151
|
Genetic targeting of sprouting angiogenesis using Apln-CreER. Nat Commun 2015; 6:6020. [PMID: 25597280 PMCID: PMC4309445 DOI: 10.1038/ncomms7020] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 12/02/2014] [Indexed: 02/07/2023] Open
Abstract
Under pathophysiological conditions in adults, endothelial cells (ECs) sprout from pre-existing blood vessels to form new ones by a process termed angiogenesis. During embryonic development, Apelin (APLN) is robustly expressed in vascular ECs. In adult mice, however, APLN expression in the vasculature is significantly reduced. Here we show that APLN expression is reactivated in adult ECs after ischaemia insults. In models of both injury ischaemia and tumor angiogenesis, we find that Apln-CreER genetically labels sprouting but not quiescent vasculature. By leveraging this specific activity, we demonstrate that abolishment of the VEGF-VEGFR2 signalling pathway as well as ablation of sprouting ECs diminished tumour vascularization and growth without compromising vascular homeostasis in other organs. Collectively, we show that Apln-CreER distinguishes sprouting vessels from stabilized vessels in multiple pathological settings. The Apln-CreER line described here will greatly aid future mechanistic studies in both vascular developmental biology and adult vascular diseases.
Collapse
|
152
|
Serpooshan V, Sivanesan S, Huang X, Mahmoudi M, Malkovskiy AV, Zhao M, Inayathullah M, Wagh D, Zhang XJ, Metzler S, Bernstein D, Wu JC, Ruiz-Lozano P, Rajadas J. [Pyr1]-Apelin-13 delivery via nano-liposomal encapsulation attenuates pressure overload-induced cardiac dysfunction. Biomaterials 2015; 37:289-98. [PMID: 25443792 PMCID: PMC5555682 DOI: 10.1016/j.biomaterials.2014.08.045] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 08/29/2014] [Indexed: 12/12/2022]
Abstract
Nanoparticle-mediated sustained delivery of therapeutics is one of the highly effective and increasingly utilized applications of nanomedicine. Here, we report the development and application of a drug delivery system consisting of polyethylene glycol (PEG)-conjugated liposomal nanoparticles as an efficient in vivo delivery approach for [Pyr1]-apelin-13 polypeptide. Apelin is an adipokine that regulates a variety of biological functions including cardiac hypertrophy and hypertrophy-induced heart failure. The clinical use of apelin has been greatly impaired by its remarkably short half-life in circulation. Here, we investigate whether [Pyr1]-apelin-13 encapsulation in liposome nanocarriers, conjugated with PEG polymer on their surface, can prolong apelin stability in the blood stream and potentiate apelin beneficial effects in cardiac function. Atomic force microscopy and dynamic light scattering were used to assess the structure and size distribution of drug-laden nanoparticles. [Pyr1]-apelin-13 encapsulation in PEGylated liposomal nanocarriers resulted in sustained and extended drug release both in vitro and in vivo. Moreover, intraperitoneal injection of [Pyr1]-apelin-13 nanocarriers in a mouse model of pressure-overload induced heart failure demonstrated a sustainable long-term effect of [Pyr1]-apelin-13 in preventing cardiac dysfunction. We concluded that this engineered nanocarrier system can serve as a delivery platform for treating heart injuries through sustained bioavailability of cardioprotective therapeutics.
Collapse
Affiliation(s)
- Vahid Serpooshan
- Stanford University, Department of Pediatrics, 300 Pasteur Dr., Stanford, CA 94305, USA; Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Senthilkumar Sivanesan
- Biomaterials and Advanced Drug Delivery Laboratory, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Xiaoran Huang
- Biomaterials and Advanced Drug Delivery Laboratory, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Morteza Mahmoudi
- Stanford University, Department of Pediatrics, 300 Pasteur Dr., Stanford, CA 94305, USA; Biomaterials and Advanced Drug Delivery Laboratory, Stanford University School of Medicine, Stanford, CA 94305, USA; Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Andrey V Malkovskiy
- Biomaterials and Advanced Drug Delivery Laboratory, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Mingming Zhao
- Stanford University, Department of Pediatrics, 300 Pasteur Dr., Stanford, CA 94305, USA
| | - Mohammed Inayathullah
- Biomaterials and Advanced Drug Delivery Laboratory, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Dhananjay Wagh
- Biomaterials and Advanced Drug Delivery Laboratory, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Xuexiang J Zhang
- Biomaterials and Advanced Drug Delivery Laboratory, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Scott Metzler
- Stanford University, Department of Pediatrics, 300 Pasteur Dr., Stanford, CA 94305, USA
| | - Daniel Bernstein
- Stanford University, Department of Pediatrics, 300 Pasteur Dr., Stanford, CA 94305, USA; Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Pilar Ruiz-Lozano
- Stanford University, Department of Pediatrics, 300 Pasteur Dr., Stanford, CA 94305, USA; Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jayakumar Rajadas
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Biomaterials and Advanced Drug Delivery Laboratory, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
153
|
Alfarano C, Foussal C, Lairez O, Calise D, Attané C, Anesia R, Daviaud D, Wanecq E, Parini A, Valet P, Kunduzova O. Transition from metabolic adaptation to maladaptation of the heart in obesity: role of apelin. Int J Obes (Lond) 2014; 39:312-20. [PMID: 25027224 PMCID: PMC4326962 DOI: 10.1038/ijo.2014.122] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Revised: 06/15/2014] [Accepted: 06/17/2014] [Indexed: 11/10/2022]
Abstract
Background/Objectives: Impaired energy metabolism is the defining characteristic of obesity-related heart failure. The adipocyte-derived peptide apelin has a role in the regulation of cardiovascular and metabolic homeostasis and may contribute to the link between obesity, energy metabolism and cardiac function. Here we investigate the role of apelin in the transition from metabolic adaptation to maladaptation of the heart in obese state. Methods: Adult male C57BL/6J, apelin knock-out (KO) or wild-type mice were fed a high-fat diet (HFD) for 18 weeks. To induce heart failure, mice were subjected to pressure overload after 18 weeks of HFD. Long-term effects of apelin on fatty acid (FA) oxidation, glucose metabolism, cardiac function and mitochondrial changes were evaluated in HFD-fed mice after 4 weeks of pressure overload. Cardiomyocytes from HFD-fed mice were isolated for analysis of metabolic responses. Results: In HFD-fed mice, pressure overload-induced transition from hypertrophy to heart failure is associated with reduced FA utilization (P<0.05), accelerated glucose oxidation (P<0.05) and mitochondrial damage. Treatment of HFD-fed mice with apelin for 4 weeks prevented pressure overload-induced decline in FA metabolism (P<0.05) and mitochondrial defects. Furthermore, apelin treatment lowered fasting plasma glucose (P<0.01), improved glucose tolerance (P<0.05) and preserved cardiac function (P<0.05) in HFD-fed mice subjected to pressure overload. In apelin KO HFD-fed mice, spontaneous cardiac dysfunction is associated with reduced FA oxidation (P<0.001) and increased glucose oxidation (P<0.05). In isolated cardiomyocytes, apelin stimulated FA oxidation in a dose-dependent manner and this effect was prevented by small interfering RNA sirtuin 3 knockdown. Conclusions: These data suggest that obesity-related decline in cardiac function is associated with defective myocardial energy metabolism and mitochondrial abnormalities. Furthermore, our work points for therapeutic potential of apelin to prevent myocardial metabolic abnormalities in heart failure paired with obesity.
Collapse
Affiliation(s)
- C Alfarano
- 1] National Institute of Health and Medical Research (INSERM) U1048, Toulouse, France [2] University of Toulouse, UPS, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France
| | - C Foussal
- 1] National Institute of Health and Medical Research (INSERM) U1048, Toulouse, France [2] University of Toulouse, UPS, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France
| | - O Lairez
- National Institute of Health and Medical Research (INSERM) U1048, Toulouse, France
| | - D Calise
- 1] University of Toulouse, UPS, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France [2] US006, Microsurgery Services, Toulouse, France
| | - C Attané
- 1] National Institute of Health and Medical Research (INSERM) U1048, Toulouse, France [2] University of Toulouse, UPS, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France
| | - R Anesia
- 1] National Institute of Health and Medical Research (INSERM) U1048, Toulouse, France [2] University of Toulouse, UPS, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France
| | - D Daviaud
- 1] National Institute of Health and Medical Research (INSERM) U1048, Toulouse, France [2] University of Toulouse, UPS, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France
| | - E Wanecq
- 1] National Institute of Health and Medical Research (INSERM) U1048, Toulouse, France [2] University of Toulouse, UPS, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France
| | - A Parini
- 1] National Institute of Health and Medical Research (INSERM) U1048, Toulouse, France [2] University of Toulouse, UPS, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France
| | - P Valet
- 1] National Institute of Health and Medical Research (INSERM) U1048, Toulouse, France [2] University of Toulouse, UPS, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France
| | - O Kunduzova
- 1] National Institute of Health and Medical Research (INSERM) U1048, Toulouse, France [2] University of Toulouse, UPS, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France
| |
Collapse
|
154
|
Ceraudo E, Galanth C, Carpentier E, Banegas-Font I, Schonegge AM, Alvear-Perez R, Iturrioz X, Bouvier M, Llorens-Cortes C. Biased signaling favoring gi over β-arrestin promoted by an apelin fragment lacking the C-terminal phenylalanine. J Biol Chem 2014; 289:24599-610. [PMID: 25012663 DOI: 10.1074/jbc.m113.541698] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Apelin plays a prominent role in body fluid and cardiovascular homeostasis. We previously showed that the C-terminal Phe of apelin 17 (K17F) is crucial for triggering apelin receptor internalization and decreasing blood pressure (BP) but is not required for apelin binding or Gi protein coupling. Based on these findings, we hypothesized that the important role of the C-terminal Phe in BP decrease may be as a Gi-independent but β-arrestin-dependent signaling pathway that could involve MAPKs. For this purpose, we have used apelin fragments K17F and K16P (K17F with the C-terminal Phe deleted), which exhibit opposite profiles on apelin receptor internalization and BP. Using BRET-based biosensors, we showed that whereas K17F activates Gi and promotes β-arrestin recruitment to the receptor, K16P had a much reduced ability to promote β-arrestin recruitment while maintaining its Gi activating property, revealing the biased agonist character of K16P. We further show that both β-arrestin recruitment and apelin receptor internalization contribute to the K17F-stimulated ERK1/2 activity, whereas the K16P-promoted ERK1/2 activity is entirely Gi-dependent. In addition to providing new insights on the structural basis underlying the functional selectivity of apelin peptides, our study indicates that the β-arrestin-dependent ERK1/2 activation and not the Gi-dependent signaling may participate in K17F-induced BP decrease.
Collapse
Affiliation(s)
- Emilie Ceraudo
- From the Laboratory of Central Neuropeptides in the Regulation of Body Fluid Homeostasis and Cardiovascular Functions, INSERM U1050, Paris F-75005, France, the Center for Interdisciplinary Research in Biology, Collège de France, Paris F-75005, France, CNRS, UMR 7241, Paris F-75005, France, and
| | - Cécile Galanth
- From the Laboratory of Central Neuropeptides in the Regulation of Body Fluid Homeostasis and Cardiovascular Functions, INSERM U1050, Paris F-75005, France, the Center for Interdisciplinary Research in Biology, Collège de France, Paris F-75005, France, CNRS, UMR 7241, Paris F-75005, France, and
| | - Eric Carpentier
- the Department of Biochemistry, Institute for Research in Immunology and Cancer, and Groupe de Recherche Universitaire sur le Médicament, Université de Montréal, Montréal, Québec H3T 1J4, Canada
| | - Inmaculada Banegas-Font
- From the Laboratory of Central Neuropeptides in the Regulation of Body Fluid Homeostasis and Cardiovascular Functions, INSERM U1050, Paris F-75005, France, the Center for Interdisciplinary Research in Biology, Collège de France, Paris F-75005, France, CNRS, UMR 7241, Paris F-75005, France, and
| | - Anne-Marie Schonegge
- the Department of Biochemistry, Institute for Research in Immunology and Cancer, and Groupe de Recherche Universitaire sur le Médicament, Université de Montréal, Montréal, Québec H3T 1J4, Canada
| | - Rodrigo Alvear-Perez
- From the Laboratory of Central Neuropeptides in the Regulation of Body Fluid Homeostasis and Cardiovascular Functions, INSERM U1050, Paris F-75005, France, the Center for Interdisciplinary Research in Biology, Collège de France, Paris F-75005, France, CNRS, UMR 7241, Paris F-75005, France, and
| | - Xavier Iturrioz
- From the Laboratory of Central Neuropeptides in the Regulation of Body Fluid Homeostasis and Cardiovascular Functions, INSERM U1050, Paris F-75005, France, the Center for Interdisciplinary Research in Biology, Collège de France, Paris F-75005, France, CNRS, UMR 7241, Paris F-75005, France, and
| | - Michel Bouvier
- the Department of Biochemistry, Institute for Research in Immunology and Cancer, and Groupe de Recherche Universitaire sur le Médicament, Université de Montréal, Montréal, Québec H3T 1J4, Canada
| | - Catherine Llorens-Cortes
- From the Laboratory of Central Neuropeptides in the Regulation of Body Fluid Homeostasis and Cardiovascular Functions, INSERM U1050, Paris F-75005, France, the Center for Interdisciplinary Research in Biology, Collège de France, Paris F-75005, France, CNRS, UMR 7241, Paris F-75005, France, and
| |
Collapse
|
155
|
Apelin increases cardiac contractility via protein kinase Cε- and extracellular signal-regulated kinase-dependent mechanisms. PLoS One 2014; 9:e93473. [PMID: 24695532 PMCID: PMC3973555 DOI: 10.1371/journal.pone.0093473] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 03/06/2014] [Indexed: 01/05/2023] Open
Abstract
Background Apelin, the endogenous ligand for the G protein-coupled apelin receptor, is an important regulator of the cardiovascular homoeostasis. We previously demonstrated that apelin is one of the most potent endogenous stimulators of cardiac contractility; however, its underlying signaling mechanisms remain largely elusive. In this study we characterized the contribution of protein kinase C (PKC), extracellular signal-regulated kinase 1/2 (ERK1/2) and myosin light chain kinase (MLCK) to the positive inotropic effect of apelin. Methods and Results In isolated perfused rat hearts, apelin increased contractility in association with activation of prosurvival kinases PKC and ERK1/2. Apelin induced a transient increase in the translocation of PKCε, but not PKCα, from the cytosol to the particulate fraction, and a sustained increase in the phosphorylation of ERK1/2 in the left ventricle. Suppression of ERK1/2 activation diminished the apelin-induced increase in contractility. Although pharmacological inhibition of PKC attenuated the inotropic response to apelin, it had no effect on ERK1/2 phosphorylation. Moreover, the apelin-induced positive inotropic effect was significantly decreased by inhibition of MLCK, a kinase that increases myofilament Ca2+ sensitivity. Conclusions Apelin increases cardiac contractility through parallel and independent activation of PKCε and ERK1/2 signaling in the adult rat heart. Additionally MLCK activation represents a downstream mechanism in apelin signaling. Our data suggest that, in addition to their role in cytoprotection, modest activation of PKCε and ERK1/2 signaling improve contractile function, therefore these pathways represent attractive possible targets in the treatment of heart failure.
Collapse
|
156
|
Margathe JF, Iturrioz X, Alvear-Perez R, Marsol C, Riché S, Chabane H, Tounsi N, Kuhry M, Heissler D, Hibert M, Llorens-Cortes C, Bonnet D. Structure-activity relationship studies toward the discovery of selective apelin receptor agonists. J Med Chem 2014; 57:2908-19. [PMID: 24625069 DOI: 10.1021/jm401789v] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Apelin is the endogenous ligand for the previously orphaned G protein-coupled receptor APJ. Apelin and its receptor are widely distributed in the brain, heart, and vasculature, and are emerging as an important regulator of body fluid homeostasis and cardiovascular functions. To further progress in the pharmacology and the physiological role of the apelin receptor, the development of small, bioavailable agonists and antagonists of the apelin receptor, is crucial. In this context, E339-3D6 (1) was described as the first nonpeptidic apelin receptor agonist. We show here that 1 is actually a mixture of polymethylated species, and we describe an alternative and versatile solid-phase approach that allows access to highly pure 27, the major component of 1. This approach was also applied to prepare a series of derivatives in order to identify the crucial structural determinants required for the ligand to maintain its affinity for the apelin receptor as well as its capacity to promote apelin receptor signaling and internalization. The study of the structure-activity relationships led to the identification of ligands 19, 21, and 38, which display an increased affinity compared to that of 27. The latter and 19 behave as full agonists with regard to cAMP production and apelin receptor internalization, whereas 21 is a biased agonist toward cAMP production. Interestingly, the three ligands display a much higher stability in mouse plasma (T1/2 > 10 h) than the endogenous apelin-17 peptide 2 (T1/2 < 4 min).
Collapse
Affiliation(s)
- Jean-François Margathe
- Laboratoire d'Innovation Thérapeutique, UMR7200 CNRS/Université de Strasbourg, Labex Medalis, Faculté de Pharmacie, 74 route du Rhin, 67401 Illkirch, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
157
|
Zhang L, Chen X, Sharma P, Moon M, Sheftel AD, Dawood F, Nghiem MP, Wu J, Li RK, Gramolini AO, Sorensen PH, Penninger JM, Brumell JH, Liu PP. HACE1-dependent protein degradation provides cardiac protection in response to haemodynamic stress. Nat Commun 2014; 5:3430. [PMID: 24614889 PMCID: PMC3959209 DOI: 10.1038/ncomms4430] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Accepted: 02/11/2014] [Indexed: 01/10/2023] Open
Abstract
The HECT E3 ubiquitin ligase HACE1
is a tumour suppressor known to regulate Rac1 activity under stress conditions. HACE1 is increased in the serum of patients
with heart failure. Here we show that HACE1 protects the heart under pressure stress by controlling
protein degradation. Hace1
deficiency in mice results in accelerated heart failure and increased mortality
under haemodynamic stress. Hearts from Hace1−/− mice
display abnormal cardiac hypertrophy, left ventricular dysfunction, accumulation of
LC3, p62 and ubiquitinated proteins enriched for
cytoskeletal species, indicating impaired autophagy. Our data suggest that
HACE1 mediates p62-dependent selective autophagic turnover
of ubiquitinated proteins by its ankyrin repeat domain through
protein–protein interaction, which is independent of its E3 ligase
activity. This would classify HACE1 as a dual-function E3 ligase. Our finding that
HACE1 has a protective
function in the heart in response to haemodynamic stress suggests that HACE1 may be a potential diagnostic and
therapeutic target for heart disease. HACE1 is an E3 ubiquitin ligase known to regulate various cell
biological processes. Here, Zhang et al. identify HACE1 as a protective factor in
the heart, demonstrating that HACE1 inhibits the development of heart failure in
response to haemodynamic stress by regulating protein degradation pathways.
Collapse
Affiliation(s)
- Liyong Zhang
- 1] University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, Ontario, Canada K1Y 4W7 [2] Heart and Stroke/Richard Lewar Centre of Excellent for Cardiovascular Research, University of Toronto and Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada M5G 2C4
| | - Xin Chen
- 1] University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, Ontario, Canada K1Y 4W7 [2] Heart and Stroke/Richard Lewar Centre of Excellent for Cardiovascular Research, University of Toronto and Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada M5G 2C4
| | - Parveen Sharma
- Heart and Stroke/Richard Lewar Centre of Excellent for Cardiovascular Research, University of Toronto and Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada M5G 2C4
| | - Mark Moon
- 1] University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, Ontario, Canada K1Y 4W7 [2] Heart and Stroke/Richard Lewar Centre of Excellent for Cardiovascular Research, University of Toronto and Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada M5G 2C4
| | - Alex D Sheftel
- University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, Ontario, Canada K1Y 4W7
| | - Fayez Dawood
- 1] University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, Ontario, Canada K1Y 4W7 [2] Heart and Stroke/Richard Lewar Centre of Excellent for Cardiovascular Research, University of Toronto and Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada M5G 2C4
| | - Mai P Nghiem
- Heart and Stroke/Richard Lewar Centre of Excellent for Cardiovascular Research, University of Toronto and Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada M5G 2C4
| | - Jun Wu
- Heart and Stroke/Richard Lewar Centre of Excellent for Cardiovascular Research, University of Toronto and Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada M5G 2C4
| | - Ren-Ke Li
- Heart and Stroke/Richard Lewar Centre of Excellent for Cardiovascular Research, University of Toronto and Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada M5G 2C4
| | - Anthony O Gramolini
- 1] Heart and Stroke/Richard Lewar Centre of Excellent for Cardiovascular Research, University of Toronto and Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada M5G 2C4 [2] Department of Physiology, University of Toronto, Toronto, Ontario, Canada M5S 1A8
| | - Poul H Sorensen
- Department of Molecular Oncology, BC Cancer Research Center, University of British Columbia, Vancouver, British Columbia, Canada V5Z 1L3
| | - Josef M Penninger
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Dr Bohrgasse 3, A-1030 Vienna, Austria
| | - John H Brumell
- 1] Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada M5S 1A8 [2] Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada M5S 1A8 [3] Cell Biology Program, Hospital for Sick Children, Toronto, Ontario, Canada M5G 1 × 8
| | - Peter P Liu
- 1] University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, Ontario, Canada K1Y 4W7 [2] Heart and Stroke/Richard Lewar Centre of Excellent for Cardiovascular Research, University of Toronto and Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada M5G 2C4 [3] Department of Physiology, University of Toronto, Toronto, Ontario, Canada M5S 1A8 [4] Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada M5S 1A8
| |
Collapse
|
158
|
Pauli A, Norris ML, Valen E, Chew GL, Gagnon JA, Zimmerman S, Mitchell A, Ma J, Dubrulle J, Reyon D, Tsai SQ, Joung JK, Saghatelian A, Schier AF. Toddler: an embryonic signal that promotes cell movement via Apelin receptors. Science 2014; 343:1248636. [PMID: 24407481 DOI: 10.1126/science.1248636] [Citation(s) in RCA: 486] [Impact Index Per Article: 44.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
It has been assumed that most, if not all, signals regulating early development have been identified. Contrary to this expectation, we identified 28 candidate signaling proteins expressed during zebrafish embryogenesis, including Toddler, a short, conserved, and secreted peptide. Both absence and overproduction of Toddler reduce the movement of mesendodermal cells during zebrafish gastrulation. Local and ubiquitous production of Toddler promote cell movement, suggesting that Toddler is neither an attractant nor a repellent but acts globally as a motogen. Toddler drives internalization of G protein-coupled APJ/Apelin receptors, and activation of APJ/Apelin signaling rescues toddler mutants. These results indicate that Toddler is an activator of APJ/Apelin receptor signaling, promotes gastrulation movements, and might be the first in a series of uncharacterized developmental signals.
Collapse
Affiliation(s)
- Andrea Pauli
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
159
|
Yu XH, Tang ZB, Liu LJ, Qian H, Tang SL, Zhang DW, Tian GP, Tang CK. Apelin and its receptor APJ in cardiovascular diseases. Clin Chim Acta 2014; 428:1-8. [DOI: 10.1016/j.cca.2013.09.001] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2013] [Revised: 08/31/2013] [Accepted: 09/01/2013] [Indexed: 12/29/2022]
|
160
|
Chng SC, Ho L, Tian J, Reversade B. ELABELA: a hormone essential for heart development signals via the apelin receptor. Dev Cell 2013; 27:672-80. [PMID: 24316148 DOI: 10.1016/j.devcel.2013.11.002] [Citation(s) in RCA: 365] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Revised: 10/06/2013] [Accepted: 11/04/2013] [Indexed: 02/06/2023]
Abstract
We report here the discovery and characterization of a gene, ELABELA (ELA), encoding a conserved hormone of 32 amino acids. Present in human embryonic stem cells, ELA is expressed at the onset of zebrafish zygotic transcription and is ubiquitous in the naive ectodermal cells of the embryo. Using zinc-finger-nuclease-mediated gene inactivation in zebrafish, we created an allelic series of ela mutants. ela null embryos have impaired endoderm differentiation potential marked by reduced gata5 and sox17 expression. Loss of Ela causes embryos to develop with a rudimentary heart or no heart at all, surprisingly phenocopying the loss of the apelin receptor (aplnr), which we show serves as Ela's cognate G protein-coupled receptor. Our results reveal the existence of a peptide hormone, ELA, which, together with APLNR, forms an essential signaling axis for early cardiovascular development.
Collapse
Affiliation(s)
- Serene C Chng
- Institute of Medical Biology, Human Genetics and Embryology Laboratory, A(∗)STAR, Singapore 138648, Singapore
| | - Lena Ho
- Institute of Medical Biology, Human Genetics and Embryology Laboratory, A(∗)STAR, Singapore 138648, Singapore
| | - Jing Tian
- Institute of Medical Biology, Human Genetics and Embryology Laboratory, A(∗)STAR, Singapore 138648, Singapore
| | - Bruno Reversade
- Institute of Medical Biology, Human Genetics and Embryology Laboratory, A(∗)STAR, Singapore 138648, Singapore; Institute of Molecular and Cellular Biology, A(∗)STAR, Singapore 138673, Singapore; Department of Pediatrics, National University of Singapore, Singapore 119260, Singapore.
| |
Collapse
|
161
|
Sato T, Suzuki T, Watanabe H, Kadowaki A, Fukamizu A, Liu PP, Kimura A, Ito H, Penninger JM, Imai Y, Kuba K. Apelin is a positive regulator of ACE2 in failing hearts. J Clin Invest 2013; 123:5203-11. [PMID: 24177423 DOI: 10.1172/jci69608] [Citation(s) in RCA: 152] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Accepted: 08/29/2013] [Indexed: 11/17/2022] Open
Abstract
Angiotensin converting enzyme 2 (ACE2) is a negative regulator of the renin-angiotensin system (RAS), catalyzing the conversion of Angiotensin II to Angiotensin 1-7. Apelin is a second catalytic substrate for ACE2 and functions as an inotropic and cardioprotective peptide. While an antagonistic relationship between the RAS and apelin has been proposed, such functional interplay remains elusive. Here we found that ACE2 was downregulated in apelin-deficient mice. Pharmacological or genetic inhibition of angiotensin II type 1 receptor (AT1R) rescued the impaired contractility and hypertrophy of apelin mutant mice, which was accompanied by restored ACE2 levels. Importantly, treatment with angiotensin 1-7 rescued hypertrophy and heart dysfunctions of apelin-knockout mice. Moreover, apelin, via activation of its receptor, APJ, increased ACE2 promoter activity in vitro and upregulated ACE2 expression in failing hearts in vivo. Apelin treatment also increased cardiac contractility and ACE2 levels in AT1R-deficient mice. These data demonstrate that ACE2 couples the RAS to the apelin system, adding a conceptual framework for the apelin-ACE2-angiotensin 1-7 axis as a therapeutic target for cardiovascular diseases.
Collapse
|
162
|
Voelkel NF, Natarajan R, Drake JI, Bogaard HJ. Right ventricle in pulmonary hypertension. Compr Physiol 2013; 1:525-40. [PMID: 23737184 DOI: 10.1002/cphy.c090008] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
During heart development chamber specification is controlled and directed by a number of genes and a fetal heart gene expression pattern is revisited during heart failure. In the setting of chronic pulmonary hypertension the right ventricle undergoes hypertrophy, which is likely initially adaptive, but often followed by decompensation, dilatation and failure. Here we discuss differences between the right ventricle and the left ventricle of the heart and begin to describe the cellular and molecular changes which characterize right heart failure. A prevention and treatment of right ventricle failure becomes a treatment goal for patients with severe pulmonary hypertension it follows that we need to understand the pathobiology of right heart hypertrophy and the transition to right heart failure.
Collapse
Affiliation(s)
- Norbert F Voelkel
- Division of Pulmonary & Critical Care Medicine, Department of Internal Medicine, The Victoria Johnson Center for Pulmonary Obstructive Disease Research, Virginia Commonwealth University, Richmond, Virginia, USA.
| | | | | | | |
Collapse
|
163
|
O'Carroll AM, Lolait SJ, Harris LE, Pope GR. The apelin receptor APJ: journey from an orphan to a multifaceted regulator of homeostasis. J Endocrinol 2013; 219:R13-35. [PMID: 23943882 DOI: 10.1530/joe-13-0227] [Citation(s) in RCA: 246] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The apelin receptor (APJ; gene symbol APLNR) is a member of the G protein-coupled receptor gene family. Neural gene expression patterns of APJ, and its cognate ligand apelin, in the brain implicate the apelinergic system in the regulation of a number of physiological processes. APJ and apelin are highly expressed in the hypothalamo-neurohypophysial system, which regulates fluid homeostasis, in the hypothalamic-pituitary-adrenal axis, which controls the neuroendocrine response to stress, and in the forebrain and lower brainstem regions, which are involved in cardiovascular function. Recently, apelin, synthesised and secreted by adipocytes, has been described as a beneficial adipokine related to obesity, and there is growing awareness of a potential role for apelin and APJ in glucose and energy metabolism. In this review we provide a comprehensive overview of the structure, expression pattern and regulation of apelin and its receptor, as well as the main second messengers and signalling proteins activated by apelin. We also highlight the physiological and pathological roles that support this system as a novel therapeutic target for pharmacological intervention in treating conditions related to altered water balance, stress-induced disorders such as anxiety and depression, and cardiovascular and metabolic disorders.
Collapse
Affiliation(s)
- Anne-Marie O'Carroll
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, School of Clinical Sciences, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol BS1 3NY, UK
| | | | | | | |
Collapse
|
164
|
|
165
|
Seifirad S, Masoudkabir F. Apelin could reduce risk of contrast-induced nephropathy in patients with congestive heart failure. Med Hypotheses 2013; 81:898-900. [PMID: 23968574 DOI: 10.1016/j.mehy.2013.08.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Accepted: 08/04/2013] [Indexed: 12/13/2022]
Abstract
Compared to the normal population, patients with congestive heart failure are at higher risk for contrast-induced nephropathy. A variety of interventions are suggested to reduce the risk for contrast-induced nephropathy. Unfortunately results of none of current protective treatments are satisfactory. Apelin a vasodilator adipocytokine, positively inotropic agent, and free radical scavenger has been recently introduced. It has been shown that endogenous apelin levels are decreased in patients with congestive heart failure. Two major mechanisms have been suggested for pathophysiology of contrast induced nephropathy including reactive oxygen species production and impaired renal perfusion due to vasoconstriction. Pretreatment with recombinant apelin (exogenous apelin-13), could compensate decreased endogenous apelin serum levels of congestive heart failure patients. Its antioxidant and cell-protective properties, decrease nephrotoxicity of contrast agent; additionally impaired renal perfusion due to malfunction of cardiac pump will refurbish, because of positively inotropic property of apelin plus its vasodilatation effect in renal arteries. We believe that the triangle of increased contractility, decreased vascular resistance and decreased contrast agent nephrotoxicity could significantly reduce risk of contrast-induced nephropathy in patients with congestive heart failure.
Collapse
Affiliation(s)
- Soroush Seifirad
- Endocrinology and Metabolism Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
166
|
Wang W, McKinnie SMK, Patel VB, Haddad G, Wang Z, Zhabyeyev P, Das SK, Basu R, McLean B, Kandalam V, Penninger JM, Kassiri Z, Vederas JC, Murray AG, Oudit GY. Loss of Apelin exacerbates myocardial infarction adverse remodeling and ischemia-reperfusion injury: therapeutic potential of synthetic Apelin analogues. J Am Heart Assoc 2013; 2:e000249. [PMID: 23817469 PMCID: PMC3828798 DOI: 10.1161/jaha.113.000249] [Citation(s) in RCA: 171] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Coronary artery disease leading to myocardial ischemia is the most common cause of heart failure. Apelin (APLN), the endogenous peptide ligand of the APJ receptor, has emerged as a novel regulator of the cardiovascular system. METHODS AND RESULTS Here we show a critical role of APLN in myocardial infarction (MI) and ischemia-reperfusion (IR) injury in patients and animal models. Myocardial APLN levels were reduced in patients with ischemic heart failure. Loss of APLN increased MI-related mortality, infarct size, and inflammation with drastic reductions in prosurvival pathways resulting in greater systolic dysfunction and heart failure. APLN deficiency decreased vascular sprouting, impaired sprouting of human endothelial progenitor cells, and compromised in vivo myocardial angiogenesis. Lack of APLN enhanced susceptibility to ischemic injury and compromised functional recovery following ex vivo and in vivo IR injury. We designed and synthesized two novel APLN analogues resistant to angiotensin converting enzyme 2 cleavage and identified one analogue, which mimicked the function of APLN, to be markedly protective against ex vivo and in vivo myocardial IR injury linked to greater activation of survival pathways and promotion of angiogenesis. CONCLUSIONS APLN is a critical regulator of the myocardial response to infarction and ischemia and pharmacologically targeting this pathway is feasible and represents a new class of potential therapeutic agents.
Collapse
Affiliation(s)
- Wang Wang
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
167
|
Affiliation(s)
- Annemarieke E. Loot
- From the Institute for Vascular Signalling, Centre for Molecular Medicine and German Centre for Cardiovascular Research (DZHK) partner site Rhine-Main, Goethe University, Theodor-Stern-Kai, Frankfurt am Main, Germany
| | - Ingrid Fleming
- From the Institute for Vascular Signalling, Centre for Molecular Medicine and German Centre for Cardiovascular Research (DZHK) partner site Rhine-Main, Goethe University, Theodor-Stern-Kai, Frankfurt am Main, Germany
| |
Collapse
|
168
|
The G-protein-coupled receptor APJ is expressed in the second heart field and regulates Cerberus–Baf60c axis in embryonic stem cell cardiomyogenesis. Cardiovasc Res 2013; 100:95-104. [DOI: 10.1093/cvr/cvt166] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
|
169
|
Barnes GD, Alam S, Carter G, Pedersen CM, Lee KM, Hubbard TJ, Veitch S, Jeong H, White A, Cruden NL, Huson L, Japp AG, Newby DE. Sustained Cardiovascular Actions of APJ Agonism During Renin–Angiotensin System Activation and in Patients With Heart Failure. Circ Heart Fail 2013; 6:482-91. [DOI: 10.1161/circheartfailure.111.000077] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Gareth D. Barnes
- From the British Heart Foundation/University of Edinburgh Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK (G.D.B., S.A., C.M.P., K.M.L., T.J.H., S.V., H.J., A.W., D.E.N.); Edinburgh Heart Centre, Royal Infirmary of Edinburgh, Edinburgh, UK (A.G.J., N.L.C., D.E.N.); and Department of Experimental Medicine, Imperial College, London, UK (L.H.)
| | - Shirjel Alam
- From the British Heart Foundation/University of Edinburgh Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK (G.D.B., S.A., C.M.P., K.M.L., T.J.H., S.V., H.J., A.W., D.E.N.); Edinburgh Heart Centre, Royal Infirmary of Edinburgh, Edinburgh, UK (A.G.J., N.L.C., D.E.N.); and Department of Experimental Medicine, Imperial College, London, UK (L.H.)
| | - Gordon Carter
- From the British Heart Foundation/University of Edinburgh Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK (G.D.B., S.A., C.M.P., K.M.L., T.J.H., S.V., H.J., A.W., D.E.N.); Edinburgh Heart Centre, Royal Infirmary of Edinburgh, Edinburgh, UK (A.G.J., N.L.C., D.E.N.); and Department of Experimental Medicine, Imperial College, London, UK (L.H.)
| | - Christian M. Pedersen
- From the British Heart Foundation/University of Edinburgh Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK (G.D.B., S.A., C.M.P., K.M.L., T.J.H., S.V., H.J., A.W., D.E.N.); Edinburgh Heart Centre, Royal Infirmary of Edinburgh, Edinburgh, UK (A.G.J., N.L.C., D.E.N.); and Department of Experimental Medicine, Imperial College, London, UK (L.H.)
| | - Kristina M. Lee
- From the British Heart Foundation/University of Edinburgh Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK (G.D.B., S.A., C.M.P., K.M.L., T.J.H., S.V., H.J., A.W., D.E.N.); Edinburgh Heart Centre, Royal Infirmary of Edinburgh, Edinburgh, UK (A.G.J., N.L.C., D.E.N.); and Department of Experimental Medicine, Imperial College, London, UK (L.H.)
| | - Thomas J. Hubbard
- From the British Heart Foundation/University of Edinburgh Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK (G.D.B., S.A., C.M.P., K.M.L., T.J.H., S.V., H.J., A.W., D.E.N.); Edinburgh Heart Centre, Royal Infirmary of Edinburgh, Edinburgh, UK (A.G.J., N.L.C., D.E.N.); and Department of Experimental Medicine, Imperial College, London, UK (L.H.)
| | - Scott Veitch
- From the British Heart Foundation/University of Edinburgh Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK (G.D.B., S.A., C.M.P., K.M.L., T.J.H., S.V., H.J., A.W., D.E.N.); Edinburgh Heart Centre, Royal Infirmary of Edinburgh, Edinburgh, UK (A.G.J., N.L.C., D.E.N.); and Department of Experimental Medicine, Imperial College, London, UK (L.H.)
| | - Herim Jeong
- From the British Heart Foundation/University of Edinburgh Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK (G.D.B., S.A., C.M.P., K.M.L., T.J.H., S.V., H.J., A.W., D.E.N.); Edinburgh Heart Centre, Royal Infirmary of Edinburgh, Edinburgh, UK (A.G.J., N.L.C., D.E.N.); and Department of Experimental Medicine, Imperial College, London, UK (L.H.)
| | - Audrey White
- From the British Heart Foundation/University of Edinburgh Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK (G.D.B., S.A., C.M.P., K.M.L., T.J.H., S.V., H.J., A.W., D.E.N.); Edinburgh Heart Centre, Royal Infirmary of Edinburgh, Edinburgh, UK (A.G.J., N.L.C., D.E.N.); and Department of Experimental Medicine, Imperial College, London, UK (L.H.)
| | - Nicholas L. Cruden
- From the British Heart Foundation/University of Edinburgh Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK (G.D.B., S.A., C.M.P., K.M.L., T.J.H., S.V., H.J., A.W., D.E.N.); Edinburgh Heart Centre, Royal Infirmary of Edinburgh, Edinburgh, UK (A.G.J., N.L.C., D.E.N.); and Department of Experimental Medicine, Imperial College, London, UK (L.H.)
| | - Les Huson
- From the British Heart Foundation/University of Edinburgh Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK (G.D.B., S.A., C.M.P., K.M.L., T.J.H., S.V., H.J., A.W., D.E.N.); Edinburgh Heart Centre, Royal Infirmary of Edinburgh, Edinburgh, UK (A.G.J., N.L.C., D.E.N.); and Department of Experimental Medicine, Imperial College, London, UK (L.H.)
| | - Alan G. Japp
- From the British Heart Foundation/University of Edinburgh Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK (G.D.B., S.A., C.M.P., K.M.L., T.J.H., S.V., H.J., A.W., D.E.N.); Edinburgh Heart Centre, Royal Infirmary of Edinburgh, Edinburgh, UK (A.G.J., N.L.C., D.E.N.); and Department of Experimental Medicine, Imperial College, London, UK (L.H.)
| | - David E. Newby
- From the British Heart Foundation/University of Edinburgh Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK (G.D.B., S.A., C.M.P., K.M.L., T.J.H., S.V., H.J., A.W., D.E.N.); Edinburgh Heart Centre, Royal Infirmary of Edinburgh, Edinburgh, UK (A.G.J., N.L.C., D.E.N.); and Department of Experimental Medicine, Imperial College, London, UK (L.H.)
| |
Collapse
|
170
|
Kang Y, Kim J, Anderson JP, Wu J, Gleim SR, Kundu RK, McLean DL, Kim JD, Park H, Jin SW, Hwa J, Quertermous T, Chun HJ. Apelin-APJ signaling is a critical regulator of endothelial MEF2 activation in cardiovascular development. Circ Res 2013; 113:22-31. [PMID: 23603510 DOI: 10.1161/circresaha.113.301324] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE The peptide ligand apelin and its receptor APJ constitute a signaling pathway with numerous effects on the cardiovascular system, including cardiovascular development in model organisms such as xenopus and zebrafish. OBJECTIVE This study aimed to characterize the embryonic lethal phenotype of the Apj-/- mice and to define the involved downstream signaling targets. METHODS AND RESULTS We report the first characterization of the embryonic lethality of the Apj-/- mice. More than half of the expected Apj-/- embryos died in utero because of cardiovascular developmental defects. Those succumbing to early embryonic death had markedly deformed vasculature of the yolk sac and the embryo, as well as poorly looped hearts with aberrantly formed right ventricles and defective atrioventricular cushion formation. Apj-/- embryos surviving to later stages demonstrated incomplete vascular maturation because of a deficiency of vascular smooth muscle cells and impaired myocardial trabeculation and ventricular wall development. The molecular mechanism implicates a novel, noncanonical signaling pathway downstream of apelin-APJ involving Gα13, which induces histone deacetylase (HDAC) 4 and HDAC5 phosphorylation and cytoplasmic translocation, resulting in activation of myocyte enhancer factor 2. Apj-/- mice have greater endocardial Hdac4 and Hdac5 nuclear localization and reduced expression of the myocyte enhancer factor 2 (MEF2) transcriptional target Krüppel-like factor 2. We identify a number of commonly shared transcriptional targets among apelin-APJ, Gα13, and MEF2 in endothelial cells, which are significantly decreased in the Apj-/- embryos and endothelial cells. CONCLUSIONS Our results demonstrate a novel role for apelin-APJ signaling as a potent regulator of endothelial MEF2 function in the developing cardiovascular system.
Collapse
Affiliation(s)
- Yujung Kang
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06511, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
171
|
Krist J, Wieder K, Klöting N, Oberbach A, Kralisch S, Wiesner T, Schön MR, Gärtner D, Dietrich A, Shang E, Lohmann T, Dreßler M, Fasshauer M, Stumvoll M, Blüher M. Effects of weight loss and exercise on apelin serum concentrations and adipose tissue expression in human obesity. Obes Facts 2013; 6:57-69. [PMID: 23429279 PMCID: PMC5644751 DOI: 10.1159/000348667] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2012] [Accepted: 08/03/2012] [Indexed: 01/17/2023] Open
Abstract
OBJECTIVE Apelin is an adipokine which plays a role in the regulation of glucose homeostasis and may contribute to the link between increased adipose tissue mass and obesity related metabolic diseases. Here we investigate the role of omental and subcutaneous (SC) adipose tissue apelin and its receptor APJ mRNA expression in human obesity and test the hypothesis that changes in circulating apelin are associated with reduced fat mass in three weight loss intervention studies. METHODS Apelin serum concentration was measured in 740 individuals in a cross-sectional (n = 629) study including a subgroup (n = 161) for which omental and SC apelin mRNA expression has been analyzed and in three interventions: 12 weeks exercise (n = 60), 6 months calorie-restricted diet (n = 19), 12 months after bariatric surgery (n = 32). RESULTS Apelin mRNA is significantly higher expressed in adipose tissue of patients with type 2 diabetes and correlates with circulating apelin, BMI, body fat, C-reactive protein, and insulin sensitivity. Obesity surgery-induced weight loss causes a significant reduction in omental and SC apelin expression. All interventions led to significantly reduced apelin serum concentrations which significantly correlate with improved insulin sensitivity, independently of changes in BMI. CONCLUSIONS Reduced apelin expression and serum concentration may contribute to improved insulin sensitivity beyond significant weight loss.
Collapse
Affiliation(s)
- Joanna Krist
- Department of Medicine, University of Leipzig, Junior Research Group Animal Models, Dresden, Germany
| | - Katharina Wieder
- Department of Medicine, University of Leipzig, Junior Research Group Animal Models, Dresden, Germany
| | - Nora Klöting
- Leipzig University Medical Center, IFB Adiposity Diseases, Junior Research Group Animal Models, Dresden, Germany
| | - Andreas Oberbach
- Department of Pediatric Surgery, University of Leipzig, Leipzig, Germany
| | - Susan Kralisch
- Department of Medicine, University of Leipzig, Junior Research Group Animal Models, Dresden, Germany
| | - Tobias Wiesner
- Department of Medicine, University of Leipzig, Junior Research Group Animal Models, Dresden, Germany
| | - Michael R. Schön
- Clinic of Visceral Surgery, Städtisches Klinikum Karlsruhe, Karlsruhe, Germany
| | - Daniel Gärtner
- Clinic of Visceral Surgery, Städtisches Klinikum Karlsruhe, Karlsruhe, Germany
| | - Arne Dietrich
- Department of Surgery, University of Leipzig, Dresden, Germany
| | - Edward Shang
- Department of Surgery, University of Leipzig, Dresden, Germany
| | | | | | - Mathias Fasshauer
- Department of Medicine, University of Leipzig, Junior Research Group Animal Models, Dresden, Germany
| | - Michael Stumvoll
- Department of Medicine, University of Leipzig, Junior Research Group Animal Models, Dresden, Germany
| | - Matthias Blüher
- Department of Medicine, University of Leipzig, Junior Research Group Animal Models, Dresden, Germany
- *Prof. Dr. med. Matthias Blüher, Department of Medicine, University of Leipzig, Liebigstraße 20, 04103 Leipzig (Germany),
| |
Collapse
|
172
|
Abstract
The discovery of leptin in 1994 sparked dramatic new interest in the study of white adipose tissue. It is now recognised to be a metabolically active endocrine organ, producing important chemical messengers - adipokines and cytokines (adipocytokines). The search for new adipocytokines or adipokines gained added fervour with the prospect of the reconciliation between cardiovascular diseases (CVDs), obesity and metabolic syndrome. The role these new chemical messengers play in inflammation, satiety, metabolism and cardiac function has paved the way for new research and theories examining the effects they have on (in this case) CVD. Adipokines are involved in a 'good-bad', yin-yang homoeostatic balance whereby there are substantial benefits: cardioprotection, promoting endothelial function, angiogenesis and reducing hypertension, atherosclerosis and inflammation. The flip side may show contrasting, detrimental effects in aggravating these cardiac parameters.
Collapse
Affiliation(s)
- Harman S Mattu
- Division of Metabolic and Vascular Health, University of Warwick Medical School, Gibbet Hill Road, Coventry CV4 7AL, UK
| | | |
Collapse
|
173
|
Jin W, Su X, Xu M, Liu Y, Shi J, Lu L, Niu W. Interactive association of five candidate polymorphisms in Apelin/APJ pathway with coronary artery disease among Chinese hypertensive patients. PLoS One 2012; 7:e51123. [PMID: 23226564 PMCID: PMC3513301 DOI: 10.1371/journal.pone.0051123] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2012] [Accepted: 10/31/2012] [Indexed: 12/29/2022] Open
Abstract
Background Via sequencing the genes of apelin/angiotensin receptor-like 1 (apelin/APJ) pathway, we have recently identified and validated four common polymorphisms (rs3761581, rs56204867, rs7119375, and rs10501367) implicated in the development of hypertension. Extending these findings, we, in Chinese hypertensive patients, sought to investigate the association of these four polymorphisms and one additional promising candidate (rs9943582) from this pathway with the risk of developing coronary artery disease (CAD). Methodology/Principal Findings Genotypes were obtained from 994 sporadic CAD patients and 708 age- and sex-matched controls. All participants were hypertensives and angiographically-confirmed. Data were analyzed by Haplo.Stats and multifactor dimensionality reduction (MDR) softwares. Genotype distributions of five examined polymorphisms satisfied Hardy-Weinberg equilibrium in controls of both genders. Single-locus analyses exhibited no significant differences in the genotype/allele frequencies of examined polymorphisms between CAD patients and controls (P>0.05), even after controlling traditional cardiovascular confounders. In haplotype analyses, low-penetrance haplotype G-A (in order of rs56204867 and rs3761581 from apelin gene) was significantly overrepresented in controls (1.73%) relative to in CAD patients (0.4%) in males (P = 0.047). Further interaction analyses suggested an overall best MDR model including rs3761581 in males (P = 0.0408) and including rs7119375 and rs9943582 in females (P<0.0001), which were further substantiated in the classical logistical regression model. Conclusions Our findings demonstrated a contributive role of low-penetrance haplotype in apelin gene on CAD in males, and more importantly, interactive effects of genetic defects in apelin/APJ pathway might confer a potential risk in Chinese hypertensive patients.
Collapse
Affiliation(s)
- Wei Jin
- Department of Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiuxiu Su
- Department of Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min Xu
- Department of Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Liu
- Department of Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jingyi Shi
- State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lin Lu
- Department of Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenquan Niu
- State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Hypertension, Shanghai, China
- * E-mail:
| |
Collapse
|
174
|
Scimia MC, Hurtado C, Ray S, Metzler S, Wei K, Wang J, Woods CE, Purcell NH, Catalucci D, Akasaka T, Bueno OF, Vlasuk GP, Kaliman P, Bodmer R, Smith LH, Ashley E, Mercola M, Brown JH, Ruiz-Lozano P. APJ acts as a dual receptor in cardiac hypertrophy. Nature 2012; 488:394-8. [PMID: 22810587 PMCID: PMC3422434 DOI: 10.1038/nature11263] [Citation(s) in RCA: 198] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2012] [Accepted: 05/25/2012] [Indexed: 01/08/2023]
Abstract
Cardiac hypertrophy is initiated as an adaptive response to sustained overload but progresses pathologically as heart failure ensues. Here we report that genetic loss of APJ, a G-protein-coupled receptor, confers resistance to chronic pressure overload by markedly reducing myocardial hypertrophy and heart failure. In contrast, mice lacking apelin (the endogenous APJ ligand) remain sensitive, suggesting an apelin-independent function of APJ. Freshly isolated APJ-null cardiomyocytes exhibit an attenuated response to stretch, indicating that APJ is a mechanosensor. Activation of APJ by stretch increases cardiomyocyte cell size and induces molecular markers of hypertrophy. Whereas apelin stimulates APJ to activate Gαi and elicits a protective response, stretch signals in an APJ-dependent, G-protein-independent fashion to induce hypertrophy. Stretch-mediated hypertrophy is prevented by knockdown of β-arrestins or by pharmacological doses of apelin acting through Gαi. Taken together, our data indicate that APJ is a bifunctional receptor for both mechanical stretch and the endogenous peptide apelin. By sensing the balance between these stimuli, APJ occupies a pivotal point linking sustained overload to cardiomyocyte hypertrophy.
Collapse
MESH Headings
- Adipokines
- Animals
- Aorta/pathology
- Apelin
- Apelin Receptors
- Arrestins/deficiency
- Arrestins/genetics
- Arrestins/metabolism
- Blood Pressure
- Cardiomegaly/metabolism
- Cardiomegaly/pathology
- Cardiomegaly/physiopathology
- Cardiomegaly/prevention & control
- Female
- GTP-Binding Protein alpha Subunits, Gi-Go/metabolism
- Intercellular Signaling Peptides and Proteins/deficiency
- Intercellular Signaling Peptides and Proteins/genetics
- Intercellular Signaling Peptides and Proteins/metabolism
- Intercellular Signaling Peptides and Proteins/pharmacology
- Male
- Mechanoreceptors/metabolism
- Mechanotransduction, Cellular/drug effects
- Mechanotransduction, Cellular/physiology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/pathology
- Receptors, G-Protein-Coupled/agonists
- Receptors, G-Protein-Coupled/deficiency
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/metabolism
- Signal Transduction/drug effects
- beta-Arrestins
Collapse
Affiliation(s)
| | - Cecilia Hurtado
- Sanford-Burnham Medical Research Institute, Stanford University, CA
| | - Saugata Ray
- Sanford-Burnham Medical Research Institute, Stanford University, CA
| | - Scott Metzler
- Department of Pediatrics, School of Medicine, Stanford University, CA 94304
| | - Ke Wei
- Sanford-Burnham Medical Research Institute, Stanford University, CA
| | - Jianming Wang
- Sanford-Burnham Medical Research Institute, Stanford University, CA
| | - Chris E. Woods
- Department of Medicine, School of Medicine, Stanford University, CA
| | | | - Daniele Catalucci
- Biomedical and Genetic Research Institute, National Research Council, via Fantoli 16/15, 20138, Milan, and Istituto Clinico Humanitas IRCSS, Rozzano, Italy
| | - Takashi Akasaka
- Sanford-Burnham Medical Research Institute, Stanford University, CA
| | | | | | - Perla Kaliman
- Institute of Biomedical Research August Pi i Sunyer (IDIBAPS) Villarroel 170, E-08036 Barcelona, Spain
| | - Rolf Bodmer
- Sanford-Burnham Medical Research Institute, Stanford University, CA
| | - Layton H. Smith
- Sanford-Burnham Medical Research Institute, Stanford University, CA
| | - Euan Ashley
- Department of Medicine, School of Medicine, Stanford University, CA
| | - Mark Mercola
- Sanford-Burnham Medical Research Institute, Stanford University, CA
| | | | - Pilar Ruiz-Lozano
- Sanford-Burnham Medical Research Institute, Stanford University, CA
- Department of Pediatrics, School of Medicine, Stanford University, CA 94304
| |
Collapse
|
175
|
ACE2 links amino acid malnutrition to microbial ecology and intestinal inflammation. Nature 2012; 487:477-81. [PMID: 22837003 PMCID: PMC7095315 DOI: 10.1038/nature11228] [Citation(s) in RCA: 980] [Impact Index Per Article: 75.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2011] [Accepted: 05/14/2012] [Indexed: 11/08/2022]
Abstract
Malnutrition affects up to one billion people in the world and is a major cause of mortality. In many cases, malnutrition is associated with diarrhoea and intestinal inflammation, further contributing to morbidity and death. The mechanisms by which unbalanced dietary nutrients affect intestinal homeostasis are largely unknown. Here we report that deficiency in murine angiotensin I converting enzyme (peptidyl-dipeptidase A) 2 (Ace2), which encodes a key regulatory enzyme of the renin-angiotensin system (RAS), results in highly increased susceptibility to intestinal inflammation induced by epithelial damage. The RAS is known to be involved in acute lung failure, cardiovascular functions and SARS infections. Mechanistically, ACE2 has a RAS-independent function, regulating intestinal amino acid homeostasis, expression of antimicrobial peptides, and the ecology of the gut microbiome. Transplantation of the altered microbiota from Ace2 mutant mice into germ-free wild-type hosts was able to transmit the increased propensity to develop severe colitis. ACE2-dependent changes in epithelial immunity and the gut microbiota can be directly regulated by the dietary amino acid tryptophan. Our results identify ACE2 as a key regulator of dietary amino acid homeostasis, innate immunity, gut microbial ecology, and transmissible susceptibility to colitis. These results provide a molecular explanation for how amino acid malnutrition can cause intestinal inflammation and diarrhoea.
Collapse
|
176
|
Targeting the ACE2 and Apelin Pathways Are Novel Therapies for Heart Failure: Opportunities and Challenges. Cardiol Res Pract 2012; 2012:823193. [PMID: 22655211 PMCID: PMC3359660 DOI: 10.1155/2012/823193] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2011] [Accepted: 02/21/2012] [Indexed: 11/17/2022] Open
Abstract
Angiotensin-converting enzyme 2 (ACE2)/Ang II/Ang 1-7 and the apelin/APJ are two important peptide systems which exert diverse effects on the cardiovascular system. ACE2 is a key negative regulator of the renin-angiotensin system (RAS) where it metabolizes angiotensin (Ang) II into Ang 1-7, an endogenous antagonist of Ang II. Both the prolonged activation of RAS and the loss of ACE2 can be detrimental as they lead to functional deterioration of the heart and progression of cardiac, renal, and vascular diseases. Recombinant human ACE2 in an animal model of ACE2 knockout mice lowers Ang II. These interactions neutralize the pressor and subpressor pathologic effects of Ang II by producing Ang 1-7 levels in vivo, that might be cardiovascular protective. ACE2 hydrolyzes apelin to Ang II and, therefore, is responsible for the degradation of both peptides. Apelin has emerged as a promising peptide biomarker of heart failure. The serum level of apelin in cardiovascular diseases tends to be decreased. Apelin is recognized as an imperative controller of systemic blood pressure and myocardium contractility. Dysregulation of the apelin/APJ system may be involved in the predisposition to cardiovascular diseases, and enhancing apelin action may have important therapeutic effects.
Collapse
|
177
|
Falcão-Pires I, Castro-Chaves P, Miranda-Silva D, Lourenço AP, Leite-Moreira AF. Physiological, pathological and potential therapeutic roles of adipokines. Drug Discov Today 2012; 17:880-9. [PMID: 22561894 DOI: 10.1016/j.drudis.2012.04.007] [Citation(s) in RCA: 96] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2011] [Revised: 03/19/2012] [Accepted: 04/19/2012] [Indexed: 02/06/2023]
Abstract
Formerly regarded purely as passive energy storage, adipose tissue is now recognized as a vital endocrine organ. Adipocytes secrete diverse peptide hormones named adipokines, which act in a autocrine, paracrine or endocrine way to influence several biological functions. Adipokines comprise diverse bioactive substances, including cytokines, growth, and complement factors, which perform essential regulatory functions related to energy balance, satiety and immunity. Presently adipokines have been widely implicated in obesity, diabetes, hypertension and cardiovascular diseases. In this article we aim to present a brief description of the roles and potential therapeutic modulation of adipokines, such as leptin, resistin, adiponectin, apelin, visfatin, FABP-4, tumor necrosis factor-α (TNF-α), interleukin-6 and plasminogen activator inhibitor-1 (PAI-1).
Collapse
Affiliation(s)
- Inês Falcão-Pires
- Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, Universidade do Porto, Porto, Portugal
| | | | | | | | | |
Collapse
|
178
|
Angiotensin-Converting Enzyme 2 (ACE2) Is a Key Modulator of the Renin Angiotensin System in Health and Disease. INTERNATIONAL JOURNAL OF PEPTIDES 2012; 2012:256294. [PMID: 22536270 PMCID: PMC3321295 DOI: 10.1155/2012/256294] [Citation(s) in RCA: 399] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2011] [Accepted: 12/05/2011] [Indexed: 12/29/2022]
Abstract
Angiotensin-converting enzyme 2 (ACE2) shares some homology with angiotensin-converting enzyme (ACE) but is not inhibited by ACE inhibitors. The main role of ACE2 is the degradation of Ang II resulting in the formation of angiotensin 1–7 (Ang 1–7) which opposes the actions of Ang II. Increased Ang II levels are thought to upregulate ACE2 activity, and in ACE2 deficient mice Ang II levels are approximately double that of wild-type mice, whilst Ang 1–7 levels are almost undetectable. Thus, ACE2 plays a crucial role in the RAS because it opposes the actions of Ang II. Consequently, it has a beneficial role in many diseases such as hypertension, diabetes, and cardiovascular disease where its expression is decreased. Not surprisingly, current therapeutic strategies for ACE2 involve augmenting its expression using ACE2 adenoviruses, recombinant ACE2 or compounds in these diseases thereby affording some organ protection.
Collapse
|
179
|
García-Hoz C, Sánchez-Fernández G, García-Escudero R, Fernández-Velasco M, Palacios-García J, Ruiz-Meana M, Díaz-Meco MT, Leitges M, Moscat J, García-Dorado D, Boscá L, Mayor F, Ribas C. Protein kinase C (PKC)ζ-mediated Gαq stimulation of ERK5 protein pathway in cardiomyocytes and cardiac fibroblasts. J Biol Chem 2012; 287:7792-7802. [PMID: 22232556 PMCID: PMC3293562 DOI: 10.1074/jbc.m111.282210] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2011] [Revised: 12/20/2011] [Indexed: 02/05/2023] Open
Abstract
Gq-coupled G protein-coupled receptors (GPCRs) mediate the actions of a variety of messengers that are key regulators of cardiovascular function. Enhanced Gα(q)-mediated signaling plays an important role in cardiac hypertrophy and in the transition to heart failure. We have recently described that Gα(q) acts as an adaptor protein that facilitates PKCζ-mediated activation of ERK5 in epithelial cells. Because the ERK5 cascade is known to be involved in cardiac hypertrophy, we have investigated the potential relevance of this pathway in cardiovascular Gq-dependent signaling using both cultured cardiac cell types and chronic administration of angiotensin II in mice. We find that PKCζ is required for the activation of the ERK5 pathway by Gq-coupled GPCR in neonatal and adult murine cardiomyocyte cultures and in cardiac fibroblasts. Stimulation of ERK5 by angiotensin II is blocked upon pharmacological inhibition or siRNA-mediated silencing of PKCζ in primary cultures of cardiac cells and in neonatal cardiomyocytes isolated from PKCζ-deficient mice. Moreover, upon chronic challenge with angiotensin II, these mice fail to promote the changes in the ERK5 pathway, in gene expression patterns, and in hypertrophic markers observed in wild-type animals. Taken together, our results show that PKCζ is essential for Gq-dependent ERK5 activation in cardiomyocytes and cardiac fibroblasts and indicate a key cardiac physiological role for the Gα(q)/PKCζ/ERK5 signaling axis.
Collapse
Affiliation(s)
- Carlota García-Hoz
- From the Departamento de Biología Molecular and Centro de Biología Molecular “Severo Ochoa,” Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), 28049 Madrid, Universidad Autónoma de Madrid, Spain
- the Instituto de Investigación Sanitaria La Princesa, 28006 Madrid, Spain
| | - Guzmán Sánchez-Fernández
- From the Departamento de Biología Molecular and Centro de Biología Molecular “Severo Ochoa,” Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), 28049 Madrid, Universidad Autónoma de Madrid, Spain
- the Instituto de Investigación Sanitaria La Princesa, 28006 Madrid, Spain
| | - Ramón García-Escudero
- the Molecular Oncology Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas, 28040 Madrid, Spain
| | | | - Julia Palacios-García
- From the Departamento de Biología Molecular and Centro de Biología Molecular “Severo Ochoa,” Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), 28049 Madrid, Universidad Autónoma de Madrid, Spain
- the Instituto de Investigación Sanitaria La Princesa, 28006 Madrid, Spain
| | - Marisol Ruiz-Meana
- the Vall d'Hebron University Hospital and Research Institute, Universitat Autonoma de Barcelona, 08035 Barcelona, Spain
| | - Maria Teresa Díaz-Meco
- the Tumor Microenvironment Program, Sanford-Burnham Medical Research Institute, La Jolla, California 92037
| | - Michael Leitges
- The Biotechnology Centre of Oslo, University of Oslo, 0317 Oslo, Norway, and
| | - Jorge Moscat
- the Tumor Microenvironment Program, Sanford-Burnham Medical Research Institute, La Jolla, California 92037
| | - David García-Dorado
- the Vall d'Hebron University Hospital and Research Institute, Universitat Autonoma de Barcelona, 08035 Barcelona, Spain
| | - Lisardo Boscá
- the Instituto de Investigaciones Biomédicas Alberto Sols, CSIC-UAM, 28029 Madrid, Spain
| | - Federico Mayor
- From the Departamento de Biología Molecular and Centro de Biología Molecular “Severo Ochoa,” Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), 28049 Madrid, Universidad Autónoma de Madrid, Spain
- the Instituto de Investigación Sanitaria La Princesa, 28006 Madrid, Spain
| | - Catalina Ribas
- From the Departamento de Biología Molecular and Centro de Biología Molecular “Severo Ochoa,” Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), 28049 Madrid, Universidad Autónoma de Madrid, Spain
- the Instituto de Investigación Sanitaria La Princesa, 28006 Madrid, Spain
| |
Collapse
|
180
|
Affiliation(s)
- Joerg Heineke
- From the Medizinische Hochschule Hannover (J.H.), Klinik für Kardiologie und Angiologie, Rebirth–Cluster of Excellence, Carl-Neuberg-Str.1, 30625 Hannover, Germany
| |
Collapse
|
181
|
Attané C, Foussal C, Le Gonidec S, Benani A, Daviaud D, Wanecq E, Guzmán-Ruiz R, Dray C, Bezaire V, Rancoule C, Kuba K, Ruiz-Gayo M, Levade T, Penninger J, Burcelin R, Pénicaud L, Valet P, Castan-Laurell I. Apelin treatment increases complete Fatty Acid oxidation, mitochondrial oxidative capacity, and biogenesis in muscle of insulin-resistant mice. Diabetes 2012; 61:310-20. [PMID: 22210322 PMCID: PMC3266414 DOI: 10.2337/db11-0100] [Citation(s) in RCA: 156] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Both acute and chronic apelin treatment have been shown to improve insulin sensitivity in mice. However, the effects of apelin on fatty acid oxidation (FAO) during obesity-related insulin resistance have not yet been addressed. Thus, the aim of the current study was to determine the impact of chronic treatment on lipid use, especially in skeletal muscles. High-fat diet (HFD)-induced obese and insulin-resistant mice treated by an apelin injection (0.1 μmol/kg/day i.p.) during 4 weeks had decreased fat mass, glycemia, and plasma levels of triglycerides and were protected from hyperinsulinemia compared with HFD PBS-treated mice. Indirect calorimetry experiments showed that apelin-treated mice had a better use of lipids. The complete FAO, the oxidative capacity, and mitochondrial biogenesis were increased in soleus of apelin-treated mice. The action of apelin was AMP-activated protein kinase (AMPK) dependent since all the effects studied were abrogated in HFD apelin-treated mice with muscle-specific inactive AMPK. Finally, the apelin-stimulated improvement of oxidative capacity led to decreased levels of acylcarnitines and enhanced insulin-stimulated glucose uptake in soleus. Thus, by promoting complete lipid use in muscle of insulin-resistant mice through mitochondrial biogenesis and tighter matching between FAO and the tricarboxylic acid cycle, apelin treatment could contribute to insulin sensitivity improvement.
Collapse
Affiliation(s)
- Camille Attané
- INSERM U1048, Toulouse, France
- Université de Toulouse, Université Paul Sabatier, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Toulouse, France
| | - Camille Foussal
- INSERM U1048, Toulouse, France
- Université de Toulouse, Université Paul Sabatier, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Toulouse, France
| | - Sophie Le Gonidec
- INSERM U1048, Toulouse, France
- Université de Toulouse, Université Paul Sabatier, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Toulouse, France
| | - Alexandre Benani
- Centre des Sciences du Goût et de l'Alimentation, Unité Mixte de Recherche 6265–Centre National de la Recherche Scientifique 13241–Institut National de la Recherche Agronomique, Université de Bourgogne, Dijon, France
| | - Danièle Daviaud
- INSERM U1048, Toulouse, France
- Université de Toulouse, Université Paul Sabatier, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Toulouse, France
| | - Estelle Wanecq
- INSERM U1048, Toulouse, France
- Université de Toulouse, Université Paul Sabatier, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Toulouse, France
| | - Rocío Guzmán-Ruiz
- Departemento de Ciencias Farmacéuticas y de la Alimentación, School of Pharmacy, University CEU–San Pablo, Madrid, Spain
| | - Cédric Dray
- INSERM U1048, Toulouse, France
- Université de Toulouse, Université Paul Sabatier, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Toulouse, France
| | - Veronic Bezaire
- INSERM U1048, Toulouse, France
- Université de Toulouse, Université Paul Sabatier, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Toulouse, France
| | - Chloé Rancoule
- INSERM U1048, Toulouse, France
- Université de Toulouse, Université Paul Sabatier, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Toulouse, France
| | - Keiji Kuba
- Department of Biological Informatics and Experimental Therapeutics, Akita University Graduate School of Medicine, Akita, Japan
| | - Mariano Ruiz-Gayo
- Departemento de Ciencias Farmacéuticas y de la Alimentación, School of Pharmacy, University CEU–San Pablo, Madrid, Spain
| | - Thierry Levade
- INSERM U1048, Toulouse, France
- Université de Toulouse, Université Paul Sabatier, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Toulouse, France
| | | | - Rémy Burcelin
- INSERM U1048, Toulouse, France
- Université de Toulouse, Université Paul Sabatier, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Toulouse, France
| | - Luc Pénicaud
- Centre des Sciences du Goût et de l'Alimentation, Unité Mixte de Recherche 6265–Centre National de la Recherche Scientifique 13241–Institut National de la Recherche Agronomique, Université de Bourgogne, Dijon, France
| | - Philippe Valet
- INSERM U1048, Toulouse, France
- Université de Toulouse, Université Paul Sabatier, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Toulouse, France
| | - Isabelle Castan-Laurell
- INSERM U1048, Toulouse, France
- Université de Toulouse, Université Paul Sabatier, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Toulouse, France
- Corresponding author: Isabelle Castan-Laurell,
| |
Collapse
|
182
|
Paskaradevan S, Scott IC. The Aplnr GPCR regulates myocardial progenitor development via a novel cell-non-autonomous, Gα(i/o) protein-independent pathway. Biol Open 2012; 1:275-85. [PMID: 23213418 PMCID: PMC3507289 DOI: 10.1242/bio.2012380] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Myocardial progenitor development involves the migration of cells to the anterior lateral plate mesoderm (ALPM) where they are exposed to the necessary signals for heart development to proceed. Whether the arrival of cells to this location is sufficient, or whether earlier signaling events are required, for progenitor development is poorly understood. Here we demonstrate that in the absence of Aplnr signaling, cells fail to migrate to the heart-forming region of the ALPM. Our work uncovers a previously uncharacterized cell-non-autonomous function for Aplnr signaling in cardiac development. Furthermore, we show that both the single known Aplnr ligand, Apelin, and the canonical Gαi/o proteins that signal downstream of Aplnr are dispensable for Aplnr function in the context of myocardial progenitor development. This novel Aplnr signal can be substituted for by activation of Gata5/Smarcd3 in myocardial progenitors, suggesting a novel mechanism for Aplnr signaling in the establishment of a niche required for the proper migration/development of myocardial progenitor cells.
Collapse
Affiliation(s)
- Sivani Paskaradevan
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, 555 University Avenue , Toronto, ON M5G 1X8 , Canada ; Department of Molecular Genetics, University of Toronto , Toronto, ON M5S 1A8 , Canada
| | | |
Collapse
|
183
|
Abdalla S, Fu X, Elzahwy SS, Klaetschke K, Streichert T, Quitterer U. Up-regulation of the cardiac lipid metabolism at the onset of heart failure. Cardiovasc Hematol Agents Med Chem 2012; 9:190-206. [PMID: 21711241 PMCID: PMC3319925 DOI: 10.2174/187152511797037583] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2011] [Accepted: 06/24/2011] [Indexed: 12/19/2022]
Abstract
Chronic pressure overload and atherosclerosis are primary etiologic factors for cardiac hypertrophy and failure. However, mechanisms underlying the transition from hypertrophy to heart failure are incompletely understood. We analyzed the development of heart failure in mice with chronic pressure overload induced by aortic constriction and compared the results with aged apolipoprotein E-deficient mice suffering from advanced atherosclerosis. We combined cardiac function analysis by echocardiography and invasive hemodynamics with a comprehensive microarray gene expression study (GSE25765-8). The microarray data showed that the onset of heart failure induced by pressure overload or advanced atherosclerosis was accompanied by a strong up-regulation of key lipid metabolizing enzymes involved in fat synthesis, storage and oxidation. Cardiac lipid overload may be involved in the progression of heart failure by enhancing cardiomyocyte death. Up-regulation of the cardiac lipid metabolism was related to oxygen and ATP depletion of failing hearts because anti-ischemic treatment with ranolazine normalized the cardiac lipid metabolism and improved cardiac function. Vice versa, inhibition of cellular respiration and ATP generation by mild thiol-blocking with cystamine triggered the cardiac lipid metabolism and caused signs of heart failure. Cardiac tissue specimens of patients with heart failure also showed high protein levels of key fat metabolizing enzymes as well as lipid accumulation. Taken together, our data strongly indicate that up-regulation of the cardiac lipid metabolism and myocardial lipid overload are underlying the development of heart failure.
Collapse
Affiliation(s)
- Said Abdalla
- Molecular Pharmacology Unit, Swiss Federal Institute of Technology and University of Zurich, Switzerland
| | | | | | | | | | | |
Collapse
|
184
|
Pope GR, Roberts EM, Lolait SJ, O’Carroll AM. Central and peripheral apelin receptor distribution in the mouse: species differences with rat. Peptides 2012; 33:139-48. [PMID: 22197493 PMCID: PMC3314948 DOI: 10.1016/j.peptides.2011.12.005] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2011] [Revised: 12/08/2011] [Accepted: 12/08/2011] [Indexed: 01/02/2023]
Abstract
The G protein-coupled apelin receptor (APJ) binds the endogenous peptide apelin and has been shown to have roles in many physiological systems. Thus far, distribution studies have predominantly been conducted in the rat and there is limited knowledge of the cellular distribution of APJ in mouse or human tissues. As recent functional studies have been conducted in APJ knock-out mice (APJ KO), in this study we undertook to characterize APJ mRNA and I(125)[Pyr(1)]apelin-13 binding site distribution in mouse tissues to enable correlation of distribution with function. We have utilized in situ hybridization histochemistry (ISHH) using APJ riboprobes, which revealed strong hybridization specifically in the paraventricular (PVN) and supraoptic (SON) nuclei of the hypothalamus and in the anterior pituitary, with marginally lower levels in the posterior pituitary. In the periphery, strong hybridization was observed in the lung, heart, adrenal cortex, renal medulla, ovary and uterus. Autoradiographic binding to APJ with I(125)[Pyr(1)]apelin-13 exhibited significant binding in the anterior pituitary, while lower levels were observed in the posterior pituitary and PVN and SON. In the periphery, strong receptor binding was observed in tissues exhibiting intense riboprobe hybridization, indicating a good correlation between receptor transcription and translation. While the distribution of APJ mRNA and functional protein in the mouse shows similarities to that of the rat, we report a species difference in central APJ distribution and in the pituitary gland.
Collapse
|
185
|
Koguchi W, Kobayashi N, Takeshima H, Ishikawa M, Sugiyama F, Ishimitsu T. Cardioprotective effect of apelin-13 on cardiac performance and remodeling in end-stage heart failure. Circ J 2011; 76:137-44. [PMID: 22082814 DOI: 10.1253/circj.cj-11-0689] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND Apelin and its cognate G protein-coupled receptor, APJ, constitute a signaling pathway with a positive inotropic effect on cardiac function. Recently, we and other investigators demonstrated that a reduction in myocardial apelin/APJ expression might play a critical role in experimental models of end-stage heart failure (HF). Therefore, we evaluated whether exogenous apelin infusion restores apelin/APJ expression and improves cardiac function in the failing heart of Dahl salt-sensitive hypertensive (DS) rats. METHODS AND RESULTS High salt-loaded DS rats were treated with vehicle and pyroglutamylated apelin-13 (Pyr-AP13; 200µg·kg(-1)·day(-1), IP) from the age of 11 to 18 weeks. Decreased end-systolic elastance and percent fractional shortening in failing rats was significantly ameliorated by Pyr-AP13. Pyr-AP13 effectively inhibited vascular lesion formation and suppressed expression of inflammation factors such as tumor necrosis factor-α and interleukin-1β protein. Downregulation of apelin and APJ expression, and phosphorylation of endothelial nitric oxide synthase at Ser(1177) and Akt at Ser(473) in failing rats was significantly increased by Pyr-AP13. Upregulation of NAD(P)H oxidase p22(phox), p47(phox), and gp91(phox) in DS rats was significantly suppressed by Pyr-AP13. CONCLUSIONS Exogenous apelin-13 may ameliorate cardiac dysfunction and remodeling and restore apelin/APJ expression in DS rats with end-stage HF. Thus, apelin-13 may have significant therapeutic potential for end-stage HF.
Collapse
Affiliation(s)
- Wataru Koguchi
- Department of Hypertension and Cardiorenal Medicine, Dokkyo Medical University School of Medicine, Tochigi, Japan
| | | | | | | | | | | |
Collapse
|
186
|
Apelin is required for non-neovascular remodeling in the retina. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 180:399-409. [PMID: 22067912 DOI: 10.1016/j.ajpath.2011.09.035] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2011] [Revised: 09/21/2011] [Accepted: 09/27/2011] [Indexed: 12/11/2022]
Abstract
Retinal pathologies are frequently accompanied by retinal vascular responses, including the formation of new vessels by angiogenesis (neovascularization). Pathological vascular changes may also include less well characterized traits of vascular remodeling that are non-neovascular, such as vessel pruning and the emergence of dilated and tortuous vessel phenotypes (telangiectasis). The molecular mechanisms underlying neovascular growth versus non-neovascular remodeling are poorly understood. We therefore undertook to identify novel regulators of non-neovascular remodeling in the retina by using the dystrophic Royal College of Surgeons (RCS) rat and the retinal dystrophy 1 (RD1) mouse, both of which display pronounced non-neovascular remodeling. Gene expression profiling of isolated retinal vessels from these mutant rodent models and wild-type controls revealed 60 differentially expressed genes. These included the genes for apelin (Apln) and for its receptor (Aplnr), both of which were strongly up-regulated in the mutants. Crossing RD1 mice into an Apln-null background substantially reduced vascular telangiectasia. In contrast, Apln gene deletion had no effect in two models of neovascular pathology [laser-induced choroidal neovascularization and the very low density lipoprotein receptor (Vldlr)-knockout mouse]. These findings suggest that in these models apelin has minimal effect on sprouting retinal angiogenesis, but contributes significantly to pathogenic non-neovascular remodeling.
Collapse
|
187
|
Pchejetski D, Foussal C, Alfarano C, Lairez O, Calise D, Guilbeau-Frugier C, Schaak S, Seguelas MH, Wanecq E, Valet P, Parini A, Kunduzova O. Apelin prevents cardiac fibroblast activation and collagen production through inhibition of sphingosine kinase 1. Eur Heart J 2011; 33:2360-9. [PMID: 22028387 DOI: 10.1093/eurheartj/ehr389] [Citation(s) in RCA: 122] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
AIMS Activation of cardiac fibroblasts and their differentiation into myofibroblasts is a key event in the progression of cardiac fibrosis that leads to end-stage heart failure. Apelin, an adipocyte-derived factor, exhibits a number of cardioprotective properties; however, whether apelin is involved in cardiac fibroblast activation and myofibroblast formation remains unknown. The aim of this study was to determine the effects of apelin in activated cardiac fibroblasts, the potential related mechanisms and impact on cardiac fibrotic remodelling process. METHODS AND RESULTS In vitro experiments were performed in mouse cardiac fibroblasts obtained from normal and pressure-overload hearts. Pretreatment of naive cardiac fibroblasts with apelin (1-100 nM) inhibited Transforming growth factor-β (TGF-β)-mediated expression of the myofibroblast marker α-smooth muscle actin (α-SMA) and collagen production. Furthermore, apelin decreased the spontaneous collagen production in cardiac fibroblasts isolated from hearts after aortic banding. Knockdown strategy and pharmacological inhibition revealed that prevention of collagen accumulation by apelin was mediated by a reduction in sphingosine kinase 1 (SphK1) activity. In vivo studies using the aortic banding model indicated that pretreatment with apelin attenuated the development of myocardial fibrotic remodelling and inhibited cardiac SphK1 activity and α-SMA expression. Moreover, administration of apelin 2 weeks after aortic banding prevented cardiac remodelling by inhibiting myocyte hypertrophy, cardiac fibrosis, and ventricular dysfunction. CONCLUSION Our data provide the first evidence that apelin inhibits TGF-β-stimulated activation of cardiac fibroblasts through a SphK1-dependent mechanism. We also demonstrated that the administration of apelin during the phase of reactive fibrosis prevents structural remodelling of the myocardium and ventricular dysfunction. These findings may have important implications for designing future therapies for myocardial performance during fibrotic remodelling, affecting the clinical management of patients with progressive heart failure.
Collapse
Affiliation(s)
- Dmitri Pchejetski
- Department of Surgery and Cancer, Imperial College London, London, UK
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
188
|
Ferris WF, Crowther NJ. Once fat was fat and that was that: our changing perspectives on adipose tissue. Cardiovasc J Afr 2011; 22:147-54. [PMID: 21713306 PMCID: PMC3721932 DOI: 10.5830/cvja-2010-083] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Abstract Past civilisations saw excess body fat as a symbol of wealth and prosperity as the general population struggled with food shortages and famine. Nowadays it is recognised that obesity is associated with co-morbidities such as cardiovascular disease and diabetes. Our views on the roll of adipose tissue have also changed, from being solely a passive energy store, to an important endocrine organ that modulates metabolism, immunity and satiety. The relationship between increased visceral adiposity and obesity-related co-morbidities has lead to the recognition that variation in fat distribution contributes to ethnic differences in the prevalence of obesity-related diseases. Our current negative view of adipose tissue may change with the use of pluripotent adipose-derived stromal cells, which may lead to future autologous stem cell therapies for bone, muscle, cardiac and cartilage disorders. Here, we briefly review the concepts that adipose tissue is an endocrine organ, that differences in body fat distribution underline the aetiology of obesity-related co-morbidities, and the use of adipose-derived stem cells for future therapies.
Collapse
Affiliation(s)
- W F Ferris
- Division of Endocrinology, Department of Medicine, Faculty of Health Sciences, University of Stellenbosch, Stellenbosch, South Africa.
| | | |
Collapse
|
189
|
Abstract
Pulmonary arterial hypertension (PAH) is a devastating disease characterized by pulmonary vasoconstriction, pulmonary arterial remodeling, abnormal angiogenesis and impaired right ventricular function. Despite progress in pharmacological therapy, there is still no cure for PAH. The peptide apelin and the G-protein coupled apelin receptor (APLNR) are expressed in several tissues throughout the organism. Apelin is localized in vascular endothelial cells while the APLNR is localized in both endothelial and smooth muscle cells in vessels and in the heart. Apelin is regulated by hypoxia inducible factor -1α and bone morphogenetic protein receptor-2. Patients with PAH have lower levels of plasma-apelin, and decreased apelin expression in pulmonary endothelial cells. Apelin has therefore been proposed as a potential biomarker for PAH. Furthermore, apelin plays a role in angiogenesis and regulates endothelial and smooth muscle cell apoptosis and proliferation complementary and opposite to vascular endothelial growth factor. In the systemic circulation, apelin modulates endothelial nitric oxide synthase (eNOS) expression, induces eNOS-dependent vasodilatation, counteracts angiotensin-II mediated vasoconstriction, and has positive inotropic and cardioprotective effects. Apelin attenuates vasoconstriction in isolated rat pulmonary arteries, and chronic treatment with apelin attenuates the development of pulmonary hypertension in animal models. The existing literature thus renders APLNR an interesting potential new therapeutic target for PH.
Collapse
Affiliation(s)
| | - Ole Hilberg
- Department of Allergology & Respiratory Diseases, Aarhus University Hospital, Denmark
| | | | | | - U. Simonsen
- Department of Biomedicine, Aarhus University, Denmark
| |
Collapse
|
190
|
Angiotensin-converting enzyme 2 is a key modulator of the renin-angiotensin system in cardiovascular and renal disease. Curr Opin Nephrol Hypertens 2011; 20:62-8. [PMID: 21099686 DOI: 10.1097/mnh.0b013e328341164a] [Citation(s) in RCA: 119] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW Angiotensin-converting enzyme 2 (ACE2) has recently emerged as a key regulator of the renin-angiotensin system in both health and disease. RECENT FINDINGS ACE2 deficiency is associated with elevated tissue and circulating levels of angiotensin II and reduced levels of angiotensin 1-7. Phenotypically, this results in a modest elevation in systolic blood pressure and left ventricular hypertrophy. In atherosclerosis-prone apolipoprotein E knockout mice, ACE2 deficiency results in augmented vascular inflammation and an inflammatory response that contributes to increased atherosclerotic plaque formation. In the kidney, ACE2 deficiency is associated with progressive glomerulosclerosis. Interventions such as ACE2 replenishment or augmentation of its actions have proven successful in reducing hypertension, plaque accumulation, and renal and cardiac damage in a range of different models. Although promising, the balance of the renin-angiotensin system remains complicated, with some evidence that overexpression of ACE2 may have adverse cardiac effects, and ACE2 and its metabolic products may promote epithelial-to-mesenchymal transition. SUMMARY Repletion of ACE2's activities offers a new strategy to complement current clinical interventions in treating hypertension, renal and cardiovascular disease. In particular conditions where ACE inhibition and angiotensin receptor blockade are partially effective, the adjunctive actions of ACE2 may not only reduce clinical escape but also augment the efficacy of interventions.
Collapse
|
191
|
Charles CJ. Update on apelin peptides as putative targets for cardiovascular drug discovery. Expert Opin Drug Discov 2011; 6:633-44. [DOI: 10.1517/17460441.2011.571251] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
192
|
Choucair-Jaafar N, Beetz N, Gilsbach R, Yalcin I, Waltisperger E, Freund-Mercier MJ, Monassier L, Hein L, Barrot M. Cardiovascular effects of chronic treatment with a β2-adrenoceptor agonist relieving neuropathic pain in mice. Neuropharmacology 2011; 61:51-60. [PMID: 21352833 DOI: 10.1016/j.neuropharm.2011.02.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2010] [Revised: 02/15/2011] [Accepted: 02/15/2011] [Indexed: 01/06/2023]
Abstract
Neuropathic pain is often a chronic condition, disabling and difficult to treat. Using a murine model of neuropathic pain induced by placing a polyethylene cuff around the main branch of the sciatic nerve, we have shown that chronic treatment with β-AR agonists is effective against neuropathic allodynia. β-mimetics are widely used against asthma and chronic obstructive pulmonary disease and may offer an interesting option for neuropathic pain management. The most prominent adverse effects of chronic treatment with β-mimetics are cardiovascular. In this study, we compared the action of low doses of the selective β(2)-AR agonist terbutaline and of a high dose of the mixed β(1)/β(2)-AR agonist isoproterenol on cardiovascular parameters in a neuropathic pain context. Isoproterenol was used as a positive control for some heart-related changes. Cardiac functions were studied by echocardiography, hemodynamic measurements, histological analysis of fibrosis and cardiac hypertrophy, and by quantitative real time PCR analysis of atrial natriuretic peptide (Nppa), periostin (Postn), connective tissue growth factor (Ctgf) and β-myosin heavy chain (Myh7). Our data show that a chronic treatment with the β(2)-AR agonist terbutaline at low antiallodynic dose does not affect cardiovascular parameters, whereas the mixed β(1)/β(2)-AR agonist isoproterenol induces cardiac hypertrophy. These data suggest that low doses of β(2)-AR agonists may provide a suitable treatment with rare side effects in neuropathic pain management. This study conducted in an animal model requires clinical confirmation in humans.
Collapse
Affiliation(s)
- Nada Choucair-Jaafar
- Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, 21 rue René Descartes, 67084 Strasbourg cedex, France
| | | | | | | | | | | | | | | | | |
Collapse
|
193
|
Validation of genetic association in apelin-AGTRL1 system with hypertension in a larger Han Chinese population. J Hypertens 2010; 28:1854-61. [PMID: 20485192 DOI: 10.1097/hjh.0b013e32833b1fad] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND AND OBJECTIVE We have recently resequenced apelin and AGTRL1 genes to identify candidate polymorphisms in family-based association with hypertension and related phenotypes. In this study, we aimed to determine and replicate these polymorphisms via a larger, independent case-control study in Shanghai Han Chinese. METHODS Two polymorphisms [rs3761581 (A/C) and T-1860C] in apelin gene and two [rs7119375 (G/A), rs10501367 (G/A)] in AGTRL1 gene were genotyped using the TaqMan assay among 969 patients diagnosed with essential hypertension and 980 age and sex-matched controls. Data were analyzed using Haplo.stats program. RESULTS In single-locus analysis, significant differences were observed in allele distribution of rs3761581 (P = 0.0156) in men and in rs7119375 (P = 0.0488) genotype distribution in women between patients and controls. Haplotype analysis indicated that haplotypes C-C-G-G (in order of T-1860C, rs3761581, rs7119375 and rs10501367) [adjusted odds ratio (ORadjusted) = 1.67, P = 0.0061] and T-A-A-A (ORadjusted = 1.62, P = 0.0008) conferred an increased risk for hypertension after adjustment for age, onset age, body mass index (BMI) and waist-to-hip ratio, whereas haplotype C-C-A-A (ORadjusted = 0.33, P = 0.0048) conferred a protective effect. In women, increased risk for hypertension was seen for haplotypes T-A-G-G (ORadjusted = 1.30, P = 0.0051), C-C-G-G (ORadjusted = 2.86, P < 0.0001), T-A-A-A (ORadjusted = 1.66, P = 0.0003) and C-C-A-A (ORadjusted = 2.65, P < 0.0001), whereas decreased risk was seen for haplotypes C-A-G-G (ORadjusted = 0.48, P < 0.0001) and T-C-G-G (ORadjusted = 0.40, P < 0.0001). Further haplotype-phenotype analyses indicated the robustness of these associations. For example, haplotype T-A-A-A was significantly associated with obesity index (BMI and waist-to-hip ratio) in both sexes and blood pressures in men (P-sim < 0.05). CONCLUSION In this exploratory pilot case-control study, we found robust haplotype-based and haplotype-phenotype associations of four well characterized polymorphisms in apelin-AGTRL1 system with hypertension and related phenotypes.
Collapse
|
194
|
Pitkin SL, Maguire JJ, Kuc RE, Davenport AP. Modulation of the apelin/APJ system in heart failure and atherosclerosis in man. Br J Pharmacol 2010; 160:1785-95. [PMID: 20649580 DOI: 10.1111/j.1476-5381.2010.00821.x] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND AND PURPOSE The aim of this study was to determine whether the apelin/APJ system is altered in human cardiovascular disease by investigating whether the expression of apelin or its receptor is altered at the protein level. EXPERIMENTAL APPROACH Radioligand binding studies were used to determine apelin receptor density in human cardiac tissues. Apelin peptide levels in cardiovascular tissues were determined by radioimmunoassay. In vitro pharmacology was used to assess vasoactive properties of apelin in human coronary artery. Localization of apelin and its receptor in coronary artery was determined using immunohistochemistry. KEY RESULTS Apelin receptor density was significantly decreased in left ventricle from patients with dilated cardiomyopathy or ischaemic heart disease compared with controls, but apelin peptide levels remained unchanged. Apelin was up-regulated in human atherosclerotic coronary artery and this additional peptide localized to the plaque, colocalizing with markers for macrophages and smooth muscle cells. Apelin potently constricted human coronary artery. CONCLUSIONS AND IMPLICATIONS We have detected changes in the apelin/APJ system in human diseased cardiac and vascular tissue. The decrease in receptor density in heart failure may limit the positive inotropic actions of apelin, contributing to contractile dysfunction. The contribution of the increased apelin levels in atherosclerotic coronary artery to disease progression remains to be determined. These data suggest a potential role for the apelin/APJ system in human cardiovascular disease.
Collapse
Affiliation(s)
- Sarah L Pitkin
- Clinical Pharmacology Unit, University of Cambridge, Level 6 Centre for Clinical Investigation, Box 110 Addenbrooke's Hospital, Cambridge, UK
| | | | | | | |
Collapse
|
195
|
Bodineau L, Hus-Citharel A, Llorens-Cortes C. Participation de l’apéline à la régulation de l’équilibre hydrique, de l’homéostasie glucidique et des fonctions cardiovasculaires. ANNALES D'ENDOCRINOLOGIE 2010; 71:249-56. [DOI: 10.1016/j.ando.2010.03.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2010] [Accepted: 03/12/2010] [Indexed: 10/19/2022]
|
196
|
Pitkin SL, Maguire JJ, Bonner TI, Davenport AP. International Union of Basic and Clinical Pharmacology. LXXIV. Apelin receptor nomenclature, distribution, pharmacology, and function. Pharmacol Rev 2010; 62:331-42. [PMID: 20605969 DOI: 10.1124/pr.110.002949] [Citation(s) in RCA: 147] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2025] Open
Abstract
A gene encoding a novel class a G-protein-coupled receptor was discovered in 1993 by homology cloning and was called APJ. It was designated an "orphan" receptor until 1998, when its endogenous ligand was identified and named apelin (for APJ endogenous ligand). Since this pairing, both apelin and its receptor have been found to have a widespread distribution in both the central nervous system and the periphery. A number of physiological and pathophysiological roles for the receptor have emerged, including regulation of cardiovascular function, fluid homeostasis, and the adipoinsular axis. This review outlines the official International Union of Pharmacology Committee on Receptor Nomenclature and Drug Classification nomenclature, designating the receptor protein as the apelin receptor, together with current knowledge of its pharmacology, distribution, and functions.
Collapse
Affiliation(s)
- Sarah L Pitkin
- Clinical Pharmacology Unit, University of Cambridge, Addenbrooke's Hospital, Cambridge, CB2 0QQ, UK
| | | | | | | |
Collapse
|
197
|
Abstract
Sprouting of developing blood vessels is mediated by specialized motile endothelial cells localized at the tips of growing capillaries. Following behind the tip cells, endothelial stalk cells form the capillary lumen and proliferate. Expression of the Notch ligand Delta-like-4 (Dll4) in tip cells suppresses tip cell fate in neighboring stalk cells via Notch signaling. In DLL4(+/-) mouse mutants, most retinal endothelial cells display morphologic features of tip cells. We hypothesized that these mouse mutants could be used to isolate tip cells and so to determine their genetic repertoire. Using transcriptome analysis of retinal endothelial cells isolated from DLL4(+/-) and wild-type mice, we identified 3 clusters of tip cell-enriched genes, encoding extracellular matrix degrading enzymes, basement membrane components, and secreted molecules. Secreted molecules endothelial-specific molecule 1, angiopoietin 2, and apelin bind to cognate receptors on endothelial stalk cells. Knockout mice and zebrafish morpholino knockdown of apelin showed delayed angiogenesis and reduced proliferation of stalk cells expressing the apelin receptor APJ. Thus, tip cells may regulate angiogenesis via matrix remodeling, production of basement membrane, and release of secreted molecules, some of which regulate stalk cell behavior.
Collapse
|
198
|
Iturrioz X, Gerbier R, Leroux V, Alvear-Perez R, Maigret B, Llorens-Cortes C. By interacting with the C-terminal Phe of apelin, Phe255 and Trp259 in helix VI of the apelin receptor are critical for internalization. J Biol Chem 2010; 285:32627-37. [PMID: 20675385 DOI: 10.1074/jbc.m110.127167] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Apelin is the endogenous ligand of the orphan seven-transmembrane domain (TM) G protein-coupled receptor APJ. Apelin is involved in the regulation of body fluid homeostasis and cardiovascular functions. We previously showed the importance of the C-terminal Phe of apelin 17 (K17F) in the hypotensive activity of this peptide. Here, we show either by deleting the Phe residue (K16P) or by substituting it by an Ala (K17A), that it plays a crucial role in apelin receptor internalization but not in apelin binding or in Gα(i)-protein coupling. Then we built a homology three-dimensional model of the human apelin receptor using the cholecystokinin receptor-1 model as a template, and we subsequently docked K17F into the binding site. We visualized a hydrophobic cavity at the bottom of the binding pocket in which the C-terminal Phe of K17F was embedded by Trp(152) in TMIV and Trp(259) and Phe(255) in TMVI. Using molecular modeling and site-directed mutagenesis studies, we further showed that Phe(255) and Trp(259) are key residues in triggering receptor internalization without playing a role in apelin binding or in Gα(i)-protein coupling. These findings bring new insights into apelin receptor activation and show that Phe(255) and Trp(259), by interacting with the C-terminal Phe of the pyroglutamyl form of apelin 13 (pE13F) or K17F, are crucial for apelin receptor internalization.
Collapse
Affiliation(s)
- Xavier Iturrioz
- INSERM, U691, Collège de France, Université Pierre et Marie-Curie Paris 6, Paris FR-75005, France
| | | | | | | | | | | |
Collapse
|
199
|
Kuba K, Imai Y, Ohto-Nakanishi T, Penninger JM. Trilogy of ACE2: a peptidase in the renin-angiotensin system, a SARS receptor, and a partner for amino acid transporters. Pharmacol Ther 2010; 128:119-28. [PMID: 20599443 PMCID: PMC7112678 DOI: 10.1016/j.pharmthera.2010.06.003] [Citation(s) in RCA: 385] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2010] [Accepted: 06/09/2010] [Indexed: 02/07/2023]
Abstract
Angiotensin-converting enzyme (ACE) 2 is a homolog to the carboxypeptidase ACE, which generates angiotensin II, the main active peptide of renin-angiotensin system (RAS). After the cloning of ACE2 in 2000, three major ACE2 functions have been described so far. First ACE2 has emerged as a potent negative regulator of the RAS counterbalancing the multiple functions of ACE. By targeting angiotensin II ACE2 exhibits a protective role in the cardiovascular system and many other organs. Second ACE2 was identified as an essential receptor for the SARS coronavirus that causes severe acute lung failure. Downregulation of ACE2 strongly contributes to the pathogenesis of severe lung failure. Third, both ACE2 and its homologue Collectrin can associate with amino acid transporters and play essential role in the absorption of amino acids in the kidney and gut. In this review, we will discuss the multiple biological functions of ACE2.
Collapse
Affiliation(s)
- Keiji Kuba
- Department of Biological Informatics and Experimental Therapeutics, Akita University Graduate School of Medicine, Akita 010-8543, Japan.
| | | | | | | |
Collapse
|
200
|
Myocardial Apelin Production is Reduced in Humans With Left Ventricular Systolic Dysfunction. J Card Fail 2010; 16:556-61. [DOI: 10.1016/j.cardfail.2010.02.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2009] [Revised: 02/09/2010] [Accepted: 02/10/2010] [Indexed: 11/21/2022]
|