1
|
Peiu SN, Zugun-Eloae F, Stoica B, Anisie E, Iosep DG, Danciu M, Silivestru-Crețu I, Akad F, Avadanei AN, Condur L, Popa RF, Mocanu V. Obesity-Induced PVAT Dysfunction and Atherosclerosis Development: The Role of GHSR-1a in Increased Macrophage Infiltration and Adipocytokine Secretion. J Cardiovasc Dev Dis 2025; 12:87. [PMID: 40137085 PMCID: PMC11942683 DOI: 10.3390/jcdd12030087] [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: 11/28/2024] [Revised: 02/03/2025] [Accepted: 02/21/2025] [Indexed: 03/27/2025] Open
Abstract
In obesity, recent research revealed that increased expression of the growth hormone secretagogue receptor (GHSR) in macrophages plays a pivotal role in the development of meta-inflammation, promoting macrophage infiltration and pro-inflammatory polarization. This study aimed to examine the association between GHSR-1a expression in atherosclerotic plaques and adjacent perivascular adipose tissue (PVAT) from 11 patients with obesity and peripheral artery disease (PAD) who underwent revascularization procedures. Immunohistochemistry was used to assess the expression of CD68, CD80, and CD14, while tissue homogenate levels of adiponectin, leptin, IL-6, and CRP were quantified via ELISA. Serum markers of inflammation were also measured. Among patients with GHSR-1a-positive (+) macrophages in atherosclerotic plaques, we observed significantly higher white blood cell counts and platelet-to-lymphocyte ratios in serum, a lower adiponectin-to-leptin ratio, and elevated IL-6 levels in both arterial and PVAT homogenates. Our findings suggest a link between GHSR-1a and macrophage/monocyte infiltration, macrophage polarization, and adipocytokine secretion in atherosclerotic plaques associated with obesity-induced PVAT dysfunction.
Collapse
Affiliation(s)
- Sorin Nicolae Peiu
- Department of Vascular Surgery, “Grigore T. Popa” University of Medicine and Pharmacy, 16, Universitatii Street, 700115 Iasi, Romania; (S.N.P.); (A.N.A.); (R.F.P.)
- Department of Morpho-Functional Sciences II (Pathophysiology), “Grigore T. Popa” University of Medicine and Pharmacy, 16, Universitatii Street, 700115 Iasi, Romania; (I.S.-C.); (F.A.)
| | - Florin Zugun-Eloae
- Department of Morpho-Functional Sciences I (Immunology), “Grigore T. Popa” University of Medicine and Pharmacy, 16, Universitatii Street, 700115 Iasi, Romania
- Regional Institute of Oncology, TRANSCEND Research Centre, 2-4, General Mathias Berthelot, 700483 Iasi, Romania;
| | - Bogdan Stoica
- Department of Morpho-Functional Sciences II (Biochemistry), “Grigore T. Popa” University of Medicine and Pharmacy, 16, Universitatii Street, 700115 Iasi, Romania;
| | - Ecaterina Anisie
- Regional Institute of Oncology, TRANSCEND Research Centre, 2-4, General Mathias Berthelot, 700483 Iasi, Romania;
| | - Diana Gabriela Iosep
- Department of Morpho-Functional Sciences I (Morphopathology), “Grigore T. Popa” University of Medicine and Pharmacy, 16, Universitatii Street, 700115 Iasi, Romania; (D.G.I.); (M.D.)
| | - Mihai Danciu
- Department of Morpho-Functional Sciences I (Morphopathology), “Grigore T. Popa” University of Medicine and Pharmacy, 16, Universitatii Street, 700115 Iasi, Romania; (D.G.I.); (M.D.)
| | - Iustina Silivestru-Crețu
- Department of Morpho-Functional Sciences II (Pathophysiology), “Grigore T. Popa” University of Medicine and Pharmacy, 16, Universitatii Street, 700115 Iasi, Romania; (I.S.-C.); (F.A.)
| | - Fawzy Akad
- Department of Morpho-Functional Sciences II (Pathophysiology), “Grigore T. Popa” University of Medicine and Pharmacy, 16, Universitatii Street, 700115 Iasi, Romania; (I.S.-C.); (F.A.)
- Department of Morpho-Functional Sciences I (Anatomy), “Grigore T. Popa” University of Medicine and Pharmacy, 16, Universitatii Street, 700115 Iasi, Romania
| | - Andrei Nicolae Avadanei
- Department of Vascular Surgery, “Grigore T. Popa” University of Medicine and Pharmacy, 16, Universitatii Street, 700115 Iasi, Romania; (S.N.P.); (A.N.A.); (R.F.P.)
| | - Laura Condur
- Department of Family Medicine, Faculty of Medicine, Ovidius University, 124, Bd. Mamaia, 900527 Constanta, Romania;
| | - Radu Florin Popa
- Department of Vascular Surgery, “Grigore T. Popa” University of Medicine and Pharmacy, 16, Universitatii Street, 700115 Iasi, Romania; (S.N.P.); (A.N.A.); (R.F.P.)
| | - Veronica Mocanu
- Department of Morpho-Functional Sciences II (Pathophysiology), “Grigore T. Popa” University of Medicine and Pharmacy, 16, Universitatii Street, 700115 Iasi, Romania; (I.S.-C.); (F.A.)
| |
Collapse
|
2
|
Deis T, Goetze JP, Kistorp C, Gustafsson F. Gut Hormones in Heart Failure. Circ Heart Fail 2024; 17:e011813. [PMID: 39498569 DOI: 10.1161/circheartfailure.124.011813] [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: 03/18/2024] [Accepted: 10/07/2024] [Indexed: 11/21/2024]
Abstract
Heart failure (HF) is a syndrome affecting all organ systems. While some organ interactions have been studied intensively in HF (such as the cardiorenal interaction), the endocrine gut has to some degree been overlooked. However, there is growing evidence of direct cardiac effects of several hormones secreted from the gastrointestinal tract. For instance, GLP-1 (glucagon-like peptide-1), an incretin hormone secreted from the distal intestine following food intake, has notable effects on the heart, impacting heart rate and contractility. GLP-1 may even possess cardioprotective abilities, such as inhibition of myocardial ischemia and cardiac remodeling. While other gut hormones have been less studied, there is evidence suggesting cardiostimulatory properties of several hormones. Moreover, it has been reported that patients with HF have altered bioavailability of numerous gastrointestinal hormones, which may have prognostic implications. This might indicate an important role of gut hormones in cardiac physiology and pathology, which may be of particular importance in the failing heart. We present an overview of the current knowledge on gut hormones in HF, focusing on HF with reduced ejection fraction, and discuss how these hormones may be regulators of cardiac function and central hemodynamics. Potential therapeutic perspectives are discussed, and knowledge gaps are highlighted herein.
Collapse
Affiliation(s)
- Tania Deis
- Department of Cardiology (T.D., F.G.), Rigshospitalet, Copenhagen, Denmark
| | - Jens P Goetze
- Department of Clinical Biochemistry (J.P.G.), Rigshospitalet, Copenhagen, Denmark
- Department of Biomedical Sciences (J.P.G.), University of Copenhagen, Denmark
| | - Caroline Kistorp
- Department of Endocrinology (C.K.), Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine (C.K., F.G.), University of Copenhagen, Denmark
| | - Finn Gustafsson
- Department of Cardiology (T.D., F.G.), Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine (C.K., F.G.), University of Copenhagen, Denmark
| |
Collapse
|
3
|
van Vugt M, Finan C, Chopade S, Providencia R, Bezzina CR, Asselbergs FW, van Setten J, Schmidt AF. Integrating metabolomics and proteomics to identify novel drug targets for heart failure and atrial fibrillation. Genome Med 2024; 16:120. [PMID: 39434187 PMCID: PMC11492627 DOI: 10.1186/s13073-024-01395-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 10/11/2024] [Indexed: 10/23/2024] Open
Abstract
BACKGROUND Altered metabolism plays a role in the pathophysiology of cardiac diseases, such as atrial fibrillation (AF) and heart failure (HF). We aimed to identify novel plasma metabolites and proteins associating with cardiac disease. METHODS Mendelian randomisation (MR) was used to assess the association of 174 metabolites measured in up to 86,507 participants with AF, HF, dilated cardiomyopathy (DCM), and non-ischemic cardiomyopathy (NICM). Subsequently, we sourced data on 1567 plasma proteins and performed cis MR to identify proteins affecting the identified metabolites as well as the cardiac diseases. Proteins were prioritised on cardiac expression and druggability, and mapped to biological pathways. RESULTS We identified 35 metabolites associating with cardiac disease. AF was affected by seventeen metabolites, HF by nineteen, DCM by four, and NCIM by taurine. HF was particularly enriched for phosphatidylcholines (p = 0.029) and DCM for acylcarnitines (p = 0.001). Metabolite involvement with AF was more uniform, spanning for example phosphatidylcholines, amino acids, and acylcarnitines. We identified 38 druggable proteins expressed in cardiac tissue, with a directionally concordant effect on metabolites and cardiac disease. We recapitulated known associations, for example between the drug target of digoxin (AT1B2), taurine and NICM risk. Additionally, we identified numerous novel findings, such as higher RET values associating with phosphatidylcholines and decreasing AF and HF. RET is targeted by drugs such as regorafenib which has known cardiotoxic side-effects. Pathway analysis implicated involvement of GDF15 signalling through RET, and ghrelin regulation of energy homeostasis in cardiac pathogenesis. CONCLUSIONS This study identified 35 plasma metabolites involved with cardiac diseases and linked these to 38 druggable proteins, providing actionable leads for drug development.
Collapse
Affiliation(s)
- Marion van Vugt
- Department of Cardiology, University Medical Center Utrecht, Utrecht University, Division Heart & Lungs, Utrecht, The Netherlands.
- Institute of Cardiovascular Science, Faculty of Population Health, University College London, London, UK.
- Department of Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centre, University of Amsterdam, Amsterdam, Netherlands.
- Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, Amsterdam, The Netherlands.
| | - Chris Finan
- Department of Cardiology, University Medical Center Utrecht, Utrecht University, Division Heart & Lungs, Utrecht, The Netherlands
- Institute of Cardiovascular Science, Faculty of Population Health, University College London, London, UK
- UCL British Heart Foundation Research Accelerator, London, UK
- Health Data Research UK and Institute of Health Informatics, University College London, London, UK
| | - Sandesh Chopade
- Institute of Cardiovascular Science, Faculty of Population Health, University College London, London, UK
- UCL British Heart Foundation Research Accelerator, London, UK
| | - Rui Providencia
- Health Data Research UK and Institute of Health Informatics, University College London, London, UK
| | - Connie R Bezzina
- Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, Amsterdam, The Netherlands
- Department of Experimental Cardiology, Amsterdam University Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
- European Reference Network for rare, low prevalence and complex diseases of the heart: ERN GUARD-Heart , Amsterdam, The Netherlands
| | - Folkert W Asselbergs
- Department of Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centre, University of Amsterdam, Amsterdam, Netherlands
- Institute of Health Informatics, University College London, London, UK
- The National Institute for Health Research University College London Hospitals Biomedical Research Centre, University College London, London, UK
| | - Jessica van Setten
- Department of Cardiology, University Medical Center Utrecht, Utrecht University, Division Heart & Lungs, Utrecht, The Netherlands
| | - A Floriaan Schmidt
- Department of Cardiology, University Medical Center Utrecht, Utrecht University, Division Heart & Lungs, Utrecht, The Netherlands
- Institute of Cardiovascular Science, Faculty of Population Health, University College London, London, UK
- Department of Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centre, University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, Amsterdam, The Netherlands
- UCL British Heart Foundation Research Accelerator, London, UK
| |
Collapse
|
4
|
Dash M, Mahajan B, Shah S, Dar GM, Sahu P, Sharma AK, Nimisha, Saluja SS. Distinct methylome profile of cfDNA in AMI patients reveals significant alteration in cAMP signaling pathway genes regulating cardiac muscle contraction. Clin Epigenetics 2024; 16:144. [PMID: 39415189 PMCID: PMC11484321 DOI: 10.1186/s13148-024-01755-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 09/30/2024] [Indexed: 10/18/2024] Open
Abstract
BACKGROUND The role of epigenetics in cardiovascular diseases has paved the way for innovative therapeutic approaches. Investigating epigenetic changes using cell-free DNA (cfDNA) holds substantial promise beyond mere diagnostics, especially for heart-related conditions like acute myocardial infarction (AMI), where obtaining tissue samples is a challenge. This study explores the methylation patterns of cfDNA in AMI patients and compares them with genomic DNA (gDNA) from the same individuals, aiming to evaluate the effectiveness of cfDNA as a valuable resource for studying heart-related diseases. METHODOLOGY We generated global methylome profiles of cfDNA and gDNA from 25 AMI patients using EM-Seq. Tissue deconvolution analysis was performed to estimate tissue specificity based on the methylation patterns. Differentially methylated loci were identified and explored to understand AMI pathophysiology. RESULTS Comparative analysis of cfDNA and gDNA methylation patterns in AMI patients reveals cfDNA holds more significance than gDNA. Principal component analysis revealed distinct clusters for cfDNA and gDNA, indicating distinct methylome profiles. cfDNA originated from multiple sources, predominantly from neutrophils (~ 75%) and about 10% from the left atrium, highlighting cardiac-specific changes. In contrast, immune cells are the major source of gDNA, indicative of inflammatory responses. Gene set enrichment analysis (GSEA) associates cfDNA methylation patterns with pathways related to cardiac muscle contraction, inflammation, hypoxia, and lipid metabolism. The affected genes include G protein-coupled receptors (GHSR, FFAR2, HTR1A, and VIPR2) that are part of the cAMP signaling pathway. CONCLUSION Epigenetic changes in cfDNA are more specific to cardiac tissue compared to those in gDNA, providing better insights into the molecular mechanisms involved in AMI. Genes that are differentially methylated in cfDNA and regulate core pathways, such as cAMP signaling, could be targeted for clinical applications, including the development of effective biomarkers and therapeutic targets.
Collapse
Grants
- ISRM/12(44)/2020 Indian Council of Medical Research
- ISRM/12(44)/2020 Indian Council of Medical Research
- ISRM/12(44)/2020 Indian Council of Medical Research
- BT/INF/22/SP33063/2019 Department of Biotechnology, Ministry of Science and Technology, India
- BT/INF/22/SP33063/2019 Department of Biotechnology, Ministry of Science and Technology, India
- BT/INF/22/SP33063/2019 Department of Biotechnology, Ministry of Science and Technology, India
- BT/INF/22/SP33063/2019 Department of Biotechnology, Ministry of Science and Technology, India
- BT/INF/22/SP33063/2019 Department of Biotechnology, Ministry of Science and Technology, India
Collapse
Affiliation(s)
- Manoswini Dash
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India
- School of Medicine, Center for Aging, Tulane University, Louisiana, USA
| | - Bhawna Mahajan
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India.
- Department of Biochemistry, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), Room No:419, Fourth Floor, Academic Block, New Delhi, India.
| | - Shobhita Shah
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India
| | - Ghulam Mehdi Dar
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India
| | - Parameswar Sahu
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India
| | - Abhay Kumar Sharma
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India
| | - Nimisha
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India
| | - Sundeep Singh Saluja
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India
- Department of GI Surgery, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India
| |
Collapse
|
5
|
Sullivan R, Hou J, Yu L, Wilk B, Sykes J, Biernaski H, Butler J, Kovacs M, Hicks J, Thiessen JD, Dharmakumar R, Prato FS, Wisenberg G, Luyt LG, Dhanvantari S. Design, Synthesis, and Preclinical Evaluation of a High-Affinity 18F-Labeled Radioligand for Myocardial Growth Hormone Secretagogue Receptor Before and After Myocardial Infarction. J Nucl Med 2024; 65:1633-1639. [PMID: 39266294 DOI: 10.2967/jnumed.124.267578] [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: 02/22/2024] [Accepted: 08/13/2024] [Indexed: 09/14/2024] Open
Abstract
The peptide hormone ghrelin is produced in cardiomyocytes and acts through the myocardial growth hormone secretagogue receptor (GHSR) to promote cardiomyocyte survival. Administration of ghrelin may have therapeutic effects on post-myocardial infarction (MI) outcomes. Therefore, there is a need to develop molecular imaging probes that can track the dynamics of GHSR in health and disease to better predict the effectiveness of ghrelin-based therapeutics. We designed a high-affinity GHSR ligand labeled with 18F for imaging by PET and characterized its in vivo properties in a canine model of MI. Methods: We rationally designed and radiolabeled with 18F a quinazolinone derivative ([18F]LCE470) with subnanomolar binding affinity to GHSR. We determined the sensitivity and in vivo and ex vivo specificity of [18F]LCE470 in a canine model of surgically induced MI using PET/MRI, which allowed for anatomic localization of tracer uptake and simultaneous determination of global cardiac function. Uptake of [18F]LCE470 was determined by time-activity curve and SUV analysis in 3 regions of the left ventricle-area of infarct, territory served by the left circumflex coronary artery, and remote myocardium-over a period of 1.5 y. Changes in cardiac perfusion were tracked by [13N]NH3 PET. Results: The receptor binding affinity of LCE470 was measured at 0.33 nM, the highest known receptor binding affinity for a radiolabeled GHSR ligand. In vivo blocking studies in healthy hounds and ex vivo blocking studies in myocardial tissue showed the specificity of [18F]LCE470, and sensitivity was demonstrated by a positive correlation between tracer uptake and GHSR abundance. Post-MI changes in [18F]LCE470 uptake occurred independently of perfusion tracer distributions and changes in global cardiac function. We found that the regional distribution of [18F]LCE470 within the left ventricle diverged significantly within 1 d after MI and remained that way throughout the 1.5-y duration of the study. Conclusion: [18F]LCE470 is a high-affinity PET tracer that can detect changes in the regional distribution of myocardial GHSR after MI. In vivo PET molecular imaging of the global dynamics of GHSR may lead to improved GHSR-based therapeutics in the treatment of post-MI remodeling.
Collapse
Affiliation(s)
- Rebecca Sullivan
- Imaging Research Program, Lawson Health Research Institute, London, Ontario, Canada
- Department of Pathology and Laboratory Medicine, Western University, London, Ontario, Canada
| | - Jinqiang Hou
- Lakehead University and Thunder Bay Regional Health Research Institute, Thunder Bay, Ontario, Canada
| | - Lihai Yu
- London Regional Cancer Program, London, Ontario, Canada
| | - Benjamin Wilk
- Imaging Research Program, Lawson Health Research Institute, London, Ontario, Canada
- Department of Medical Biophysics, Western University, London, Ontario, Canada
| | - Jane Sykes
- Imaging Research Program, Lawson Health Research Institute, London, Ontario, Canada
| | - Heather Biernaski
- Imaging Research Program, Lawson Health Research Institute, London, Ontario, Canada
| | - John Butler
- Imaging Research Program, Lawson Health Research Institute, London, Ontario, Canada
| | - Michael Kovacs
- Imaging Research Program, Lawson Health Research Institute, London, Ontario, Canada
- Department of Medical Biophysics, Western University, London, Ontario, Canada
| | - Justin Hicks
- Imaging Research Program, Lawson Health Research Institute, London, Ontario, Canada
- Department of Medical Biophysics, Western University, London, Ontario, Canada
| | - Jonathan D Thiessen
- Imaging Research Program, Lawson Health Research Institute, London, Ontario, Canada
- Department of Medical Biophysics, Western University, London, Ontario, Canada
| | | | - Frank S Prato
- Imaging Research Program, Lawson Health Research Institute, London, Ontario, Canada
- Department of Medical Biophysics, Western University, London, Ontario, Canada
| | - Gerald Wisenberg
- Imaging Research Program, Lawson Health Research Institute, London, Ontario, Canada
| | - Leonard G Luyt
- London Regional Cancer Program, London, Ontario, Canada
- Departments of Chemistry, Oncology, and Medical Imaging, Western University, London, Ontario, Canada
| | - Savita Dhanvantari
- Imaging Research Program, Lawson Health Research Institute, London, Ontario, Canada;
- Department of Pathology and Laboratory Medicine, Western University, London, Ontario, Canada
- Department of Medical Biophysics, Western University, London, Ontario, Canada
| |
Collapse
|
6
|
Ngernsoungnern P, Rungsawang P, Janthaweera A, Duangsuwan P, Saowakon N, Sritangos P, Ngernsoungnern A. Ultrastructural study of neuronal cells and localization of ghrelin-like peptide and its receptor in the ganglia of the golden apple snail (Pomacea canaliculata). Tissue Cell 2024; 88:102348. [PMID: 38493758 DOI: 10.1016/j.tice.2024.102348] [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: 10/29/2023] [Revised: 03/02/2024] [Accepted: 03/04/2024] [Indexed: 03/19/2024]
Abstract
Pomacea canaliculata is an invasive snail species causing major problems in agriculture. The snail biology was then investigated. The main objective of the present study was to investigate the nervous system of the snail. The nervous system comprises pairs of cerebral, buccal, pedal, pleural, parietal ganglia and an unpaired visceral ganglion. Most neurons were concentrated at the periphery of the ganglia. The neurons were classified into four types: NR1, NR2, NR3, and NR4. The percentages of the NR3 and NR4 in the pleural and pedal ganglia were significantly higher than those of other ganglia. Ultrastructural study revealed that nuclei of all neuronal types exhibited mostly euchromatins. Many organelles including ribosomes and endoplasmic reticulum were found in their cytoplasm. However, various mitochondria were found in the NR2 and NR3. The immunohistochemistry revealed immunoreactivity of ghrelin-like peptide in the neurons of the cerebral, pleural and pedal ganglia. However, immunoreactivity of GHS-R1a-like peptide existed only in the neurons of the pleural and pedal ganglia. The present study is the first to demonstrate the existence of ghrelin-like peptide and its receptor in P. canaliculata nervous system.
Collapse
Affiliation(s)
- Piyada Ngernsoungnern
- School of Preclinical Sciences, Institute of Science, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand
| | - Piyachat Rungsawang
- School of Preclinical Sciences, Institute of Science, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand
| | | | - Pornsawan Duangsuwan
- Anatomy Program, Division of Health and Applied Sciences, Faculty of Science, Prince of Songkla University, Hat Yai, Songkhla 90112, Thailand
| | - Naruwan Saowakon
- School of Preclinical Sciences, Institute of Science, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand
| | - Pishyaporn Sritangos
- School of Preclinical Sciences, Institute of Science, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand
| | - Apichart Ngernsoungnern
- School of Preclinical Sciences, Institute of Science, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand.
| |
Collapse
|
7
|
Rathore M, Das N, Ghosh N, Guha R. Insights on discovery, efficacy, safety and clinical applications of ghrelin receptor agonist capromorelin in veterinary medicine. Vet Res Commun 2024; 48:1-10. [PMID: 37493940 DOI: 10.1007/s11259-023-10184-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 07/20/2023] [Indexed: 07/27/2023]
Abstract
Growth hormone and insulin like growth factor-1 plays an important role in the regulation of body composition and metabolism. Growth Hormone is released from the pituitary through a specific G-protein coupled receptor (GPCR) called growth hormone secretagogue receptor 1a expressed in the hypothalamus. Ghrelin is a peptide hormone released from the cells in the stomach, which stimulates appetite and food intake in mammals, regulates gut motility, gastric acid secretion, taste sensation, circadian rhythm, learning and memory, oxidative stress, autophagy, glucose metabolism etc. When the release of the endogenous ligand GHSR-1a, i.e., ghrelin is malfunctioned or stopped, external substitutes are administrated to induce the stimulation of growth hormone and appetite. A class of compound known as ghrelin receptor agonists are developed as an external substitute of ghrelin for regulation and stimulation of growth hormone in frailty, for body weight gain, muscle mass gain, prevention of cachexia and for the treatment of chronic fatigue syndromes. Capromorelin [Entyce™ (Aratana Therapeutics, Leawood, KS, USA)] is the only FDA (Food and Drug Administration) approved (May 2016) drug used for stimulating appetite in dogs and was marketed in the fall of 2017. In 2020, USFDA approved Capromorelin [Elura™ (Elanco US Inc.)] for the management of weight loss in chronic kidney disease of cats. This article reviews the discovery of the ghrelin receptor agonist capromorelin, its efficacy, safety, clinical applications and aims to delineate its further scope of use in veterinary practice.
Collapse
Affiliation(s)
- Manisha Rathore
- Laboratory Animal Facility, CSIR-Central Drug Research Institute, Lucknow, India
| | - Nabanita Das
- National Institute of Pharmaceutical Education and Research, Raebareli, India
| | - Nayan Ghosh
- Division of Medicinal and Process Chemistry, CSIR-Central Drug Research Institute, Lucknow, India
| | - Rajdeep Guha
- Laboratory Animal Facility, CSIR-Central Drug Research Institute, Lucknow, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India.
| |
Collapse
|
8
|
Kim DM, Lee JH, Pan Q, Han HW, Shen Z, Eshghjoo S, Wu CS, Yang W, Noh JY, Threadgill DW, Guo S, Wright G, Alaniz R, Sun Y. Nutrient-sensing growth hormone secretagogue receptor in macrophage programming and meta-inflammation. Mol Metab 2024; 79:101852. [PMID: 38092245 PMCID: PMC10772824 DOI: 10.1016/j.molmet.2023.101852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 12/03/2023] [Accepted: 12/08/2023] [Indexed: 12/20/2023] Open
Abstract
OBJECTIVE Obesity-associated chronic inflammation, aka meta-inflammation, is a key pathogenic driver for obesity-associated comorbidity. Growth hormone secretagogue receptor (GHSR) is known to mediate the effects of nutrient-sensing hormone ghrelin in food intake and fat deposition. We previously reported that global Ghsr ablation protects against diet-induced inflammation and insulin resistance, but the site(s) of action and mechanism are unknown. Macrophages are key drivers of meta-inflammation. To unravel the role of GHSR in macrophages, we generated myeloid-specific Ghsr knockout mice (LysM-Cre;Ghsrf/f). METHODS LysM-Cre;Ghsrf/f and control Ghsrf/f mice were subjected to 5 months of high-fat diet (HFD) feeding to induce obesity. In vivo, metabolic profiling of food intake, physical activity, and energy expenditure, as well as glucose and insulin tolerance tests (GTT and ITT) were performed. At termination, peritoneal macrophages (PMs), epididymal white adipose tissue (eWAT), and liver were analyzed by flow cytometry and histology. For ex vivo studies, bone marrow-derived macrophages (BMDMs) were generated from the mice and treated with palmitic acid (PA) or lipopolysaccharide (LPS). For in vitro studies, macrophage RAW264.7 cells with Ghsr overexpression or Insulin receptor substrate 2 (Irs2) knockdown were studied. RESULTS We found that Ghsr expression in PMs was increased under HFD feeding. In vivo, HFD-fed LysM-Cre;Ghsrf/f mice exhibited significantly attenuated systemic inflammation and insulin resistance without affecting food intake or body weight. Tissue analysis showed that HFD-fed LysM-Cre;Ghsrf/f mice have significantly decreased monocyte/macrophage infiltration, pro-inflammatory activation, and lipid accumulation, showing elevated lipid-associated macrophages (LAMs) in eWAT and liver. Ex vivo, Ghsr-deficient macrophages protected against PA- or LPS-induced pro-inflammatory polarization, showing reduced glycolysis, increased fatty acid oxidation, and decreased NF-κB nuclear translocation. At molecular level, GHSR metabolically programs macrophage polarization through PKA-CREB-IRS2-AKT2 signaling pathway. CONCLUSIONS These novel results demonstrate that macrophage GHSR plays a key role in the pathogenesis of meta-inflammation, and macrophage GHSR promotes macrophage infiltration and induces pro-inflammatory polarization. These exciting findings suggest that GHSR may serve as a novel immunotherapeutic target for the treatment of obesity and its associated comorbidity.
Collapse
Affiliation(s)
- Da Mi Kim
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA
| | - Jong Han Lee
- Department of Marine Bioindustry, Hanseo University, Seosan 31962, South Korea; USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College Medicine, Houston, TX 77030, USA
| | - Quan Pan
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA
| | - Hye Won Han
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA
| | - Zheng Shen
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA
| | - Sahar Eshghjoo
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX 77807, USA; Agilent technologies, Aanta Clara, CA 95051, USA
| | - Chia-Shan Wu
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA; USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College Medicine, Houston, TX 77030, USA
| | - Wanbao Yang
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA
| | - Ji Yeon Noh
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA
| | - David W Threadgill
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA; Texas A&M Institute for Genome Sciences and Society, Department of Cell Biology and Genetics, Texas A&M University, College Station, TX 77843, USA
| | - Shaodong Guo
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA
| | - Gus Wright
- Department of Veterinary Pathobiology, Texas A&M University, College Station, TX 77843, USA
| | - Robert Alaniz
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX 77807, USA; Tlaloc Therapeutics Inc., College Station, TX 77845, USA
| | - Yuxiang Sun
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA; USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College Medicine, Houston, TX 77030, USA.
| |
Collapse
|
9
|
Zhu L, Dou Z, Wu W, Hou Q, Wang S, Yuan Z, Li B, Liu J. Ghrelin/GHSR Axis Induced M2 Macrophage and Alleviated Intestinal Barrier Dysfunction in a Sepsis Rat Model by Inactivating E2F1/NF- κB Signaling. Can J Gastroenterol Hepatol 2023; 2023:1629777. [PMID: 38187112 PMCID: PMC10769719 DOI: 10.1155/2023/1629777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 10/20/2023] [Accepted: 12/11/2023] [Indexed: 01/09/2024] Open
Abstract
Sepsis is an inflammatory reaction disorder state that is induced by infection. The activation and regulation of the immune system play an essential role in the development of sepsis. Our previous studies have shown that ghrelin ameliorates intestinal dysfunction in sepsis. Very little is known about the mechanism of ghrelin and its receptor (GHSR) on the intestinal barrier and the immune function of macrophage regulation. Our research is to investigate the regulatory effect and molecular mechanism of the ghrelin/GHSR axis on intestinal dysfunction and macrophage polarization in septic rats. A rat model of sepsis was established by cecal ligation and puncture (CLP) operation. Then, the sepsis rats were treated with a ghrelin receptor agonist (TZP-101) or ghrelin inhibitor (obestatin). The results suggested that TZP-101 further enhanced ghrelin and GHSR expressions in the colon and spleen of septic rats and obestatin showed the opposite results. Ghrelin/GHSR axis ameliorated colonic structural destruction and intestinal epithelial tight junction injury in septic rats. In addition, the ghrelin/GHSR axis promoted M2-type polarization of macrophages, which was characterized by the decreases of IL-1β, IL-6, and TNF-α, as well as the increase of IL-10. Mechanistically, the ghrelin/GHSR axis promoted E2F2 expression and suppressed the activation of the NF-κB signaling pathway in septic rats. Collectively, targeting ghrelin/GHSR during sepsis may represent a novel therapeutic approach for the treatment of intestinal barrier injury.
Collapse
Affiliation(s)
- Lei Zhu
- Department of Intensive Care Medicine, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Zhimin Dou
- Department of Intensive Care Medicine, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Wei Wu
- Department of Intensive Care Medicine, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Qiliang Hou
- Department of Intensive Care Medicine, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Sen Wang
- Department of Intensive Care Medicine, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Ziqian Yuan
- Department of Intensive Care Medicine, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Bin Li
- Department of Intensive Care Medicine, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Jian Liu
- Department of Intensive Care Medicine, The First Hospital of Lanzhou University, Lanzhou 730000, China
| |
Collapse
|
10
|
Hage C, Ståhlberg M, Thorvaldsen T, Faxén UL, Pironti G, Webb DL, Hellström PM, Andersson DC, Lund LH. Acyl ghrelin infusion increases circulating growth hormone in patients with heart failure and reduced ejection fraction. Eur J Heart Fail 2023; 25:2093-2095. [PMID: 37655533 DOI: 10.1002/ejhf.3019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 08/24/2023] [Accepted: 08/29/2023] [Indexed: 09/02/2023] Open
Affiliation(s)
- Camilla Hage
- Unit of Cardiology, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
- Karolinska University Hospital, Heart, Vascular and Neuro Theme, Stockholm, Sweden
| | - Marcus Ståhlberg
- Unit of Cardiology, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
- Karolinska University Hospital, Heart, Vascular and Neuro Theme, Stockholm, Sweden
| | - Tonje Thorvaldsen
- Unit of Cardiology, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
- Karolinska University Hospital, Heart, Vascular and Neuro Theme, Stockholm, Sweden
| | - Ulrika L Faxén
- Unit of Cardiology, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
- Perioperative Medicine and Intensive Care, Karolinska University Hospital, Stockholm, Sweden
| | - Gianluigi Pironti
- Unit of Cardiology, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Dominic-Luc Webb
- Department of Medical Sciences, Gastroenterology and Hepatology, Uppsala University, Uppsala, Sweden
| | - Per M Hellström
- Department of Medical Sciences, Gastroenterology and Hepatology, Uppsala University, Uppsala, Sweden
| | - Daniel C Andersson
- Karolinska University Hospital, Heart, Vascular and Neuro Theme, Stockholm, Sweden
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Lars H Lund
- Unit of Cardiology, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
- Karolinska University Hospital, Heart, Vascular and Neuro Theme, Stockholm, Sweden
| |
Collapse
|
11
|
Inceu AI, Neag MA, Craciun AE, Buzoianu AD. Gut Molecules in Cardiometabolic Diseases: The Mechanisms behind the Story. Int J Mol Sci 2023; 24:3385. [PMID: 36834796 PMCID: PMC9965280 DOI: 10.3390/ijms24043385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/03/2023] [Accepted: 02/06/2023] [Indexed: 02/10/2023] Open
Abstract
Atherosclerotic cardiovascular disease is the most common cause of morbidity and mortality worldwide. Diabetes mellitus increases cardiovascular risk. Heart failure and atrial fibrillation are associated comorbidities that share the main cardiovascular risk factors. The use of incretin-based therapies promoted the idea that activation of alternative signaling pathways is effective in reducing the risk of atherosclerosis and heart failure. Gut-derived molecules, gut hormones, and gut microbiota metabolites showed both positive and detrimental effects in cardiometabolic disorders. Although inflammation plays a key role in cardiometabolic disorders, additional intracellular signaling pathways are involved and could explain the observed effects. Revealing the involved molecular mechanisms could provide novel therapeutic strategies and a better understanding of the relationship between the gut, metabolic syndrome, and cardiovascular diseases.
Collapse
Affiliation(s)
- Andreea-Ioana Inceu
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
| | - Maria-Adriana Neag
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
| | - Anca-Elena Craciun
- Department of Diabetes, and Nutrition Diseases, Iuliu Hatieganu University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania
| | - Anca-Dana Buzoianu
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
| |
Collapse
|
12
|
Childs MD, Yu L, Kovacs MS, Luyt LG. Radiofluorination of non-activated aromatic prosthetic groups for synthesis and evaluation of fluorine-18 labelled ghrelin(1-8) analogues. Org Biomol Chem 2021; 19:8812-8820. [PMID: 34590643 DOI: 10.1039/d1ob01023a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The growth hormone secretagogue receptor 1a (GHSR) is differentially expressed in various disease states compared to healthy tissues and thus is a target for molecular imaging. The endogenous ligand for the GHSR is ghrelin, a 28 amino acid peptide with a unique octanoyl group on the serine-3 residue. A recently reported ghrelin analogue revealed the successful use of fluorine-containing, polycyclic aromatic groups in place of the octanoyl side chain, thereby providing potential access to new 18F-PET imaging probes. The peptide [Inp1,Dpr3(6-FN),1Nal4,Thr8]ghrelin(1-8) amide (1) showed sub-nanomolar receptor affinity (IC50 = 0.11 nM) toward the GHSR making it the strongest affinity ghrelin analogue reported to date. However, attempts to label such non-activated aromatic groups with fluoride-18 through conventional substitution methods resulted in low radiochemical yields, impractical for use in vivo. Since larger, non-activated aromatic groups appear to be of value for incorporating fluorine into ghrelin(1-8) analogues, an additional peptide bearing a 4'-fluorobiphenyl-4-carboxyl (4'-FBC) group in place of the octanoyl side chain was also of interest. Herein, we describe the radiosynthesis of [Inp1,Dpr3([18F]6-FN),1Nal4,Thr8]ghrelin(1-8) amide ([18F]1) and [Inp1,Dpr3([18F]4'-FBC),1Nal4,Thr8]ghrelin(1-8) amide ([18F]2) using a prosthetic group approach from iodonium ylide precursors as well as initial in vitro and in vivo evaluation of [18F]1 as a potential PET tracer for targeted imaging of the GHSR.
Collapse
Affiliation(s)
- Marina D Childs
- Department of Chemistry, University of Western Ontario, 1151 Richmond Street, London, Ontario, N6A 3 K7, Canada.
| | - Lihai Yu
- London Regional Cancer Program, Lawson Health Research Institute, 800 Commissioners Road East, London, Ontario, N6A 4L6, Canada
| | - Michael S Kovacs
- Department of Medical Imaging, University of Western Ontario, 1151 Richmond Street, London, Ontario, N6A 3 K7, Canada
- Department of Medical Biophysics, University of Western Ontario, 1151 Richmond Street, London, Ontario, N6A 3 K7, Canada
- Lawson Cyclotron & PET Radiochemistry Facility, Lawson Health Research Institute, 268 Grosvenor Street, London, Ontario, N6A 5 W9, Canada
| | - Leonard G Luyt
- Department of Chemistry, University of Western Ontario, 1151 Richmond Street, London, Ontario, N6A 3 K7, Canada.
- London Regional Cancer Program, Lawson Health Research Institute, 800 Commissioners Road East, London, Ontario, N6A 4L6, Canada
- Department of Medical Imaging, University of Western Ontario, 1151 Richmond Street, London, Ontario, N6A 3 K7, Canada
- Department of Oncology, University of Western Ontario, 1151 Richmond Street, London, Ontario, N6A 3 K7, Canada
| |
Collapse
|
13
|
Chen Y, Zhou S, Zhang A, Huang J, Zhang G, Cui L. Temporal changes and prognostic value of plasma ghrelin level in patients with acute heart failure: a prospective study. Heart Vessels 2021; 37:419-425. [PMID: 34533592 DOI: 10.1007/s00380-021-01935-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 08/27/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND Plasma ghrelin levels can be elevated in patients with acute heart failure (AHF). This study aimed to analyze the temporal changes and prognostic value of ghrelin levels in patients with AHF. METHODS This prospective study included patients with AHF at the Cardiology Department, Weifang People's Hospital (May 2018-October 2019), and age- and sex-matched healthy controls. Plasma ghrelin levels were measured. Multivariable logistic regression and receiver operating characteristic (ROC) curve analyses were used to evaluate whether ghrelin levels could predict major cardiac adverse events (MACEs) during a 1-year follow-up. RESULTS Finally, 92 patients with AHF and 50 healthy controls were enrolled. Ghrelin levels were higher in patients with AHF at 1, 3, 12, and 24 h compared with controls (all P < 0.01). Ghrelin levels in the AHF group were higher at 3 and 12 h than at 1 and 24 h (P < 0.001). Ghrelin level at 3 h in patients with AHF was negatively correlated with the left ventricular end-diastolic diameter and left ventricular ejection fraction (both P < 0.05). MACEs occurred in 48 patients with AHF. Ghrelin levels were higher in the MACE group than in the non-MACE group at 1 (P = 0.011) and 3 h (P = 0.034). Multivariable regression showed that ghrelin level at 3 h was independently associated with MACEs [OR = 0.629, 95% confidence interval (CI): 0.515-0.742, P = 0.010], but the area under the ROC curve was only 0.629 (95% CI 0.515-0.742). CONCLUSIONS Plasma ghrelin levels are elevated in AHF and patients with MACEs during follow-up.
Collapse
Affiliation(s)
- Yanbo Chen
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China.,Department of Cardioangiology, Weifang People's Hospital, Weifang, 261041, Shandong, China
| | - Sani Zhou
- Central Sterile Supply Department, Weifang People's Hospital, Weifang, 261041, Shandong, China
| | - Aiyuan Zhang
- Department of Cardioangiology, Weifang People's Hospital, Weifang, 261041, Shandong, China
| | - Jing Huang
- Department of Cardioangiology, Weifang People's Hospital, Weifang, 261041, Shandong, China
| | - Guangfang Zhang
- Department of Cardioangiology, Weifang People's Hospital, Weifang, 261041, Shandong, China
| | - Lianqun Cui
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China.
| |
Collapse
|
14
|
Wang M, Qian L, Li J, Ming H, Fang L, Li Y, Zhang M, Xu Y, Ban Y, Zhang W, Zhang Y, Liu Y, Wang N. GHSR deficiency exacerbates cardiac fibrosis: role in macrophage inflammasome activation and myofibroblast differentiation. Cardiovasc Res 2021; 116:2091-2102. [PMID: 31790138 DOI: 10.1093/cvr/cvz318] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 08/06/2019] [Accepted: 11/28/2019] [Indexed: 12/19/2022] Open
Abstract
AIMS Sustained activation of β-adrenergic signalling induces cardiac fibrosis, which marks progression to heart failure. GHSR (growth hormone secretagogue receptor) is the receptor for ghrelin, which is an orexigenic gastric hormone with newly defined cardiovascular effects. The present study determined the effects of GHSR deficiency in a mouse model of isoproterenol (ISO)-induced cardiac fibrosis and examined the underlying mechanism. METHODS AND RESULTS Histochemical studies showed that GHSR deficiency exacerbated cardiac fibrosis. Quantitative RT-PCR, western blotting, and immunofluorescence staining demonstrated that cardiac fibroblasts isolated from GHSR-/- mice exhibited increased expression of marker genes for myofibroblast trans-differentiation (α-SMA, SM22, and calponin) upon transforming growth factor-β treatment compared to wild-type mice. RNA-sequencing of heart transcriptomes revealed that differentially expressed genes in GHSR-/- hearts were enriched in such biological processes as extracellular matrix organization, inflammatory response, lipid metabolism, cell cycle, migration, and adhesion. Particularly, GHSR deficiency increased Wnt/β-catenin pathway activation in ISO-induced myocardial fibrosis. In addition, loss of GHSR in macrophages instigated inflammasome activation with increased cleavage and release of interleukin-18. CONCLUSION These results for the first time demonstrated that GHSR deficiency aggravated ISO-induced cardiac fibrosis, suggesting that GHSR was a potential target for the intervention of cardiac fibrosis.
Collapse
Affiliation(s)
- Mo Wang
- Institute of Cardiovascular Sciences, Peking University Health Science Center, Beijing 100191, China.,Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China
| | - Lei Qian
- The Advanced Institute of Medical Sciences, Dalian Medical University, Dalian 116044, China
| | - Jing Li
- Institute of Cardiovascular Sciences, Peking University Health Science Center, Beijing 100191, China.,Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China
| | - Hao Ming
- Institute of Cardiovascular Sciences, Peking University Health Science Center, Beijing 100191, China.,Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China
| | - Li Fang
- Institute of Cardiovascular Sciences, Peking University Health Science Center, Beijing 100191, China.,Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China
| | - Yingjia Li
- Institute of Cardiovascular Sciences, Peking University Health Science Center, Beijing 100191, China.,Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China
| | - Man Zhang
- Institute of Cardiovascular Sciences, Peking University Health Science Center, Beijing 100191, China.,Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China
| | - Yaohua Xu
- Institute of Cardiovascular Sciences, Peking University Health Science Center, Beijing 100191, China.,Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China
| | - Yiqian Ban
- Institute of Cardiovascular Sciences, Peking University Health Science Center, Beijing 100191, China.,Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China
| | - Weizhen Zhang
- Institute of Cardiovascular Sciences, Peking University Health Science Center, Beijing 100191, China.,Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China
| | - Youyi Zhang
- Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China.,Institute of Vascular Medicine, The Third Hospital, Peking University, Beijing, China
| | - Yahan Liu
- Institute of Cardiovascular Sciences, Peking University Health Science Center, Beijing 100191, China.,Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China
| | - Nanping Wang
- Institute of Cardiovascular Sciences, Peking University Health Science Center, Beijing 100191, China.,Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China.,The Advanced Institute of Medical Sciences, Dalian Medical University, Dalian 116044, China
| |
Collapse
|
15
|
Devesa J. The Complex World of Regulation of Pituitary Growth Hormone Secretion: The Role of Ghrelin, Klotho, and Nesfatins in It. Front Endocrinol (Lausanne) 2021; 12:636403. [PMID: 33776931 PMCID: PMC7991839 DOI: 10.3389/fendo.2021.636403] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 02/12/2021] [Indexed: 12/27/2022] Open
Abstract
The classic concept of how pituitary GH is regulated by somatostatin and GHRH has changed in recent years, following the discovery of peripheral hormones involved in the regulation of energy homeostasis and mineral homeostasis. These hormones are ghrelin, nesfatins, and klotho. Ghrelin is an orexigenic hormone, released primarily by the gastric mucosa, although it is widely expressed in many different tissues, including the central nervous system and the pituitary. To be active, ghrelin must bind to an n-octanoyl group (n = 8, generally) on serine 3, forming acyl ghrelin which can then bind and activate a G-protein-coupled receptor leading to phospholipase C activation that induces the formation of inositol 1,4,5-triphosphate and diacylglycerol that produce an increase in cytosolic calcium that allows the release of GH. In addition to its direct action on somatotrophs, ghrelin co-localizes with GHRH in several neurons, facilitating its release by inhibiting somatostatin, and acts synergistically with GHRH stimulating the synthesis and secretion of pituitary GH. Gastric ghrelin production declines with age, as does GH. Klotho is an anti-aging agent, produced mainly in the kidneys, whose soluble circulating form directly induces GH secretion through the activation of ERK1/2 and inhibits the inhibitory effect that IGF-I exerts on GH. Children and adults with untreated GH-deficiency show reduced plasma levels of klotho, but treatment with GH restores them to normal values. Deletions or mutations of the Klotho gene affect GH production. Nesfatins 1 and 2 are satiety hormones, they inhibit food intake. They have been found in GH3 cell cultures where they significantly reduce the expression of gh mRNA and that of pituitary-specific positive transcription factor 1, consequently acting as inhibitors of GH production. This is a consequence of the down-regulation of the cAMP/PKA/CREB signaling pathway. Interestingly, nesfatins eliminate the strong positive effect that ghrelin has on GH synthesis and secretion. Throughout this review, we will attempt to broadly analyze the role of these hormones in the complex world of GH regulation, a world in which these hormones already play a very important role.
Collapse
Affiliation(s)
- Jesús Devesa
- Scientific and Medical Direction, Medical Center Foltra, Teo, Spain
| |
Collapse
|
16
|
Research progress of ghrelin on cardiovascular disease. Biosci Rep 2021; 41:227556. [PMID: 33427286 PMCID: PMC7823193 DOI: 10.1042/bsr20203387] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 01/04/2021] [Accepted: 01/08/2021] [Indexed: 01/04/2023] Open
Abstract
Ghrelin, a 28-aminoacid peptide, was isolated from the human and rat stomach and identified in 1999 as an endogenous ligand for the growth hormone secretagogue-receptor (GHS-R). In addition to stimulating appetite and regulating energy balance, ghrelin and its receptor GHS-R1a have a direct effect on the cardiovascular system. In recent years, it has been shown that ghrelin exerts cardioprotective effects, including the modulation of sympathetic activity and hypertension, enhancement of the vascular activity and angiogenesis, inhibition of arrhythmias, reduction in heart failure and inhibition of cardiac remodeling after myocardial infarction (MI). The cardiovascular protective effect of ghrelin may be associated with anti-inflammation, anti-apoptosis, inhibited sympathetic nerve activation, regulated autophagy, and endothelial dysfunction. However, the molecular mechanisms underlying the effects of ghrelin on the cardiovascular system have not been fully elucidated, and no specific therapeutic agent has been established. It is important to further explore the pharmacological potential of ghrelin pathway modulation for the treatment of cardiovascular diseases.
Collapse
|
17
|
Liang Y, Yin W, Yin Y, Zhang W. Ghrelin Based Therapy of Metabolic Diseases. Curr Med Chem 2021; 28:2565-2576. [PMID: 32538716 PMCID: PMC11213490 DOI: 10.2174/0929867327666200615152804] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 05/04/2020] [Accepted: 05/18/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Ghrelin, a unique 28 amino acid peptide hormone secreted by the gastric X/A like cells, is an endogenous ligand of the growth hormone secretagogue receptor (GHSR). Ghrelin-GHSR signaling has been found to exert various physiological functions, including stimulation of appetite, regulation of body weight, lipid and glucose metabolism, and increase of gut motility and secretion. This system is thus critical for energy homeostasis. OBJECTIVE The objective of this review is to highlight the strategies of ghrelin-GHSR based intervention for therapy of obesity and its related metabolic diseases. RESULTS Therapeutic strategies of metabolic disorders targeting the ghrelin-GHSR pathway involve neutralization of circulating ghrelin by antibodies and RNA spiegelmers, antagonism of ghrelin receptor by its antagonists and inverse agonists, inhibition of ghrelin O-acyltransferase (GOAT), as well as potential pharmacological approach to decrease ghrelin synthesis and secretion. CONCLUSION Various compounds targeting the ghrelin-GHSR system have shown promising efficacy for the intervention of obesity and relevant metabolic disorders in animals and in vitro. Further clinical trials to validate their efficacy in human beings are urgently needed.
Collapse
Affiliation(s)
- Yuan Liang
- Key Laboratory of Molecular Cardiovascular Science, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Wenzhen Yin
- Key Laboratory of Molecular Cardiovascular Science, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Yue Yin
- Key Laboratory of Molecular Cardiovascular Science, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Weizhen Zhang
- Key Laboratory of Molecular Cardiovascular Science, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
- Department of Surgery, University of Michigan Medical Center, Ann Arbor, MI 48109-0346, USA
| |
Collapse
|
18
|
Regional Differences in the Ghrelin-Growth Hormone Secretagogue Receptor Signalling System in Human Heart Disease. CJC Open 2020; 3:182-194. [PMID: 33644732 PMCID: PMC7893201 DOI: 10.1016/j.cjco.2020.10.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Accepted: 10/27/2020] [Indexed: 12/20/2022] Open
Abstract
Background The hormone ghrelin and its receptor, the growth hormone secretagogue receptor (GHSR) are expressed in myocardium. GHSR binding activates signalling pathways coupled to cardiomyocyte survival and contractility. These properties have made the ghrelin-GHSR axis a candidate for a biomarker of cardiac function. The dynamics of ghrelin-GHSR are altered significantly in late stages of heart failure (HF) and cardiomyopathy, when left ventricular (LV) function is failing. We examined the relationship of GHSR with ghrelin in cardiac tissue from patients with valvular disease with no detectable changes in LV function. Methods Biopsy samples from the left ventricle and left atrium were obtained from 25 patients with valvular disease (of whom 13 also had coronary artery disease) and preserved LV ejection fraction, and compared to control samples obtained via autopsy. Using quantitative confocal fluorescence microscopy, levels of GHSR were determined using [Dpr3(n-octanoyl),Lys19(sulfo-Cy5)]ghrelin(1-19), and immunofluorescence determined ghrelin, the heart failure marker natriuretic peptide type-B (BNP), and contractility marker sarcoplasmic reticulum ATPase pump (SERCA2a). Results A positive correlation between GHSR and ghrelin was apparent in only diseased tissue. Ghrelin and BNP significantly correlated in the left ventricle and strongly colocalized to the same intracellular compartment in diseased and control tissue. GHSR, ghrelin, and BNP all strongly and significantly correlated with SERCA2a in the left ventricle of diseased tissue only. Conclusions Our results suggest that the dynamics of the myocardial ghrelin-GHSR axis is altered in cardiovascular disease in the absence of measurable changes in heart function, and might accompany a regional shift in endocrine programming.
Collapse
|
19
|
Childs MD, Luyt LG. A Decade's Progress in the Development of Molecular Imaging Agents Targeting the Growth Hormone Secretagogue Receptor. Mol Imaging 2020; 19:1536012120952623. [PMID: 33104445 PMCID: PMC8865914 DOI: 10.1177/1536012120952623] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
The growth hormone secretagogue receptor 1a (GHSR), also called the ghrelin receptor, is a G protein-coupled receptor known to play an important metabolic role in the regulation of various physiological processes, including energy expenditure, growth hormone secretion, and cell proliferation. This receptor has been implicated in numerous health issues including obesity, gastrointestinal disorders, type II diabetes, and regulation of body weight in patients with Prader-Willi syndrome, and there has been growing interest in studying its mechanism of behavior to unlock further applications of GHSR-targeted therapeutics. In addition, the GHSR is expressed in various types of cancer including prostate, breast, and testicular cancers, while aberrant expression has been reported in cardiac disease. Targeted molecular imaging of the GHSR could provide insights into its role in biological processes related to these disease states. Over the past decade, imaging probes targeting this receptor have been discovered for the imaging modalities PET, SPECT, and optical imaging. High-affinity analogues of ghrelin, the endogenous ligand for the GHSR, as well as small molecule inhibitors have been developed and evaluated both in vitro and in pre-clinical models. This review provides a comprehensive overview of the molecular imaging agents targeting the GHSR reported to the end of 2019.
Collapse
Affiliation(s)
- Marina D Childs
- Department of Chemistry, University of Western Ontario, London, Ontario, Canada
| | - Leonard G Luyt
- Department of Chemistry, University of Western Ontario, London, Ontario, Canada.,Lawson Health Research Institute, London, Ontario, Canada.,Department of Oncology, University of Western Ontario, London, Ontario, Canada.,Department of Medical Imaging, University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
20
|
Gupta S, Mitra A. Heal the heart through gut (hormone) ghrelin: a potential player to combat heart failure. Heart Fail Rev 2020; 26:417-435. [PMID: 33025414 DOI: 10.1007/s10741-020-10032-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/21/2020] [Indexed: 12/17/2022]
Abstract
Ghrelin, a small peptide hormone (28 aa), secreted mainly by X/A-like cells of gastric mucosa, is also locally produced in cardiomyocytes. Being an orexigenic factor (appetite stimulant), it promotes release of growth hormone (GH) and exerts diverse physiological functions, viz. regulation of energy balance, glucose, and/or fat metabolism for body weight maintenance. Interestingly, administration of exogenous ghrelin significantly improves cardiac functions in CVD patients as well as experimental animal models of heart failure. Ghrelin ameliorates pathophysiological condition of the heart in myocardial infarction, cardiac hypertrophy, fibrosis, cachexia, and ischemia reperfusion injury. This peptide also exerts significant impact at the level of vasculature leading to lowering high blood pressure and reversal of endothelial dysfunction and atherosclerosis. However, the molecular mechanism of actions elucidating the healing effects of ghrelin on the cardiovascular system is still a matter of conjecture. Some experimental data indicate its beneficial effects via complex cellular cross talks between autonomic nervous system and cardiovascular cells, some other suggest more direct receptor-mediated molecular actions via autophagy or ionotropic regulation and interfering with apoptotic and inflammatory pathways of cardiomyocytes and vascular endothelial cells. Here, in this review, we summarise available recent data to encourage more research to find the missing links of unknown ghrelin receptor-mediated pathways as we see ghrelin as a future novel therapy in cardiovascular protection.
Collapse
Affiliation(s)
- Shreyasi Gupta
- Department of Zoology, Triveni Devi Bhalotia College, Raniganj, Paschim Bardhaman, 713347, India
| | - Arkadeep Mitra
- Department of Zoology, City College , 102/1, Raja Rammohan Sarani, Kolkata, 700009, India.
| |
Collapse
|
21
|
Yuan Y, Huang F, Deng C, Zhu P. The Additional Prognostic Value of Ghrelin for Mortality and Readmission in Elderly Patients with Acute Heart Failure. Clin Interv Aging 2020; 15:1353-1363. [PMID: 32848376 PMCID: PMC7429106 DOI: 10.2147/cia.s259889] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 07/20/2020] [Indexed: 12/20/2022] Open
Abstract
Purpose To evaluate the prognostic value of ghrelin, a growth hormone-releasing peptide, for mortality and readmission in elderly patients with acute heart failure (AHF). Patients and Methods We measured plasma ghrelin and pro B-type natriuretic peptide (NT-proBNP) levels upon emergency admission in 241 prospectively recruited elderly AHF patients (61.0% men). The outcomes were all-cause mortality and/or readmission due to heart failure (HF). Multivariate Cox proportional hazards regression analyses were used to evaluate the prognostic value of ghrelin. Discrimination, calibration, and reclassification indices were compared between models, with or without ghrelin. Results During 1.2 years of follow-up, we observed 90 events (57 deaths and 33 readmissions due to HF). Plasma ghrelin levels were significantly elevated in elderly AHF patients, when compared to healthy control subjects (P < 0.001). Patients with events had significantly higher baseline ghrelin levels, when compared to those without (P < 0.001). Ghrelin levels were positively correlated with NT-proBNP levels and HF severity, whereas they were negatively correlated with nutritional status (all P < 0.05). Log transformed ghrelin levels were independently associated with AHF events (hazard ratio = 2.64, 95% confidence interval = 1.11–6.25, P = 0.028). The incorporation of ghrelin into the reference model, or reference with the NT-proBNP model, both improved C-statistics (from 0.742–0.780 and 0.836–0.857; P = 0.074 and 0.044, respectively), resulting in an improvement in net reclassification index (14.42% and 10.45%, P = 0.020 and 0.025, respectively), and integrated discrimination index (5.64% and 3.60%, both P < 0.001). Patients who displayed the above NT-proBNP and ghrelin median levels had a markedly higher risk of AHF adverse events (P < 0.001). Conclusion Plasma ghrelin is an independent predictor of adverse events in elderly AHF patients. Ghrelin may provide additional value to clinical parameters or NT-proBNP for prognostic risk stratification in AHF.
Collapse
Affiliation(s)
- Yin Yuan
- The Shengli Clinical Medical College, Fujian Medical University, Fuzhou, Fujian, People's Republic of China
| | - Feng Huang
- The Shengli Clinical Medical College, Fujian Medical University, Fuzhou, Fujian, People's Republic of China.,Department of Geriatric Medicine, Fujian Provincial Hospital, Fuzhou, Fujian, People's Republic of China.,Fujian Provincial Institute of Clinical Geriatrics, Fuzhou, Fujian, People's Republic of China.,Fujian Provincial Key Laboratory of Geriatrics, Fuzhou, Fujian, People's Republic of China
| | - Chaochao Deng
- Fujian Provincial Institute of Clinical Geriatrics, Fuzhou, Fujian, People's Republic of China
| | - Pengli Zhu
- The Shengli Clinical Medical College, Fujian Medical University, Fuzhou, Fujian, People's Republic of China.,Department of Geriatric Medicine, Fujian Provincial Hospital, Fuzhou, Fujian, People's Republic of China
| |
Collapse
|
22
|
Maekawa T, Tamura H, Domon H, Hiyoshi T, Isono T, Yonezawa D, Hayashi N, Takahashi N, Tabeta K, Maeda T, Oda M, Ziogas A, Alexaki VI, Chavakis T, Terao Y, Hajishengallis G. Erythromycin inhibits neutrophilic inflammation and mucosal disease by upregulating DEL-1. JCI Insight 2020; 5:136706. [PMID: 32603314 DOI: 10.1172/jci.insight.136706] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 06/24/2020] [Indexed: 02/06/2023] Open
Abstract
Macrolide antibiotics exert antiinflammatory effects; however, little is known regarding their immunomodulatory mechanisms. In this study, using 2 distinct mouse models of mucosal inflammatory disease (LPS-induced acute lung injury and ligature-induced periodontitis), we demonstrated that the antiinflammatory action of erythromycin (ERM) is mediated through upregulation of the secreted homeostatic protein developmental endothelial locus-1 (DEL-1). Consistent with the anti-neutrophil recruitment action of endothelial cell-derived DEL-1, ERM inhibited neutrophil infiltration in the lungs and the periodontium in a DEL-1-dependent manner. Whereas ERM (but not other antibiotics, such as josamycin and penicillin) protected against lethal pulmonary inflammation and inflammatory periodontal bone loss, these protective effects of ERM were abolished in Del1-deficient mice. By interacting with the growth hormone secretagogue receptor and activating JAK2 in human lung microvascular endothelial cells, ERM induced DEL-1 transcription that was mediated by MAPK p38 and was CCAAT/enhancer binding protein-β dependent. Moreover, ERM reversed IL-17-induced inhibition of DEL-1 transcription, in a manner that was dependent not only on JAK2 but also on PI3K/AKT signaling. Because DEL-1 levels are severely reduced in inflammatory conditions and with aging, the ability of ERM to upregulate DEL-1 may lead to a novel approach for the treatment of inflammatory and aging-related diseases.
Collapse
Affiliation(s)
- Tomoki Maekawa
- Center for Advanced Oral Science.,Division of Microbiology and Infectious Diseases.,Division of Periodontology, and
| | - Hikaru Tamura
- Center for Advanced Oral Science.,Division of Microbiology and Infectious Diseases.,Division of Periodontology, and
| | - Hisanori Domon
- Center for Advanced Oral Science.,Division of Microbiology and Infectious Diseases
| | - Takumi Hiyoshi
- Center for Advanced Oral Science.,Division of Microbiology and Infectious Diseases
| | | | - Daisuke Yonezawa
- Center for Advanced Oral Science.,Division of Oral Science for Health Promotion, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Naoki Hayashi
- Department of Microbiology and Infection Control Sciences, Kyoto Pharmaceutical University, Yamashina, Japan
| | | | | | - Takeyasu Maeda
- Center for Advanced Oral Science.,Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Masataka Oda
- Department of Microbiology and Infection Control Sciences, Kyoto Pharmaceutical University, Yamashina, Japan
| | - Athanasios Ziogas
- Institute of Clinical Chemistry and Laboratory Medicine, Faculty of Medicine and University Clinic Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Vasileia Ismini Alexaki
- Institute of Clinical Chemistry and Laboratory Medicine, Faculty of Medicine and University Clinic Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Triantafyllos Chavakis
- Institute of Clinical Chemistry and Laboratory Medicine, Faculty of Medicine and University Clinic Carl Gustav Carus, TU Dresden, Dresden, Germany.,Centre for Cardiovascular Science, Queen's Medical Research Institute, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Yutaka Terao
- Center for Advanced Oral Science.,Division of Microbiology and Infectious Diseases
| | - George Hajishengallis
- Laboratory of Innate Immunity and Inflammation, Department of Basic and Translational Sciences, Penn Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
23
|
A traditional herbal medicine rikkunshito prevents angiotensin II-Induced atrial fibrosis and fibrillation. J Cardiol 2020; 76:626-635. [PMID: 32682626 DOI: 10.1016/j.jjcc.2020.07.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 05/26/2020] [Accepted: 06/15/2020] [Indexed: 12/27/2022]
Abstract
BACKGROUND Rikkunshito (RKT), a traditional herbal medicine, has been demonstrated to exert anti-inflammatory, anti-apoptotic, and anti-fibrotic effects in several organs. This study tested the hypothesis that RKT can suppress angiotensin II (AngII)-induced inflammatory atrial fibrosis and ameliorate enhanced vulnerability to atrial fibrillation (AF). METHODS Eight-week-old male C57BL/6 mice were subcutaneously infused with either vehicle or AngII (2.0 mg/kg/day) for 2 weeks. Water or RKT at a dose of 1000 mg/kg/day were orally administered once daily for 2 weeks. Morphological, histological, and biochemical analyses were performed. AF was induced either by transesophageal burst pacing in vivo or by burst/extrastimuli in isolated perfused hearts using a Langendorff apparatus. RESULTS RKT at a dose of 1000 mg/kg/day for 2 weeks attenuated atrial interstitial fibrosis and profibrotic and proinflammatory signals induced by continuous infusion of AngII. RKT attenuated AngII-induced enhanced vulnerability to AF in in vivo experiments and in isolated perfused hearts. Atractylodin, an active component of RKT, exhibited antifibrotic activity comparable to that of RKT. RKT reversed AngII-induced suppression of sirtuin 1 (Sirt1) translocation to the nuclei. RKT suppressed AngII-induced phosphorylation of IκB, overexpression of p53, and cellular apoptotic signals and apoptosis. All of the antagonizing effects of RKT against AngII were attenuated by a concomitant treatment with a growth hormone secretagogue receptor (GHSR)-inhibitor. CONCLUSION Our results demonstrated that RKT prevented atrial fibrosis and attenuated enhanced vulnerability to AF induced by AngII. The results also suggested that potentiating the GHSR-Sirt1 pathway is involved in these processes.
Collapse
|
24
|
Chen Y, Wang H, Zhang Y, Wang Z, Liu S, Cui L. Pretreatment of ghrelin protects H9c2 cells against hypoxia/reoxygenation-induced cell death via PI3K/AKT and AMPK pathways. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:2179-2187. [PMID: 31159591 DOI: 10.1080/21691401.2019.1620253] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Ghrelin has been widely recognized as a key peptide in the cardiovascular system. This study detected the potential of ghrelin in MI management and tried to decode one of the possible underlying mechanisms. H9c2 cells were pretreated with ghrelin and were subjected to hypoxia/reoxygenation (H/R). CCK-8, flow cytometry, Western blot and LDH analysis were conducted to assess the changes in cell survival. LY294002 and Compound C were used to treat H9c2 cells for blocking PI3K/AKT and AMPK pathways, respectively. Ghrelin expression in H9c2 cells was suppressed by siRNA-mediated silencing to see the effects of endogenous ghrelin. We found that, following H/R, H9c2 cells viability was decreased, CyclinD1 and CDK4 were down-regulated, apoptosis was induced, the release of LDH was enhanced, and the expression levels of Cox-2 and iNOS were up-regulated. Ghrelin protected H9c2 cells against H/R induced these alterations. Besides, ghrelin activated PI3K/AKT and AMPK pathways even in H/R-stimulated cells. The protective effects of ghrelin against H/R-induced cell damage were all attenuated by the addition of LY294002 or Compound C. Moreover, endogenous inhibition of ghrelin significantly induced cell death of H9c2 cells. In conclusion, this study demonstrated that ghrelin pretreatment protected H9c2 cells against H/R-induced cell damage, possibly via PI3K/AKT and AMPK pathways.
Collapse
Affiliation(s)
- Yanbo Chen
- a Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong University , Jinan , Shandong , China.,b Department of Cardiology, Weifang People's Hospital , Weifang , Shandong , China
| | - Honggang Wang
- c Department of Clinical Laboratory, Weifang People's Hospital , Weifang , Shandong , China
| | - Yong Zhang
- b Department of Cardiology, Weifang People's Hospital , Weifang , Shandong , China
| | - Zhengping Wang
- d Department of Ultrasonography, Weifang People's Hospital , Weifang , Shandong , China
| | - Shanshan Liu
- b Department of Cardiology, Weifang People's Hospital , Weifang , Shandong , China
| | - Lianqun Cui
- a Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong University , Jinan , Shandong , China
| |
Collapse
|
25
|
Ghrelin Derangements in Idiopathic Dilated Cardiomyopathy: Impact of Myocardial Disease Duration and Left Ventricular Ejection Fraction. J Clin Med 2019; 8:jcm8081152. [PMID: 31375017 PMCID: PMC6723091 DOI: 10.3390/jcm8081152] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 07/29/2019] [Accepted: 07/31/2019] [Indexed: 11/30/2022] Open
Abstract
Background: Ghrelin may exert positive effects on cardiac structure and function in heart failure (HF) patients. Methods: We assessed ghrelin levels in 266 dilated cardiomyopathy (DCM) patients and in 200 age, gender and body mass index (BMI) matched controls. Further, we evaluated the expression of ghrelin and growth hormone secretagogue-receptor (GHSR) in the myocardium of 41 DCM patients and in 11 controls. Results: DCM patients had significantly lower levels of total, acylated and unacylated ghrelin when compared to controls (p < 0.05 for all). In controls, we observed a negative correlation of ghrelin with age, male gender and BMI. These correlations were lost in the DCM group, except for male gender. Total ghrelin was higher in patients with more recent diagnosis when compared to patients with longer duration of the DCM (p = 0.033). Further, total ghrelin was higher in patients with lower left ventricular systolic function (<40% LVEF, vs. 40% ≤ LVEF < 49% vs. LVEF ≥ 50%: 480.8, vs. 429.7, vs. 329.5 pg/mL, respectively, p = 0.05). Ghrelin prepropeptide was expressed more in DCM patients than in controls (p = 0.0293) while GHSR was expressed less in DCM patients (p < 0.001). Furthermore, ghrelin showed an inverse correlation with its receptor (ρ = −0.406, p = 0.009), and this receptor showed a significant inverse correlation with Interleukin-1β (ρ = −0.422, p = 0.0103). Conclusion: DCM duration and severity are accompanied by alterations in the ghrelin–GHSR system.
Collapse
|
26
|
Sakata N, Yoshimatsu G, Kodama S. Development and Characteristics of Pancreatic Epsilon Cells. Int J Mol Sci 2019; 20:ijms20081867. [PMID: 31014006 PMCID: PMC6514973 DOI: 10.3390/ijms20081867] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 04/11/2019] [Accepted: 04/12/2019] [Indexed: 12/19/2022] Open
Abstract
Pancreatic endocrine cells expressing the ghrelin gene and producing the ghrelin hormone were first identified in 2002. These cells, named ε cells, were recognized as the fifth type of endocrine cells. Differentiation of ε cells is induced by various transcription factors, including Nk2 homeobox 2, paired box proteins Pax-4 and Pax6, and the aristaless-related homeobox. Ghrelin is generally considered to be a "hunger hormone" that stimulates the appetite and is produced mainly by the stomach. Although the population of ε cells is small in adults, they play important roles in regulating other endocrine cells, especially β cells, by releasing ghrelin. However, the roles of ghrelin in β cells are complex. Ghrelin contributes to increased blood glucose levels by suppressing insulin release from β cells and is also involved in the growth and proliferation of β cells and the prevention of β cell apoptosis. Despite increasing evidence and clarification of the mechanisms of ε cells over the last 20 years, many questions remain to be answered. In this review, we present the current evidence for the participation of ε cells in differentiation and clarify their characteristics by focusing on the roles of ghrelin.
Collapse
Affiliation(s)
- Naoaki Sakata
- Department of Regenerative Medicine and Transplantation, Faculty of Medicine, Fukuoka University, Fukuoka 814-0180, Japan.
- Center for Regenerative Medicine, Fukuoka University Hospital, Fukuoka 814-0180, Japan.
| | - Gumpei Yoshimatsu
- Department of Regenerative Medicine and Transplantation, Faculty of Medicine, Fukuoka University, Fukuoka 814-0180, Japan.
- Center for Regenerative Medicine, Fukuoka University Hospital, Fukuoka 814-0180, Japan.
| | - Shohta Kodama
- Department of Regenerative Medicine and Transplantation, Faculty of Medicine, Fukuoka University, Fukuoka 814-0180, Japan.
- Center for Regenerative Medicine, Fukuoka University Hospital, Fukuoka 814-0180, Japan.
| |
Collapse
|
27
|
Sullivan R, Randhawa VK, Stokes A, Wu D, Lalonde T, Kiaii B, Luyt L, Wisenberg G, Dhanvantari S. Dynamics of the Ghrelin/Growth Hormone Secretagogue Receptor System in the Human Heart Before and After Cardiac Transplantation. J Endocr Soc 2019; 3:748-762. [PMID: 30937420 PMCID: PMC6438351 DOI: 10.1210/js.2018-00393] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 02/11/2019] [Indexed: 12/14/2022] Open
Abstract
Currently, the early preclinical detection of left ventricular dysfunction is difficult because biomarkers are not specific for the cardiomyopathic process. The underlying molecular mechanisms leading to heart failure remain elusive, highlighting the need for identification of cardiac-specific markers. The growth hormone secretagogue receptor (GHSR) and its ligand ghrelin are present in cardiac tissue and are known to contribute to myocardial energetics. Here, we examined tissue ghrelin-GHSR levels as specific markers of cardiac dysfunction in patients who underwent cardiac transplantation. Samples of cardiac tissue were obtained from 10 patients undergoing cardiac transplant at the time of organ harvesting and during serial posttransplant biopsies. Quantitative fluorescence microscopy using a fluorescent ghrelin analog was used to measure levels of GHSR, and immunofluorescence was used to measure levels of ghrelin, B-type natriuretic peptide (BNP), and tissue markers of cardiomyocyte contractility and growth. GHSR and ghrelin expression levels were highly variable in the explanted heart, less in the grafted heart biopsies. GHSR and ghrelin were strongly positively correlated, and both markers were negatively correlated with left ventricular ejection fraction. Ghrelin had stronger positive correlations than BNP with the signaling markers for contractility and growth. These data suggest that GHSR-ghrelin have potential use as an integrated marker of cardiac dysfunction. Interestingly, tissue ghrelin appeared to be a more sensitive indicator than BNP to the biochemical processes that are characteristic of heart failure. This work allows for further use of ghrelin-GHSR to interrogate cardiac-specific biochemical mechanisms in preclinical stages of heart failure (HF).
Collapse
Affiliation(s)
- Rebecca Sullivan
- Pathology and Laboratory Medicine, Western University, London, Ontario, Canada
| | - Varinder K Randhawa
- Cardiac Imaging Research, Lawson Health Research Institute, London, Ontario, Canada
| | - Anne Stokes
- Metabolism and Diabetes, Lawson Health Research Institute, London, Ontario, Canada
| | - Derek Wu
- Pathology and Laboratory Medicine, Western University, London, Ontario, Canada
| | - Tyler Lalonde
- Chemistry, Western University, London, Ontario, Canada
| | - Bob Kiaii
- Cardiac Surgery, Western University, London, Ontario, Canada
| | - Leonard Luyt
- Chemistry, Western University, London, Ontario, Canada
- Imaging Program, Lawson Health Research Institute, London, Ontario, Canada
- Department of Oncology, London Regional Cancer Program, Western University, London, Ontario, Canada
| | - Gerald Wisenberg
- Imaging Program, Lawson Health Research Institute, London, Ontario, Canada
- Medical Biophysics, Western University, London, Ontario, Canada
| | - Savita Dhanvantari
- Pathology and Laboratory Medicine, Western University, London, Ontario, Canada
- Metabolism and Diabetes, Lawson Health Research Institute, London, Ontario, Canada
- Imaging Program, Lawson Health Research Institute, London, Ontario, Canada
- Medical Biophysics, Western University, London, Ontario, Canada
| |
Collapse
|
28
|
Franco Machado J, Silva RD, Melo R, G Correia JD. Less Exploited GPCRs in Precision Medicine: Targets for Molecular Imaging and Theranostics. Molecules 2018; 24:E49. [PMID: 30583594 PMCID: PMC6337414 DOI: 10.3390/molecules24010049] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Revised: 12/07/2018] [Accepted: 12/09/2018] [Indexed: 12/18/2022] Open
Abstract
Precision medicine relies on individually tailored therapeutic intervention taking into account individual variability. It is strongly dependent on the availability of target-specific drugs and/or imaging agents that recognize molecular targets and patient-specific disease mechanisms. The most sensitive molecular imaging modalities, Single Photon Emission Computed Tomography (SPECT) and Positron Emission Tomography (PET), rely on the interaction between an imaging radioprobe and a target. Moreover, the use of target-specific molecular tools for both diagnostics and therapy, theranostic agents, represent an established methodology in nuclear medicine that is assuming an increasingly important role in precision medicine. The design of innovative imaging and/or theranostic agents is key for further accomplishments in the field. G-protein-coupled receptors (GPCRs), apart from being highly relevant drug targets, have also been largely exploited as molecular targets for non-invasive imaging and/or systemic radiotherapy of various diseases. Herein, we will discuss recent efforts towards the development of innovative imaging and/or theranostic agents targeting selected emergent GPCRs, namely the Frizzled receptor (FZD), Ghrelin receptor (GHSR-1a), G protein-coupled estrogen receptor (GPER), and Sphingosine-1-phosphate receptor (S1PR). The pharmacological and clinical relevance will be highlighted, giving particular attention to the studies on the synthesis and characterization of targeted molecular imaging agents, biological evaluation, and potential clinical applications in oncology and non-oncology diseases. Whenever relevant, supporting computational studies will be also discussed.
Collapse
Affiliation(s)
- João Franco Machado
- Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, CTN, Estrada Nacional 10 (km 139,7), 2695-066 Bobadela LRS, Portugal.
- Centro de Química Estrutural, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisboa, Portugal.
| | - Rúben D Silva
- Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, CTN, Estrada Nacional 10 (km 139,7), 2695-066 Bobadela LRS, Portugal.
| | - Rita Melo
- Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, CTN, Estrada Nacional 10 (km 139,7), 2695-066 Bobadela LRS, Portugal.
- Center for Neuroscience and Cell Biology; Rua Larga, Faculdade de Medicina, Polo I, 1ºandar, Universidade de Coimbra, 3004-504 Coimbra, Portugal.
| | - João D G Correia
- Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, CTN, Estrada Nacional 10 (km 139,7), 2695-066 Bobadela LRS, Portugal.
| |
Collapse
|
29
|
Abbas A, Yu L, Lalonde T, Wu D, Thiessen JD, Luyt LG, Dhanvantari S. Development and Characterization of an 18F-labeled Ghrelin Peptidomimetic for Imaging the Cardiac Growth Hormone Secretagogue Receptor. Mol Imaging 2018; 17:1536012118809587. [PMID: 30394854 PMCID: PMC6236854 DOI: 10.1177/1536012118809587] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
One-third of patients with heart disease develop heart failure, which is diagnosed
through imaging and detection of circulating biomarkers. Imaging strategies reveal
morphologic and functional changes but fall short of detecting molecular abnormalities
that can lead to heart failure, and circulating biomarkers are not cardiac specific. Thus,
there is critical need for biomarkers that are endogenous to myocardial tissues. The
cardiac growth hormone secretagogue receptor 1a (GHSR1a), which binds the hormone ghrelin,
is a potential biomarker for heart failure. We have synthesized and characterized a novel
ghrelin peptidomimetic tracer, an 18F-labeled analogue of G-7039, for positron
emission tomography (PET) imaging of cardiac GHSR1a. In vitro analysis showed enhanced
serum stability compared to natural ghrelin and significantly increased cellular uptake in
GHSR1a-expressing OVCAR cells. Biodistribution studies in mice showed that tissue uptake
of the tracer was independent of circulating ghrelin levels, and there was negligible
cardiac uptake and high uptake in the liver, intestines, and kidneys. Specificity of
tracer uptake was assessed using ghsr −/− mice; both static and dynamic PET imaging revealed no difference in cardiac
uptake, and there was no significant correlation between cardiac standardized uptake
values and GHSR1a expression. Our study lays the groundwork for further refinement of
peptidomimetic PET tracers targeting cardiac GHSR1a.
Collapse
Affiliation(s)
- Ahmed Abbas
- 1 Department of Medical Biophysics, Western University, London, Ontario, Canada
| | - Lihai Yu
- 2 Department of Chemistry, Western University, London, Ontario, Canada
| | - Tyler Lalonde
- 2 Department of Chemistry, Western University, London, Ontario, Canada
| | - Derek Wu
- 3 Department of Pathology and Laboratory Medicine, Western University, London, Ontario, Canada
| | - Jonathan D Thiessen
- 1 Department of Medical Biophysics, Western University, London, Ontario, Canada.,4 Imaging Research, Lawson Health Research Institute, London, Ontario, Canada
| | - Leonard G Luyt
- 2 Department of Chemistry, Western University, London, Ontario, Canada.,4 Imaging Research, Lawson Health Research Institute, London, Ontario, Canada.,5 Department of oncology, Western University, London, Ontario, Canada
| | - Savita Dhanvantari
- 1 Department of Medical Biophysics, Western University, London, Ontario, Canada.,3 Department of Pathology and Laboratory Medicine, Western University, London, Ontario, Canada.,4 Imaging Research, Lawson Health Research Institute, London, Ontario, Canada.,6 Metabolism/Diabetes, Lawson Health Research Institute, London, Ontario, Canada
| |
Collapse
|
30
|
Posttraumatic Stress Disorder Augments Plasma Triglycerides in TT Homozygotes of rs495225 at Growth Hormone Secretagogue Receptor Gene. Biochem Genet 2018; 57:273-288. [PMID: 30269201 DOI: 10.1007/s10528-018-9890-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 09/25/2018] [Indexed: 10/28/2022]
Abstract
Posttraumatic stress disorder (PTSD) and growth hormone secretagogue receptor (GHSR) were reported to be associated with plasma lipid and glucose levels. However, interplays of PTSD with GHSR on plasma lipid and glucose levels have not been explored yet. This study was to investigate the interplays of PTSD and GHSR rs495225 on plasma glucose and lipid profiles. A total of 709 high school students were recruited at 6 months after the 2008 Wenchuan earthquake. Variants of GHSR rs495225 were identified by polymerase chain reaction-restriction fragment length polymorphism analyses and verified by DNA sequencing. The PTSD Checklist Civilian Version (PCL-C) was used to assess PTSD. There was no significant difference of PTSD prevalence between the TT homozygotes and the C allele carriers. However, the students with PTSD had significantly lower levels of glucose, insulin and homeostasis model assessment of insulin resistance (HOMA-IR) than the students without PTSD in the C allele carriers of GHSR rs495225 after the adjustment for age, gender and body mass index (BMI), but higher levels of TG and TG/HDL-C in the TT homozygotes. Meanwhile, the TT homozygotes had lower levels of HDL-C than the C allele carriers in the students without PTSD, but higher levels of insulin and HOMA-IR in the subjects with PTSD. After the adjustment of age and gender, and additional adjustment for BMI, the results were not changed except the difference of insulin was only a tendency (p = 0.054) after the additional adjustment for BMI. PTSD may augment TG levels and the related lipid ratio TG/HDL-C in the TT homozygotes of GHSR rs495225 but decrease the levels of glucose, insulin and HOMA-IR in the C allele carriers.
Collapse
|
31
|
Napolitano T, Silvano S, Vieira A, Balaji S, Garrido-Utrilla A, Friano ME, Atlija J, Collombat P. Role of ghrelin in pancreatic development and function. Diabetes Obes Metab 2018; 20 Suppl 2:3-10. [PMID: 30230184 DOI: 10.1111/dom.13385] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 05/23/2018] [Accepted: 05/25/2018] [Indexed: 11/28/2022]
Abstract
Ghrelin is a gastric peptide with anabolic functions. It acutely stimulates growth hormone (GH) secretion from the anterior pituitary glands and modulates hypothalamic circuits that control food intake and energy expenditure. Besides its central activity, ghrelin is also involved in the regulation of pancreatic development and physiology. Particularly, several studies highlighted the ability of ghrelin to sustain β-cell viability and proliferation. Furthermore, ghrelin seems to exert inhibitory effects on pancreatic acinar and endocrine secretory functions. Due to its pleiotropic activity on energy metabolism, ghrelin has become a topic of great interest for experimental research focused on type II diabetes and obesity. The aim of this review is to illustrate the complex and not fully understood interplay between ghrelin, pancreas and glucose homeostasis.
Collapse
Affiliation(s)
- Tiziana Napolitano
- Inserm, CNRS, iBV, University of Nice Sophia Antipolis, Nice, France
- iBV, Institut de Biologie Valrose, Centre de Biochimie, University of Nice Sophia Antipolis, Nice Cedex 2, France
| | - Serena Silvano
- Inserm, CNRS, iBV, University of Nice Sophia Antipolis, Nice, France
- iBV, Institut de Biologie Valrose, Centre de Biochimie, University of Nice Sophia Antipolis, Nice Cedex 2, France
| | - Andhira Vieira
- Inserm, CNRS, iBV, University of Nice Sophia Antipolis, Nice, France
- iBV, Institut de Biologie Valrose, Centre de Biochimie, University of Nice Sophia Antipolis, Nice Cedex 2, France
| | - Shruti Balaji
- Inserm, CNRS, iBV, University of Nice Sophia Antipolis, Nice, France
- iBV, Institut de Biologie Valrose, Centre de Biochimie, University of Nice Sophia Antipolis, Nice Cedex 2, France
| | - Anna Garrido-Utrilla
- Inserm, CNRS, iBV, University of Nice Sophia Antipolis, Nice, France
- iBV, Institut de Biologie Valrose, Centre de Biochimie, University of Nice Sophia Antipolis, Nice Cedex 2, France
| | - Marika E Friano
- Inserm, CNRS, iBV, University of Nice Sophia Antipolis, Nice, France
- iBV, Institut de Biologie Valrose, Centre de Biochimie, University of Nice Sophia Antipolis, Nice Cedex 2, France
| | - Josipa Atlija
- Inserm, CNRS, iBV, University of Nice Sophia Antipolis, Nice, France
- iBV, Institut de Biologie Valrose, Centre de Biochimie, University of Nice Sophia Antipolis, Nice Cedex 2, France
| | - Patrick Collombat
- Inserm, CNRS, iBV, University of Nice Sophia Antipolis, Nice, France
- iBV, Institut de Biologie Valrose, Centre de Biochimie, University of Nice Sophia Antipolis, Nice Cedex 2, France
| |
Collapse
|
32
|
Fowkes MM, Lalonde T, Yu L, Dhanvantari S, Kovacs MS, Luyt LG. Peptidomimetic growth hormone secretagogue derivatives for positron emission tomography imaging of the ghrelin receptor. Eur J Med Chem 2018; 157:1500-1511. [PMID: 30282322 DOI: 10.1016/j.ejmech.2018.08.062] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Revised: 08/16/2018] [Accepted: 08/22/2018] [Indexed: 02/06/2023]
Abstract
The ghrelin receptor is a seven-transmembrane (7-TM) receptor known to have an increased level of expression in human carcinoma and heart failure. Recent work has focused on the synthesis of positron emission tomography (PET) probes designed to target and image this receptor for disease diagnosis and staging. However, these probes have been restricted to small-molecule quinalizonones and peptide derivatives of the endogenous ligand ghrelin. We describe the design, synthesis and biological evaluation of a series of 4-fluorobenzoylated growth hormone secretagogues (GHSs) derived from peptidic (GHRP-1, GHPR-2 and GHRP-6) and peptidomimetic (G-7039, [1-Nal4]G-7039 and ipamorelin) families in order to test locations for the insertion of fluorine-18 for PET imaging. The peptidomimetic G-7039 was found to be the most suitable for 18F-radiolabelling as its non-radioactive 4-fluorobenzoylated analogue ([1-Nal4,Lys5(4-FB)]G-7039), had both a high binding affinity (IC50 = 69 nM) and promising in vitro efficacy (EC50 = 1.1 nM). Prosthetic group radiolabelling of the precursor compound [1-Nal4]G-7039 using N-succinimidyl-4-[18F]fluorobenzoate ([18F]SFB) delivered the PET probe [1-Nal4,Lys5(4-[18F]-FB)]G-7039 in an average decay-corrected radiochemical yield of 48%, a radio-purity ≥ 99% and an average molar activity of >34 GBq/μmol. This compound could be investigated as a PET probe for the detection of diseases that are characterised by overexpression of the ghrelin receptor.
Collapse
Affiliation(s)
- Milan M Fowkes
- Department of Chemistry, Western University, 1151 Richmond Street, London, Ontario, N6A 5B7, Canada; London Regional Cancer Program, Lawson Health Research Institute, 790 Commissioners Road East, London, Ontario, N6A 4L6, Canada
| | - Tyler Lalonde
- Department of Chemistry, Western University, 1151 Richmond Street, London, Ontario, N6A 5B7, Canada; London Regional Cancer Program, Lawson Health Research Institute, 790 Commissioners Road East, London, Ontario, N6A 4L6, Canada
| | - Lihai Yu
- Department of Chemistry, Western University, 1151 Richmond Street, London, Ontario, N6A 5B7, Canada; London Regional Cancer Program, Lawson Health Research Institute, 790 Commissioners Road East, London, Ontario, N6A 4L6, Canada
| | - Savita Dhanvantari
- Imaging Program, Lawson Health Research Institute, 268 Grosvenor Street, London, Ontario, N6A 4V2, Canada
| | - Michael S Kovacs
- Imaging Program, Lawson Health Research Institute, 268 Grosvenor Street, London, Ontario, N6A 4V2, Canada
| | - Leonard G Luyt
- Department of Chemistry, Western University, 1151 Richmond Street, London, Ontario, N6A 5B7, Canada; London Regional Cancer Program, Lawson Health Research Institute, 790 Commissioners Road East, London, Ontario, N6A 4L6, Canada; Imaging Program, Lawson Health Research Institute, 268 Grosvenor Street, London, Ontario, N6A 4V2, Canada.
| |
Collapse
|
33
|
Ghrelin Ameliorates Angiotensin II-Induced Myocardial Fibrosis by Upregulating Peroxisome Proliferator-Activated Receptor Gamma in Young Male Rats. BIOMED RESEARCH INTERNATIONAL 2018; 2018:9897581. [PMID: 30175152 PMCID: PMC6098901 DOI: 10.1155/2018/9897581] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 07/06/2018] [Accepted: 07/25/2018] [Indexed: 01/07/2023]
Abstract
Angiotensin (Ang) II contributes to the formation and development of myocardial fibrosis. Ghrelin, a gut peptide, has demonstrated beneficial effects against cardiovascular disease. In the present study, we explored the effect and related mechanism of Ghrelin on myocardial fibrosis in Ang II-infused rats. Adult Sprague-Dawley (SD) rats were divided into 6 groups: Control, Ang II (200ng/kg/min, microinfusion), Ang II+Ghrelin (100 μg/kg, subcutaneously twice daily), Ang II+Ghrelin+GW9662 (a specific PPAR-γ inhibitor, 1 mg/kg/d, orally), Ang II+GW9662, and Ghrelin for 4 wks. In vitro, adult rat cardiac fibroblasts (CFs) were pretreated with or without Ghrelin, Ghrelin+GW9662, or anti-Transforming growth factor (TGF)-β1 antibody and then stimulated with or without Ang II (100 nmol/L) for 24 h. Ang II infusion significantly increased myocardial fibrosis, expression of collagen I, collagen III, and TGF-β1, as well as TGF-β1 downstream proteins p-Smad2, p-Smad3, TRAF6, and p-TAK1 (all p<0.05). Ghrelin attenuated these effects. Similar results were seen in Ang II-stimulated rat cardiac fibroblasts in vitro. In addition, Ghrelin upregulated PPAR-γ expression in vivo and in vitro, and treatment with GW9662 counteracted the effects of Ghrelin. In conclusion, Ghrelin ameliorated Ang II-induced myocardial fibrosis by upregulating PPAR-γ and in turn inhibiting TGF-β1signaling.
Collapse
|
34
|
Sullivan R, McGirr R, Hu S, Tan A, Wu D, Charron C, Lalonde T, Arany E, Chakrabarti S, Luyt L, Dhanvantari S. Changes in the Cardiac GHSR1a-Ghrelin System Correlate With Myocardial Dysfunction in Diabetic Cardiomyopathy in Mice. J Endocr Soc 2017; 2:178-189. [PMID: 29450407 PMCID: PMC5799831 DOI: 10.1210/js.2017-00433] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 12/19/2017] [Indexed: 01/16/2023] Open
Abstract
Ghrelin and its receptor, the growth hormone secretagogue receptor 1a (GHSR1a), are present in cardiac tissue. Activation of GHSR1a by ghrelin promotes cardiomyocyte contractility and survival, and changes in myocardial GHSR1a and circulating ghrelin track with end-stage heart failure, leading to the hypothesis that GHSR1a is a biomarker for heart failure. We hypothesized that GHSR1a could also be a biomarker for diabetic cardiomyopathy (DCM). We used two models of streptozotocin (STZ)-induced DCM: group 1, adult mice treated with 35 mg/kg STZ for 3 days; and group 2, neonatal mice treated with 70 mg/kg STZ at days 2 and 5 after birth. In group 1, mild fasting hyperglycemia (11 mM) was first detected 8 weeks after the last injection, and in group 2, severe fasting hyperglycemia (20 mM) was first detected 1 to 3 weeks after the last injection. In group 1, left ventricular function was slightly impaired as measured by echocardiography, and Western blot analysis showed a significant decrease in myocardial GHSR1a. In group 2, GHSR1a levels were also decreased as assessed by Cy5-ghrelin(1–19) fluorescence microscopy, and there was a significant negative correlation between GHSR1a levels and glucose tolerance. There were significant positive correlations between GHSR1a and ghrelin and between GHSR1a and sarcoplasmic reticulum Ca2+-ATPase 2a (SERCA2a), a marker for contractility, but not between GHSR1a and B-type natriuretic peptide, a marker for heart failure. We conclude that the subclinical stage of DCM is accompanied by alterations in the myocardial ghrelin-GHSR1a system, suggesting the possibility of a biomarker for DCM.
Collapse
Affiliation(s)
- Rebecca Sullivan
- Imaging Research, Lawson Health Research Institute, London, Ontario N6A 4V2, Canada.,Department of Pathology and Laboratory Medicine, Western University, London, Ontario N6A 4V2, Canada
| | - Rebecca McGirr
- Imaging Research, Lawson Health Research Institute, London, Ontario N6A 4V2, Canada
| | - Shirley Hu
- Department of Physiology and Pharmacology, Western University, London, Ontario N6A 3K7, Canada
| | - Alice Tan
- Department of Pathology and Laboratory Medicine, Western University, London, Ontario N6A 4V2, Canada
| | - Derek Wu
- Department of Pathology and Laboratory Medicine, Western University, London, Ontario N6A 4V2, Canada
| | - Carlie Charron
- Department of Chemistry, Western University, London, Ontario N6A 5B7, Canada
| | - Tyler Lalonde
- Department of Chemistry, Western University, London, Ontario N6A 5B7, Canada
| | - Edith Arany
- Department of Pathology and Laboratory Medicine, Western University, London, Ontario N6A 4V2, Canada
| | - Subrata Chakrabarti
- Department of Pathology and Laboratory Medicine, Western University, London, Ontario N6A 4V2, Canada
| | - Leonard Luyt
- Department of Chemistry, Western University, London, Ontario N6A 5B7, Canada.,Departments of Oncology and Medical Imaging, Western University, London, Ontario N6A 4L6, Canada.,London Regional Cancer Program, Lawson Health Research Institute, London, Ontario N6A 4V2, Canada
| | - Savita Dhanvantari
- Imaging Research, Lawson Health Research Institute, London, Ontario N6A 4V2, Canada.,Department of Pathology and Laboratory Medicine, Western University, London, Ontario N6A 4V2, Canada.,Department of Medical Biophysics, Western University, London, Ontario N6A 5C1, Canada
| |
Collapse
|
35
|
Crotta K, Casnici C, Tonna N, Lattuada D, Bianco F, Marelli O. Characterization of a Monoclonal Antibody Specific for the Growth Hormone Secretagogue Receptor. Monoclon Antib Immunodiagn Immunother 2017; 36:37-43. [PMID: 28409695 DOI: 10.1089/mab.2016.0053] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Ghrelin is an orexigenic peptide hormone that primarily regulates growth hormone secretion, food intake, and energy homeostasis. It has been shown to also play a role in numerous higher brain functions, such as the regulation of inflammation and cell proliferation. Ghrelin is the endogenous ligand of the growth hormone secretagogue receptor (GHSR), a G-protein-coupled receptor highly expressed in brain and detectable in some peripheral tissues. The wide distribution of ghrelin receptor and the number of tissues and cell types known to respond to ghrelin suggest that a number of systems may be affected by treatment with this hormone or its analogues. In this study, we characterized a new GHSR specific monoclonal antibody recognizing specifically the ghrelin receptor. This could be a useful tool for immunoassays aimed at obtaining insights into the physiological and pathological significance of the GHSR/ghrelin system.
Collapse
Affiliation(s)
- Katia Crotta
- 1 Department of Medical Biotechnologies and Translational Medicine, School of Medicine, University of Milan , Milan, Italy
| | - Claudia Casnici
- 1 Department of Medical Biotechnologies and Translational Medicine, School of Medicine, University of Milan , Milan, Italy .,2 Ferdinando Santarelli Foundation , Milan, Italy
| | | | - Donatella Lattuada
- 1 Department of Medical Biotechnologies and Translational Medicine, School of Medicine, University of Milan , Milan, Italy
| | - Fabio Bianco
- 2 Ferdinando Santarelli Foundation , Milan, Italy .,4 Sanipedia srl , OpenZone, Bresso, Italy
| | - Ornella Marelli
- 1 Department of Medical Biotechnologies and Translational Medicine, School of Medicine, University of Milan , Milan, Italy
| |
Collapse
|
36
|
Colldén G, Tschöp MH, Müller TD. Therapeutic Potential of Targeting the Ghrelin Pathway. Int J Mol Sci 2017; 18:ijms18040798. [PMID: 28398233 PMCID: PMC5412382 DOI: 10.3390/ijms18040798] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 04/03/2017] [Accepted: 04/06/2017] [Indexed: 02/07/2023] Open
Abstract
Ghrelin was discovered in 1999 as the endogenous ligand of the growth-hormone secretagogue receptor 1a (GHSR1a). Since then, ghrelin has been found to exert a plethora of physiological effects that go far beyond its initial characterization as a growth hormone (GH) secretagogue. Among the numerous well-established effects of ghrelin are the stimulation of appetite and lipid accumulation, the modulation of immunity and inflammation, the stimulation of gastric motility, the improvement of cardiac performance, the modulation of stress, anxiety, taste sensation and reward-seeking behavior, as well as the regulation of glucose metabolism and thermogenesis. Due to a variety of beneficial effects on systems’ metabolism, pharmacological targeting of the endogenous ghrelin system is widely considered a valuable approach to treat metabolic complications, such as chronic inflammation, gastroparesis or cancer-associated anorexia and cachexia. The aim of this review is to discuss and highlight the broad pharmacological potential of ghrelin pathway modulation for the treatment of anorexia, cachexia, sarcopenia, cardiopathy, neurodegenerative disorders, renal and pulmonary disease, gastrointestinal (GI) disorders, inflammatory disorders and metabolic syndrome.
Collapse
Affiliation(s)
- Gustav Colldén
- Institute for Diabetes and Obesity & Helmholtz Diabetes Center, Helmholtz Zentrum München German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany.
| | - Matthias H Tschöp
- Institute for Diabetes and Obesity & Helmholtz Diabetes Center, Helmholtz Zentrum München German Research Center for Environmental Health (GmbH), 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, Helmholtz Zentrum München German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany.
- Institute for Diabetes and Obesity (IDO), Business Campus Garching-Hochbrück, Parkring 13, 85748 Garching, Germany.
| |
Collapse
|
37
|
Abstract
Ghrelin is a small peptide released primarily from the stomach. It is a potent stimulator of growth hormone secretion from the pituitary gland and is well known for its regulation of metabolism and appetite. There is also a strong relationship between ghrelin and the cardiovascular system. Ghrelin receptors are present throughout the heart and vasculature and have been linked with molecular pathways, including, but not limited to, the regulation of intracellular calcium concentration, inhibition of proapoptotic cascades, and protection against oxidative damage. Ghrelin shows robust cardioprotective effects including enhancing endothelial and vascular function, preventing atherosclerosis, inhibiting sympathetic drive, and decreasing blood pressure. After myocardial infarction, exogenous administration of ghrelin preserves cardiac function, reduces the incidence of fatal arrhythmias, and attenuates apoptosis and ventricular remodeling, leading to improvements in heart failure. It ameliorates cachexia in end-stage congestive heart failure patients and has shown clinical benefit in pulmonary hypertension. Nonetheless, since ghrelin's discovery is relatively recent, there remains a substantial amount of research needed to fully understand its clinical significance in cardiovascular disease.
Collapse
|
38
|
Cardioprotection Effects of Sevoflurane by Regulating the Pathway of Neuroactive Ligand-Receptor Interaction in Patients Undergoing Coronary Artery Bypass Graft Surgery. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2017; 2017:3618213. [PMID: 28348638 PMCID: PMC5350303 DOI: 10.1155/2017/3618213] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Revised: 02/10/2017] [Accepted: 02/19/2017] [Indexed: 01/17/2023]
Abstract
This study was designed to identify attractor modules and further reveal the potential biological processes involving in sevoflurane-induced anesthesia in patients treated with coronary artery bypass graft (CABG) surgery. Microarray profile data (ID: E-GEOD-4386) on atrial samples obtained from patients receiving anesthetic gas sevoflurane prior to and following CABG procedure were downloaded from EMBL-EBI database for further analysis. Protein-protein interaction (PPI) networks of baseline and sevoflurane groups were inferred and reweighted according to Spearman correlation coefficient (SCC), followed by systematic modules inference using clique-merging approach. Subsequently, attract method was utilized to explore attractor modules. Finally, pathway enrichment analyses for genes in the attractor modules were implemented to illuminate the biological processes in sevoflurane group. Using clique-merging approach, 27 and 36 modules were obtained from the PPI networks of baseline and sevoflurane-treated samples, respectively. By comparing with the baseline condition, 5 module pairs with the same gene composition were identified. Subsequently, 1 out of 5 modules was identified as an attractor based on attract method. Additionally, pathway analysis indicated that genes in the attractor module were associated with neuroactive ligand-receptor interaction. Accordingly, sevoflurane might exert important functions in cardioprotection in patients following CABG, partially through regulating the pathway of neuroactive ligand-receptor interaction.
Collapse
|
39
|
GHSR-1a is a novel pro-angiogenic and anti-remodeling target in rats after myocardial infarction. Eur J Pharmacol 2016; 788:218-225. [DOI: 10.1016/j.ejphar.2016.06.032] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 06/10/2016] [Accepted: 06/21/2016] [Indexed: 12/19/2022]
|
40
|
Junqueira A, Cicogna AC, Engel LE, Aldá MA, de Tomasi LC, Giuffrida R, Giometti IC, Freire APCF, Aguiar AF, Pacagnelli FL. Effects of Growth Hormone on Cardiac Remodeling During Resistance Training in Rats. Arq Bras Cardiol 2015; 106:18-25. [PMID: 26647722 PMCID: PMC4728591 DOI: 10.5935/abc.20160003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 09/04/2015] [Indexed: 11/30/2022] Open
Abstract
Background Although the beneficial effects of resistance training (RT) on the cardiovascular
system are well established, few studies have investigated the effects of the
chronic growth hormone (GH) administration on cardiac remodeling during an RT
program. Objective To evaluate the effects of GH on the morphological features of cardiac remodeling
and Ca2+ transport gene expression in rats submitted to RT. Methods Male Wistar rats were divided into 4 groups (n = 7 per group): control (CT), GH,
RT and RT with GH (RTGH). The dose of GH was 0.2 IU/kg every other day for 30
days. The RT model used was the vertical jump in water (4 sets of 10 jumps, 3
bouts/wk) for 30 consecutive days. After the experimental period, the following
variables were analyzed: final body weight (FBW), left ventricular weight (LVW),
LVW/FBW ratio, cardiomyocyte cross-sectional area (CSA), collagen fraction,
creatine kinase muscle-brain fraction (CK-MB) and gene expressions of SERCA2a,
phospholamban (PLB) and ryanodine (RyR). Results There was no significant (p > 0.05) difference among groups for FBW, LVW,
LVW/FBW ratio, cardiomyocyte CSA, and SERCA2a, PLB and RyR gene expressions. The
RT group showed a significant (p < 0.05) increase in collagen fraction compared
to the other groups. Additionally, the trained groups (RT and RTGH) had greater
CK-MB levels compared to the untrained groups (CT and GH). Conclusion GH may attenuate the negative effects of RT on cardiac remodeling by counteracting
the increased collagen synthesis, without affecting the gene expression that
regulates cardiac Ca2+ transport.
Collapse
|
41
|
Zhang L, Gao L, Bi HM. New perspectives of incretin research. Shijie Huaren Xiaohua Zazhi 2015; 23:4473-4481. [DOI: 10.11569/wcjd.v23.i28.4473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Incretins are a group of hormones released into the blood stream by gastrointestinal cells after food stimulation, including glucagon like peptide and glucose-dependent insulinotropic polypeptide (GIP), which can promote insulin secretion and regulate blood sugar. GLP-1 is secreted by intestinal L cells, and fulfills its function through the specific GLP-1 receptor (GLP-1R). GLP-1R is widely distributed in the pancreas and non-pancreatic tissues such as central nervous system, gastrointestinal tract, cardiovascular system, lungs, and kidneys. In recent years, GLP-1 drugs have been used for the treatment of diabetes, but it has attracted more interest because of its beneficiary effects on β cell function, weight reduction, endothelial function, and Alzheimer's disease. This article will review the recent progress in research of GLP-1 with regards to its synthesis and secretion, its effects on taste and Alzheimer's disease, and its relationship with other gastrointestinal hormones, with an aim to illuminate the future clinical use and research of GLP-1.
Collapse
|
42
|
Müller TD, Nogueiras R, Andermann ML, Andrews ZB, Anker SD, Argente J, Batterham RL, Benoit SC, Bowers CY, Broglio F, Casanueva FF, D'Alessio D, Depoortere I, Geliebter A, Ghigo E, Cole PA, Cowley M, Cummings DE, Dagher A, Diano S, Dickson SL, Diéguez C, Granata R, Grill HJ, Grove K, Habegger KM, Heppner K, Heiman ML, Holsen L, Holst B, Inui A, Jansson JO, Kirchner H, Korbonits M, Laferrère B, LeRoux CW, Lopez M, Morin S, Nakazato M, Nass R, Perez-Tilve D, Pfluger PT, Schwartz TW, Seeley RJ, Sleeman M, Sun Y, Sussel L, Tong J, Thorner MO, van der Lely AJ, van der Ploeg LHT, Zigman JM, Kojima M, Kangawa K, Smith RG, Horvath T, Tschöp MH. Ghrelin. Mol Metab 2015; 4:437-60. [PMID: 26042199 PMCID: PMC4443295 DOI: 10.1016/j.molmet.2015.03.005] [Citation(s) in RCA: 771] [Impact Index Per Article: 77.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Revised: 03/11/2015] [Accepted: 03/11/2015] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The gastrointestinal peptide hormone ghrelin was discovered in 1999 as the endogenous ligand of the growth hormone secretagogue receptor. Increasing evidence supports more complicated and nuanced roles for the hormone, which go beyond the regulation of systemic energy metabolism. SCOPE OF REVIEW In this review, we discuss the diverse biological functions of ghrelin, the regulation of its secretion, and address questions that still remain 15 years after its discovery. MAJOR CONCLUSIONS In recent years, ghrelin has been found to have a plethora of central and peripheral actions in distinct areas including learning and memory, gut motility and gastric acid secretion, sleep/wake rhythm, reward seeking behavior, taste sensation and glucose metabolism.
Collapse
Affiliation(s)
- T D Müller
- Institute for Diabetes and Obesity, Helmholtz Zentrum München, München, Germany
| | - R Nogueiras
- Department of Physiology, Centro de Investigación en Medicina Molecular y Enfermedades Crónicas, University of Santiago de Compostela (CIMUS)-Instituto de Investigación Sanitaria (IDIS)-CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, Spain
| | - M L Andermann
- Division of Endocrinology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Z B Andrews
- Department of Physiology, Faculty of Medicine, Monash University, Melbourne, Victoria, Australia
| | - S D Anker
- Applied Cachexia Research, Department of Cardiology, Charité Universitätsmedizin Berlin, Germany
| | - J Argente
- Department of Pediatrics and Pediatric Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain ; Department of Pediatrics, Universidad Autónoma de Madrid and CIBER Fisiopatología de la obesidad y nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - R L Batterham
- Centre for Obesity Research, University College London, London, United Kingdom
| | - S C Benoit
- Metabolic Disease Institute, Division of Endocrinology, Department of Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - C Y Bowers
- Tulane University Health Sciences Center, Endocrinology and Metabolism Section, Peptide Research Section, New Orleans, LA, USA
| | - F Broglio
- Division of Endocrinology, Diabetes and Metabolism, Dept. of Medical Sciences, University of Torino, Torino, Italy
| | - F F Casanueva
- Department of Medicine, Santiago de Compostela University, Complejo Hospitalario Universitario de Santiago (CHUS), CIBER de Fisiopatologia Obesidad y Nutricion (CB06/03), Instituto Salud Carlos III, Santiago de Compostela, Spain
| | - D D'Alessio
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA
| | - I Depoortere
- Translational Research Center for Gastrointestinal Disorders, University of Leuven, Leuven, Belgium
| | - A Geliebter
- New York Obesity Nutrition Research Center, Department of Medicine, St Luke's-Roosevelt Hospital Center, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - E Ghigo
- Department of Pharmacology & Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - P A Cole
- Monash Obesity & Diabetes Institute, Monash University, Clayton, Victoria, Australia
| | - M Cowley
- Department of Physiology, Faculty of Medicine, Monash University, Melbourne, Victoria, Australia ; Monash Obesity & Diabetes Institute, Monash University, Clayton, Victoria, Australia
| | - D E Cummings
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - A Dagher
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - S Diano
- Dept of Neurobiology, Yale University School of Medicine, New Haven, CT, USA
| | - S L Dickson
- Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - C Diéguez
- Department of Physiology, School of Medicine, Instituto de Investigacion Sanitaria (IDIS), University of Santiago de Compostela, Spain
| | - R Granata
- Division of Endocrinology, Diabetes and Metabolism, Dept. of Medical Sciences, University of Torino, Torino, Italy
| | - H J Grill
- Department of Psychology, Institute of Diabetes, Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, USA
| | - K Grove
- Department of Diabetes, Obesity and Metabolism, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA
| | - K M Habegger
- Comprehensive Diabetes Center, University of Alabama School of Medicine, Birmingham, AL, USA
| | - K Heppner
- Division of Diabetes, Obesity, and Metabolism, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - M L Heiman
- NuMe Health, 1441 Canal Street, New Orleans, LA 70112, USA
| | - L Holsen
- Departments of Psychiatry and Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - B Holst
- Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen N, Denmark
| | - A Inui
- Department of Psychosomatic Internal Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - J O Jansson
- Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - H Kirchner
- Medizinische Klinik I, Universitätsklinikum Schleswig-Holstein Campus Lübeck, Lübeck, Germany
| | - M Korbonits
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London, Queen Mary University of London, London, UK
| | - B Laferrère
- New York Obesity Research Center, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - C W LeRoux
- Diabetes Complications Research Centre, Conway Institute, University College Dublin, Ireland
| | - M Lopez
- Department of Physiology, Centro de Investigación en Medicina Molecular y Enfermedades Crónicas, University of Santiago de Compostela (CIMUS)-Instituto de Investigación Sanitaria (IDIS)-CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, Spain
| | - S Morin
- Institute for Diabetes and Obesity, Helmholtz Zentrum München, München, Germany
| | - M Nakazato
- Division of Neurology, Respirology, Endocrinology and Metabolism, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Kiyotake, Miyazaki, Japan
| | - R Nass
- Division of Endocrinology and Metabolism, University of Virginia, Charlottesville, VA, USA
| | - D Perez-Tilve
- Department of Internal Medicine, Department of Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - P T Pfluger
- Institute for Diabetes and Obesity, Helmholtz Zentrum München, München, Germany
| | - T W Schwartz
- Department of Neuroscience and Pharmacology, Laboratory for Molecular Pharmacology, The Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | - R J Seeley
- Department of Surgery, University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - M Sleeman
- Department of Physiology, Faculty of Medicine, Monash University, Melbourne, Victoria, Australia
| | - Y Sun
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - L Sussel
- Department of Genetics and Development, Columbia University, New York, NY, USA
| | - J Tong
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA
| | - M O Thorner
- Division of Endocrinology and Metabolism, University of Virginia, Charlottesville, VA, USA
| | - A J van der Lely
- Department of Medicine, Erasmus University MC, Rotterdam, The Netherlands
| | | | - J M Zigman
- Departments of Internal Medicine and Psychiatry, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - M Kojima
- Molecular Genetics, Institute of Life Science, Kurume University, Kurume, Japan
| | - K Kangawa
- National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
| | - R G Smith
- The Scripps Research Institute, Florida Department of Metabolism & Aging, Jupiter, FL, USA
| | - T Horvath
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Section of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - M H Tschöp
- Institute for Diabetes and Obesity, Helmholtz Zentrum München, München, Germany ; Division of Metabolic Diseases, Department of Medicine, Technical University Munich, Munich, Germany
| |
Collapse
|
43
|
Callaghan B, Furness JB. Novel and conventional receptors for ghrelin, desacyl-ghrelin, and pharmacologically related compounds. Pharmacol Rev 2014; 66:984-1001. [PMID: 25107984 DOI: 10.1124/pr.113.008433] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2025] Open
Abstract
The only molecularly identified ghrelin receptor is the growth hormone secretagogue receptor GHSR1a. Its natural ligand, ghrelin, is an acylated peptide whose unacylated counterpart (UAG) is almost inactive at GHSR1a. A truncated, nonfunctional receptor, GHSR1b, derives from the same gene. We have critically evaluated evidence for effects of ghrelin receptor ligands that are not consistent with actions at GHSR1a. Effects of ghrelin are observed in cells or tissues where the expression of GHSR1a is not detectable or after the Ghsr gene has been inactivated. In several, effects of ghrelin are mimicked by UAG, and ghrelin binding is competitively reduced by UAG. Effects in the absence of GHSR1a and sites at which ghrelin and UAG have similar potency suggest the presence of novel nonspecific ghrelin receptors (ghrelin receptor-like receptors [GRLRs]). A third class of receptor, the UAG receptors, at which UAG, but not ghrelin, is an agonist has been proposed. None of the novel receptors, with the exception of the glycoprotein CD36, which accounts for ghrelin action at a limited number of sites, have been identified. GHSR1a and GHSR1b combine with other G protein-coupled receptors to form heterodimers, whose pharmacologies differ from their components. Thus, it is feasible some GRLRs and some UAG receptors are heterodimers. Effects mediated through GRLRs or UAG receptors include adipocyte lipid accumulation, myoblast differentiation, osteoblast proliferation, insulin release, cardioprotection, coronary artery constriction, vascular endothelial cell proliferation, and tumor cell proliferation. The molecular identification and pharmacologic characterization of novel ghrelin receptors are thus important objectives.
Collapse
Affiliation(s)
- Brid Callaghan
- Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Victoria, Australia
| | - John B Furness
- Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
44
|
Yuan MJ, Huang H, Quan L, Tang YH, Wang X, Jiang H, Huang CX. Expression of ghrelin and its receptor in rats after coronary artery ligation. ACTA ACUST UNITED AC 2014; 192-193:1-5. [PMID: 25058156 DOI: 10.1016/j.regpep.2014.07.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Revised: 06/18/2014] [Accepted: 07/14/2014] [Indexed: 11/26/2022]
Abstract
Ghrelin is a novel growth hormone-releasing peptide, which has been shown to exert beneficial effects on cardiac function and ventricular remodeling. The present study aimed to investigate the expression of ghrelin and the growth hormone (GH) secretagogue receptor 1a (GHSR-1a), and the association with cardiac remodeling in rats with myocardial infarction (MI). Twenty-four hours after ligation of the anterior descending artery (LAD), adult male Sprague-Dawley rats were randomized to 3 d, 7 d and 28 d group. Sham animals underwent thoracotomy and pericardiotomy, but not LAD ligation. Expression of both ghrelin and GHSR-1a was assessed by means of immunohistochemistry and real-time PCR. Plasma ghrelin levels were measured by ELISA kit. In addition, cardiac remodeling was assessed by echocardiographic and hemodynamic measurements. Plasma and cardiac expression of ghrelin decreased on days 3, 7 and 28 compared with the sham group (P<0.05). In contrast the GHSR-1a mRNA levels increased during the same days (P<0.05). Decreased positive immunoreaction for ghrelin and increased positive GHSR-1a were also observed in the infarcted heart. Interestingly, plasma ghrelin correlated negatively with left ventricular end-diastolic pressure (r=-0.59, P=0.002) and left ventricular end-diastolic dimension (r=-0.73, P<0.01). The ghrelin system may play an important role regulating cardiac remodeling after MI and present as a potential significant target for pharmacological modulation and treating cardiac remodeling.
Collapse
Affiliation(s)
- Ming-Jie Yuan
- Dept of Cardiology, Wuhan Univ, Renmin Hospital, Wuhan 430060, Hubei Province, PR China
| | - He Huang
- Dept of Cardiology, Wuhan Univ, Renmin Hospital, Wuhan 430060, Hubei Province, PR China
| | - Li Quan
- Dept of Cardiology, Wuhan Univ, Renmin Hospital, Wuhan 430060, Hubei Province, PR China
| | - Yan-Hong Tang
- Dept of Cardiology, Wuhan Univ, Renmin Hospital, Wuhan 430060, Hubei Province, PR China
| | - Xi Wang
- Dept of Cardiology, Wuhan Univ, Renmin Hospital, Wuhan 430060, Hubei Province, PR China
| | - Hong Jiang
- Dept of Cardiology, Wuhan Univ, Renmin Hospital, Wuhan 430060, Hubei Province, PR China
| | - Cong-Xin Huang
- Dept of Cardiology, Wuhan Univ, Renmin Hospital, Wuhan 430060, Hubei Province, PR China.
| |
Collapse
|
45
|
Yuan MJ, Huang H, Huang CX. Potential new role of the GHSR-1a-mediated signaling pathway in cardiac remodeling after myocardial infarction (Review). Oncol Lett 2014; 8:969-971. [PMID: 25120643 PMCID: PMC4114710 DOI: 10.3892/ol.2014.2245] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2013] [Accepted: 05/15/2014] [Indexed: 11/17/2022] Open
Abstract
The gastrointestinal hormone ghrelin has important cardiovascular protective effects, however, its specific mechanisms are not yet completely understood. Recent studies have shown that the ghrelin receptor, growth hormone secretagogue receptor type 1a (GHSR-1a), regulates cell proliferation, apoptosis and inflammation-related signaling pathways. In human aortic endothelial cells, ghrelin activates NO production through AMP-activated protein kinase (AMPK) and Akt activation, and these effects can be blocked by knockdown of GHSR-1a. Obese mice have been found to exhibit an increased GHSR-1a content and expression in the heart, associated with an increase in phosphatidylinositol 3-kinase (PI3K) content and an increase AKT content and phosphorylation. Furthermore, GHSR-1a expression was observed to be increased in heart failure after myocardial infarction (MI) in rats. Given such complexity in GHSR-1a signaling and crosstalk with the AMPK and PI3K/Akt signaling pathways, both of which are well-known factors involved in cardiac remodeling after MI, we speculate that GHSR-1a signaling may play a regulatory role in cardiac protection and hope to identify new drugs targets. However, to date, no direct association between GHSR-1a and cardiac remodeling has been found. Therefore, further studies are required.
Collapse
Affiliation(s)
- Ming-Jie Yuan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - He Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Cong-Xin Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
46
|
Soeki T. Ghrelin and its receptor: The role of the ghrelin signaling system in regulating cardiac function. ACTA ACUST UNITED AC 2014; 192-193:57-8. [DOI: 10.1016/j.regpep.2014.08.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2014] [Revised: 08/21/2014] [Accepted: 08/24/2014] [Indexed: 10/24/2022]
|
47
|
Mitacchione G, Powers JC, Grifoni G, Woitek F, Lam A, Ly L, Settanni F, Makarewich CA, McCormick R, Trovato L, Houser SR, Granata R, Recchia FA. The gut hormone ghrelin partially reverses energy substrate metabolic alterations in the failing heart. Circ Heart Fail 2014; 7:643-51. [PMID: 24855152 DOI: 10.1161/circheartfailure.114.001167] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND The gut-derived hormone ghrelin, especially its acylated form, plays a major role in the regulation of systemic metabolism and exerts also relevant cardioprotective effects; hence, it has been proposed for the treatment of heart failure (HF). We tested the hypothesis that ghrelin can directly modulate cardiac energy substrate metabolism. METHODS AND RESULTS We used chronically instrumented dogs, 8 with pacing-induced HF and 6 normal controls. Human des-acyl ghrelin [1.2 nmol/kg per hour] was infused intravenously for 15 minutes, followed by washout (rebaseline) and infusion of acyl ghrelin at the same dose. (3)H-oleate and (14)C-glucose were coinfused and arterial and coronary sinus blood sampled to measure cardiac free fatty acid and glucose oxidation and lactate uptake. As expected, cardiac substrate metabolism was profoundly altered in HF because baseline oxidation levels of free fatty acids and glucose were, respectively, >70% lower and >160% higher compared with control. Neither des-acyl ghrelin nor acyl ghrelin significantly affected function and metabolism in normal hearts. However, in HF, des-acyl and acyl ghrelin enhanced myocardial oxygen consumption by 10.2±3.5% and 9.9±3.7%, respectively (P<0.05), and cardiac mechanical efficiency was not significantly altered. This was associated, respectively, with a 41.3±6.7% and 32.5±10.9% increase in free fatty acid oxidation and a 31.3±9.2% and 41.4±8.9% decrease in glucose oxidation (all P<0.05). CONCLUSIONS Acute increases in des-acyl or acyl ghrelin do not interfere with cardiac metabolism in normal dogs, whereas they enhance free fatty acid oxidation and reduce glucose oxidation in HF dogs, thus partially correcting metabolic alterations in HF. This novel mechanism might contribute to the cardioprotective effects of ghrelin in HF.
Collapse
Affiliation(s)
- Gianfranco Mitacchione
- From the Department of Physiology, Temple University School of Medicine, Philadelphia, PA (G.M., J.C.P., G.G., F.W., A.L., L.L., C.A.M., R.M., S.R.H., F.A.R.); Department of Medical Sciences, University of Turin, Turin, Italy (F.S., L.T., R.G.); and Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy (F.A.R.)
| | - Jeffrey C Powers
- From the Department of Physiology, Temple University School of Medicine, Philadelphia, PA (G.M., J.C.P., G.G., F.W., A.L., L.L., C.A.M., R.M., S.R.H., F.A.R.); Department of Medical Sciences, University of Turin, Turin, Italy (F.S., L.T., R.G.); and Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy (F.A.R.)
| | - Gino Grifoni
- From the Department of Physiology, Temple University School of Medicine, Philadelphia, PA (G.M., J.C.P., G.G., F.W., A.L., L.L., C.A.M., R.M., S.R.H., F.A.R.); Department of Medical Sciences, University of Turin, Turin, Italy (F.S., L.T., R.G.); and Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy (F.A.R.)
| | - Felix Woitek
- From the Department of Physiology, Temple University School of Medicine, Philadelphia, PA (G.M., J.C.P., G.G., F.W., A.L., L.L., C.A.M., R.M., S.R.H., F.A.R.); Department of Medical Sciences, University of Turin, Turin, Italy (F.S., L.T., R.G.); and Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy (F.A.R.)
| | - Amy Lam
- From the Department of Physiology, Temple University School of Medicine, Philadelphia, PA (G.M., J.C.P., G.G., F.W., A.L., L.L., C.A.M., R.M., S.R.H., F.A.R.); Department of Medical Sciences, University of Turin, Turin, Italy (F.S., L.T., R.G.); and Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy (F.A.R.)
| | - Lien Ly
- From the Department of Physiology, Temple University School of Medicine, Philadelphia, PA (G.M., J.C.P., G.G., F.W., A.L., L.L., C.A.M., R.M., S.R.H., F.A.R.); Department of Medical Sciences, University of Turin, Turin, Italy (F.S., L.T., R.G.); and Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy (F.A.R.)
| | - Fabio Settanni
- From the Department of Physiology, Temple University School of Medicine, Philadelphia, PA (G.M., J.C.P., G.G., F.W., A.L., L.L., C.A.M., R.M., S.R.H., F.A.R.); Department of Medical Sciences, University of Turin, Turin, Italy (F.S., L.T., R.G.); and Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy (F.A.R.)
| | - Catherine A Makarewich
- From the Department of Physiology, Temple University School of Medicine, Philadelphia, PA (G.M., J.C.P., G.G., F.W., A.L., L.L., C.A.M., R.M., S.R.H., F.A.R.); Department of Medical Sciences, University of Turin, Turin, Italy (F.S., L.T., R.G.); and Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy (F.A.R.)
| | - Ryan McCormick
- From the Department of Physiology, Temple University School of Medicine, Philadelphia, PA (G.M., J.C.P., G.G., F.W., A.L., L.L., C.A.M., R.M., S.R.H., F.A.R.); Department of Medical Sciences, University of Turin, Turin, Italy (F.S., L.T., R.G.); and Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy (F.A.R.)
| | - Letizia Trovato
- From the Department of Physiology, Temple University School of Medicine, Philadelphia, PA (G.M., J.C.P., G.G., F.W., A.L., L.L., C.A.M., R.M., S.R.H., F.A.R.); Department of Medical Sciences, University of Turin, Turin, Italy (F.S., L.T., R.G.); and Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy (F.A.R.)
| | - Steven R Houser
- From the Department of Physiology, Temple University School of Medicine, Philadelphia, PA (G.M., J.C.P., G.G., F.W., A.L., L.L., C.A.M., R.M., S.R.H., F.A.R.); Department of Medical Sciences, University of Turin, Turin, Italy (F.S., L.T., R.G.); and Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy (F.A.R.)
| | - Riccarda Granata
- From the Department of Physiology, Temple University School of Medicine, Philadelphia, PA (G.M., J.C.P., G.G., F.W., A.L., L.L., C.A.M., R.M., S.R.H., F.A.R.); Department of Medical Sciences, University of Turin, Turin, Italy (F.S., L.T., R.G.); and Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy (F.A.R.)
| | - Fabio A Recchia
- From the Department of Physiology, Temple University School of Medicine, Philadelphia, PA (G.M., J.C.P., G.G., F.W., A.L., L.L., C.A.M., R.M., S.R.H., F.A.R.); Department of Medical Sciences, University of Turin, Turin, Italy (F.S., L.T., R.G.); and Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy (F.A.R.).
| |
Collapse
|
48
|
Douglas GAF, McGirr R, Charlton CL, Kagan DB, Hoffman LM, Luyt LG, Dhanvantari S. Characterization of a far-red analog of ghrelin for imaging GHS-R in P19-derived cardiomyocytes. Peptides 2014; 54:81-8. [PMID: 24468548 DOI: 10.1016/j.peptides.2014.01.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Revised: 01/16/2014] [Accepted: 01/16/2014] [Indexed: 11/29/2022]
Abstract
Ghrelin and its receptor, the growth hormone secretagogue receptor (GHS-R), are expressed in the heart, and may function to promote cardiomyocyte survival, differentiation and contractility. Previously, we had generated a truncated analog of ghrelin conjugated to fluorescein isothiocyanate for the purposes of determining GHS-R expression in situ. We now report the generation and characterization of a far-red ghrelin analog, [Dpr(3)(octanoyl), Lys(19)(Cy5)]ghrelin (1-19), and show that it can be used to image changes in GHS-R in developing cardiomyocytes. We also generated the des-acyl analog, des-acyl [Lys(19)(Cy5)]ghrelin (1-19) and characterized its binding to mouse heart sections. Receptor binding affinity of Cy5-ghrelin as measured in HEK293 cells overexpressing GHS-R1a was within an order of magnitude of that of fluorescein-ghrelin and native human ghrelin, while the des-acyl Cy5-ghrelin did not bind GHS-R1a. Live cell imaging in HEK293/GHS-R1a cells showed cell surface labeling that was displaced by excess ghrelin. Interestingly, Cy5-ghrelin, but not the des-acyl analog, showed concentration-dependent binding in mouse heart tissue sections. We then used Cy5-ghrelin to track GHS-R expression in P19-derived cardiomyocytes. Live cell imaging at different time points after DMSO-induced differentiation showed that GHS-R expression preceded that of the differentiation marker aMHC and tracked with the contractility marker SERCA 2a. Our far-red analog of ghrelin adds to the tools we are developing to map GHS-R in developing and diseased cardiac tissues.
Collapse
Affiliation(s)
- Gregory A F Douglas
- Departments of Pathology, Chemistry, Medical Imaging, Medical Biophysics, and Oncology, Western University, Richmond Street, London, Ontario, Canada; Imaging Program, Lawson Health Research Institute, 268 Grosvenor Street, London, Ontario N6A 4V2, Canada
| | - Rebecca McGirr
- Departments of Pathology, Chemistry, Medical Imaging, Medical Biophysics, and Oncology, Western University, Richmond Street, London, Ontario, Canada; Imaging Program, Lawson Health Research Institute, 268 Grosvenor Street, London, Ontario N6A 4V2, Canada
| | - Carlie L Charlton
- Departments of Pathology, Chemistry, Medical Imaging, Medical Biophysics, and Oncology, Western University, Richmond Street, London, Ontario, Canada; Imaging Program, Lawson Health Research Institute, 268 Grosvenor Street, London, Ontario N6A 4V2, Canada
| | - Dov B Kagan
- Departments of Pathology, Chemistry, Medical Imaging, Medical Biophysics, and Oncology, Western University, Richmond Street, London, Ontario, Canada; Imaging Program, Lawson Health Research Institute, 268 Grosvenor Street, London, Ontario N6A 4V2, Canada
| | - Lisa M Hoffman
- Departments of Pathology, Chemistry, Medical Imaging, Medical Biophysics, and Oncology, Western University, Richmond Street, London, Ontario, Canada; Imaging Program, Lawson Health Research Institute, 268 Grosvenor Street, London, Ontario N6A 4V2, Canada
| | - Leonard G Luyt
- Departments of Pathology, Chemistry, Medical Imaging, Medical Biophysics, and Oncology, Western University, Richmond Street, London, Ontario, Canada; Imaging Program, Lawson Health Research Institute, 268 Grosvenor Street, London, Ontario N6A 4V2, Canada
| | - Savita Dhanvantari
- Departments of Pathology, Chemistry, Medical Imaging, Medical Biophysics, and Oncology, Western University, Richmond Street, London, Ontario, Canada; Imaging Program, Lawson Health Research Institute, 268 Grosvenor Street, London, Ontario N6A 4V2, Canada.
| |
Collapse
|
49
|
The growth hormone secretagogue receptor: its intracellular signaling and regulation. Int J Mol Sci 2014; 15:4837-55. [PMID: 24651458 PMCID: PMC3975427 DOI: 10.3390/ijms15034837] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Revised: 03/06/2014] [Accepted: 03/11/2014] [Indexed: 01/29/2023] Open
Abstract
The growth hormone secretagogue receptor (GHSR), also known as the ghrelin receptor, is involved in mediating a wide variety of biological effects of ghrelin, including: stimulation of growth hormone release, increase of food intake and body weight, modulation of glucose and lipid metabolism, regulation of gastrointestinal motility and secretion, protection of neuronal and cardiovascular cells, and regulation of immune function. Dependent on the tissues and cells, activation of GHSR may trigger a diversity of signaling mechanisms and subsequent distinct physiological responses. Distinct regulation of GHSR occurs at levels of transcription, receptor interaction and internalization. Here we review the current understanding on the intracellular signaling pathways of GHSR and its modulation. An overview of the molecular structure of GHSR is presented first, followed by the discussion on its signaling mechanisms. Finally, potential mechanisms regulating GHSR are reviewed.
Collapse
|
50
|
MicroRNA-150 inhibits expression of adiponectin receptor 2 and is a potential therapeutic target in patients with chronic heart failure. J Heart Lung Transplant 2014; 33:252-60. [DOI: 10.1016/j.healun.2013.10.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Revised: 07/14/2013] [Accepted: 10/09/2013] [Indexed: 11/20/2022] Open
|