1
|
Zhang S, Kaiya H, Kitazawa T. Does ghrelin regulate intestinal motility in rabbits? An in vitro study using isolated duodenal strips. Gen Comp Endocrinol 2023; 344:114384. [PMID: 37722460 DOI: 10.1016/j.ygcen.2023.114384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 09/07/2023] [Accepted: 09/15/2023] [Indexed: 09/20/2023]
Abstract
Rabbit duodenum has been used for examining the ability of motilin to cause muscle contraction in vitro. A motilin-related peptide, ghrelin, is known to be involved in the regulation of gastrointestinal (GI) motility in various animals, but its ability to cause rabbit GI contraction have not been well examined. The aim of this study is to clarify the action of rat ghrelin and its interaction with motilin in the rabbit duodenum. The mRNA expression of ghrelin and motilin receptors was also examined using RT-PCR. Rat ghrelin (10-9-10-6 M) did not change the contractile activity of the duodenum measured by the mean muscle tonus and area under the curve of contraction waves. In agreement with this result, the distribution of ghrelin receptor mRNA in the rabbit GI tract varied depending on the GI region from which the samples were taken; the expression level in the duodenum was negligible, but that in the esophagus or stomach was significant. On the other hand, motilin (10-10-10-6 M) caused a concentration-dependent contraction by means of increased mean muscle tonus, and consistently, motilin receptor mRNA was expressed heterogeneously depending on the GI region (esophagus = stomach = colon = rectum < duodenum = jejunum = ileum < cecum). Expression level of motilin receptor was comparable to that of ghrelin receptor in the esophagus and stomach. Pretreatment with ghrelin (10-6 M) prior to motilin did not affect the contractile activity of motilin in the duodenum. In conclusion, ghrelin does not affect muscle contractility or motilin-induced contraction in the rabbit duodenum, which is due to the lack of ghrelin receptors. The present in vitro results suggest that ghrelin might not be a regulator of intestinal motility in rabbits.
Collapse
Affiliation(s)
- Shuangyi Zhang
- Laboratory of Veterinary Physiology, College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010018, China; School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Hokkaido 069-8501, Japan
| | - Hiroyuki Kaiya
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka 564-8565, Japan; Faculty of Science, University of Toyama, Toyama, Toyama 933-8555, Japan; Grandsoul Research Institute for Immunology, Inc., Uda, Nara 633-2221, Japan
| | - Takio Kitazawa
- School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Hokkaido 069-8501, Japan.
| |
Collapse
|
2
|
Kaplan A, Lakkis B, El-Samadi L, Karaayvaz EB, Booz GW, Zouein FA. Cooling Down Inflammation in the Cardiovascular System via the Nicotinic Acetylcholine Receptor. J Cardiovasc Pharmacol 2023; 82:241-265. [PMID: 37539950 DOI: 10.1097/fjc.0000000000001455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 07/06/2023] [Indexed: 08/05/2023]
Abstract
ABSTRACT Inflammation is a major player in many cardiovascular diseases including hypertension, atherosclerosis, myocardial infarction, and heart failure. In many individuals, these conditions coexist and mutually exacerbate each other's progression. The pathophysiology of these diseases entails the active involvement of both innate and adaptive immune cells. Immune cells that possess the α7 subunit of the nicotinic acetylcholine receptor on their surface have the potential to be targeted through both pharmacological and electrical stimulation of the cholinergic system. The cholinergic system regulates the inflammatory response to various stressors in different organ systems by systematically suppressing spleen-derived monocytes and chemokines and locally improving immune cell function. Research on the cardiovascular system has demonstrated the potential for atheroma plaque stabilization and regression as favorable outcomes. Smaller infarct size and reduced fibrosis have been associated with improved cardiac function and a decrease in adverse cardiac remodeling. Furthermore, enhanced electrical stability of the myocardium can lead to a reduction in the incidence of ventricular tachyarrhythmia. In addition, improving mitochondrial dysfunction and decreasing oxidative stress can result in less myocardial tissue damage caused by reperfusion injury. Restoring baroreflex activity and reduction in renal damage can promote blood pressure regulation and help counteract hypertension. Thus, the present review highlights the potential of nicotinic acetylcholine receptor activation as a natural approach to alleviate the adverse consequences of inflammation in the cardiovascular system.
Collapse
Affiliation(s)
- Abdullah Kaplan
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Riad El-Solh, Beirut, Lebanon
- Department of Cardiology, Kemer Public Hospital, Kemer, Antalya, Turkey
- The Cardiovascular, Renal, and Metabolic Diseases Research Center of Excellence, American University of Beirut Medical Center, Riad El-Solh, Beirut, Lebanon
| | - Bachir Lakkis
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Riad El-Solh, Beirut, Lebanon
| | - Lana El-Samadi
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Riad El-Solh, Beirut, Lebanon
| | - Ekrem Bilal Karaayvaz
- Department of Cardiology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - George W Booz
- Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center, Jackson, MS; and
| | - Fouad A Zouein
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Riad El-Solh, Beirut, Lebanon
- The Cardiovascular, Renal, and Metabolic Diseases Research Center of Excellence, American University of Beirut Medical Center, Riad El-Solh, Beirut, Lebanon
- Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center, Jackson, MS; and
- Department of Signaling and Cardiovascular Pathophysiology, UMR-S 1180, Inserm, Université Paris-Saclay, France
| |
Collapse
|
3
|
Effect of Ghrelin on the Cardiovascular System. BIOLOGY 2022; 11:biology11081190. [PMID: 36009817 PMCID: PMC9405061 DOI: 10.3390/biology11081190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 08/03/2022] [Accepted: 08/05/2022] [Indexed: 11/19/2022]
Abstract
Simple Summary Ghrelin is an octanoylated peptide that was initially isolated from rat and human stomachs in the process of searching for an endogenous ligand to the orphan growth hormone secretagogue receptor (GHS-R), a G-protein-coupled receptor. Exogenous or endogenous ghrelin secreted from the stomach binds to GHS-R on gastric vagal nerve terminals, and the signals are transmitted to the central nervous system via the vagal afferent nerve to facilitate growth hormone (GH) secretion, feeding, sympathetic inhibition, parasympathetic activation, and anabolic effects. Ghrelin also binds directly to the pituitary GHS-R and stimulates GH secretion. Ghrelin has beneficial effects on the cardiovascular system, including cardioprotective effects such as anti-heart failure, anti-arrhythmic, and anti-inflammatory actions, and it enhances vascular activity via GHS-R-dependent stimulation of GH/IGF-1 (insulin-like growth factor-1) and modulation of the autonomic nervous system. The anti-heart failure effects of ghrelin could be useful as a new therapeutic strategy for chronic heart failure. Abstract Ghrelin, an n-octanoyl-modified 28-amino-acid-peptide, was first discovered in the human and rat stomach as an endogenous ligand for the growth hormone secretagogue receptor (GHS-R). Ghrelin-GHS-R1a signaling regulates feeding behavior and energy balance, promotes vascular activity and angiogenesis, improves arrhythmia and heart failure, and also protects against cardiovascular disease by suppressing cardiac remodeling after myocardial infarction. Ghrelin’s cardiovascular protective effects are mediated by the suppression of sympathetic activity; activation of parasympathetic activity; alleviation of vascular endothelial dysfunction; and regulation of inflammation, apoptosis, and autophagy. The physiological functions of ghrelin should be clarified to determine its pharmacological potential as a cardiovascular medication.
Collapse
|
4
|
Perelló M, Cornejo MP, De Francesco PN, Fernandez G, Gautron L, Valdivia LS. The controversial role of the vagus nerve in mediating ghrelin´s actions: gut feelings and beyond. IBRO Neurosci Rep 2022; 12:228-239. [PMID: 35746965 PMCID: PMC9210457 DOI: 10.1016/j.ibneur.2022.03.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 03/08/2022] [Accepted: 03/08/2022] [Indexed: 12/26/2022] Open
Abstract
Ghrelin is a stomach-derived peptide hormone that acts via the growth hormone secretagogue receptor (GHSR) and displays a plethora of neuroendocrine, metabolic, autonomic and behavioral actions. It has been proposed that some actions of ghrelin are exerted via the vagus nerve, which provides a bidirectional communication between the central nervous system and peripheral systems. The vagus nerve comprises sensory fibers, which originate from neurons of the nodose and jugular ganglia, and motor fibers, which originate from neurons of the medulla. Many anatomical studies have mapped GHSR expression in vagal sensory or motor neurons. Also, numerous functional studies investigated the role of the vagus nerve mediating specific actions of ghrelin. Here, we critically review the topic and discuss the available evidence supporting, or not, a role for the vagus nerve mediating some specific actions of ghrelin. We conclude that studies using rats have provided the most congruent evidence indicating that the vagus nerve mediates some actions of ghrelin on the digestive and cardiovascular systems, whereas studies in mice resulted in conflicting observations. Even considering exclusively studies performed in rats, the putative role of the vagus nerve in mediating the orexigenic and growth hormone (GH) secretagogue properties of ghrelin remains debated. In humans, studies are still insufficient to draw definitive conclusions regarding the role of the vagus nerve mediating most of the actions of ghrelin. Thus, the extent to which the vagus nerve mediates ghrelin actions, particularly in humans, is still uncertain and likely one of the most intriguing unsolved aspects of the field.
Collapse
|
5
|
Hu Z, Zhang T, Mei Y, Sun N, Lv K, Wang D. Impact of Ghrelin on Ventricular Arrhythmia and Related Mechanism After Myocardial Infarction. Pharmacology 2021; 107:102-110. [PMID: 34718242 DOI: 10.1159/000519330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Accepted: 08/26/2021] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Ghrelin is an endogenous peptide with potential protective effects on ischemic heart. METHODS Synthetic ghrelin was administered (100 μg·kg-1 subcutaneous injection, twice daily) for 4 weeks in a rat model of myocardial infarction (MI) with coronary artery occlusion. At the 5th week, electrocardiogram, monophasic action potentials and autonomic nerve function were evaluated. Cardiac tyrosine hydroxylase (TH) was determined by immunofluorescence staining. RESULTS MI significantly increased sympathetic nerve activity (SNA) and ventricular arrhythmias, and prolonged APD dispersion and APD alternans (p < 0.01). Ghrelin treatment significantly increased ventricular fibrillation threshold (VFT), shortened APD dispersion and APD alternans, inhibited SNA and promoted vagus nerve activities (p < 0.01). Ghrelin also markedly reversed abnormal expression of TH in the peri-infarcted area of the heart (p < 0.01). DISCUSSION/CONCLUSION Ghrelin provides a sustained electrophysiological protection by the increase of VFT and improvement of APD dispersion and APD alternans. The mechanism may be related to the regulation of autonomic nerve and sympathetic nerve remodeling. Thus, ghrelin represents a novel drug to prevent ventricular arrhythmia in ischemic heart disease.
Collapse
Affiliation(s)
- Zhengtao Hu
- Department of Gerontology, First Affiliated Hospital of Wannan Medical College (Yijishan Hospital), Wuhu, China
| | - Ting Zhang
- Department of Gerontology, First Affiliated Hospital of Wannan Medical College (Yijishan Hospital), Wuhu, China
- Department of Psychology, Wannan Medical College, Wuhu, China
| | - Yong Mei
- Department of Gerontology, First Affiliated Hospital of Wannan Medical College (Yijishan Hospital), Wuhu, China
| | - Nan Sun
- Department of Gerontology, First Affiliated Hospital of Wannan Medical College (Yijishan Hospital), Wuhu, China
| | - Kun Lv
- Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institution (Wannan Medical College), Wuhu, China
| | - Deguo Wang
- Department of Gerontology, First Affiliated Hospital of Wannan Medical College (Yijishan Hospital), Wuhu, China
- Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institution (Wannan Medical College), Wuhu, China
| |
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
|
McDonald H, Peart J, Kurniawan ND, Galloway G, Royce SG, Samuel CS, Chen C. Hexarelin targets neuroinflammatory pathways to preserve cardiac morphology and function in a mouse model of myocardial ischemia-reperfusion. Biomed Pharmacother 2020; 127:110165. [PMID: 32403043 DOI: 10.1016/j.biopha.2020.110165] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 03/15/2020] [Accepted: 04/13/2020] [Indexed: 11/17/2022] Open
Abstract
Acute myocardial ischemia and reperfusion injury (IRI) underly the detrimental effects of coronary heart disease on the myocardium. Despite the ongoing advances in reperfusion therapies, there remains a lack of effective therapeutic strategies for preventing IRI. Growth hormone secretagogues (GHS) have been demonstrated to improve cardiac function, attenuate inflammation and modulate the autonomic nervous system (ANS) in models of cardiovascular disease. Recently, we demonstrated a reduction in infarct size after administration of hexarelin (HEX), in a murine model of myocardial infarction. In the present study we employed a reperfused ischemic (IR) model, to determine whether HEX would continue to have a cardioprotective influence in a model of higher clinical relevance. Myocardial ischemia was induced by transient ligation of the left descending coronary artery (tLAD) in C57BL/6 J mice followed by HEX (0.3 mg/kg/day; n = 20) or vehicle (VEH) (n = 18) administration for 21 days, first administered immediately prior-to reperfusion. IR-injured and sham mice were subjected to high-field magnetic resonance imaging to assess left ventricular (LV) function, with HEX-treated mice demonstrating a significant improvement in LV function compared with VEH-treated mice. A significant decrease in interstitial collagen, TGF-β1 expression and myofibroblast differentiation was also seen in the HEX-treated mice after 21 days. HEX treatment shifted the ANS balance towards a parasympathetic predominance; combined with a significant decrease in cardiac troponin-I and TNF-α levels, these findings were suggestive of an anti-inflammatory action on the myocardium mediated via HEX. In this model of IR, HEX appeared to rebalance the deregulated ANS and activate vagal anti-inflammatory pathways to prevent adverse remodelling and LV dysfunction. There are limited interventions focusing on IRI that have been successful in improving clinical outcome in acute myocardial infarction (AMI) patients, this study provides compelling evidence towards the translational potential of HEX where all others have largely failed.
Collapse
Affiliation(s)
- H McDonald
- School of Biomedical Science, University of Queensland, Brisbane, Australia
| | - J Peart
- Menzies Health Institute of Queensland, Griffith University, Gold Coast, Australia
| | - N D Kurniawan
- Centre for Advanced Imaging, University of Queensland, Brisbane, Australia
| | - G Galloway
- Centre for Advanced Imaging, University of Queensland, Brisbane, Australia
| | - S G Royce
- Cardiovascular Disease Program, Biomedical Discovery Institute and Department of Pharmacology, Australia; Central Clinical School, Monash University, Victoria, Australia
| | - C S Samuel
- Cardiovascular Disease Program, Biomedical Discovery Institute and Department of Pharmacology, Australia
| | - C Chen
- School of Biomedical Science, University of Queensland, Brisbane, Australia.
| |
Collapse
|
8
|
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
|
9
|
Shimizu S, Akiyama T, Kawada T, Sata Y, Turner MJ, Fukumitsu M, Yamamoto H, Kamiya A, Shishido T, Sugimachi M. Sodium ion transport participates in non-neuronal acetylcholine release in the renal cortex of anesthetized rabbits. J Physiol Sci 2017; 67:587-593. [PMID: 27660058 PMCID: PMC10717196 DOI: 10.1007/s12576-016-0489-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2016] [Accepted: 09/13/2016] [Indexed: 12/12/2022]
Abstract
This study examined the mechanism of release of endogenous acetylcholine (ACh) in rabbit renal cortex by applying a microdialysis technique. In anesthetized rabbits, a microdialysis probe was implanted into the renal cortex and perfused with Ringer's solution containing high potassium concentration, high sodium concentration, a Na+/K+-ATPase inhibitor (ouabain), or an epithelial Na+ channel blocker (benzamil). Dialysate samples were collected at baseline and during exposure to each agent, and ACh concentrations in the samples were measured by high-performance liquid chromatography. High potassium had no effect on renal ACh release. High sodium increased dialysate ACh concentrations significantly. Ouabain increased dialysate ACh concentration significantly. Benzamil decreased dialysate ACh concentrations significantly both at baseline and under high sodium. The finding that high potassium-induced depolarization does not increase ACh release suggests that endogenous ACh is released in renal cortex mainly by non-neuronal mechanism. Sodium ion transport may be involved in the non-neuronal ACh release.
Collapse
Affiliation(s)
- Shuji Shimizu
- Department of Cardiovascular Dynamics, National Cerebral and Cardiovascular Center, 5-7-1 Fujishiro-dai, Suita, Osaka, 565-8565, Japan.
| | - Tsuyoshi Akiyama
- Department of Cardiac Physiology, National Cerebral and Cardiovascular Center, Osaka, 565-8565, Japan
| | - Toru Kawada
- Department of Cardiovascular Dynamics, National Cerebral and Cardiovascular Center, 5-7-1 Fujishiro-dai, Suita, Osaka, 565-8565, Japan
| | - Yusuke Sata
- Department of Cardiovascular Dynamics, National Cerebral and Cardiovascular Center, 5-7-1 Fujishiro-dai, Suita, Osaka, 565-8565, Japan
| | - Michael James Turner
- Department of Cardiovascular Dynamics, National Cerebral and Cardiovascular Center, 5-7-1 Fujishiro-dai, Suita, Osaka, 565-8565, Japan
| | - Masafumi Fukumitsu
- Department of Cardiovascular Dynamics, National Cerebral and Cardiovascular Center, 5-7-1 Fujishiro-dai, Suita, Osaka, 565-8565, Japan
| | - Hiromi Yamamoto
- Division of Cardiology, Department of Medicine, Faculty of Medicine, Kindai University, Osaka, 589-8511, Japan
| | - Atsunori Kamiya
- Department of Cardiovascular Dynamics, National Cerebral and Cardiovascular Center, 5-7-1 Fujishiro-dai, Suita, Osaka, 565-8565, Japan
| | - Toshiaki Shishido
- Department of Research Promotion, National Cerebral and Cardiovascular Center, Osaka, 565-8565, Japan
| | - Masaru Sugimachi
- Department of Cardiovascular Dynamics, National Cerebral and Cardiovascular Center, 5-7-1 Fujishiro-dai, Suita, Osaka, 565-8565, Japan
| |
Collapse
|
10
|
Denney WS, Sonnenberg GE, Carvajal-Gonzalez S, Tuthill T, Jackson VM. Pharmacokinetics and pharmacodynamics of PF-05190457: The first oral ghrelin receptor inverse agonist to be profiled in healthy subjects. Br J Clin Pharmacol 2016; 83:326-338. [PMID: 27621150 DOI: 10.1111/bcp.13127] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 08/25/2016] [Accepted: 09/08/2016] [Indexed: 02/06/2023] Open
Abstract
AIM To evaluate safety, tolerability and pharmacokinetics of oral PF-05190457, an oral ghrelin receptor inverse agonist, in healthy adults. METHODS Single (SAD) and multiple ascending dose (MAD) studies were randomised, placebo-controlled, double-blind studies. Thirty-five healthy men (age 38.2 ± 10.4 years; body mass index 24.8 ± 3.1 kg m-2 [mean ± standard deviation]) received ≥1 dose (2, 10, 40 [divided], 50, 100, 150, and 300 [single or divided] mg) of PF-05190457 and/or placebo in the SAD. In the MAD study, 35 healthy men (age 39.7 ± 10.1 years; body mass index 25.9 ± 3.3 kg m-2 ) received ≥1 dose (2, 10, 40 and 100 mg twice daily) of PF-05190457 and/or placebo daily for 2 weeks. RESULTS PF-05190457 absorption was rapid with a Tmax of 0.5-3 hours and a half-life between 8.2-9.8 hours. PF-05190457 dose-dependently blocked ghrelin (1 pmol kg-1 min-1 )-induced growth hormone (GH) release with (mean [90% confidence interval]) 77% [63-85%] inhibition at 100 mg. PF-05190457 (150 mg) delayed gastric emptying lag time by 30% [7-58%] and half emptying time by 20% [7-35%] with a corresponding decrease in postprandial glucose by 9 mg dL-1 . The most frequent adverse event reported by 30 subjects at doses ≥50 mg was somnolence. PF-05190457 plasma concentrations also increased heart rate up to 13.4 [4.8-58.2] beats min-1 and, similar to the effect on glucose and ghrelin-induced GH, was lost within 2 weeks. CONCLUSIONS PF-05190457 is a well-tolerated first-in-class ghrelin receptor inverse agonist with acceptable pharmacokinetics for oral daily dosing. Blocking ghrelin receptors inhibits ghrelin-induced GH, and increases heart rate, effects that underwent tachyphylaxis with chronic dosing. PF-051940457 has the potential to treat centrally-acting disorders such as insomnia.
Collapse
Affiliation(s)
- William S Denney
- Biotherapeutics Clinical Pharmacology, Pfizer Worldwide Research and Development, Cambridge, Massachusetts, 02139, USA
| | - Gabriele E Sonnenberg
- Cardiovascular, Metabolic, and Endocrine Diseases Research Unit, Pfizer Worldwide Research and Development, Cambridge, Massachusetts, 02139, USA
| | - Santos Carvajal-Gonzalez
- Cardiovascular, Metabolic, and Endocrine Diseases Research Unit, Pfizer Worldwide Research and Development, Cambridge, Massachusetts, 02139, USA
| | - Theresa Tuthill
- Cardiovascular, Metabolic, and Endocrine Diseases Research Unit, Pfizer Worldwide Research and Development, Cambridge, Massachusetts, 02139, USA
| | - V Margaret Jackson
- Cardiovascular, Metabolic, and Endocrine Diseases Research Unit, Pfizer Worldwide Research and Development, Cambridge, Massachusetts, 02139, USA
| |
Collapse
|
11
|
Shirai M, Joe N, Tsuchimochi H, Sonobe T, Schwenke DO. Ghrelin Supresses Sympathetic Hyperexcitation in Acute Heart Failure in Male Rats: Assessing Centrally and Peripherally Mediated Pathways. Endocrinology 2015; 156:3309-16. [PMID: 26121343 DOI: 10.1210/en.2015-1333] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The hormone ghrelin prevents a dangerous increase in cardiac sympathetic nerve activity (SNA) after acute myocardial infarction (MI), although the underlying mechanisms remain unknown. This study aimed to determine whether ghrelin's sympathoinhibitory properties stem either from directly within the central nervous system, or via modulation of specific cardiac vagal inhibitory afferents. Cardiac SNA was recorded in urethane-anesthetized rats for 3 hours after the ligation of the left anterior descending coronary artery (ie, MI). Rats received ghrelin either sc (150 μg/kg) or intracerebroventricularly (5 μg/kg) immediately after the MI. In another two groups, the cervical vagi were denervated prior to the MI, followed by sc injection of either ghrelin or placebo. Acute MI induced a 188% increase in cardiac SNA, which was significantly attenuated in ghrelin-treated rats for both sc or intracerebroventricularly administration (36% and 76% increase, respectively). Consequently, mortality (47%) and the incidence of arrhythmic episodes (12 per 2 h) were improved with both routes of ghrelin administration (<13% and less than five per 2 h, respectively). Bilateral vagotomy significantly attenuated the cardiac SNA response to acute MI (99% increase). Ghrelin further attenuated the sympathetic response to MI in vagotomized rats so that the SNA response was comparable between vagotomized and vagal-intact MI rats treated with ghrelin. These results suggest that ghrelin may act primarily via a central pathway within the brain to suppress SNA after MI, although peripheral vagal afferent pathways may also contribute in part. The exact region(s) within the central nervous system whereby ghrelin inhibits SNA remains to be fully elucidated.
Collapse
Affiliation(s)
- Mikiyasu Shirai
- Department of Cardiac Physiology (M.S., H.T., T.S.), National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka 565-8565, Japan; and Department of Physiology-Heart Otago (N.J., D.O.S.), University of Otago, Dunedin 9054, New Zealand
| | - Natalie Joe
- Department of Cardiac Physiology (M.S., H.T., T.S.), National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka 565-8565, Japan; and Department of Physiology-Heart Otago (N.J., D.O.S.), University of Otago, Dunedin 9054, New Zealand
| | - Hirotsugu Tsuchimochi
- Department of Cardiac Physiology (M.S., H.T., T.S.), National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka 565-8565, Japan; and Department of Physiology-Heart Otago (N.J., D.O.S.), University of Otago, Dunedin 9054, New Zealand
| | - Takashi Sonobe
- Department of Cardiac Physiology (M.S., H.T., T.S.), National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka 565-8565, Japan; and Department of Physiology-Heart Otago (N.J., D.O.S.), University of Otago, Dunedin 9054, New Zealand
| | - Daryl O Schwenke
- Department of Cardiac Physiology (M.S., H.T., T.S.), National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka 565-8565, Japan; and Department of Physiology-Heart Otago (N.J., D.O.S.), University of Otago, Dunedin 9054, New Zealand
| |
Collapse
|
12
|
Mao Y, Tokudome T, Kishimoto I, Otani K, Nishimura H, Yamaguchi O, Otsu K, Miyazato M, Kangawa K. Endogenous ghrelin attenuates pressure overload-induced cardiac hypertrophy via a cholinergic anti-inflammatory pathway. Hypertension 2015; 65:1238-44. [PMID: 25870195 DOI: 10.1161/hypertensionaha.114.04864] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 03/01/2015] [Indexed: 01/20/2023]
Abstract
Cardiac hypertrophy, which is commonly caused by hypertension, is a major risk factor for heart failure and sudden death. Endogenous ghrelin has been shown to exert a beneficial effect on cardiac dysfunction and postinfarction remodeling via modulation of the autonomic nervous system. However, ghrelin's ability to attenuate cardiac hypertrophy and its potential mechanism of action are unknown. In this study, cardiac hypertrophy was induced by transverse aortic constriction in ghrelin knockout mice and their wild-type littermates. After 12 weeks, the ghrelin knockout mice showed significantly increased cardiac hypertrophy compared with wild-type mice, as evidenced by their significantly greater heart weight/tibial length ratios (9.2±1.9 versus 7.9±0.8 mg/mm), left ventricular anterior wall thickness (1.3±0.2 versus 1.0±0.2 mm), and posterior wall thickness (1.1±0.3 versus 0.9±0.1 mm). Furthermore, compared with wild-type mice, ghrelin knockout mice showed suppression of the cholinergic anti-inflammatory pathway, as indicated by reduced parasympathetic nerve activity and higher plasma interleukin-1β and interleukin-6 levels. The administration of either nicotine or ghrelin activated the cholinergic anti-inflammatory pathway and attenuated cardiac hypertrophy in ghrelin knockout mice. In conclusion, our results show that endogenous ghrelin plays a crucial role in the progression of pressure overload-induced cardiac hypertrophy via a mechanism that involves the activation of the cholinergic anti-inflammatory pathway.
Collapse
Affiliation(s)
- Yuanjie Mao
- From the Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan (Y.M., T.T., I.K., H.N., M.M., K.K.); Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center Research Institute (K.O.), Department of Cardiovascular Medicine, Graduate School of Medicine (O.Y.), Osaka University, Suita, Japan; and Cardiovascular Division, King's College London British Heart Foundation Centre, London, United Kingdom (K.O.)
| | - Takeshi Tokudome
- From the Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan (Y.M., T.T., I.K., H.N., M.M., K.K.); Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center Research Institute (K.O.), Department of Cardiovascular Medicine, Graduate School of Medicine (O.Y.), Osaka University, Suita, Japan; and Cardiovascular Division, King's College London British Heart Foundation Centre, London, United Kingdom (K.O.)
| | - Ichiro Kishimoto
- From the Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan (Y.M., T.T., I.K., H.N., M.M., K.K.); Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center Research Institute (K.O.), Department of Cardiovascular Medicine, Graduate School of Medicine (O.Y.), Osaka University, Suita, Japan; and Cardiovascular Division, King's College London British Heart Foundation Centre, London, United Kingdom (K.O.).
| | - Kentaro Otani
- From the Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan (Y.M., T.T., I.K., H.N., M.M., K.K.); Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center Research Institute (K.O.), Department of Cardiovascular Medicine, Graduate School of Medicine (O.Y.), Osaka University, Suita, Japan; and Cardiovascular Division, King's College London British Heart Foundation Centre, London, United Kingdom (K.O.)
| | - Hirohito Nishimura
- From the Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan (Y.M., T.T., I.K., H.N., M.M., K.K.); Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center Research Institute (K.O.), Department of Cardiovascular Medicine, Graduate School of Medicine (O.Y.), Osaka University, Suita, Japan; and Cardiovascular Division, King's College London British Heart Foundation Centre, London, United Kingdom (K.O.)
| | - Osamu Yamaguchi
- From the Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan (Y.M., T.T., I.K., H.N., M.M., K.K.); Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center Research Institute (K.O.), Department of Cardiovascular Medicine, Graduate School of Medicine (O.Y.), Osaka University, Suita, Japan; and Cardiovascular Division, King's College London British Heart Foundation Centre, London, United Kingdom (K.O.)
| | - Kinya Otsu
- From the Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan (Y.M., T.T., I.K., H.N., M.M., K.K.); Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center Research Institute (K.O.), Department of Cardiovascular Medicine, Graduate School of Medicine (O.Y.), Osaka University, Suita, Japan; and Cardiovascular Division, King's College London British Heart Foundation Centre, London, United Kingdom (K.O.)
| | - Mikiya Miyazato
- From the Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan (Y.M., T.T., I.K., H.N., M.M., K.K.); Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center Research Institute (K.O.), Department of Cardiovascular Medicine, Graduate School of Medicine (O.Y.), Osaka University, Suita, Japan; and Cardiovascular Division, King's College London British Heart Foundation Centre, London, United Kingdom (K.O.)
| | - Kenji Kangawa
- From the Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan (Y.M., T.T., I.K., H.N., M.M., K.K.); Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center Research Institute (K.O.), Department of Cardiovascular Medicine, Graduate School of Medicine (O.Y.), Osaka University, Suita, Japan; and Cardiovascular Division, King's College London British Heart Foundation Centre, London, United Kingdom (K.O.)
| |
Collapse
|
13
|
De Raedt S, De Vos A, De Keyser J. Autonomic dysfunction in acute ischemic stroke: an underexplored therapeutic area? J Neurol Sci 2014; 348:24-34. [PMID: 25541326 DOI: 10.1016/j.jns.2014.12.007] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Revised: 11/30/2014] [Accepted: 12/02/2014] [Indexed: 01/04/2023]
Abstract
Impaired autonomic function, characterized by a predominance of sympathetic activity, is common in patients with acute ischemic stroke. This review describes methods to measure autonomic dysfunction in stroke patients. It summarizes a potential relationship between ischemic stroke-associated autonomic dysfunction and factors that have been associated with worse outcome, including cardiac complications, blood pressure variability changes, hyperglycemia, immune depression, sleep disordered breathing, thrombotic effects, and malignant edema. Involvement of the insular cortex has been suspected to play an important role in causing sympathovagal imbalance, but its exact role and that of other brain regions remain unclear. Although sympathetic overactivity in patients with ischemic stroke appears to be a negative prognostic factor, it remains to be seen whether therapeutic strategies that reduce sympathetic activity or increase parasympathetic activity might improve outcome.
Collapse
Affiliation(s)
- Sylvie De Raedt
- Department of Neurology, Universitair Ziekenhuis Brussel, Center for Neurosciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium.
| | - Aurelie De Vos
- Department of Neurology, Universitair Ziekenhuis Brussel, Center for Neurosciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium.
| | - Jacques De Keyser
- Department of Neurology, Universitair Ziekenhuis Brussel, Center for Neurosciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium; Department of Neurology, Universitair Medisch Centrum Groningen, Groningen, The Netherlands.
| |
Collapse
|
14
|
Soeki T, Koshiba K, Niki T, Kusunose K, Yamaguchi K, Yamada H, Wakatsuki T, Shimabukuro M, Minakuchi K, Kishimoto I, Kangawa K, Sata M. Effect of ghrelin on autonomic activity in healthy volunteers. Peptides 2014; 62:1-5. [PMID: 25265271 DOI: 10.1016/j.peptides.2014.09.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Revised: 09/18/2014] [Accepted: 09/18/2014] [Indexed: 02/06/2023]
Abstract
Ghrelin is a novel growth hormone (GH)-releasing peptide originally isolated from the stomach. Recently, we have shown that ghrelin suppresses cardiac sympathetic activity and prevents early left ventricular remodeling in rats with myocardial infarction. In the present study, we evaluated the effect of ghrelin on autonomic nerve activity in healthy human subjects. An intravenous bolus of human synthetic ghrelin (10μg/kg) was administered to 10 healthy men (mean age, 33 years). Holter monitoring assessment was performed before and during 2h after the ghrelin therapy. The standard deviation of normal RR intervals (SDNN), square root of the mean of the sum of the squares of differences between adjacent RR intervals (rMSSD), high-frequency power (HF), and low-frequency power (LF) were analyzed. Blood samples were also obtained before and after the therapy. A single administration of ghrelin decreased both heart rate and blood pressure. Interestingly, ghrelin significantly decreased the LF and LF/HF ratio of heart rate variability and increased the SDNN, rMSSD, and HF. Ghrelin also elicited a marked increase in circulating GH, but not insulin-like growth factor-1. These data suggest that ghrelin might suppress cardiac sympathetic nerve activity and stimulate cardiac parasympathetic nerve activity.
Collapse
Affiliation(s)
- Takeshi Soeki
- Department of Cardiovascular Medicine, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Japan.
| | - Kunihiko Koshiba
- Department of Cardiovascular Medicine, Anan Central Hospital of the Medical Association, Anan, Japan
| | - Toshiyuki Niki
- Department of Cardiovascular Medicine, Shikoku Medical Center for Children and Adults, Zentsuji, Japan
| | - Kenya Kusunose
- Department of Cardiovascular Medicine, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Japan
| | - Koji Yamaguchi
- Department of Cardiovascular Medicine, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Japan
| | - Hirotsugu Yamada
- Department of Cardiovascular Medicine, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Japan
| | - Tetsuzo Wakatsuki
- Department of Cardiovascular Medicine, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Japan
| | - Michio Shimabukuro
- Department of Cardio-Diabetes Medicine, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Japan
| | - Kazuo Minakuchi
- Department of Clinical Pharmacy, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Japan
| | - Ichiro Kishimoto
- National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan
| | - Kenji Kangawa
- National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan
| | - Masataka Sata
- Department of Cardiovascular Medicine, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Japan
| |
Collapse
|
15
|
Du CK, Zhan DY, Morimoto S, Akiyama T, Schwenke DO, Hosoda H, Kangawa K, Shirai M. Survival benefit of ghrelin in the heart failure due to dilated cardiomyopathy. Pharmacol Res Perspect 2014; 2:e00064. [PMID: 25505608 PMCID: PMC4186424 DOI: 10.1002/prp2.64] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Accepted: 06/23/2014] [Indexed: 11/07/2022] Open
Abstract
Although ghrelin has been demonstrated to improve cardiac function in heart failure, its therapeutic efficacy on the life expectancy remains unknown. We aim to examine whether ghrelin can improve the life survival in heart failure using a mouse model of inherited dilated cardiomyopathy (DCM) caused by a deletion mutation ΔK210 in cardiac troponin T (cTnT). From 30 days of age, ghrelin (150 μg/kg) was administered subcutaneously to DCM mice once daily, control mice received saline only. The survival rates were compared between the two groups for 30 days. After 30-day treatment, functional and morphological measurements were conducted. Ghrelin-treated DCM mice had significantly prolonged life spans compared with saline-treated control DCM mice. Echocardiography showed that ghrelin reduced left ventricular (LV) end-diastolic dimensions and increased LV ejection fraction. Moreover, histoanatomical data revealed that ghrelin decreased the heart-to-body weight ratio, prevented cardiac remodeling and fibrosis, and markedly decreased the expression of brain natriuretic peptide. Telemetry recording and heart rate variability analysis showed that ghrelin suppressed the excessive cardiac sympathetic nerve activity (CSNA) and recovered the cardiac parasympathetic nerve activity. These results suggest that ghrelin has therapeutic benefits for survival as well as for the cardiac function and remodeling in heart failure probably through suppression of CSNA and recovery of cardiac parasympathetic nerve activity.
Collapse
Affiliation(s)
- Cheng-Kun Du
- Department of Cardiac Physiology, National Cerebral and Cardiovascular Center Research Institute Osaka, Japan
| | - Dong-Yun Zhan
- Department of Cardiac Physiology, National Cerebral and Cardiovascular Center Research Institute Osaka, Japan
| | - Sachio Morimoto
- Department of Clinical Pharmacology, Graduate School of Medical Sciences, Kyushu University Fukuoka, Japan
| | - Tsuyoshi Akiyama
- Department of Cardiac Physiology, National Cerebral and Cardiovascular Center Research Institute Osaka, Japan
| | - Daryl O Schwenke
- Department of Physiology, University of Otago Otago, New Zealand
| | - Hiroshi Hosoda
- Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center Research Institute Osaka, Japan
| | - Kenji Kangawa
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute Osaka, Japan
| | - Mikiyasu Shirai
- Department of Cardiac Physiology, National Cerebral and Cardiovascular Center Research Institute Osaka, Japan
| |
Collapse
|
16
|
Mao Y, Tokudome T, Kishimoto I. Ghrelin as a treatment for cardiovascular diseases. Hypertension 2014; 64:450-4. [PMID: 24958496 DOI: 10.1161/hypertensionaha.114.03726] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Affiliation(s)
- Yuanjie Mao
- From the Department of Biochemistry (Y.M., T.T.) and Department of Endocrinology and Metabolism (I.K.), National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Takeshi Tokudome
- From the Department of Biochemistry (Y.M., T.T.) and Department of Endocrinology and Metabolism (I.K.), National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Ichiro Kishimoto
- From the Department of Biochemistry (Y.M., T.T.) and Department of Endocrinology and Metabolism (I.K.), National Cerebral and Cardiovascular Center, Osaka, Japan.
| |
Collapse
|
17
|
Ulrich-Lai YM, Ryan KK. Neuroendocrine circuits governing energy balance and stress regulation: functional overlap and therapeutic implications. Cell Metab 2014; 19:910-25. [PMID: 24630812 PMCID: PMC4047143 DOI: 10.1016/j.cmet.2014.01.020] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Significant comorbidities between obesity-related metabolic disease and stress-related psychological disorders suggest important functional interactions between energy balance and brain stress integration. Largely overlapping neural circuits control these systems, and this anatomical arrangement optimizes opportunities for mutual influence. Here we first review the current literature identifying effects of metabolic neuroendocrine signals on stress regulation, and vice versa. Next, the contributions of reward-driven food intake to these metabolic and stress interactions are discussed. Lastly, we consider the interrelationships between metabolism, stress, and reward in light of their important implications in the development of therapies for metabolism- or stress-related disease.
Collapse
Affiliation(s)
- Yvonne M Ulrich-Lai
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, OH 45237, USA
| | - Karen K Ryan
- Department of Internal Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Cincinnati College of Medicine, Cincinnati, OH 45237, USA.
| |
Collapse
|
18
|
Mao Y, Tokudome T, Kishimoto I, Otani K, Hosoda H, Nagai C, Minamino N, Miyazato M, Kangawa K. Hexarelin treatment in male ghrelin knockout mice after myocardial infarction. Endocrinology 2013; 154:3847-54. [PMID: 23861368 DOI: 10.1210/en.2013-1291] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Both ghrelin and the synthetic analog hexarelin are reported to possess cardioprotective actions that are mainly exerted through different receptors. However, their effects on acute myocardial infarction have not been compared in vivo. This study aimed to clarify whether hexarelin treatment can compensate for ghrelin deficiency in ghrelin-knockout mice and to compare the effects of hexarelin (400 nmol/kg/d, sc) and equimolar ghrelin treatment after myocardial infarction. Myocardial infarction was produced by left coronary artery ligation in male ghrelin-knockout mice, which then received ghrelin, hexarelin, or vehicle treatment for 2 weeks. The mortality within 2 weeks was significantly lower in the hexarelin group (6.7%) and ghrelin group (14.3%) than in the vehicle group (50%) (P < .05). A comparison of cardiac function 2 weeks after infarction showed that in the ghrelin and hexarelin treatment groups, cardiac output was greater, whereas systolic function, represented by ejection fraction, and diastolic function, represented by dP/dt min (peak rate of pressure decline), were significantly superior compared with the vehicle group (P < .05). Hexarelin treatment was more effective than ghrelin treatment, as indicated by the ejection fraction, dP/dt max (peak rate of pressure rise), and dP/dt min. Telemetry recording and heart rate variability analysis demonstrated that sympathetic nervous activity was clearly suppressed in the hexarelin and ghrelin groups relative to the vehicle group. Our data demonstrated that hexarelin treatment can result in better heart function than ghrelin treatment 2 weeks after myocardial infarction in ghrelin-knockout mice, although both hormones have similar effects on heart rate variability and mortality.
Collapse
Affiliation(s)
- Yuanjie Mao
- Department of Biochemistry, National Cardiovascular Center Research Institute, 5-7-1 Fujishiro-dai, Suita, Osaka 565-8565, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Mao Y, Tokudome T, Otani K, Kishimoto I, Miyazato M, Kangawa K. Excessive sympathoactivation and deteriorated heart function after myocardial infarction in male ghrelin knockout mice. Endocrinology 2013; 154:1854-63. [PMID: 23515286 DOI: 10.1210/en.2012-2132] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We have previously demonstrated the protective role of endogenous ghrelin against malignant arrhythmias in the very acute phase of myocardial infarction (MI). However, the role of endogenous ghrelin in the chronic phase is unknown. Therefore, the aim of the current study was to focus on the effects of endogenous ghrelin on cardiac function and sympathetic activation after acute MI. In 46 ghrelin-knockout (KO) and 41 wild-type (WT) male mice, MI was produced by left coronary artery ligation. The mortality due to heart failure within 2 weeks was 0% in WT and 10.9% in KO (P < 0.05). At the end of this period, lung weight/tibial length, atrial natriuretic peptide and brain natriuretic peptide transcripts, end-systolic and end-diastolic volumes were all significantly greater in KO mice, whereas systolic function, represented by ejection fraction (16.4 ± 4.7% vs 25.3 ± 5.1%), end-systolic elastance, and preload-recruitable stroke work, was significantly inferior to that in WT mice (P < 0.05). Telemetry recording and heart rate variability analysis showed that KO mice had stronger sympathetic activation after MI than did WT mice. Metoprolol treatment and ghrelin treatment in KO mice prevented excessive sympathetic activation, decreased plasma epinephrine and norepinephrine levels, and improved heart function and survival rate after MI. Our data demonstrate that endogenous ghrelin plays a crucial role in protecting heart function and reducing mortality after myocardial infarction, and that these effects seem to be partly the result of sympathetic inhibition.
Collapse
Affiliation(s)
- Yuanjie Mao
- Department of Biochemistry, National Cardiovascular Center Research Institute, 5-7-1 Fujishiro-dai, Suita, Osaka 565-8565, Japan
| | | | | | | | | | | |
Collapse
|
20
|
May CN, Calzavacca P, Ishikawa K, Langenberg C, Wan L, Ramchandra R, Bellomo R. Novel targets for sepsis-induced kidney injury: the glomerular arterioles and the sympathetic nervous system. Exp Physiol 2012; 97:1168-77. [DOI: 10.1113/expphysiol.2011.061804] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
21
|
Savalle M, Gillaizeau F, Maruani G, Puymirat E, Bellenfant F, Houillier P, Fagon JY, Faisy C. Assessment of body cell mass at bedside in critically ill patients. Am J Physiol Endocrinol Metab 2012; 303:E389-96. [PMID: 22649067 DOI: 10.1152/ajpendo.00502.2011] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Critical illness affects body composition profoundly, especially body cell mass (BCM). BCM loss reflects lean tissue wasting and could be a nutritional marker in critically ill patients. However, BCM assessment with usual isotopic or tracer methods is impractical in intensive care units (ICUs). We aimed to modelize the BCM of critically ill patients using variables available at bedside. Fat-free mass (FFM), bone mineral (Mo), and extracellular water (ECW) of 49 critically ill patients were measured prospectively by dual-energy X-ray absorptiometry and multifrequency bioimpedance. BCM was estimated according to the four-compartment cellular level: BCM = FFM - (ECW/0.98) - (0.73 × Mo). Variables that might influence the BCM were assessed, and multivariable analysis using fractional polynomials was conducted to determine the relations between BCM and these data. Bootstrap resampling was then used to estimate the most stable model predicting BCM. BCM was 22.7 ± 5.4 kg. The most frequent model included height (cm), leg circumference (cm), weight shift (Δ) between ICU admission and body composition assessment (kg), and trunk length (cm) as a linear function: BCM (kg) = 0.266 × height + 0.287 × leg circumference + 0.305 × Δweight - 0.406 × trunk length - 13.52. The fraction of variance explained by this model (adjusted r(2)) was 46%. Including bioelectrical impedance analysis variables in the model did not improve BCM prediction. In summary, our results suggest that BCM can be estimated at bedside, with an error lower than ±20% in 90% subjects, on the basis of static (height, trunk length), less stable (leg circumference), and dynamic biometric variables (Δweight) for critically ill patients.
Collapse
|
22
|
Mao Y, Tokudome T, Otani K, Kishimoto I, Nakanishi M, Hosoda H, Miyazato M, Kangawa K. Ghrelin prevents incidence of malignant arrhythmia after acute myocardial infarction through vagal afferent nerves. Endocrinology 2012; 153:3426-34. [PMID: 22535766 DOI: 10.1210/en.2012-1065] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Ghrelin is a GH-releasing peptide mainly excreted from the stomach. Ghrelin administration has been shown to inhibit cardiac sympathetic nerve activity (CSNA), reduce malignant arrhythmia, and improve prognosis after acute myocardial infarction (MI). We therefore investigated the effects and potential mechanisms of the action of endogenous ghrelin on survival rate and CSNA after MI by using ghrelin-knockout (KO) mice. MI was induced by left coronary artery ligation in 46 KO mice and 41 wild-type mice. On the first day, malignant arrhythmia-induced mortality was observed within 30 min of the ligation and had an incidence of 2.4% in wild-type and 17.4% in KO mice (P < 0.05). We next evaluated CSNA by spectral analysis of heart rate variability. CSNA, represented by the low frequency/high frequency ratio, was higher in KO mice at baseline (2.18 ± 0.43 vs. 0.98 ± 0.09; P < 0.05), and especially after MI (25.5 ± 11.8 vs. 1.4 ± 0.3; P < 0.05), than in wild-type mice. Ghrelin (150 μg/kg, s.c.) 15 min before ligation suppressed the activation of CSNA and reduced mortality in KO mice. Further, this effect of ghrelin was inhibited by methylatropine bromide (1 mg/kg, i.p.) or by perineural treatment of both cervical vagal trunks with capsaicin (a specific afferent neurotoxin). Our data demonstrated that both exogenous and endogenous ghrelin suppressed CSNA, prevented the incidence of malignant arrhythmia, and improved the prognosis after acute MI. These effects are likely to be via the vagal afferent nerves.
Collapse
Affiliation(s)
- Yuanjie Mao
- Department of Biochemistry, National Cerebral and Cardiovascular Center, Suita, Osaka 565-8565, Japan
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Schwenke DO, Tokudome T, Kishimoto I, Horio T, Cragg PA, Shirai M, Kangawa K. One dose of ghrelin prevents the acute and sustained increase in cardiac sympathetic tone after myocardial infarction. Endocrinology 2012; 153:2436-43. [PMID: 22434083 DOI: 10.1210/en.2011-2057] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Acute myocardial infarction (MI) increases sympathetic nerve activity (SNA) to the heart, which exacerbates chronic cardiac deterioration. The hormone ghrelin, if administered soon after an MI, prevents the increase in cardiac SNA and improves early survival prognosis. Whether these early beneficial effects of ghrelin also impact on cardiac function in chronic heart failure has not yet been addressed and thus was the aim of this study. MI was induced in Sprague Dawley rats by ligating the left coronary artery. One bolus of saline (n = 7) or ghrelin (150 μg/kg, sc, n = 9) was administered within 30 min of MI. Two weeks after the infarct (or sham; n = 7), rats were anesthetized and cardiac function was evaluated using a Millar pressure-volume conductance catheter. Cardiac SNA was measured using whole-nerve electrophysiological techniques. Untreated-MI rats had a high mortality rate (50%), evidence of severe cardiac dysfunction (ejection fraction 28%; P < 0.001), and SNA was significantly elevated (102% increase; P = 0.03). In comparison, rats that received a single dose of ghrelin after the MI tended to have a lower mortality rate (25%; P = NS) and no increase in SNA, and cardiac dysfunction was attenuated (ejection fraction of 43%; P = 0.014). This study implicates ghrelin as a potential clinical treatment for acute MI but also highlights the importance of therapeutic intervention in the early stages after acute MI. Moreover, these results uncover an intricate causal relationship between early and chronic changes in the neural control of cardiac function in heart failure.
Collapse
Affiliation(s)
- Daryl O Schwenke
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, 5-7-1 Fujishirodai, Suita, Osaka 565-8565, Japan
| | | | | | | | | | | | | |
Collapse
|
24
|
Abstract
Sepsis, a systemic inflammatory response to infection, continues to carry a high mortality despite advances in critical care medicine. Elevated sympathetic nerve activity in sepsis has been shown to contribute to early hepatocellular dysfunction and subsequently multiple organ failure, resulting in a poor prognosis, especially in the elderly. Thus, suppression of sympathetic nerve activity represents a novel therapeutic option for sepsis. Ghrelin is a 28-amino acid peptide shown to inhibit sympathetic nerve activity and inflammation in animal models of tissue injury. Age-related ghrelin hyporesponsiveness has also been shown to exacerbate sepsis. However, the mechanistic relationship between ghrelin-mediated sympathoinhibition and suppression of inflammation remains poorly understood. This review assesses the therapeutic potential of ghrelin in sepsis in the context of the neuroanatomical and molecular basis of ghrelin-mediated suppression of inflammation through inhibition of central sympathetic outflow.
Collapse
Affiliation(s)
- Cletus Cheyuo
- Elmezzi Graduate School of Molecular Medicine, Hofstra North Shore-LIJ Medical School, Manhasset, New York, USA
| | | | | |
Collapse
|
25
|
Shimizu S, Akiyama T, Kawada T, Sata Y, Mizuno M, Kamiya A, Shishido T, Inagaki M, Shirai M, Sano S, Sugimachi M. Medetomidine, an α(2)-adrenergic agonist, activates cardiac vagal nerve through modulation of baroreflex control. Circ J 2011; 76:152-9. [PMID: 22040937 DOI: 10.1253/circj.cj-11-0574] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND Although α(2)-adrenergic agonists have been reported to induce a vagal-dominant condition through suppression of sympathetic nerve activity, there is little direct evidence that they directly increase cardiac vagal nerve activity. Using a cardiac microdialysis technique, we investigated the effects of medetomidine, an α(2)-adrenergic agonist, on norepinephrine (NE) and acetylcholine (ACh) release from cardiac nerve endings. METHODS AND RESULTS A microdialysis probe was implanted into the right atrial wall near the sinoatrial node in anesthetized rabbits and perfused with Ringer's solution containing eserine. Dialysate NE and ACh concentrations were measured using high-performance liquid chromatography. Both 10 and 100µg/kg of intravenous medetomidine significantly decreased mean blood pressure (BP) and the dialysate NE concentration, but only 100µg/kg of medetomidine enhanced ACh release. Combined administration of medetomidine and phenylephrine maintained mean BP at baseline level, and augmented the medetomidine-induced ACh release. When we varied the mean BP using intravenous administration of phenylephrine, treatment with medetomidine significantly steepened the slope of the regression line between mean BP and log ACh concentration. CONCLUSIONS Medetomidine increased ACh release from cardiac vagal nerve endings and augmented baroreflex control of vagal nerve activity.
Collapse
Affiliation(s)
- Shuji Shimizu
- Department of Cardiovascular Dynamics, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|