1
|
Lund LH, Hage C, Pironti G, Thorvaldsen T, Ljung-Faxén U, Zabarovskaja S, Shahgaldi K, Webb DL, Hellström PM, Andersson DC, Ståhlberg M. Acyl ghrelin improves cardiac function in heart failure and increases fractional shortening in cardiomyocytes without calcium mobilization. Eur Heart J 2023; 44:2009-2025. [PMID: 36916707 PMCID: PMC10256198 DOI: 10.1093/eurheartj/ehad100] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 01/05/2023] [Accepted: 02/13/2023] [Indexed: 03/16/2023] Open
Abstract
BACKGROUND AND AIMS Ghrelin is an endogenous appetite-stimulating peptide hormone with potential cardiovascular benefits. Effects of acylated (activated) ghrelin were assessed in patients with heart failure and reduced ejection fraction (HFrEF) and in ex vivo mouse cardiomyocytes. METHODS AND RESULTS In a randomized placebo-controlled double-blind trial, 31 patients with chronic HFrEF were randomized to synthetic human acyl ghrelin (0.1 µg/kg/min) or placebo intravenously over 120 min. The primary outcome was change in cardiac output (CO). Isolated mouse cardiomyocytes were treated with acyl ghrelin and fractional shortening and calcium transients were assessed. Acyl ghrelin but not placebo increased cardiac output (acyl ghrelin: 4.08 ± 1.15 to 5.23 ± 1.98 L/min; placebo: 4.26 ± 1.23 to 4.11 ± 1.99 L/min, P < 0.001). Acyl ghrelin caused a significant increase in stroke volume and nominal increases in left ventricular ejection fraction and segmental longitudinal strain and tricuspid annular plane systolic excursion. There were no effects on blood pressure, arrhythmias, or ischaemia. Heart rate decreased nominally (acyl ghrelin: 71 ± 11 to 67 ± 11 b.p.m.; placebo 69 ± 8 to 68 ± 10 b.p.m.). In cardiomyocytes, acyl ghrelin increased fractional shortening, did not affect cellular Ca2+ transients, and reduced troponin I phosphorylation. The increase in fractional shortening and reduction in troponin I phosphorylation was blocked by the acyl ghrelin antagonist D-Lys 3. CONCLUSION In patients with HFrEF, acyl ghrelin increased cardiac output without causing hypotension, tachycardia, arrhythmia, or ischaemia. In isolated cardiomyocytes, acyl ghrelin increased contractility independently of preload and afterload and without Ca2+ mobilization, which may explain the lack of clinical side effects. Ghrelin treatment should be explored in additional randomized trials. CLINICAL TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT05277415.
Collapse
Affiliation(s)
- Lars H Lund
- Department of Medicine, Unit of Cardiology, Karolinska Institutet, D1:04, 171 76 Stockholm, Sweden
- Heart and Vascular Theme, Karolinska University Hospital, Norrbacka, S1:02, 171 76 Stockholm, Sweden
| | - Camilla Hage
- Department of Medicine, Unit of Cardiology, Karolinska Institutet, D1:04, 171 76 Stockholm, Sweden
- Heart and Vascular Theme, Karolinska University Hospital, Norrbacka, S1:02, 171 76 Stockholm, Sweden
| | - Gianluigi Pironti
- Department of Medicine, Unit of Cardiology, Karolinska Institutet, D1:04, 171 76 Stockholm, Sweden
- Department of Physiology and Pharmacology, Karolinska Institutet, Biomedicum, Solnavägen 9 171 65 Solna, Sweden
| | - Tonje Thorvaldsen
- Department of Medicine, Unit of Cardiology, Karolinska Institutet, D1:04, 171 76 Stockholm, Sweden
- Heart and Vascular Theme, Karolinska University Hospital, Norrbacka, S1:02, 171 76 Stockholm, Sweden
| | - Ulrika Ljung-Faxén
- Department of Medicine, Unit of Cardiology, Karolinska Institutet, D1:04, 171 76 Stockholm, Sweden
- Perioperative Medicine and Intensive Care, Karolinska University Hospital, 171 76 Stockholm, Sweden
| | - Stanislava Zabarovskaja
- Department of Medicine, Unit of Cardiology, Karolinska Institutet, D1:04, 171 76 Stockholm, Sweden
| | - Kambiz Shahgaldi
- Department of Clinical Physiology, Sunderby Hospital, 971 80 Luleå, Sweden
| | - Dominic-Luc Webb
- Department of Medical Sciences, Gastroenterology and Hepatology, Uppsala University, 751 05 Uppsala, Sweden
| | - Per M Hellström
- Department of Medical Sciences, Gastroenterology and Hepatology, Uppsala University, 751 05 Uppsala, Sweden
| | - Daniel C Andersson
- Department of Medicine, Unit of Cardiology, Karolinska Institutet, D1:04, 171 76 Stockholm, Sweden
- Heart and Vascular Theme, Karolinska University Hospital, Norrbacka, S1:02, 171 76 Stockholm, Sweden
- Department of Physiology and Pharmacology, Karolinska Institutet, Biomedicum, Solnavägen 9 171 65 Solna, Sweden
| | - Marcus Ståhlberg
- Department of Medicine, Unit of Cardiology, Karolinska Institutet, D1:04, 171 76 Stockholm, Sweden
- Heart and Vascular Theme, Karolinska University Hospital, Norrbacka, S1:02, 171 76 Stockholm, Sweden
| |
Collapse
|
2
|
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: 2.5] [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
|
3
|
Speer KE, Koenig J, Telford RM, Olive LS, Mara JK, Semple S, Naumovski N, Telford RD, McKune AJ. Relationship between heart rate variability and body mass index: A cross-sectional study of preschool children. Prev Med Rep 2021; 24:101638. [PMID: 34976689 PMCID: PMC8684011 DOI: 10.1016/j.pmedr.2021.101638] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 10/02/2021] [Accepted: 11/13/2021] [Indexed: 11/29/2022] Open
Abstract
Heart rate variability and BMI are inversely related in preschool children. One unit increase in BMI resulted in a reduction in RMSSD(ln) of 0.06% Age, sex and physical activity levels did not influence this relationship.
Reduced heart rate variability (HRV) is associated with overweight and obesity in adults. However, little is known about this relationship in early childhood. We investigated the relationship between resting vagally-mediated HRV and body mass index (BMI) in Australian preschool children. Children were recruited from 13 non-government early learning centres located in Queensland and New South Wales, Australia. From this population-based sample, data from 146 healthy children (58 females) between 3 and 5 years of age (mean age 4.35 ± 0.44 years) were analysed. BMI was calculated from child body weight and height. Physical activity was recorded using an Actigraph wGT3x accelerometer worn at the waist of participants over 3 consecutive days. A Polar H10 chest strap measured seated, resting RR intervals for the calculation of HRV with the root mean square of successive differences (RMSSD) reflecting vagally-mediated activity. The relationship between HRV and BMI was analysed using a linear mixed model adjusted for age, sex and physical activity. Analysis revealed that RMSSD (ln) demonstrated a significant inverse relationship with BMI (β = -0.06; 95% CI = -0.12 – −0.01; p = 0.032), and the model accounted for 23% of the variance in RMSSD (ln). Notably, a one unit increase in BMI resulted in a reduction in RMSDD (ln) of 0.06. This investigation demonstrated evidence for a significant inverse linear relationship between vagally-mediated HRV and BMI in 3 – 5-year-old Australian children, similar to that of adults. Furthermore, this relationship was independent of age, sex and physical activity levels. Results may indicate that the cardiometabolic health of preschool children is, in part, influenced by the relationship between vagally-mediated HRV and weight status.
Collapse
Affiliation(s)
- Kathryn E Speer
- Faculty of Health, Discipline of Sport and Exercise Science/University of Canberra, Canberra, ACT, Australia.,Research Institute for Sport and Exercise/University of Canberra, Canberra, ACT, Australia
| | - Julian Koenig
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Cologne, Germany
| | - Rohan M Telford
- Research Institute for Sport and Exercise/University of Canberra, Canberra, ACT, Australia
| | - Lisa S Olive
- Centre for Social and Early Emotional Development, Faculty of Health, Deakin University, Geelong, Victoria, Australia.,IMPACT, The Institute for Mental and Physical Health and Clinical Translation, Faculty of Health, Deakin University, Geelong, Victoria, Australia
| | - Jocelyn K Mara
- Faculty of Health, Discipline of Sport and Exercise Science/University of Canberra, Canberra, ACT, Australia.,Research Institute for Sport and Exercise/University of Canberra, Canberra, ACT, Australia
| | - Stuart Semple
- Faculty of Health, Discipline of Sport and Exercise Science/University of Canberra, Canberra, ACT, Australia.,Research Institute for Sport and Exercise/University of Canberra, Canberra, ACT, Australia
| | - Nenad Naumovski
- Faculty of Health, University of Canberra, Discipline of Nutrition and Dietetics/University of Canberra, Canberra, ACT, Australia.,Functional Foods and Nutrition Research (FFNR) Laboratory, University of Canberra, Bruce, ACT, Australia.,Department of Nutrition and Dietetics, Harokopio University, Athens 17671, Greece
| | - Richard D Telford
- Research Institute for Sport and Exercise/University of Canberra, Canberra, ACT, Australia
| | - Andrew J McKune
- Faculty of Health, Discipline of Sport and Exercise Science/University of Canberra, Canberra, ACT, Australia.,Research Institute for Sport and Exercise/University of Canberra, Canberra, ACT, Australia.,Discipline of Biokinetics, Exercise and Leisure Sciences, School of Health Sciences/ University of KwaZulu-Natal, Durban, KwaZulu-Natal, South Africa
| |
Collapse
|
4
|
Sales da Silva E, Ferreira PM, Castro CH, Pacheco LF, Graziani D, Pontes CNR, Bessa ADSMD, Fernandes E, Naves LM, Ribeiro LCDS, Mendonça MM, Gomes RM, Pedrino GR, Ferreira RN, Xavier CH. Brain and kidney GHS-R1a underexpression is associated with changes in renal function and hemodynamics during neurogenic hypertension. Mol Cell Endocrinol 2020; 518:110984. [PMID: 32814069 DOI: 10.1016/j.mce.2020.110984] [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: 12/14/2019] [Revised: 08/07/2020] [Accepted: 08/07/2020] [Indexed: 10/23/2022]
Abstract
Ghrelin is a peptide hormone whose effects are mediated by the growth hormone secretagogue receptor subtype 1a (GHS-R1a), mainly expressed in the brain but also in kidneys. The hypothesis herein raised is that GHS-R1a would be player in the renal contribution to the neurogenic hypertension pathophysiology. To investigate GHS-R1a role on renal function and hemodynamics, we used Wistar (WT) and spontaneously hypertensive rats (SHR). First, we assessed the effect of systemically injected vehicle, ghrelin, GHS-R1a antagonist PF04628935, ghrelin plus PF04628935 or GHS-R1a synthetic agonist MK-677 in WT and SHR rats housed in metabolic cages (24 h). Blood and urine samples were also analyzed. Then, we assessed the GHS-R1a contribution to the control of renal vasomotion and hemodynamics in WT and SHR. Finally, we assessed the GHS-R1a levels in brain areas, aorta, renal artery, renal cortex and medulla of WT and SHR rats using western blot. We found that ghrelin and MK-677 changed osmolarity parameters of SHR, in a GHS-R1a-dependent manner. GHS-R1a antagonism reduced the urinary Na+ and K+ and creatinine clearance in WT but not in SHR. Ghrelin reduced arterial pressure and increased renal artery conductance in SHR. GHS-R1a protein levels were decreased in the kidney and brain areas of SHR when compared to WT. Therefore, GHS-R1a role in the control of renal function and hemodynamics during neurogenic hypertension seem to be different, and this may be related to brain and kidney GHS-R1a downregulation.
Collapse
Affiliation(s)
- Elder Sales da Silva
- Systems Neurobiology Laboratory, Department of Physiology, Institute of Biological Sciences, Room 203, Federal University of Goiás, Goiânia, GO, 74690-900, Brazil.
| | - Patrícia Maria Ferreira
- Systems Neurobiology Laboratory, Department of Physiology, Institute of Biological Sciences, Room 203, Federal University of Goiás, Goiânia, GO, 74690-900, Brazil.
| | - Carlos Henrique Castro
- Systems Neurobiology Laboratory, Department of Physiology, Institute of Biological Sciences, Room 203, Federal University of Goiás, Goiânia, GO, 74690-900, Brazil.
| | - Lilian Fernanda Pacheco
- Systems Neurobiology Laboratory, Department of Physiology, Institute of Biological Sciences, Room 203, Federal University of Goiás, Goiânia, GO, 74690-900, Brazil.
| | - Daniel Graziani
- Systems Neurobiology Laboratory, Department of Physiology, Institute of Biological Sciences, Room 203, Federal University of Goiás, Goiânia, GO, 74690-900, Brazil.
| | - Carolina Nobre Ribeiro Pontes
- Systems Neurobiology Laboratory, Department of Physiology, Institute of Biological Sciences, Room 203, Federal University of Goiás, Goiânia, GO, 74690-900, Brazil.
| | - Amanda de Sá Martins de Bessa
- Systems Neurobiology Laboratory, Department of Physiology, Institute of Biological Sciences, Room 203, Federal University of Goiás, Goiânia, GO, 74690-900, Brazil.
| | - Erika Fernandes
- Systems Neurobiology Laboratory, Department of Physiology, Institute of Biological Sciences, Room 203, Federal University of Goiás, Goiânia, GO, 74690-900, Brazil.
| | - Lara Marques Naves
- Systems Neurobiology Laboratory, Department of Physiology, Institute of Biological Sciences, Room 203, Federal University of Goiás, Goiânia, GO, 74690-900, Brazil.
| | - Larissa Cristina Dos Santos Ribeiro
- Systems Neurobiology Laboratory, Department of Physiology, Institute of Biological Sciences, Room 203, Federal University of Goiás, Goiânia, GO, 74690-900, Brazil.
| | - Michelle Mendanha Mendonça
- Systems Neurobiology Laboratory, Department of Physiology, Institute of Biological Sciences, Room 203, Federal University of Goiás, Goiânia, GO, 74690-900, Brazil.
| | - Rodrigo Mello Gomes
- Systems Neurobiology Laboratory, Department of Physiology, Institute of Biological Sciences, Room 203, Federal University of Goiás, Goiânia, GO, 74690-900, Brazil.
| | - Gustavo Rodrigues Pedrino
- Systems Neurobiology Laboratory, Department of Physiology, Institute of Biological Sciences, Room 203, Federal University of Goiás, Goiânia, GO, 74690-900, Brazil.
| | - Reginaldo Nassar Ferreira
- Systems Neurobiology Laboratory, Department of Physiology, Institute of Biological Sciences, Room 203, Federal University of Goiás, Goiânia, GO, 74690-900, Brazil.
| | - Carlos Henrique Xavier
- Systems Neurobiology Laboratory, Department of Physiology, Institute of Biological Sciences, Room 203, Federal University of Goiás, Goiânia, GO, 74690-900, Brazil.
| |
Collapse
|
5
|
Shati AA, Dallak M. Acylated Ghrelin Protects the Hearts of Rats from Doxorubicin-Induced Fas/FasL Apoptosis by Stimulating SERCA2a Mediated by Activation of PKA and Akt. Cardiovasc Toxicol 2020; 19:529-547. [PMID: 31093930 DOI: 10.1007/s12012-019-09527-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
This study investigated if the cardioprotective effect of acylated ghrelin (AG) against doxorubicin (DOX)-induced cardiac toxicity in rats involves inhibition of Fas/FasL-mediated cell death. It also investigated if such an effect is mediated by restoring Ca+2 homeostasis from the aspect of stimulation of SERCA2a receptors. Adult male Wistar rats were divided into 4 groups (20 rats/each) as control, control + AG, DOX, and DOX + AG. AG was co-administered to all rats consecutively for 35 days. In addition, isolated cardiomyocytes were cultured and treated with AG in the presence or absence of DOX with or without pre-incubation with [D-Lys3]-GHRP-6 (a AG receptor antagonist), VIII (]an Akt inhibitor), or KT-5720 (a PKA inhibitor). AG increased LVSP, dp/dtmax, and dp/dtmin in both control and DOX-treated animals and improved cardiac ultrastructural changes in DOX-treated rats. It also inhibited ROS in control rats and lowered LVEDP, intracellular levels of ROS and Ca2+, and activity of calcineurin in LVs of DOX-treated rats. Concomitantly, it inhibited LV NFAT-4 nuclear translocation and downregulated their protein levels of Fas and FasL. Mechanistically, in control or DOX-treated hearts or cells, AG upregulated the levels of SERCA2a and increased the activities of PKA and Akt, leading to increase phosphorylation of phospholamban at Ser16 and Thr17. All these effects were abolished by D-Lys3-GHRP-6, VIII, or KT-5720 and were independent of food intake or GH/IGF-1. In conclusion, AG cardioprotection against DOX involves inhibition of extrinsic cell death and restoring normal Ca+2 homeostasis.
Collapse
Affiliation(s)
- Ali A Shati
- Department of Biology, College of Science, King Khalid University, Abha, Saudi Arabia.
| | - M Dallak
- Department of Physiology, College of Medicine, King Khalid University, Abha, Saudi Arabia
| |
Collapse
|
6
|
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: 1.0] [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
|
7
|
Gray SM, Page LC, Tong J. Ghrelin regulation of glucose metabolism. J Neuroendocrinol 2019; 31:e12705. [PMID: 30849212 PMCID: PMC6688917 DOI: 10.1111/jne.12705] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 03/04/2019] [Accepted: 03/05/2019] [Indexed: 12/14/2022]
Abstract
Ghrelin and its receptor, the growth hormone secretagogue receptor 1a (GHSR1a), are implicated in the regulation of glucose metabolism via direct actions in the pancreatic islet, as well as peripheral insulin-sensitive tissues and the brain. Although many studies have explored the role of ghrelin in glucose tolerance and insulin secretion, a complete mechanistic understanding remains to be clarified. This review highlights the local expression and function of ghrelin and GHSR1a in pancreatic islets and how this axis may modulate insulin secretion from pancreatic β-cells. Additionally, we discuss the effect of ghrelin on in vivo glucose metabolism in rodents and humans, as well as the metabolic circumstances under which the action of ghrelin may predominate.
Collapse
Affiliation(s)
- Sarah. M. Gray
- Duke Molecular Physiology Institute, Duke University, Durham, NC 27701
| | - Laura C. Page
- Division of Endocrinology, Department of Pediatrics, Duke University, Durham, NC 27701
| | - Jenny Tong
- Duke Molecular Physiology Institute, Duke University, Durham, NC 27701
- Division of Endocrinology, Department of Pediatrics, Duke University, Durham, NC 27701
- Division of Endocrinology, Metabolism, and Nutrition, Department of Medicine, Duke University, Durham, NC 27701
| |
Collapse
|
8
|
Perello M, Cabral A, Cornejo MP, De Francesco PN, Fernandez G, Uriarte M. Brain accessibility delineates the central effects of circulating ghrelin. J Neuroendocrinol 2019; 31:e12677. [PMID: 30582239 DOI: 10.1111/jne.12677] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 12/04/2018] [Accepted: 12/04/2018] [Indexed: 12/27/2022]
Abstract
Ghrelin is a hormone produced in the gastrointestinal tract that acts via the growth hormone secretagogue receptor. In the central nervous system, ghrelin signalling is able to recruit different neuronal targets that regulate the behavioural, neuroendocrine, metabolic and autonomic effects of the hormone. Notably, several studies using radioactive or fluorescent variants of ghrelin have found that the accessibility of circulating ghrelin into the mouse brain is both strikingly low and restricted to some specific brain areas. A variety of studies addressing central effects of systemically injected ghrelin in mice have also provided indirect evidence that the accessibility of plasma ghrelin into the brain is limited. Here, we review these previous observations and discuss the putative pathways that would allow plasma ghrelin to gain access into the brain together with their physiological implications. Additionally, we discuss some potential features regarding the accessibility of plasma ghrelin into the human brain based on the observations reported by studies that investigate the consequences of ghrelin administration to humans.
Collapse
Affiliation(s)
- Mario Perello
- Laboratorio de Neurofisiología del Instituto Multidisciplinario de Biología Celular, Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, Universidad Nacional de La Plata y Comisión de Investigaciones Científicas-Provincia de Buenos Aires, Buenos Aires, Argentina
| | - Agustina Cabral
- Laboratorio de Neurofisiología del Instituto Multidisciplinario de Biología Celular, Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, Universidad Nacional de La Plata y Comisión de Investigaciones Científicas-Provincia de Buenos Aires, Buenos Aires, Argentina
| | - María P Cornejo
- Laboratorio de Neurofisiología del Instituto Multidisciplinario de Biología Celular, Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, Universidad Nacional de La Plata y Comisión de Investigaciones Científicas-Provincia de Buenos Aires, Buenos Aires, Argentina
| | - Pablo N De Francesco
- Laboratorio de Neurofisiología del Instituto Multidisciplinario de Biología Celular, Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, Universidad Nacional de La Plata y Comisión de Investigaciones Científicas-Provincia de Buenos Aires, Buenos Aires, Argentina
| | - Gimena Fernandez
- Laboratorio de Neurofisiología del Instituto Multidisciplinario de Biología Celular, Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, Universidad Nacional de La Plata y Comisión de Investigaciones Científicas-Provincia de Buenos Aires, Buenos Aires, Argentina
| | - Maia Uriarte
- Laboratorio de Neurofisiología del Instituto Multidisciplinario de Biología Celular, Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, Universidad Nacional de La Plata y Comisión de Investigaciones Científicas-Provincia de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
9
|
Farokhnia M, Faulkner ML, Piacentino D, Lee MR, Leggio L. Ghrelin: From a gut hormone to a potential therapeutic target for alcohol use disorder. Physiol Behav 2019; 204:49-57. [DOI: 10.1016/j.physbeh.2019.02.008] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 01/24/2019] [Accepted: 02/06/2019] [Indexed: 12/22/2022]
|
10
|
Farokhnia M, Lee MR, Farinelli LA, Ramchandani VA, Akhlaghi F, Leggio L. Pharmacological manipulation of the ghrelin system and alcohol hangover symptoms in heavy drinking individuals: Is there a link? Pharmacol Biochem Behav 2018; 172:39-49. [PMID: 30030128 DOI: 10.1016/j.pbb.2018.07.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 06/23/2018] [Accepted: 07/16/2018] [Indexed: 12/14/2022]
Abstract
Ghrelin, an orexigenic peptide synthesized in the stomach, is a key player in the gut-brain axis. In addition to its role in regulating food intake and energy homeostasis, ghrelin has been shown to modulate alcohol-related behaviors. Alcohol consumption frequently results in hangover, an underexplored phenomenon with considerable medical, psychological, and socioeconomic consequences. While the pathophysiology of hangover is not clear, contributions of mechanisms such as alcohol-induced metabolic/endocrine changes, inflammatory/immune response, oxidative stress, and gut dysbiosis have been reported. Interestingly, these mechanisms considerably overlap with ghrelin's physiological functions. Here, we investigated whether pharmacological manipulation of the ghrelin system may affect alcohol hangover symptoms. Data were obtained from two placebo-controlled laboratory studies. The first study tested the effects of intravenous (IV) ghrelin and consisted of two experiments: a progressive-ratio IV alcohol self-administration (IV-ASA) and a fixed-dose IV alcohol clamp. The second study tested the effects of an oral ghrelin receptor inverse agonist (PF-5190457) and included a fixed-dose oral alcohol administration experiment. Alcohol hangover data were collected the morning after each alcohol administration experiment using the Acute Hangover Scale (AHS). IV ghrelin, compared to placebo, significantly reduced alcohol hangover after IV-ASA (p = 0.04) and alcohol clamp (p = 0.04); PF-5190457 had no significant effect on AHS scores. Females reported significantly higher hangover symptoms than males following the IV-ASA experiment (p = 0.04), but no gender × drug condition (ghrelin vs. placebo) effect was found. AHS total scores were positively correlated with peak subjective responses, including 'stimulation' (p = 0.08), 'sedation' (p = 0.009), 'feel high' (p = 0.05), and 'feel intoxicated' (p = 0.03) during the IV-ASA. IV ghrelin blunted the positive association between alcohol sedation and hangover as shown by trend-level drug × sedation effect (p = 0.08). This is the first study showing that exogenous ghrelin administration, but not ghrelin receptor inverse agonism, affects hangover symptoms. Future research should investigate the potential mechanism(s) underlying this effect.
Collapse
Affiliation(s)
- Mehdi Farokhnia
- Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology, National Institute on Alcohol Abuse and Alcoholism and National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD, USA
| | - Mary R Lee
- Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology, National Institute on Alcohol Abuse and Alcoholism and National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD, USA
| | - Lisa A Farinelli
- Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology, National Institute on Alcohol Abuse and Alcoholism and National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD, USA
| | - Vijay A Ramchandani
- Section on Human Psychopharmacology, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Fatemeh Akhlaghi
- Clinical Pharmacokinetics Research Laboratory, Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, USA
| | - Lorenzo Leggio
- Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology, National Institute on Alcohol Abuse and Alcoholism and National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD, USA; Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, Brown University, Providence, RI, USA.
| |
Collapse
|
11
|
Denney WS, Sonnenberg GE, Carvajal-Gonzalez S, Tuthill T, Jackson VM. Pharmacokinetics and pharmacodynamics of PF-05190457: The first oral ghrelin receptor inverse agonist to be profiled in healthy subjects. Br J Clin Pharmacol 2016; 83:326-338. [PMID: 27621150 DOI: 10.1111/bcp.13127] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 08/25/2016] [Accepted: 09/08/2016] [Indexed: 02/06/2023] Open
Abstract
AIM To evaluate safety, tolerability and pharmacokinetics of oral PF-05190457, an oral ghrelin receptor inverse agonist, in healthy adults. METHODS Single (SAD) and multiple ascending dose (MAD) studies were randomised, placebo-controlled, double-blind studies. Thirty-five healthy men (age 38.2 ± 10.4 years; body mass index 24.8 ± 3.1 kg m-2 [mean ± standard deviation]) received ≥1 dose (2, 10, 40 [divided], 50, 100, 150, and 300 [single or divided] mg) of PF-05190457 and/or placebo in the SAD. In the MAD study, 35 healthy men (age 39.7 ± 10.1 years; body mass index 25.9 ± 3.3 kg m-2 ) received ≥1 dose (2, 10, 40 and 100 mg twice daily) of PF-05190457 and/or placebo daily for 2 weeks. RESULTS PF-05190457 absorption was rapid with a Tmax of 0.5-3 hours and a half-life between 8.2-9.8 hours. PF-05190457 dose-dependently blocked ghrelin (1 pmol kg-1 min-1 )-induced growth hormone (GH) release with (mean [90% confidence interval]) 77% [63-85%] inhibition at 100 mg. PF-05190457 (150 mg) delayed gastric emptying lag time by 30% [7-58%] and half emptying time by 20% [7-35%] with a corresponding decrease in postprandial glucose by 9 mg dL-1 . The most frequent adverse event reported by 30 subjects at doses ≥50 mg was somnolence. PF-05190457 plasma concentrations also increased heart rate up to 13.4 [4.8-58.2] beats min-1 and, similar to the effect on glucose and ghrelin-induced GH, was lost within 2 weeks. CONCLUSIONS PF-05190457 is a well-tolerated first-in-class ghrelin receptor inverse agonist with acceptable pharmacokinetics for oral daily dosing. Blocking ghrelin receptors inhibits ghrelin-induced GH, and increases heart rate, effects that underwent tachyphylaxis with chronic dosing. PF-051940457 has the potential to treat centrally-acting disorders such as insomnia.
Collapse
Affiliation(s)
- William S Denney
- Biotherapeutics Clinical Pharmacology, Pfizer Worldwide Research and Development, Cambridge, Massachusetts, 02139, USA
| | - Gabriele E Sonnenberg
- Cardiovascular, Metabolic, and Endocrine Diseases Research Unit, Pfizer Worldwide Research and Development, Cambridge, Massachusetts, 02139, USA
| | - Santos Carvajal-Gonzalez
- Cardiovascular, Metabolic, and Endocrine Diseases Research Unit, Pfizer Worldwide Research and Development, Cambridge, Massachusetts, 02139, USA
| | - Theresa Tuthill
- Cardiovascular, Metabolic, and Endocrine Diseases Research Unit, Pfizer Worldwide Research and Development, Cambridge, Massachusetts, 02139, USA
| | - V Margaret Jackson
- Cardiovascular, Metabolic, and Endocrine Diseases Research Unit, Pfizer Worldwide Research and Development, Cambridge, Massachusetts, 02139, USA
| |
Collapse
|
12
|
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: 1.9] [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
|
13
|
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: 4.8] [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
|
14
|
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: 6.7] [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
|
15
|
Heppner KM, Tong J. Mechanisms in endocrinology: regulation of glucose metabolism by the ghrelin system: multiple players and multiple actions. Eur J Endocrinol 2014; 171:R21-32. [PMID: 24714083 DOI: 10.1530/eje-14-0183] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Ghrelin is a 28-amino acid peptide secreted mainly from the X/A-like cells of the stomach. Ghrelin is found in circulation in both des-acyl (dAG) and acyl forms (AG). Acylation is catalyzed by the enzyme ghrelin O-acyltransferase (GOAT). AG acts on the GH secretagogue receptor (GHSR) in the CNS to promote feeding and adiposity and also acts on GHSR in the pancreas to inhibit glucose-stimulated insulin secretion. These well-described actions of AG have made it a popular target for obesity and type 2 diabetes mellitus pharmacotherapies. However, despite the lack of a cognate receptor, dAG appears to have gluco-regulatory action, which adds an additional layer of complexity to ghrelin's regulation of glucose metabolism. This review discusses the current literature on the gluco-regulatory action of the ghrelin system (dAG, AG, GHSR, and GOAT) with specific emphasis aimed toward distinguishing AG vs dAG action.
Collapse
Affiliation(s)
- Kristy M Heppner
- Division of DiabetesObesity and Metabolism, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, Oregon, USA andDivision of EndocrinologyDiabetes and Metabolism, Department of Medicine, University of Cincinnati, 260 Stetson Street, Suite 4200, Cincinnati, Ohio 45219-0547, USA
| | - Jenny Tong
- Division of DiabetesObesity and Metabolism, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, Oregon, USA andDivision of EndocrinologyDiabetes and Metabolism, Department of Medicine, University of Cincinnati, 260 Stetson Street, Suite 4200, Cincinnati, Ohio 45219-0547, USA
| |
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: 3.1] [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: 73] [Impact Index Per Article: 7.3] [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
|
Garin MC, Burns CM, Kaul S, Cappola AR. Clinical review: The human experience with ghrelin administration. J Clin Endocrinol Metab 2013; 98:1826-37. [PMID: 23533240 PMCID: PMC3644599 DOI: 10.1210/jc.2012-4247] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
CONTEXT Ghrelin is an endogenous stimulator of GH and is implicated in a number of physiological processes. Clinical trials have been performed in a variety of patient populations, but there is no comprehensive review of the beneficial and adverse consequences of ghrelin administration to humans. EVIDENCE ACQUISITION PubMed was utilized, and the reference list of each article was screened. We included 121 published articles in which ghrelin was administered to humans. EVIDENCE SYNTHESIS Ghrelin has been administered as an infusion or a bolus in a variety of doses to 1850 study participants, including healthy participants and patients with obesity, prior gastrectomy, cancer, pituitary disease, diabetes mellitus, eating disorders, and other conditions. There is strong evidence that ghrelin stimulates appetite and increases circulating GH, ACTH, cortisol, prolactin, and glucose across varied patient populations. There is a paucity of evidence regarding the effects of ghrelin on LH, FSH, TSH, insulin, lipolysis, body composition, cardiac function, pulmonary function, the vasculature, and sleep. Adverse effects occurred in 20% of participants, with a predominance of flushing and gastric rumbles and a mild degree of severity. The few serious adverse events occurred in patients with advanced illness and were not clearly attributable to ghrelin. Route of administration may affect the pattern of adverse effects. CONCLUSIONS Existing literature supports the short-term safety of ghrelin administration and its efficacy as an appetite stimulant in diverse patient populations. There is some evidence to suggest that ghrelin has wider ranging therapeutic effects, although these areas require further investigation.
Collapse
Affiliation(s)
- Margaret C Garin
- Division of Endocrinology, Diabetes, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, Pennsylvania 19104-5160, USA
| | | | | | | |
Collapse
|
19
|
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: 2.3] [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
|
20
|
Gautron L, Sakata I, Udit S, Zigman JM, Wood JN, Elmquist JK. Genetic tracing of Nav1.8-expressing vagal afferents in the mouse. J Comp Neurol 2012; 519:3085-101. [PMID: 21618224 DOI: 10.1002/cne.22667] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Nav1.8 is a tetrodotoxin-resistant sodium channel present in large subsets of peripheral sensory neurons, including both spinal and vagal afferents. In spinal afferents, Nav1.8 plays a key role in signaling different types of pain. Little is known, however, about the exact identity and role of Nav1.8-expressing vagal neurons. Here we generated mice with restricted expression of tdTomato fluorescent protein in all Nav1.8-expressing afferent neurons. As a result, intense fluorescence was visible in the cell bodies, central relays, and sensory endings of these neurons, revealing the full extent of their innervation sites in thoracic and abdominal viscera. For instance, vagal and spinal Nav1.8-expressing endings were seen clearly within the gastrointestinal mucosa and myenteric plexus, respectively. In the gastrointestinal muscle wall, labeled endings included a small subset of vagal tension receptors but not any stretch receptors. We also examined the detailed innervation of key metabolic tissues such as liver and pancreas and evaluated the anatomical relationship of Nav1.8-expressing vagal afferents with select enteroendocrine cells (i.e., ghrelin, glucagon, GLP-1). Specifically, our data revealed the presence of Nav1.8-expressing vagal afferents in several metabolic tissues and varying degrees of proximity between Nav1.8-expressing mucosal afferents and enteroendocrine cells, including apparent neuroendocrine apposition. In summary, this study demonstrates the power and versatility of the Cre-LoxP technology to trace identified visceral afferents, and our data suggest a previously unrecognized role for Nav1.8-expressing vagal neurons in gastrointestinal functions.
Collapse
Affiliation(s)
- Laurent Gautron
- Division of Hypothalamic Research, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA.
| | | | | | | | | | | |
Collapse
|
21
|
Kishimoto I, Tokudome T, Hosoda H, Miyazato M, Kangawa K. Ghrelin and cardiovascular diseases. J Cardiol 2011; 59:8-13. [PMID: 22178336 DOI: 10.1016/j.jjcc.2011.11.002] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2011] [Accepted: 11/14/2011] [Indexed: 11/17/2022]
Abstract
In 1999, a peptide from the stomach called ghrelin was discovered, which exerts potent growth hormone releasing powers. Subsequent studies revealed that it exerts a potent orexigenic action. In addition, the beneficial effects of ghrelin in cardiovascular diseases have been recently suggested. In humans as well as in animals, administration of ghrelin improves cardiac function and remodeling in chronic heart failure. In an animal model for myocardial infarction, ghrelin treatment early after coronary ligation effectively reduces fatal arrhythmia and, consequently, mortality, suggesting the potential therapeutic role of the peptide in acute myocardial infarction. Although how ghrelin may influence the cardiovascular system is not fully understood, the cardiovascular beneficial effects are mediated possibly through a combination of various actions, such as an increase in growth hormone level, an improvement in energy balance, direct actions to the cardiovascular cells, and regulation of the autonomic nervous activity. Of note, current experimental evidence suggests that ghrelin may act centrally to decrease sympathetic nervous system activity through peripheral afferent nerve. Thus, administration of ghrelin might become a unique new therapy for cardiovascular diseases.
Collapse
Affiliation(s)
- Ichiro Kishimoto
- National Cardiovascular Center Research Institute, Osaka, Japan.
| | | | | | | | | |
Collapse
|