational Bed-to-Bench Approach. Drug Metab Dispos 2019; 47:874-882. [PMID: 31182423 DOI: 10.1124/dmd.119.087015] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Accepted: 05/28/2019] [Indexed: 12/29/2022] Open
Abstract
(R)-2-(2-methylimidazo[2,1-b]thiazol-6-yl)-1-(2-(5-(6-methylpyrimidin-4-yl)-2,3-dihydro-1H-inden-1-yl)-2,7-diazaspiro[3.5]nonan-7-yl)ethan-1-one (PF-5190457) was identified as a potent and selective inverse agonist of the ghrelin receptor [growth hormone secretagogue receptor 1a (GHS-R1a)]. The present translational bed-to-bench work characterizes the biotransformation of this compound in vivo and then further explores in vitro metabolism in fractions of human liver and primary hepatocytes. Following oral administration of PF-5190457 in a phase 1b clinical study, hydroxyl metabolites of the compound were observed, including one that had not been observed in previously performed human liver microsomal incubations. PF-6870961 was biosynthesized using liver cytosol, and the site of hydroxylation was shown to be on the pyrimidine using nuclear magnetic resonance spectroscopy. The aldehyde oxidase (AO) inhibitor raloxifene and the xanthine oxidase inhibitor febuxostat inhibited the formation of PF-6870961 in human liver cytosol, suggesting both enzymes were involved in the metabolism of the drug. However, greater inhibition was observed with raloxifene, indicating AO is a dominant enzyme in the biotransformation. The intrinsic clearance of the drug in human liver cytosol was estimated to be 0.002 ml/min per milligram protein. This study provides important novel information at three levels: 1) it provides additional new information on the recently developed novel compound PF-5190457, the first GHS-R1a blocker that has moved to development in humans; 2) it provides an example of a reverse translational approach where a discovery in humans was brought back, validated, and further investigated at the bench level; and 3) it demonstrates the importance of considering the molybdenum-containing oxidases during the development of new drug entities. SIGNIFICANCE STATEMENT: PF-5190457 is a novel ghrelin receptor inverse agonist that is currently undergoing clinical development for treatment of alcohol use disorder. PF-6870961, a major hydroxyl metabolite of the compound, was observed in human plasma, but was absent in human liver microsomal incubations. PF-6870961 was biosynthesized using liver cytosol, and the site of hydroxylation on the pyrimidine ring was characterized. Inhibitors of aldehyde oxidase and xanthine oxidase inhibited the formation of PF-6870961 in human liver cytosol, suggesting both enzymes were involved in the metabolism of the drug. This information is important for patient selection in subsequent clinical studies.
Collapse
Affiliation(s)
- Sravani Adusumalli
- Clinical Pharmacokinetics Research Laboratory, Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island (S.A., R.J., F.A.); Department of Pharmacokinetics, Dynamics, and Metabolism, Pfizer, Inc., Groton, Connecticut (R.S.O., T.F.R.); 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, Bethesda, Maryland (L.L.); Medication Development Program, National Institute on Drug Abuse Intramural Research Program, Baltimore, Maryland (L.L.); and Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, Brown University, Providence, Rhode Island (L.L.)
| | - Rohitash Jamwal
- Clinical Pharmacokinetics Research Laboratory, Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island (S.A., R.J., F.A.); Department of Pharmacokinetics, Dynamics, and Metabolism, Pfizer, Inc., Groton, Connecticut (R.S.O., T.F.R.); 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, Bethesda, Maryland (L.L.); Medication Development Program, National Institute on Drug Abuse Intramural Research Program, Baltimore, Maryland (L.L.); and Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, Brown University, Providence, Rhode Island (L.L.)
| | - R Scott Obach
- Clinical Pharmacokinetics Research Laboratory, Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island (S.A., R.J., F.A.); Department of Pharmacokinetics, Dynamics, and Metabolism, Pfizer, Inc., Groton, Connecticut (R.S.O., T.F.R.); 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, Bethesda, Maryland (L.L.); Medication Development Program, National Institute on Drug Abuse Intramural Research Program, Baltimore, Maryland (L.L.); and Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, Brown University, Providence, Rhode Island (L.L.)
| | - Tim F Ryder
- Clinical Pharmacokinetics Research Laboratory, Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island (S.A., R.J., F.A.); Department of Pharmacokinetics, Dynamics, and Metabolism, Pfizer, Inc., Groton, Connecticut (R.S.O., T.F.R.); 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, Bethesda, Maryland (L.L.); Medication Development Program, National Institute on Drug Abuse Intramural Research Program, Baltimore, Maryland (L.L.); and Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, Brown University, Providence, Rhode Island (L.L.)
| | - Lorenzo Leggio
- Clinical Pharmacokinetics Research Laboratory, Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island (S.A., R.J., F.A.); Department of Pharmacokinetics, Dynamics, and Metabolism, Pfizer, Inc., Groton, Connecticut (R.S.O., T.F.R.); 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, Bethesda, Maryland (L.L.); Medication Development Program, National Institute on Drug Abuse Intramural Research Program, Baltimore, Maryland (L.L.); and Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, Brown University, Providence, Rhode Island (L.L.)
| | - Fatemeh Akhlaghi
- Clinical Pharmacokinetics Research Laboratory, Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island (S.A., R.J., F.A.); Department of Pharmacokinetics, Dynamics, and Metabolism, Pfizer, Inc., Groton, Connecticut (R.S.O., T.F.R.); 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, Bethesda, Maryland (L.L.); Medication Development Program, National Institute on Drug Abuse Intramural Research Program, Baltimore, Maryland (L.L.); and Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, Brown University, Providence, Rhode Island (L.L.)
| |
Collapse
|
33
|
Farokhnia M, Faulkner ML, Piacentino D, Lee MR, Leggio L. Ghrelin: From a gut hormone to a potential therapeutic target for alcohol use disorder. Physiol Behav 2019; 204:49-57. [DOI: 10.1016/j.physbeh.2019.02.008] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 01/24/2019] [Accepted: 02/06/2019] [Indexed: 12/22/2022]
|
34
|
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: 56] [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
|
35
|
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: 3.4] [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
|
36
|
Daina A, Giuliano C, Pietra C, Wang J, Chi Y, Zou Z, Li F, Yan Z, Zhou Y, Guainazzi A, Garcia Rubio S, Zoete V. Rational Design, Synthesis, and Pharmacological Characterization of Novel Ghrelin Receptor Inverse Agonists as Potential Treatment against Obesity-Related Metabolic Diseases. J Med Chem 2018; 61:11039-11060. [DOI: 10.1021/acs.jmedchem.8b00794] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Affiliation(s)
- Antoine Daina
- Molecular Modeling Group, SIB Swiss Institute of Bioinformatics, CH-1015 Lausanne, Switzerland
| | - Claudio Giuliano
- Research and Preclinical Development Department, Helsinn Healthcare, CH-6912 Lugano, Switzerland
| | - Claudio Pietra
- Research and Preclinical Development Department, Helsinn Healthcare, CH-6912 Lugano, Switzerland
| | - Junbo Wang
- Department of Medicinal Chemistry, Pharmacokinetics and Metabolism, Sundia MediTech, 388 Jialilue Road, Zhangjiang Hi-Tech Park, Shanghai 201203, China
| | - Yushi Chi
- Department of Medicinal Chemistry, Pharmacokinetics and Metabolism, Sundia MediTech, 388 Jialilue Road, Zhangjiang Hi-Tech Park, Shanghai 201203, China
| | - Zack Zou
- Department of Medicinal Chemistry, Pharmacokinetics and Metabolism, Sundia MediTech, 388 Jialilue Road, Zhangjiang Hi-Tech Park, Shanghai 201203, China
| | - Fugang Li
- Department of Discovery Biology, HD Biosciences, 590 Ruiqing Road Zhangjiang East Campus, Shanghai 201201, China
| | - Zhonghua Yan
- Department of Discovery Biology, HD Biosciences, 590 Ruiqing Road Zhangjiang East Campus, Shanghai 201201, China
| | - Yifan Zhou
- Department of Discovery Biology, HD Biosciences, 590 Ruiqing Road Zhangjiang East Campus, Shanghai 201201, China
| | - Angelo Guainazzi
- Research and Development Department, Helsinn Therapeutics (US), Inc., Iselin, New Jersey 08830, United-States
| | - Silvina Garcia Rubio
- Research and Development Department, Helsinn Therapeutics (US), Inc., Iselin, New Jersey 08830, United-States
| | - Vincent Zoete
- Molecular Modeling Group, SIB Swiss Institute of Bioinformatics, CH-1015 Lausanne, Switzerland
- Department of Fundamental Oncology, Lausanne University, Ludwig Institute for Cancer Research, Route de la Corniche 9A, 1066 Epalinges, Switzerland
| |
Collapse
|
37
|
Ghrelin, a gastrointestinal hormone, regulates energy balance and lipid metabolism. Biosci Rep 2018; 38:BSR20181061. [PMID: 30177523 PMCID: PMC6153372 DOI: 10.1042/bsr20181061] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 08/13/2018] [Accepted: 08/28/2018] [Indexed: 01/06/2023] Open
Abstract
Ghrelin, an acylated peptide hormone of 28 amino acids, is an endogenous ligand of the released growth hormone secretagogue receptor (GHSR). Ghrelin has been isolated from human and rat stomach and is also detected in the hypothalamic arcuate nucleus. Ghrelin receptor is primarily located in the neuropeptide Y and agouti-related protein neurons. Many previous studies have shown that ghrelin and GHSR are involved in the regulation of energy homeostasis, and its administration can increase food intake and body weight gain. AMP-activated protein kinase is activated by ghrelin in the hypothalamus, which contributes to lower intracellular long-chain fatty acid level. Ghrelin appears to modulate the response to food cues via a neural network involved in the regulation of feeding and in the appetitive response to food cues. It also increases the response of brain areas involved in visual processing, attention, and memory to food pictures. Ghrelin is also an important factor linking the central nervous system with peripheral tissues that regulate lipid metabolism. It promotes adiposity by the activation of hypothalamic orexigenic neurons and stimulates the expression of fat storage-related proteins in adipocytes. Meanwhile, ghrelin exerts direct peripheral effects on lipid metabolism, including increase in white adipose tissue mass, stimulation of lipogenesis in the liver, and taste sensitivity modulation.
Collapse
|
38
|
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: 3.0] [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
|
39
|
Fernandez G, Cabral A, Andreoli MF, Labarthe A, M'Kadmi C, Ramos JG, Marie J, Fehrentz JA, Epelbaum J, Tolle V, Perello M. Evidence Supporting a Role for Constitutive Ghrelin Receptor Signaling in Fasting-Induced Hyperphagia in Male Mice. Endocrinology 2018; 159:1021-1034. [PMID: 29300858 DOI: 10.1210/en.2017-03101] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 12/21/2017] [Indexed: 01/22/2023]
Abstract
Ghrelin is a potent orexigenic peptide hormone that acts through the growth hormone secretagogue receptor (GHSR), a G protein-coupled receptor highly expressed in the hypothalamus. In vitro studies have shown that GHSR displays a high constitutive activity, whose physiological relevance is uncertain. As GHSR gene expression in the hypothalamus is known to increase in fasting conditions, we tested the hypothesis that constitutive GHSR activity at the hypothalamic level drives the fasting-induced hyperphagia. We found that refed wild-type (WT) mice displayed a robust hyperphagia that continued for 5 days after refeeding and changed their food intake daily pattern. Fasted WT mice showed an increase in plasma ghrelin levels, as well as in GHSR expression levels and ghrelin binding sites in the hypothalamic arcuate nucleus. When fasting-refeeding responses were evaluated in ghrelin- or GHSR-deficient mice, only the latter displayed an ∼15% smaller hyperphagia, compared with WT mice. Finally, fasting-induced hyperphagia of WT mice was significantly smaller in mice centrally treated with the GHSR inverse agonist K-(D-1-Nal)-FwLL-NH2, compared with mice treated with vehicle, whereas it was unaffected in mice centrally treated with the GHSR antagonists D-Lys3-growth hormone-releasing peptide 6 or JMV2959. Taken together, genetic models and pharmacological results support the notion that constitutive GHSR activity modulates the magnitude of the compensatory hyperphagia triggered by fasting. Thus, the hypothalamic GHSR signaling system could affect the set point of daily food intake, independently of plasma ghrelin levels, in situations of negative energy balance.
Collapse
Affiliation(s)
- Gimena Fernandez
- Laboratory of Neurophysiology of the Multidisciplinary Institute of Cell Biology (Argentine Research Council, Scientific Research Commission of the Province of Buenos Aires and National University of La Plata), La Plata, Buenos Aires, Argentina
| | - Agustina Cabral
- Laboratory of Neurophysiology of the Multidisciplinary Institute of Cell Biology (Argentine Research Council, Scientific Research Commission of the Province of Buenos Aires and National University of La Plata), La Plata, Buenos Aires, Argentina
| | - María F Andreoli
- School of Biochemistry and Biological Sciences, National University of Litoral and Institute of Environmental Health, Santa Fe, Argentina
| | - Alexandra Labarthe
- Centre de Psychiatrie et Neurosciences Unité Mixte de Recherche Scientifique_S894 INSERM Université Paris Descartes, Sorbonne Paris-Cité, Paris, France
| | - Céline M'Kadmi
- Institut des Biomolécules Max Mousseron, Unité Mixte de Recherche Scientifique 5247 Centre National de la Recherche Scientifique-Université Montpellier-École Nationale Supérieure de Chimie de Montpellier, Faculté de Pharmacie, Montpellier, France
| | - Jorge G Ramos
- School of Biochemistry and Biological Sciences, National University of Litoral and Institute of Environmental Health, Santa Fe, Argentina
| | - Jacky Marie
- Institut des Biomolécules Max Mousseron, Unité Mixte de Recherche Scientifique 5247 Centre National de la Recherche Scientifique-Université Montpellier-École Nationale Supérieure de Chimie de Montpellier, Faculté de Pharmacie, Montpellier, France
| | - Jean-Alain Fehrentz
- Institut des Biomolécules Max Mousseron, Unité Mixte de Recherche Scientifique 5247 Centre National de la Recherche Scientifique-Université Montpellier-École Nationale Supérieure de Chimie de Montpellier, Faculté de Pharmacie, Montpellier, France
| | - Jacques Epelbaum
- Centre de Psychiatrie et Neurosciences Unité Mixte de Recherche Scientifique_S894 INSERM Université Paris Descartes, Sorbonne Paris-Cité, Paris, France
- Mécanismes Adaptatifs et Evolution, Unité Mixte de Recherche Scientifique 7179 Centre National de la Recherche Scientifique, Muséum National d'Histoire Naturelle France, Brunoy, France
| | - Virginie Tolle
- Centre de Psychiatrie et Neurosciences Unité Mixte de Recherche Scientifique_S894 INSERM Université Paris Descartes, Sorbonne Paris-Cité, Paris, France
| | - Mario Perello
- Laboratory of Neurophysiology of the Multidisciplinary Institute of Cell Biology (Argentine Research Council, Scientific Research Commission of the Province of Buenos Aires and National University of La Plata), La Plata, Buenos Aires, Argentina
| |
Collapse
|
40
|
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: 99] [Impact Index Per Article: 16.5] [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
|
41
|
AZP-531, an unacylated ghrelin analog, improves food-related behavior in patients with Prader-Willi syndrome: A randomized placebo-controlled trial. PLoS One 2018; 13:e0190849. [PMID: 29320575 PMCID: PMC5761957 DOI: 10.1371/journal.pone.0190849] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 12/18/2017] [Indexed: 11/19/2022] Open
Abstract
Context and objective Prader-Willi syndrome (PWS) is characterized by early-onset hyperphagia and increased circulating levels of the orexigenic Acylated Ghrelin (AG) hormone with a relative deficit of Unacylated Ghrelin (UAG). AZP-531, a first-in-class UAG analog, was shown to inhibit the orexigenic effect of AG in animals, to improve glycemic control and decrease body weight in humans. We aimed to investigate the safety and efficacy of AZP-531 in patients with PWS for whom no approved treatment for hyperphagia is currently available. Methods and design Multi-center, randomized, double-blind, placebo-controlled trial. Forty-seven patients with genetically confirmed PWS and evidence of hyperphagia received daily subcutaneous injections of AZP-531 (3 and 4 mg for 50–70 kg and >70 kg body weight, respectively) or matching placebo for 14 days. Assessments included adverse events, vital signs, safety laboratory tests, the Hyperphagia Questionnaire (HQ), patient-reported appetite, body composition and glycemic measures. Results AZP-531 was well tolerated. There was a significant improvement with AZP-531 versus placebo in the mean total score, the 9-item score and the severity domain score of the HQ (p < .05). The highest reduction in the total and 9-item scores was observed in AZP-531 subjects with the highest hyperphagia score at baseline. Findings were supported by a reduction in appetite scores observed with AZP-531 only. Body weight did not change in both groups while a significant reduction in waist circumference and fat mass was observed only with AZP-531. AZP-531 significantly decreased post-prandial glucose levels in a baseline glucose dependent fashion. Conclusions AZP-531 may constitute a new treatment strategy to improve hyperphagia and metabolic issues in patients with PWS. These findings support further investigation in longer-term clinical trials.
Collapse
|
42
|
Denney WS, Sonnenberg GE, Carvajal-Gonzalez S, Tuthill T, Jackson VM. Pharmacokinetics and pharmacodynamics of PF-05190457: The first oral ghrelin receptor inverse agonist to be profiled in healthy subjects. Br J Clin Pharmacol 2016; 83:326-338. [PMID: 27621150 DOI: 10.1111/bcp.13127] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 08/25/2016] [Accepted: 09/08/2016] [Indexed: 02/06/2023] Open
Abstract
AIM To evaluate safety, tolerability and pharmacokinetics of oral PF-05190457, an oral ghrelin receptor inverse agonist, in healthy adults. METHODS Single (SAD) and multiple ascending dose (MAD) studies were randomised, placebo-controlled, double-blind studies. Thirty-five healthy men (age 38.2 ± 10.4 years; body mass index 24.8 ± 3.1 kg m-2 [mean ± standard deviation]) received ≥1 dose (2, 10, 40 [divided], 50, 100, 150, and 300 [single or divided] mg) of PF-05190457 and/or placebo in the SAD. In the MAD study, 35 healthy men (age 39.7 ± 10.1 years; body mass index 25.9 ± 3.3 kg m-2 ) received ≥1 dose (2, 10, 40 and 100 mg twice daily) of PF-05190457 and/or placebo daily for 2 weeks. RESULTS PF-05190457 absorption was rapid with a Tmax of 0.5-3 hours and a half-life between 8.2-9.8 hours. PF-05190457 dose-dependently blocked ghrelin (1 pmol kg-1 min-1 )-induced growth hormone (GH) release with (mean [90% confidence interval]) 77% [63-85%] inhibition at 100 mg. PF-05190457 (150 mg) delayed gastric emptying lag time by 30% [7-58%] and half emptying time by 20% [7-35%] with a corresponding decrease in postprandial glucose by 9 mg dL-1 . The most frequent adverse event reported by 30 subjects at doses ≥50 mg was somnolence. PF-05190457 plasma concentrations also increased heart rate up to 13.4 [4.8-58.2] beats min-1 and, similar to the effect on glucose and ghrelin-induced GH, was lost within 2 weeks. CONCLUSIONS PF-05190457 is a well-tolerated first-in-class ghrelin receptor inverse agonist with acceptable pharmacokinetics for oral daily dosing. Blocking ghrelin receptors inhibits ghrelin-induced GH, and increases heart rate, effects that underwent tachyphylaxis with chronic dosing. PF-051940457 has the potential to treat centrally-acting disorders such as insomnia.
Collapse
Affiliation(s)
- William S Denney
- Biotherapeutics Clinical Pharmacology, Pfizer Worldwide Research and Development, Cambridge, Massachusetts, 02139, USA
| | - Gabriele E Sonnenberg
- Cardiovascular, Metabolic, and Endocrine Diseases Research Unit, Pfizer Worldwide Research and Development, Cambridge, Massachusetts, 02139, USA
| | - Santos Carvajal-Gonzalez
- Cardiovascular, Metabolic, and Endocrine Diseases Research Unit, Pfizer Worldwide Research and Development, Cambridge, Massachusetts, 02139, USA
| | - Theresa Tuthill
- Cardiovascular, Metabolic, and Endocrine Diseases Research Unit, Pfizer Worldwide Research and Development, Cambridge, Massachusetts, 02139, USA
| | - V Margaret Jackson
- Cardiovascular, Metabolic, and Endocrine Diseases Research Unit, Pfizer Worldwide Research and Development, Cambridge, Massachusetts, 02139, USA
| |
Collapse
|