1
|
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
|
2
|
Chen C, Wang J, Liu C, Hu J, Liu L. Pioneering therapies for post-infarction angiogenesis: Insight into molecular mechanisms and preclinical studies. Biomed Pharmacother 2023; 166:115306. [PMID: 37572633 DOI: 10.1016/j.biopha.2023.115306] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 08/01/2023] [Accepted: 08/07/2023] [Indexed: 08/14/2023] Open
Abstract
Acute myocardial infarction (MI), despite significant progress in its treatment, remains a leading cause of chronic heart failure and cardiovascular events such as cardiac arrest. Promoting angiogenesis in the myocardial tissue after MI to restore blood flow in the ischemic and hypoxic tissue is considered an effective treatment strategy. The repair of the myocardial tissue post-MI involves a robust angiogenic response, with mechanisms involved including endothelial cell proliferation and migration, capillary growth, changes in the extracellular matrix, and stabilization of pericytes for neovascularization. In this review, we provide a detailed overview of six key pathways in angiogenesis post-MI: the PI3K/Akt/mTOR signaling pathway, the Notch signaling pathway, the Wnt/β-catenin signaling pathway, the Hippo signaling pathway, the Sonic Hedgehog signaling pathway, and the JAK/STAT signaling pathway. We also discuss novel therapeutic approaches targeting these pathways, including drug therapy, gene therapy, protein therapy, cell therapy, and extracellular vesicle therapy. A comprehensive understanding of these key pathways and their targeted therapies will aid in our understanding of the pathological and physiological mechanisms of angiogenesis after MI and the development and application of new treatment strategies.
Collapse
Affiliation(s)
- Cong Chen
- Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China
| | - Jie Wang
- Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China.
| | - Chao Liu
- Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China
| | - Jun Hu
- Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China
| | - Lanchun Liu
- Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China
| |
Collapse
|
3
|
Signaling pathways and targeted therapy for myocardial infarction. Signal Transduct Target Ther 2022; 7:78. [PMID: 35273164 PMCID: PMC8913803 DOI: 10.1038/s41392-022-00925-z] [Citation(s) in RCA: 374] [Impact Index Per Article: 124.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 01/28/2022] [Accepted: 02/08/2022] [Indexed: 02/07/2023] Open
Abstract
Although the treatment of myocardial infarction (MI) has improved considerably, it is still a worldwide disease with high morbidity and high mortality. Whilst there is still a long way to go for discovering ideal treatments, therapeutic strategies committed to cardioprotection and cardiac repair following cardiac ischemia are emerging. Evidence of pathological characteristics in MI illustrates cell signaling pathways that participate in the survival, proliferation, apoptosis, autophagy of cardiomyocytes, endothelial cells, fibroblasts, monocytes, and stem cells. These signaling pathways include the key players in inflammation response, e.g., NLRP3/caspase-1 and TLR4/MyD88/NF-κB; the crucial mediators in oxidative stress and apoptosis, for instance, Notch, Hippo/YAP, RhoA/ROCK, Nrf2/HO-1, and Sonic hedgehog; the controller of myocardial fibrosis such as TGF-β/SMADs and Wnt/β-catenin; and the main regulator of angiogenesis, PI3K/Akt, MAPK, JAK/STAT, Sonic hedgehog, etc. Since signaling pathways play an important role in administering the process of MI, aiming at targeting these aberrant signaling pathways and improving the pathological manifestations in MI is indispensable and promising. Hence, drug therapy, gene therapy, protein therapy, cell therapy, and exosome therapy have been emerging and are known as novel therapies. In this review, we summarize the therapeutic strategies for MI by regulating these associated pathways, which contribute to inhibiting cardiomyocytes death, attenuating inflammation, enhancing angiogenesis, etc. so as to repair and re-functionalize damaged hearts.
Collapse
|
4
|
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
|
5
|
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
|
6
|
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
|
7
|
Audebrand A, Désaubry L, Nebigil CG. Targeting GPCRs Against Cardiotoxicity Induced by Anticancer Treatments. Front Cardiovasc Med 2020; 6:194. [PMID: 32039239 PMCID: PMC6993588 DOI: 10.3389/fcvm.2019.00194] [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] [Received: 11/13/2019] [Accepted: 12/23/2019] [Indexed: 01/01/2023] Open
Abstract
Novel anticancer medicines, including targeted therapies and immune checkpoint inhibitors, have greatly improved the management of cancers. However, both conventional and new anticancer treatments induce cardiac adverse effects, which remain a critical issue in clinic. Cardiotoxicity induced by anti-cancer treatments compromise vasospastic and thromboembolic ischemia, dysrhythmia, hypertension, myocarditis, and cardiac dysfunction that can result in heart failure. Importantly, none of the strategies to prevent cardiotoxicity from anticancer therapies is completely safe and satisfactory. Certain clinically used cardioprotective drugs can even contribute to cancer induction. Since G protein coupled receptors (GPCRs) are target of forty percent of clinically used drugs, here we discuss the newly identified cardioprotective agents that bind GPCRs of adrenalin, adenosine, melatonin, ghrelin, galanin, apelin, prokineticin and cannabidiol. We hope to provoke further drug development studies considering these GPCRs as potential targets to be translated to treatment of human heart failure induced by anticancer drugs.
Collapse
Affiliation(s)
| | | | - Canan G. Nebigil
- Laboratory of CardioOncology and Therapeutic Innovation, CNRS, Illkirch, France
| |
Collapse
|
8
|
Liu L, Gan S, Li B, Ge X, Yu H, Zhou H. Fisetin Alleviates Atrial Inflammation, Remodeling, and Vulnerability to Atrial Fibrillation after Myocardial Infarction. Int Heart J 2019; 60:1398-1406. [DOI: 10.1536/ihj.19-131] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- Liang Liu
- Department of Cardiology, Xianning Central Hospital, The First Affiliated Hospital of Hubei University of Science And Technology
| | - Shouyi Gan
- Department of Cardiology, Xianning Central Hospital, The First Affiliated Hospital of Hubei University of Science And Technology
| | - Bin Li
- Department of Cardiology, Xianning Central Hospital, The First Affiliated Hospital of Hubei University of Science And Technology
| | - Xiong Ge
- Department of Cardiology, Xianning Central Hospital, The First Affiliated Hospital of Hubei University of Science And Technology
| | - Hui Yu
- Department of Cardiology, Xianning Central Hospital, The First Affiliated Hospital of Hubei University of Science And Technology
| | - Huiliang Zhou
- Department of Cardiology, Xianning Central Hospital, The First Affiliated Hospital of Hubei University of Science And Technology
| |
Collapse
|
9
|
Liu M, Li W, Wang H, Yin L, Ye B, Tang Y, Huang C. CTRP9 Ameliorates Atrial Inflammation, Fibrosis, and Vulnerability to Atrial Fibrillation in Post-Myocardial Infarction Rats. J Am Heart Assoc 2019; 8:e013133. [PMID: 31623508 PMCID: PMC6898814 DOI: 10.1161/jaha.119.013133] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background Inflammation and fibrosis play an important role in the pathogenesis of atrial fibrillation (AF) after myocardial infarction (MI). CTRP9 (C1q/tumor necrosis factor‐related protein‐9) as a secreted glycoprotein can reverse left ventricle remodeling post‐MI, but its effects on MI‐induced atrial inflammation, fibrosis, and associated AF are unknown. Methods and Results MI model rats received adenoviral supplementation of CTRP9 (Ad‐CTRP9) by jugular‐vein injection. Cardiac function, inflammatory, and fibrotic indexes and related signaling pathways, electrophysiological properties, and AF inducibility of atria in vivo and ex vivo were detected in 3 or 7 days after MI. shCTRP9 (short hairpin CTRP9) and shRNA were injected into rat and performed similar detection at day 5 or 10. Adverse atrial inflammation and fibrosis, cardiac dysfunction were induced in both MI and Ad‐GFP (adenovirus‐encoding green fluorescent protein)+MI rats. Systemic CTRP9 treatment improved cardiac dysfunction post‐MI. CTRP9 markedly ameliorated macrophage infiltration and attenuated the inflammatory responses by downregulating interleukin‐1β and interleukin‐6, and upregulating interleukin‐10, in 3 days post‐MI; depressed left atrial fibrosis by decreasing the expressions of collagen types I and III, α‐SMA, and transforming growth factor β1 in 7 days post‐MI possibly through depressing the Toll‐like receptor 4/nuclear factor‐κB and Smad2/3 signaling pathways. Electrophysiologic recordings showed that increased AF inducibility and duration, and prolongation of interatrial conduction time induced by MI were attenuated by CTRP9; moreover, CTRP9 was negatively correlated with interleukin‐1β and AF duration. Downregulation of CTRP9 aggravated atrial inflammation, fibrosis, susceptibility of AF and prolonged interatrial conduction time, without affecting cardiac function. Conclusions CTRP9 is effective at attenuating atrial inflammation and fibrosis, possibly via its inhibitory effects on the Toll‐like receptor 4/nuclear factor‐κB and Smad2/3 signaling pathways, and may be an original upstream therapy for AF in early phase of MI.
Collapse
Affiliation(s)
- Mingxin Liu
- Department of Cardiology Renmin Hospital of Wuhan University Hubei China.,Cardiovascular Research Institute of Wuhan University Hubei China.,Hubei Key Laboratory of Cardiology Hubei China
| | - Wei Li
- Department of Cardiology Renmin Hospital of Wuhan University Hubei China.,Cardiovascular Research Institute of Wuhan University Hubei China.,Hubei Key Laboratory of Cardiology Hubei China
| | - Huibo Wang
- Department of Cardiology Renmin Hospital of Wuhan University Hubei China.,Cardiovascular Research Institute of Wuhan University Hubei China.,Hubei Key Laboratory of Cardiology Hubei China
| | - Lin Yin
- Department of Cardiology Renmin Hospital of Wuhan University Hubei China.,Cardiovascular Research Institute of Wuhan University Hubei China.,Hubei Key Laboratory of Cardiology Hubei China
| | - Bingjie Ye
- Department of Cardiology Renmin Hospital of Wuhan University Hubei China.,Cardiovascular Research Institute of Wuhan University Hubei China.,Hubei Key Laboratory of Cardiology Hubei China
| | - Yanhong Tang
- Department of Cardiology Renmin Hospital of Wuhan University Hubei China.,Cardiovascular Research Institute of Wuhan University Hubei China.,Hubei Key Laboratory of Cardiology Hubei China
| | - Congxin Huang
- Department of Cardiology Renmin Hospital of Wuhan University Hubei China.,Cardiovascular Research Institute of Wuhan University Hubei China.,Hubei Key Laboratory of Cardiology Hubei China
| |
Collapse
|
10
|
Huang J, Liu W, Doycheva DM, Gamdzyk M, Lu W, Tang J, Zhang JH. Ghrelin attenuates oxidative stress and neuronal apoptosis via GHSR-1α/AMPK/Sirt1/PGC-1α/UCP2 pathway in a rat model of neonatal HIE. Free Radic Biol Med 2019; 141:322-337. [PMID: 31279091 PMCID: PMC6718314 DOI: 10.1016/j.freeradbiomed.2019.07.001] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 07/01/2019] [Accepted: 07/02/2019] [Indexed: 11/26/2022]
Abstract
Neuronal apoptosis induced by oxidative stress is one of the major pathological processes involved in neurological impairment after hypoxic-ischemic encephalopathy (HIE). Ghrelin, the unique endogenous ligand for the growth hormone secretagogue receptor-1α (GHSR-1α), could take an anti-apoptotic role in the brain. However, whether ghrelin can attenuate neuronal apoptosis by attenuating oxidative stress after hypoxia-ischemia (HI) insult remains unknown. To investigate the beneficial effects of ghrelin on oxidative stress injury and neuronal apoptosis induced by HI, ten-day old unsexed rat pups were subjected to HI injury and exogenous recombinant human ghrelin(rh-Ghrelin) was administered intranasally at 1 h and 24 h after HI induction. [D-Lys3]-GHRP-6, a selective inhibitor of GHSR-1α and Ex527, a selective inhibitor of GHSR-1α were administered intranasally at 1 h before HI induction respectively. Small interfering ribonucleic acid (siRNA) for GHSR-1α were administered by intracerebroventricular (i.c.v) injection at 24 h before HI induction. Neurological tests, immunofluorescence, MitoSox staining, Fluoro-Jade C staining, terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining, and western blot experiments were performed. Our results indicated that ghrelin significantly improved neurobehavioral outcomes and reduced oxidative stress and neuronal apoptosis. Moreover, ghrelin treatment significantly promoted phosphorylation of AMPK, upregulated the expression of Sirt1, PGC-1α, UCP2 and the ratio of Bcl2/Bax, while it downregulated cleaved caspase-3 levels. The protective effects of ghrelin were reversed by [D-Lys3]-GHRP-6, GHSR-1α siRNA or Ex527. In conclusion, our data demonstrated that ghrelin reduced oxidative stress injury and neuronal apoptosis which was in part via the GHSR-1α/AMPK/Sirt1/PGC-1α/UCP2 signalling pathway after HI. Ghrelin may be a novel therapeutic target for treatment after neonatasl HI injury.
Collapse
Affiliation(s)
- Juan Huang
- Institute of Neuroscience, Chongqing Medical University, Chongqing, 400016, China; Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Wei Liu
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, 92350, USA; Department of Physiology, School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Desislava Met Doycheva
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Marcin Gamdzyk
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Weitian Lu
- Institute of Neuroscience, Chongqing Medical University, Chongqing, 400016, China; Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Jiping Tang
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, 92350, USA
| | - John H Zhang
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, 92350, USA; Department of Anesthesiology, Loma Linda University, Loma Linda, CA, 92350, USA; Department of Neurosurgery, Loma Linda University, Loma Linda, CA, 92350, USA.
| |
Collapse
|
11
|
Liu M, Yin L, Li W, Hu J, Wang H, Ye B, Tang Y, Huang C. C1q/TNF-related protein-9 promotes macrophage polarization and improves cardiac dysfunction after myocardial infarction. J Cell Physiol 2019; 234:18731-18747. [PMID: 30953351 PMCID: PMC6618013 DOI: 10.1002/jcp.28513] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 02/13/2019] [Accepted: 02/14/2019] [Indexed: 12/11/2022]
Abstract
The timely regulation of inflammatory M1 macrophage polarization toward regenerative M2 macrophages suggests the possibility of immunotherapy after myocardial infarction (MI). C1q/TNF‐related protein‐9 (CTRP9) has anti‐inflammatory effects and can ameliorate heart function in mice after long‐term myocardial infarction. The role of CTRP9 in macrophage polarization remains completely unclear. This study determined whether CTRP9 can preserve post‐MI early cardiac function through the regulation of macrophage polarization. In the present study, an adenovirus‐delivered CTRP9 supplement promoted macrophage polarization at Day 3 post MI and improved cardiac function at Day 7 post MI. Pretreatment with gCTRP9 promoted the M1 to M2 polarization transition and attenuated inflammation after lipopolysaccharide + interferon‐γ stimulation; the effects were partly abrogated by the adenosine monophosphate kinase (AMPK) inhibitor compound C and were obviously reinforced by pyrrolidine dithiocarbamate, a nuclear factor‐κB (NF‐κB) inhibitor. Meanwhile, CTPR9 markedly reduced the expression of toll‐like receptor 4 (TLR4), myeloid differentiation factor 88 (MyD88), and NF‐κB p65 phosphorylation by promoting AMPK phosphorylation in vivo and in vitro. Moreover, the competitive binding of gCTRP9 and LPS to the myeloid differentiation protein 2 (MD2)/TLR4 complex was associated with direct binding to MD2, thereby inhibiting the downstream signaling molecule MyD88. Taken together, we demonstrated that CTRP9 improved post‐MI early cardiac function, at least in part, by modulating M1/M2 macrophage polarization, largely via the TLR4/MD2/MyD88 and AMPK‐NF‐κB pathways.
Collapse
Affiliation(s)
- Mingxin Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.,Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei, China.,Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Lin Yin
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.,Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei, China.,Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Wei Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.,Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei, China.,Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Juan Hu
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China.,Institute of Hypertension, Central South University, Changsha, Hunan, China
| | - Huibo Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.,Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei, China.,Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Bingjie Ye
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.,Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei, China.,Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Yanhong Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.,Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei, China.,Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Congxin Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.,Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei, China.,Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| |
Collapse
|
12
|
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
|
13
|
Chen H, Liu Y, Gui Q, Zhu X, Zeng L, Meng J, Qing J, Gao L, Jackson AO, Feng J, Li Y, He J, Yin K. Ghrelin attenuates myocardial fibrosis after acute myocardial infarction via inhibiting endothelial-to mesenchymal transition in rat model. Peptides 2019; 111:118-126. [PMID: 30218693 DOI: 10.1016/j.peptides.2018.09.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 09/08/2018] [Accepted: 09/10/2018] [Indexed: 12/17/2022]
Abstract
Ghrelin, a peptide hormone produced in the gastrointestinal tract, has recently been found to be associated with the onset of myocardial fibrosis (MF). The exact mechanism, however, remains elusive. This study sought to identify the function and mechanism of ghrelin on MF after acute myocardial infarction (AMI). AMI was established in Spraque-Dawley rats by ligation of the left anterior descending (LAD). Ghrelin or saline was intraperitoneally injected two times per day for 8 weeks after ligation. The weight of heart (mg) and the weight ratio of heart to body (mg/g) as well as the fibrotic area were increased, while serum level of ghrelin was decreased after AMI. Ghrelin significantly ameliorated MF and decreased deposition of collagens in perivascular fibrosis area. In addition, ghrelin inhibited Endothelial-to-mesenchymal transition (EndMT), a crucial process for MF, in perivascular fibrosis area and TGF-β1-induced human coronary artery endothelial cells (HCAECs). Mechanistically, ghrelin persistently decreased the phosphorylation of Smad2/3 and enhanced the expression of Smad7 and p-AMPK in vivo and in vitro. After the abolition of Smad7, GHSR-1a and AMPK pathway, the effect of ghrelin on EndMT was significantly inhibited. In conclusion, these results presented a novel finding that ghrelin attenuated MF after AMI via regulation EndMT in a GHSR-1a/AMPK/Smad7- dependent manner.
Collapse
Affiliation(s)
- Hainan Chen
- Research Lab for Clinical & Translational Medicine, Medical school, University of South China, Hengyang 421001, China; Institute of Cardiovascular Research, Key Laboratory Atherosclerology of Hunan Province, University of South China, Hengyang 421001, China
| | - Yijian Liu
- The Third Hospital of Changsha, Changsha 410000, China
| | - Qingjun Gui
- Research Lab for Clinical & Translational Medicine, Medical school, University of South China, Hengyang 421001, China
| | - Xiao Zhu
- Research Lab for Clinical & Translational Medicine, Medical school, University of South China, Hengyang 421001, China; Institute of Cardiovascular Research, Key Laboratory Atherosclerology of Hunan Province, University of South China, Hengyang 421001, China
| | - Lin Zeng
- Department of Neurology, First Affiliated Hospital of University of South China, University of South China, Hengyang 421001, China
| | - Jun Meng
- Functional Department, the First Affiliated Hospital of University of South China, Hengyang, Hunan 421001, China
| | - Jina Qing
- Research Lab for Clinical & Translational Medicine, Medical school, University of South China, Hengyang 421001, China
| | - Ling Gao
- Research Lab for Clinical & Translational Medicine, Medical school, University of South China, Hengyang 421001, China
| | - Ampadu O Jackson
- Research Lab for Clinical & Translational Medicine, Medical school, University of South China, Hengyang 421001, China; International College, University of South China, Hengyang 421001, China
| | - Juling Feng
- Research Lab for Clinical & Translational Medicine, Medical school, University of South China, Hengyang 421001, China
| | - Yi Li
- Research Lab for Clinical & Translational Medicine, Medical school, University of South China, Hengyang 421001, China
| | - Jin He
- Functional Department, the First Affiliated Hospital of University of South China, Hengyang, Hunan 421001, China.
| | - Kai Yin
- Research Lab for Clinical & Translational Medicine, Medical school, University of South China, Hengyang 421001, China; Institute of Cardiovascular Research, Key Laboratory Atherosclerology of Hunan Province, University of South China, Hengyang 421001, China.
| |
Collapse
|
14
|
Tokudome T, Otani K, Miyazato M, Kangawa K. Ghrelin and the heart. Peptides 2019; 111:42-46. [PMID: 29791869 DOI: 10.1016/j.peptides.2018.05.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 05/11/2018] [Accepted: 05/15/2018] [Indexed: 12/14/2022]
Abstract
Ghrelin, a growth hormone-releasing peptide that was first discovered in the stomach of rats in 1999, is an endogenous ligand of growth hormone secretagogue receptor. Ghrelin exerts its potent growth hormone-releasing and orexigenic activities by binding to specific receptors in the brain. Subsequent studies showed that ghrelin participates in the regulation of diverse processes, including energy balance, body weight maintenance, and glucose and fat metabolism, and demonstrated that ghrelin is beneficial for treatment of cardiac diseases. In animal models of chronic heart failure, administration of ghrelin improves cardiac function and remodeling, and these findings were recapitulated in human patients with heart failure. Also in animal models, ghrelin administration effectively diminishes pulmonary hypertension induced by monocrotaline or chronic hypoxia. In addition, repeated administration of ghrelin to cachectic chronic obstructive pulmonary disease patients has positive effects on body composition, including amelioration of muscle wasting, improvement of functional capacity, and sympathetic activity. Moreover, administration of ghrelin early after myocardial infarction decreases the frequency of fatal arrhythmia and improved the survival rate. In ghrelin-deficient mice, both exogenous and endogenous ghrelin protects against fatal arrhythmia and promotes remodeling after myocardial infarction. Although the mechanisms underlying the effects of ghrelin on the cardiovascular system have not been fully elucidated, some evidence suggests that its beneficial effects are mediated through both direct actions on cardiovascular cells and regulation of autonomic nervous system activity. Therefore, ghrelin is a promising novel therapeutic agent for cardiac disease.
Collapse
Affiliation(s)
- Takeshi Tokudome
- Department of Biochemistry (T.T, M.M), Regenerative Medicine and Tissue Engineering (K.O), and Trustee (K.K), National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan.
| | - Kentaro Otani
- Department of Biochemistry (T.T, M.M), Regenerative Medicine and Tissue Engineering (K.O), and Trustee (K.K), National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan
| | - Mikiya Miyazato
- Department of Biochemistry (T.T, M.M), Regenerative Medicine and Tissue Engineering (K.O), and Trustee (K.K), National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan
| | - Kenji Kangawa
- Department of Biochemistry (T.T, M.M), Regenerative Medicine and Tissue Engineering (K.O), and Trustee (K.K), National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan
| |
Collapse
|
15
|
Pearson JT, Collie N, Lamberts RR, Inagaki T, Yoshimoto M, Umetani K, Davis P, Wilkins G, Jones PP, Shirai M, Schwenke DO. Ghrelin Preserves Ischemia-Induced Vasodilation of Male Rat Coronary Vessels Following β-Adrenergic Receptor Blockade. Endocrinology 2018; 159:1763-1773. [PMID: 29325034 DOI: 10.1210/en.2017-03070] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 12/20/2017] [Indexed: 12/24/2022]
Abstract
Acute myocardial infarction (MI) triggers an adverse increase in cardiac sympathetic nerve activity (SNA). Whereas β-adrenergic receptor (β-AR) blockers are routinely used for the management of MI, they may also counter β-AR-mediated vasodilation of coronary vessels. We have reported that ghrelin prevents sympathetic activation following MI. Whether ghrelin modulates coronary vascular tone following MI, either through the modulation of SNA or directly as a vasoactive mediator, has never been addressed. We used synchrotron microangiography to image coronary perfusion and vessel internal diameter (ID) in anesthetized Sprague-Dawley rats, before and then again 30 minutes after induction of an MI (left coronary artery ligation). Rats were injected with either saline or ghrelin (150 µg/kg, subcutaneously), immediately following the MI or sham surgery. Coronary angiograms were also recorded following β-AR blockade (propranolol, 2 mg/kg, intravenously). Finally, wire myography was used to assess the effect of ghrelin on vascular tone in isolated human internal mammary arteries (IMAs). Acute MI enhanced coronary perfusion to nonischemicregions through dilation of small arterioles (ID 50 to 250 µm) and microvessel recruitment, irrespective of ghrelin treatment. In ghrelin-treated rats, β-AR blockade did not alter the ischemia-induced vasodilation, yet in saline-treated rats, β-AR blockade abolished the vasodilation of small arterioles. Finally, ghrelin caused a dose-dependent vasodilation of IMA rings (preconstricted with phenylephrine). In summary, this study highlights ghrelin as a promising adjunct therapy that can be used in combination with routine β-AR blockade treatment for preserving coronary blood flow and cardiac performance in patients who suffer an acute MI.
Collapse
Affiliation(s)
- James T Pearson
- Department of Cardiac Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan
| | - Nicola Collie
- Department of Physiology, School of Biomedical Sciences, HeartOtago University of Otago, Dunedin, New Zealand
| | - Regis R Lamberts
- Department of Physiology, School of Biomedical Sciences, HeartOtago University of Otago, Dunedin, New Zealand
| | - Tadakatsu Inagaki
- Department of Vascular Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan
| | - Misa Yoshimoto
- Department of Health Sciences, Nara Women's University, Nara, Japan
| | - Keiji Umetani
- Japan Synchrotron Radiation Research Institute, Hyogo, Japan
| | - Philip Davis
- Department of Cardiothoracic Surgery, HeartOtago, University of Otago, Dunedin, New Zealand
| | - Gerard Wilkins
- Department of Medicine Surgery, HeartOtago, University of Otago, Dunedin, New Zealand
| | - Pete P Jones
- Department of Physiology, School of Biomedical Sciences, HeartOtago University of Otago, Dunedin, New Zealand
| | - Mikiyasu Shirai
- Department of Advanced Medical Research for Pulmonary Hypertension, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan
| | - Daryl O Schwenke
- Department of Physiology, School of Biomedical Sciences, HeartOtago University of Otago, Dunedin, New Zealand
| |
Collapse
|
16
|
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
|
17
|
Effect of a single nucleotide polymorphism in the growth hormone secretagogue receptor (GHSR) gene on growth rate in pigs. Gene 2017; 634:68-73. [PMID: 28887157 DOI: 10.1016/j.gene.2017.09.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 08/26/2017] [Accepted: 09/04/2017] [Indexed: 01/28/2023]
Abstract
The growth hormone secretagogue receptor (GHSR) gene controls growth hormone (GH) release by inducing a strong stimulatory effect on the endogenous ligand, ghrelin. In this study, we examined the possible role of GHSR in the growth traits of four pig breeds, namely Tibetan pigs (n=45), Diannan small-eared pigs (n=40), Yorkshire pigs (n=45), and New Huai pigs (n=122). Single nucleotide polymorphisms (SNPs) in these pigs were identified by polymerase chain reaction (PCR) sequencing and genotyping was performed using PCR-restriction fragment length polymorphisms (PCR-RFLPs). A SNP (C/A) named C-1595A (the "C" allele), which is located 1595bp upstream of the initiation codon of the GHSR gene, was found at a higher frequency in the fast-growing Yorkshire pigs than in the slow-growing Tibetan and Diannan small-eared pigs. In preliminary assays, the C-1595A genotype was found to be associated with growth traits in New Huai pigs. Quantitative real-time PCR and western blotting assays were used to measure the levels of GHSR1a, a functionally active form of the GHSR protein, in the tissues of the growth axis. The estimated levels of mRNA and protein in pituitary and liver tissues were significantly higher in Yorkshire pigs than in Diannan small-eared or Tibetan pigs (P<0.05). The results indicated that GHSR had a positive influence on the growth rate of pigs and suggested that the C-1595A SNP could be of value as a molecular marker for improving the production performance of pig breeds.
Collapse
|
18
|
Danshensu accelerates angiogenesis after myocardial infarction in rats and promotes the functions of endothelial progenitor cells through SDF-1α/CXCR4 axis. Eur J Pharmacol 2017; 814:274-282. [PMID: 28864209 DOI: 10.1016/j.ejphar.2017.08.035] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 08/25/2017] [Accepted: 08/28/2017] [Indexed: 01/16/2023]
Abstract
The present study was performed to investigate the potential role of Danshensu in therapeutic angiogenesis in ischemic myocardium and endothelial progenitor cells (EPCs) function. The rat model of myocardial infarction (MI) injury was induced by left anterior descending coronary artery ligation for 14 days. Danshensu significantly alleviated myocardial ischemia injury by ameliorating left ventricular function and reducing infarct size. Furthermore, Danshensu potentiated post-ischemia neovascularization as evidenced by increased microvessel density in infarction boundary zone, as well as the expression of marker proteins vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF). Moreover, Danshensu notably promoted stromal cell-derived factor-1α (SDF-1α) level in plasma and C-X-C chemokine receptor type 4 (CXCR4) expression in peri-infarction myocardium, and AMD3100 (CXCR4 antagonist) could reverse the angiogenic and cardioprotective effects of Danshensu. For in vitro study, EPCs were isolated from bone marrow of rats. On the one hand, Danshensu provided significant cytoprotection against hypoxia insult by boosting EPCs viability and inhibiting apoptosis, and upregulated Akt phosphorylation. On the other hand, Danshensu enhanced proangiogenic functions of EPCs on cell migration and tube formation, and increased SDF-1α and CXCR4 expression. Likewise, the cytoprotection and proangiogenic functions of Danshensu on EPCs were partly negated by LY294002 (PI3K antagonist) and CXCR4 siRNA, respectively. Taken together, our results suggested that the cardioprotection of Danshensu in MI rats may be related to promoting myocardial neovascularization. The possible mechanisms may involve improving EPCs survival in hypoxia condition through Akt phosphorylation, and accelerating EPCs proangiogenic functions through SDF-1α/CXCR4 axis.
Collapse
|
19
|
Wang N, Chen C, Yang D, Liao Q, Luo H, Wang X, Zhou F, Yang X, Yang J, Zeng C, Wang WE. Mesenchymal stem cells-derived extracellular vesicles, via miR-210, improve infarcted cardiac function by promotion of angiogenesis. Biochim Biophys Acta Mol Basis Dis 2017; 1863:2085-2092. [PMID: 28249798 DOI: 10.1016/j.bbadis.2017.02.023] [Citation(s) in RCA: 172] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 01/18/2017] [Accepted: 02/21/2017] [Indexed: 01/11/2023]
Abstract
Mesenchymal stem cells (MSCs) exert therapeutic effect on treating acute myocardial infarction. Recent evidence showed that paracrine function rather than direct differentiation predominately contributes to the beneficial effects of MSCs, but how the paracrine factors function are not fully elucidated. In the present study, we tested if extracellular vesicles (EVs) secreted by MSC promotes angiogenesis in infracted heart via microRNAs. Immunostaining of CD31 and matrigel plug assay were performed to detect angiogenesis in a mouse myocardial infarction (MI) model. The cardiac function and structure was examined with echocardiographic analysis. Capillary-like tube formation, migration and proliferation of human umbilical vein endothelial cells (HUVECs) were determined. As a result, MSC-EVs significantly improved angiogenesis and cardiac function in post-MI heart. MSC-EVs increased the proliferation, migration and tube formation capacity of HUVECs. MicroRNA (miR)-210 was found to be enriched in MSC-EVs. The EVs collected from MSCs with miR-210 silence largely lost the pro-angiogenic effect both in-vitro and in-vivo. The miR-210 target gene Efna3, which plays a role in angiogenesis, was down-regulated by MSC-EVs treatment in HUVECs. In conclusion, MSC-EVs are sufficient to improve angiogenesis and exert therapeutic effect on MI, its pro- angiogenesis effect might be associated with a miR-210-Efna3 dependent mechanism. This article is part of a Special Issue entitled: Genetic and epigenetic control of heart failure - edited by Jun Ren & Megan Yingmei Zhang.
Collapse
Affiliation(s)
- Na Wang
- Department of Cardiology, Chongqing Institute of Cardiology, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Caiyu Chen
- Department of Cardiology, Chongqing Institute of Cardiology, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Dezhong Yang
- Department of Cardiology, Chongqing Institute of Cardiology, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Qiao Liao
- Department of Cardiology, Chongqing Institute of Cardiology, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Hao Luo
- Department of Cardiology, Chongqing Institute of Cardiology, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Xinquan Wang
- Department of Cardiology, Chongqing Institute of Cardiology, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Faying Zhou
- Department of Cardiology, Chongqing Institute of Cardiology, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Xiaoli Yang
- Department of Cardiology, Chongqing Institute of Cardiology, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Jian Yang
- Department of Cardiology, Chongqing Institute of Cardiology, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Chunyu Zeng
- Department of Cardiology, Chongqing Institute of Cardiology, Daping Hospital, Third Military Medical University, Chongqing 400042, China.
| | - Wei Eric Wang
- Department of Cardiology, Chongqing Institute of Cardiology, Daping Hospital, Third Military Medical University, Chongqing 400042, China.
| |
Collapse
|