1
|
Wei W, Smrcka AV. Internalized β2-Adrenergic Receptors Oppose PLC-Dependent Hypertrophic Signaling. Circ Res 2024; 135:e24-e38. [PMID: 38813686 PMCID: PMC11223973 DOI: 10.1161/circresaha.123.323201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 05/16/2024] [Indexed: 05/31/2024]
Abstract
BACKGROUND Chronically elevated neurohumoral drive, and particularly elevated adrenergic tone leading to β-adrenergic receptor (β-AR) overstimulation in cardiac myocytes, is a key mechanism involved in the progression of heart failure. β1-AR (β1-adrenergic receptor) and β2-ARs (β2-adrenergic receptor) are the 2 major subtypes of β-ARs present in the human heart; however, they elicit different or even opposite effects on cardiac function and hypertrophy. For example, chronic activation of β1-ARs drives detrimental cardiac remodeling while β2-AR signaling is protective. The underlying molecular mechanisms for cardiac protection through β2-ARs remain unclear. METHODS β2-AR signaling mechanisms were studied in isolated neonatal rat ventricular myocytes and adult mouse ventricular myocytes using live cell imaging and Western blotting methods. Isolated myocytes and mice were used to examine the roles of β2-AR signaling mechanisms in the regulation of cardiac hypertrophy. RESULTS Here, we show that β2-AR activation protects against hypertrophy through inhibition of phospholipaseCε signaling at the Golgi apparatus. The mechanism for β2-AR-mediated phospholipase C inhibition requires internalization of β2-AR, activation of Gi and Gβγ subunit signaling at endosome and ERK (extracellular regulated kinase) activation. This pathway inhibits both angiotensin II and Golgi-β1-AR-mediated stimulation of phosphoinositide hydrolysis at the Golgi apparatus ultimately resulting in decreased PKD (protein kinase D) and histone deacetylase 5 phosphorylation and protection against cardiac hypertrophy. CONCLUSIONS This reveals a mechanism for β2-AR antagonism of the phospholipase Cε pathway that may contribute to the known protective effects of β2-AR signaling on the development of heart failure.
Collapse
Affiliation(s)
- Wenhui Wei
- Department of Pharmacology, University of Michigan School of Medicine, Ann Arbor, United States
| | - Alan V. Smrcka
- Department of Pharmacology, University of Michigan School of Medicine, Ann Arbor, United States
| |
Collapse
|
2
|
Lymperopoulos A. Clinical pharmacology of cardiac cyclic AMP in human heart failure: too much or too little? Expert Rev Clin Pharmacol 2023; 16:623-630. [PMID: 37403791 PMCID: PMC10529896 DOI: 10.1080/17512433.2023.2233891] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 07/04/2023] [Indexed: 07/06/2023]
Abstract
INTRODUCTION Cyclic 3', 5'-adenosine monophosphate (cAMP) is a major signaling hub in cardiac physiology. Although cAMP signaling has been extensively studied in cardiac cells and animal models of heart failure (HF), not much is known about its actual amount present inside human failing or non-failing cardiomyocytes. Since many drugs used in HF work via cAMP, it is crucial to determine the status of its intracellular levels in failing vs. normal human hearts. AREAS COVERED Only studies performed on explanted/excised cardiac tissues from patients were examined. Studies that contained no data from human hearts or no data on cAMP levels per se were excluded from this perspective's analysis. EXPERT OPINION Currently, there is no consensus on the status of cAMP levels in human failing vs. non-failing hearts. Several studies on animal models may suggest maladaptive (e.g. pro-apoptotic) effects of cAMP on HF, advocating for cAMP lowering for therapy, but human studies almost universally indicate that myocardial cAMP levels are deficient in human failing hearts. It is the expert opinion of this perspective that intracellular cAMP levels are too low in human failing hearts, contributing to the disease. Strategies to increase (restore), not decrease, these levels should be pursued in human HF.
Collapse
Affiliation(s)
- Anastasios Lymperopoulos
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, Nova Southeastern University Barry and Judy Silverman College of Pharmacy, Fort Lauderdale, FL, USA
| |
Collapse
|
3
|
Wei W, Smrcka AV. Internalized β2-Adrenergic Receptors Inhibit Subcellular Phospholipase C-Dependent Cardiac Hypertrophic Signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.07.544153. [PMID: 37333278 PMCID: PMC10274790 DOI: 10.1101/2023.06.07.544153] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Chronically elevated neurohumoral drive, and particularly elevated adrenergic tone leading to β-adrenergic receptor (β-AR) overstimulation in cardiac myocytes, is a key mechanism involved in the progression of heart failure. β1-AR and β2-ARs are the two major subtypes of β-ARs present in the human heart, however, they elicit different or even opposite effects on cardiac function and hypertrophy. For example, chronic activation of β1ARs drives detrimental cardiac remodeling while β2AR signaling is protective. The underlying molecular mechanisms for cardiac protection through β2ARs remain unclear. Here we show that β2-AR protects against hypertrophy through inhibition of PLCε signaling at the Golgi apparatus. The mechanism for β2AR-mediated PLC inhibition requires internalization of β2AR, activation of Gi and Gβγ subunit signaling at endosomes and ERK activation. This pathway inhibits both angiotensin II and Golgi-β1-AR-mediated stimulation of phosphoinositide hydrolysis at the Golgi apparatus ultimately resulting in decreased PKD and HDAC5 phosphorylation and protection against cardiac hypertrophy. This reveals a mechanism for β2-AR antagonism of the PLCε pathway that may contribute to the known protective effects of β2-AR signaling on the development of heart failure.
Collapse
Affiliation(s)
- Wenhui Wei
- Department of Pharmacology, University of Michigan School of Medicine, Ann Arbor, United States
| | - Alan V. Smrcka
- Department of Pharmacology, University of Michigan School of Medicine, Ann Arbor, United States
| |
Collapse
|
4
|
Moustafa A, Hashemi S, Brar G, Grigull J, Ng SHS, Williams D, Schmitt-Ulms G, McDermott JC. The MEF2A transcription factor interactome in cardiomyocytes. Cell Death Dis 2023; 14:240. [PMID: 37019881 PMCID: PMC10076289 DOI: 10.1038/s41419-023-05665-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 12/02/2022] [Accepted: 02/08/2023] [Indexed: 04/07/2023]
Abstract
Transcriptional regulators encoded by the Myocyte Enhancer Factor 2 (MEF2) gene family play a fundamental role in cardiac development, homeostasis and pathology. Previous studies indicate that MEF2A protein-protein interactions serve as a network hub in several cardiomyocyte cellular processes. Based on the idea that interactions with regulatory protein partners underly the diverse roles of MEF2A in cardiomyocyte gene expression, we undertook a systematic unbiased screen of the MEF2A protein interactome in primary cardiomyocytes using an affinity purification-based quantitative mass spectrometry approach. Bioinformatic processing of the MEF2A interactome revealed protein networks involved in the regulation of programmed cell death, inflammatory responses, actin dynamics and stress signaling in primary cardiomyocytes. Further biochemical and functional confirmation of specific protein-protein interactions documented a dynamic interaction between MEF2A and STAT3 proteins. Integration of transcriptome level data from MEF2A and STAT3-depleted cardiomyocytes reveals that the balance between MEF2A and STAT3 activity exerts a level of executive control over the inflammatory response and cardiomyocyte cell survival and experimentally ameliorates Phenylephrine induced cardiomyocyte hypertrophy. Lastly, we identified several MEF2A/STAT3 co-regulated genes, including the MMP9 gene. Herein, we document the cardiomyocyte MEF2A interactome, which furthers our understanding of protein networks involved in the hierarchical control of normal and pathophysiological cardiomyocyte gene expression in the mammalian heart.
Collapse
Affiliation(s)
- Amira Moustafa
- Department of Biology, York University, Toronto, ON, M3J 1P3, Canada
- Muscle Health Research Centre (MHRC), York University, Toronto, ON, M3J 1P3, Canada
- Centre for Research in Biomolecular Interactions (CRBI), York University, Toronto, ON, M3J 1P3, Canada
| | - Sara Hashemi
- Analytical Sciences, Sanofi, Toronto, ON, M2R 3T4, Canada
- Seneca College, School of Health Sciences, King City, ON, L7B 1B3, Canada
| | - Gurnoor Brar
- Department of Biology, York University, Toronto, ON, M3J 1P3, Canada
- Muscle Health Research Centre (MHRC), York University, Toronto, ON, M3J 1P3, Canada
- Centre for Research in Biomolecular Interactions (CRBI), York University, Toronto, ON, M3J 1P3, Canada
| | - Jörg Grigull
- Department of Mathematics and Statistics, York University, Toronto, ON, M3J1P3, Canada
| | - Siemon H S Ng
- Analytical Sciences, Sanofi, Toronto, ON, M2R 3T4, Canada
- Analytical Development, Notch Therapeutics, Toronto, ON, M5G 1M1, Canada
| | - Declan Williams
- Tanz Centre for Research in Neurodegenerative Diseases, and Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5T 0S8, Canada
| | - Gerold Schmitt-Ulms
- Tanz Centre for Research in Neurodegenerative Diseases, and Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5T 0S8, Canada
| | - John C McDermott
- Department of Biology, York University, Toronto, ON, M3J 1P3, Canada.
- Muscle Health Research Centre (MHRC), York University, Toronto, ON, M3J 1P3, Canada.
- Centre for Research in Biomolecular Interactions (CRBI), York University, Toronto, ON, M3J 1P3, Canada.
| |
Collapse
|
5
|
Borges JI, Suster MS, Lymperopoulos A. Cardiac RGS Proteins in Human Heart Failure and Atrial Fibrillation: Focus on RGS4. Int J Mol Sci 2023; 24:6136. [PMID: 37047106 PMCID: PMC10147095 DOI: 10.3390/ijms24076136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/22/2023] [Accepted: 03/23/2023] [Indexed: 04/14/2023] Open
Abstract
The regulator of G protein signaling (RGS) proteins are crucial for the termination of G protein signals elicited by G protein-coupled receptors (GPCRs). This superfamily of cell membrane receptors, by far the largest and most versatile in mammals, including humans, play pivotal roles in the regulation of cardiac function and homeostasis. Perturbations in both the activation and termination of their G protein-mediated signaling underlie numerous heart pathologies, including heart failure (HF) and atrial fibrillation (AFib). Therefore, RGS proteins play important roles in the pathophysiology of these two devasting cardiac diseases, and several of them could be targeted therapeutically. Although close to 40 human RGS proteins have been identified, each RGS protein seems to interact only with a specific set of G protein subunits and GPCR types/subtypes in any given tissue or cell type. Numerous in vitro and in vivo studies in animal models, and also in diseased human heart tissue obtained from transplantations or tissue banks, have provided substantial evidence of the roles various cardiomyocyte RGS proteins play in cardiac normal homeostasis as well as pathophysiology. One RGS protein in particular, RGS4, has been reported in what are now decades-old studies to be selectively upregulated in human HF. It has also been implicated in protection against AFib via knockout mice studies. This review summarizes the current understanding of the functional roles of cardiac RGS proteins and their implications for the treatment of HF and AFib, with a specific focus on RGS4 for the aforementioned reasons but also because it can be targeted successfully with small organic molecule inhibitors.
Collapse
Affiliation(s)
| | | | - Anastasios Lymperopoulos
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, Barry and Judy Silverrman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328-2018, USA
| |
Collapse
|
6
|
Del Calvo G, Baggio Lopez T, Lymperopoulos A. The therapeutic potential of targeting cardiac RGS4. Ther Adv Cardiovasc Dis 2023; 17:17539447231199350. [PMID: 37724539 PMCID: PMC10510358 DOI: 10.1177/17539447231199350] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 08/16/2023] [Indexed: 09/21/2023] Open
Abstract
G protein-coupled receptors (GPCRs) play pivotal roles in regulation of cardiac function and homeostasis. To function properly, every cell needs these receptors to be stimulated only when a specific extracellular stimulus is present, and to be silenced the moment that stimulus is removed. The regulator of G protein signaling (RGS) proteins are crucial for the latter to occur at the cell membrane, where the GPCR normally resides. Perturbations in both activation and termination of G protein signaling underlie numerous heart pathologies. Although more than 30 mammalian RGS proteins have been identified, each RGS protein seems to interact only with a specific set of G protein subunits and GPCR types/subtypes in any given tissue or cell type, and this applies to the myocardium as well. A large number of studies have provided substantial evidence for the roles various RGS proteins expressed in cardiomyocytes play in cardiac physiology and heart disease pathophysiology. This review summarizes the current understanding of the functional roles of cardiac RGS proteins and their implications for the treatment of specific heart diseases, such as heart failure and atrial fibrillation. We focus on cardiac RGS4 in particular, since this isoform appears to be selectively (among the RGS protein family) upregulated in human heart failure and is also the target of ongoing drug discovery efforts for the treatment of a variety of diseases.
Collapse
Affiliation(s)
- Giselle Del Calvo
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, USA
| | - Teresa Baggio Lopez
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, USA
| | - Anastasios Lymperopoulos
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, 3200 South University Drive, HPD (Terry) Building/Room 1350, Fort Lauderdale, FL 33328-2018, USA
| |
Collapse
|
7
|
Sabbah HN, Zhang K, Gupta RC, Xu J, Singh-Gupta V, Ma M, Stauber K, Nguyen N, Adams J. Intravenous Infusion of the β 3-Adrenergic Receptor Antagonist APD418 Improves Left Ventricular Systolic Function in Dogs With Systolic Heart Failure. J Card Fail 2020; 27:242-252. [PMID: 33352205 DOI: 10.1016/j.cardfail.2020.12.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 11/08/2020] [Accepted: 12/08/2020] [Indexed: 11/27/2022]
Abstract
BACKGROUND Unlike β1- and β2-adrenergic receptors (ARs), β3-AR stimulation inhibits cardiac contractility and relaxation. In the failing left ventricular (LV) myocardium, β3-ARs are upregulated, and can be maladaptive in the setting of decompensation by contributing to LV dysfunction. This study examined the effects of intravenous infusions of the β3-AR antagonist APD418 on cardiovascular function and safety in dogs with systolic heart failure (HF). METHODS AND RESULTS Three separate studies were performed in 21 dogs with coronary microembolization-induced HF (LV ejection fraction [LVEF] of approximately 35%). Studies 1 and 2 (n = 7 dogs each) were APD418 dose escalation studies (dosing range, 0.35-15.00 mg/kg/h) designed to identify an effective dose of APD418 to be used in study 3. Study 3, the sustained efficacy study, (n = 7 dogs) was a 6-hour constant intravenous infusion of APD418 at a dose of 4.224 mg/kg (0.70 mg/kg/h) measuring key hemodynamic endpoints (e.g., EF, cardiac output, the time velocity integral of the mitral inflow velocity waveform representing early filling to time-velocity integral representing left atrial contraction [Ei/Ai]). Studies 1 and 2 showed a dose-dependent increase of LVEF and Ei/Ai, the latter being an index of LV diastolic function. In study 3, infusion of APD418 over 6 hours increased LVEF from 31 ± 1% to 38 ± 1% (P < .05) and increased Ei/Ai from 3.4 ± 0.4 to 4.9 ± 0.5 (P < .05). Vehicle had no effect on the LVEF or Ei/Ai. In study 3, APD418 had no significant effects on the HR or the systemic blood pressure. CONCLUSIONS Intravenous infusions of APD418 in dogs with systolic HF elicit significant positive inotropic and lusitropic effects. These findings support the development of APD418 for the in-hospital treatment of patients with an acute exacerbation of chronic HF.
Collapse
Affiliation(s)
- Hani N Sabbah
- Department of Medicine, Division of Cardiovascular Medicine, Henry Ford Hospital, Detroit, Michigan.
| | - Kefei Zhang
- Department of Medicine, Division of Cardiovascular Medicine, Henry Ford Hospital, Detroit, Michigan
| | - Ramesh C Gupta
- Department of Medicine, Division of Cardiovascular Medicine, Henry Ford Hospital, Detroit, Michigan
| | - Jiang Xu
- Department of Medicine, Division of Cardiovascular Medicine, Henry Ford Hospital, Detroit, Michigan
| | - Vinita Singh-Gupta
- Department of Medicine, Division of Cardiovascular Medicine, Henry Ford Hospital, Detroit, Michigan
| | - Michael Ma
- Arena Pharmaceuticals, Inc., San Diego, California
| | | | | | - John Adams
- Arena Pharmaceuticals, Inc., San Diego, California
| |
Collapse
|
8
|
Mora MT, Gong JQX, Sobie EA, Trenor B. The role of β-adrenergic system remodeling in human heart failure: A mechanistic investigation. J Mol Cell Cardiol 2020; 153:14-25. [PMID: 33326834 DOI: 10.1016/j.yjmcc.2020.12.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 12/02/2020] [Accepted: 12/08/2020] [Indexed: 01/01/2023]
Abstract
β-adrenergic receptor antagonists (β-blockers) are extensively used to improve cardiac performance in heart failure (HF), but the electrical improvements with these clinical treatments are not fully understood. The aim of this study was to analyze the electrophysiological effects of β-adrenergic system remodeling in heart failure with reduced ejection fraction and the underlying mechanisms. We used a combined mathematical model that integrated β-adrenergic signaling with electrophysiology and calcium cycling in human ventricular myocytes. HF remodeling, both in the electrophysiological and signaling systems, was introduced to quantitatively analyze changes in electrophysiological properties due to the stimulation of β-adrenergic receptors in failing myocytes. We found that the inotropic effect of β-adrenergic stimulation was reduced in HF due to the altered Ca2+ dynamics resulting from the combination of structural, electrophysiological and signaling remodeling. Isolated cells showed proarrhythmic risk after sympathetic stimulation because early afterdepolarizations appeared, and the vulnerability was greater in failing myocytes. When analyzing coupled cells, β-adrenergic stimulation reduced transmural repolarization gradients between endocardium and epicardium in normal tissue, but was less effective at reducing these gradients after HF remodeling. The comparison of the selective activation of β-adrenergic isoforms revealed that the response to β2-adrenergic receptors stimulation was blunted in HF while β1-adrenergic receptors downstream effectors regulated most of the changes observed after sympathetic stimulation. In conclusion, this study was able to reproduce an altered β-adrenergic activity on failing myocytes and to explain the mechanisms involved. The derived predictions could help in the treatment of HF and guide in the design of future experiments.
Collapse
Affiliation(s)
- Maria T Mora
- Centro de Investigación e Innovación en Bioingeniería, Universitat Politècnica de València, Valencia, Spain
| | - Jingqi Q X Gong
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Eric A Sobie
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Beatriz Trenor
- Centro de Investigación e Innovación en Bioingeniería, Universitat Politècnica de València, Valencia, Spain.
| |
Collapse
|
9
|
Garland H, Vuylsteke A. Biased Agonism: The Future (and Present) of Inotropic Support. J Cardiothorac Vasc Anesth 2020; 34:3449-3451. [PMID: 32921607 DOI: 10.1053/j.jvca.2020.08.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 07/09/2020] [Accepted: 08/03/2020] [Indexed: 11/11/2022]
Abstract
Biased agonism, which is the concept that different ligands activate different downstream signalling partners in different ratios to cause different functional effects, is yet to gain appropriate appreciation in the field of inotropic pharmacology. Biased agonism has already proven to be a clinically translatable technology in analgesic pharmacology, but this development is yet to be translated into inotropes. A better appreciation of bias in clinically used inotropes and a focus on bias when developing novel inotropes has the potential to lead to more targeted, personalized, and cleaner inotropes.
Collapse
Affiliation(s)
- Huw Garland
- West Hertfordshire Hospital, Watford, United Kingdom.
| | | |
Collapse
|
10
|
Man KNM, Navedo MF, Horne MC, Hell JW. β 2 Adrenergic Receptor Complexes with the L-Type Ca 2+ Channel Ca V1.2 and AMPA-Type Glutamate Receptors: Paradigms for Pharmacological Targeting of Protein Interactions. Annu Rev Pharmacol Toxicol 2019; 60:155-174. [PMID: 31561738 DOI: 10.1146/annurev-pharmtox-010919-023404] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Formation of signaling complexes is crucial for the orchestration of fast, efficient, and specific signal transduction. Pharmacological disruption of defined signaling complexes has the potential for specific intervention in selected regulatory pathways without affecting organism-wide disruption of parallel pathways. Signaling by epinephrine and norepinephrine through α and β adrenergic receptors acts on many signaling pathways in many cell types. Here, we initially provide an overview of the signaling complexes formed between the paradigmatic β2 adrenergic receptor and two of its most important targets, the L-type Ca2+ channel CaV1.2 and the AMPA-type glutamate receptor. Importantly, both complexes contain the trimeric Gs protein, adenylyl cyclase, and the cAMP-dependent protein kinase, PKA. We then discuss the functional implications of the formation of these complexes, how those complexes can be specifically disrupted, and how such disruption could be utilized in the pharmacological treatment of disease.
Collapse
Affiliation(s)
- Kwun Nok Mimi Man
- Department of Pharmacology, University of California, Davis, California 95616, USA;
| | - Manuel F Navedo
- Department of Pharmacology, University of California, Davis, California 95616, USA;
| | - Mary C Horne
- Department of Pharmacology, University of California, Davis, California 95616, USA;
| | - Johannes W Hell
- Department of Pharmacology, University of California, Davis, California 95616, USA;
| |
Collapse
|
11
|
Discovery of β-arrestin-biased β 2-adrenoceptor agonists from 2-amino-2-phenylethanol derivatives. Acta Pharmacol Sin 2019; 40:1095-1105. [PMID: 30643208 DOI: 10.1038/s41401-018-0200-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 11/26/2018] [Indexed: 01/08/2023]
Abstract
β-Arrestins are a small family of proteins important for signal transduction at G protein-coupled receptors (GPCRs). β-Arrestins are involved in the desensitization of GPCRs. Recently, biased ligands possessing different efficacies in activating the G protein- versus the β-arrestin-dependent signals downstream of a single GPCR have emerged, which can be used to selectively modulate GPCR signal transduction in such a way that desirable signals are enhanced to produce therapeutic effects while undesirable signals of the same GPCR are suppressed to avoid side effects. In the present study, we evaluated agonist bias for compounds developed along a drug discovery project of β2-adrenoceptor agonists. About 150 compounds, including derivatives of fenoterol, 2-amino-1-phenylethanol and 2-amino-2-phenylethanol, were obtained or synthesized, and initially screened for their β-adrenoceptor-mediated activities in the guinea pig tracheal smooth muscle relaxation assay or the cardiomyocyte contractility assay. Nineteen bioactive compounds were further assessed using both the HTRF cAMP assay and the PathHunter β-arrestin assay. Their concentration-response data in stimulating cAMP synthesis and β-arrestin recruitment were applied to the Black-Leff operational model for ligand bias quantitation. As a result, three compounds (L-2, L-4, and L-12) with the core structure of 5-(1-amino-2-hydroxyethyl)-8-hydroxyquinolin-2(1H)-one were identified as a new series of β-arrestin-biased β2-adrenoceptor agonists, whereas salmeterol was found to be Gs-biased. These findings would facilitate the development of novel drugs for the treatment of both heart failure and asthma.
Collapse
|
12
|
Local membrane charge regulates β 2 adrenergic receptor coupling to G i3. Nat Commun 2019; 10:2234. [PMID: 31110175 PMCID: PMC6527575 DOI: 10.1038/s41467-019-10108-0] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Accepted: 04/18/2019] [Indexed: 11/13/2022] Open
Abstract
The β2 adrenergic receptor (β2AR) signals through both Gs and Gi in cardiac myocytes, and the Gi pathway counteracts the Gs pathway. However, Gi coupling is much less efficient than Gs coupling in most cell-based and biochemical assays, making it difficult to study β2AR−Gi interactions. Here we investigate the role of phospholipid composition on Gs and Gi coupling. While negatively charged phospholipids are known to enhance agonist affinity and stabilize an active state of the β2AR, we find that they impair coupling to Gi3 and facilitate coupling to Gs. Positively charged Ca2+ and Mg2+, known to interact with the negative charge on phospholipids, facilitates Gi3 coupling. Mutational analysis suggests that Ca2+ coordinates an interaction between phospholipid and the negatively charged EDGE motif on the amino terminal helix of Gi3. Taken together, our observations suggest that local membrane charge modulates the interaction between β2AR and competing G protein subtypes. In the healthy heart, the β2 adrenergic receptor (β2AR) signals through Gs and Gi proteins but the mechanism underlying G protein selectivity is not fully understood. Here, the authors show that membrane charge and intracellular cations modulate the β2AR−Gi3 interaction.
Collapse
|
13
|
Zhang R, Kang X, Wang Y, Wang F, Yu P, Shen J, Fu L. Effects of carvedilol on ventricular remodeling and the expression of β3-adrenergic receptor in a diabetic rat model subjected myocardial infarction. Int J Cardiol 2016; 222:178-184. [PMID: 27497092 DOI: 10.1016/j.ijcard.2016.07.188] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 07/28/2016] [Indexed: 10/21/2022]
Abstract
BACKGROUND This study was to assess effects of carvedilol on ventricular remodeling and expression of β3-adrenergic receptor (β3-AR) and Gi protein in a rat model of diabetes subjected to myocardial infarction (MI). METHODS Rat model of type II diabetes was established by injection of streptozotion. MI was then induced by ligating the left anterior descending coronary artery. Rats were then randomly divided into two groups treated with either placebo (PL) or carvedilol (CA - 10mg·kg(-1)·d(-)(1)). Additional controls consisted of sham-operated rats with diabetes (DS) and rats fed a normal diet subjected to myocardial infarction (NM). Echocardiographic and hemodynamic studies were performed to assess the structural and functional changes. β3-AR and Gi mRNA in the myocardium distal from the infarction region were measured, and β3-AR and Gi protein were measured with western blot. RESULTS There were no significant differences in MI size among the three MI groups. In the PL group, LVEDd, LVWI, E/A and CVF were significantly increased, while LVEF and PW% significantly decreased as compared with the DS and NM groups. Compared with the DS group, the expression of β3-AR and Gi mRNA and protein in the PL group was significantly increased, however, in the CA group, β3-AR and Gi mRNA and protein were decreased. CONCLUSIONS The expression of β3-AR and Gi mRNA and protein was increased in diabetic rats subjected to MI as compared with rats subject to either condition alone. Carvedilol treatment prevented many of these deleterious effects.
Collapse
Affiliation(s)
- Ruiying Zhang
- Cardiovascular Department, The First Affliated Hospital of Harbin Medical University, Harbin 150001, China.
| | - Xiaoning Kang
- Cardiovascular Department, The First Affliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Yumei Wang
- Cardiovascular Department, The First Affliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Fei Wang
- Cardiovascular Department, The First Affliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Ping Yu
- Cardiovascular Department, The First Affliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Jingxia Shen
- Cardiovascular Department, The First Affliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Lu Fu
- Cardiovascular Department, The First Affliated Hospital of Harbin Medical University, Harbin 150001, China
| |
Collapse
|
14
|
β-arrestin-biased signaling through the β2-adrenergic receptor promotes cardiomyocyte contraction. Proc Natl Acad Sci U S A 2016; 113:E4107-16. [PMID: 27354517 DOI: 10.1073/pnas.1606267113] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
β-adrenergic receptors (βARs) are critical regulators of acute cardiovascular physiology. In response to elevated catecholamine stimulation during development of congestive heart failure (CHF), chronic activation of Gs-dependent β1AR and Gi-dependent β2AR pathways leads to enhanced cardiomyocyte death, reduced β1AR expression, and decreased inotropic reserve. β-blockers act to block excessive catecholamine stimulation of βARs to decrease cellular apoptotic signaling and normalize β1AR expression and inotropy. Whereas these actions reduce cardiac remodeling and mortality outcomes, the effects are not sustained. Converse to G-protein-dependent signaling, β-arrestin-dependent signaling promotes cardiomyocyte survival. Given that β2AR expression is unaltered in CHF, a β-arrestin-biased agonist that operates through the β2AR represents a potentially useful therapeutic approach. Carvedilol, a currently prescribed nonselective β-blocker, has been classified as a β-arrestin-biased agonist that can inhibit basal signaling from βARs and also stimulate cell survival signaling pathways. To understand the relative contribution of β-arrestin bias to the efficacy of select β-blockers, a specific β-arrestin-biased pepducin for the β2AR, intracellular loop (ICL)1-9, was used to decouple β-arrestin-biased signaling from occupation of the orthosteric ligand-binding pocket. With similar efficacy to carvedilol, ICL1-9 was able to promote β2AR phosphorylation, β-arrestin recruitment, β2AR internalization, and β-arrestin-biased signaling. Interestingly, ICL1-9 was also able to induce β2AR- and β-arrestin-dependent and Ca(2+)-independent contractility in primary adult murine cardiomyocytes, whereas carvedilol had no efficacy. Thus, ICL1-9 is an effective tool to access a pharmacological profile stimulating cardioprotective signaling and inotropic effects through the β2AR and serves as a model for the next generation of cardiovascular drug development.
Collapse
|
15
|
Halls ML, Bathgate RAD, Sutton SW, Dschietzig TB, Summers RJ. International Union of Basic and Clinical Pharmacology. XCV. Recent advances in the understanding of the pharmacology and biological roles of relaxin family peptide receptors 1-4, the receptors for relaxin family peptides. Pharmacol Rev 2015; 67:389-440. [PMID: 25761609 PMCID: PMC4394689 DOI: 10.1124/pr.114.009472] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Relaxin, insulin-like peptide 3 (INSL3), relaxin-3, and INSL5 are the cognate ligands for the relaxin family peptide (RXFP) receptors 1-4, respectively. RXFP1 activates pleiotropic signaling pathways including the signalosome protein complex that facilitates high-sensitivity signaling; coupling to Gα(s), Gα(i), and Gα(o) proteins; interaction with glucocorticoid receptors; and the formation of hetero-oligomers with distinctive pharmacological properties. In addition to relaxin-related ligands, RXFP1 is activated by Clq-tumor necrosis factor-related protein 8 and by small-molecular-weight agonists, such as ML290 [2-isopropoxy-N-(2-(3-(trifluoromethylsulfonyl)phenylcarbamoyl)phenyl)benzamide], that act allosterically. RXFP2 activates only the Gα(s)- and Gα(o)-coupled pathways. Relaxin-3 is primarily a neuropeptide, and its cognate receptor RXFP3 is a target for the treatment of depression, anxiety, and autism. A variety of peptide agonists, antagonists, biased agonists, and an allosteric modulator target RXFP3. Both RXFP3 and the related RXFP4 couple to Gα(i)/Gα(o) proteins. INSL5 has the properties of an incretin; it is secreted from the gut and is orexigenic. The expression of RXFP4 in gut, adipose tissue, and β-islets together with compromised glucose tolerance in INSL5 or RXFP4 knockout mice suggests a metabolic role. This review focuses on the many advances in our understanding of RXFP receptors in the last 5 years, their signal transduction mechanisms, the development of novel compounds that target RXFP1-4, the challenges facing the field, and current prospects for new therapeutics.
Collapse
MESH Headings
- Allosteric Regulation
- Animals
- Cell Membrane/enzymology
- Cell Membrane/metabolism
- Cyclic AMP/physiology
- Humans
- International Agencies
- Ligands
- Models, Molecular
- Pharmacology/trends
- Pharmacology, Clinical/trends
- Protein Isoforms/agonists
- Protein Isoforms/chemistry
- Protein Isoforms/classification
- Protein Isoforms/metabolism
- Receptors, G-Protein-Coupled/agonists
- Receptors, G-Protein-Coupled/chemistry
- Receptors, G-Protein-Coupled/classification
- Receptors, G-Protein-Coupled/metabolism
- Receptors, Peptide/agonists
- Receptors, Peptide/chemistry
- Receptors, Peptide/classification
- Receptors, Peptide/metabolism
- Relaxin/agonists
- Relaxin/analogs & derivatives
- Relaxin/antagonists & inhibitors
- Relaxin/metabolism
- Second Messenger Systems
- Societies, Scientific
- Terminology as Topic
Collapse
Affiliation(s)
- Michelle L Halls
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, Australia (M.L.H., R.J.S.); Neuropeptides Division, Florey Institute of Neuroscience and Mental Health and Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, Victoria, Australia (R.A.D.B.); Neuroscience Drug Discovery, Janssen Research & Development, LLC, San Diego, California (S.W.S.); Immundiagnostik AG, Bensheim, Germany (T.B.D.); and Charité-University Medicine Berlin, Campus Mitte, Medical Clinic for Cardiology and Angiology, Berlin, Germany (T.B.D.)
| | - Ross A D Bathgate
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, Australia (M.L.H., R.J.S.); Neuropeptides Division, Florey Institute of Neuroscience and Mental Health and Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, Victoria, Australia (R.A.D.B.); Neuroscience Drug Discovery, Janssen Research & Development, LLC, San Diego, California (S.W.S.); Immundiagnostik AG, Bensheim, Germany (T.B.D.); and Charité-University Medicine Berlin, Campus Mitte, Medical Clinic for Cardiology and Angiology, Berlin, Germany (T.B.D.)
| | - Steve W Sutton
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, Australia (M.L.H., R.J.S.); Neuropeptides Division, Florey Institute of Neuroscience and Mental Health and Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, Victoria, Australia (R.A.D.B.); Neuroscience Drug Discovery, Janssen Research & Development, LLC, San Diego, California (S.W.S.); Immundiagnostik AG, Bensheim, Germany (T.B.D.); and Charité-University Medicine Berlin, Campus Mitte, Medical Clinic for Cardiology and Angiology, Berlin, Germany (T.B.D.)
| | - Thomas B Dschietzig
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, Australia (M.L.H., R.J.S.); Neuropeptides Division, Florey Institute of Neuroscience and Mental Health and Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, Victoria, Australia (R.A.D.B.); Neuroscience Drug Discovery, Janssen Research & Development, LLC, San Diego, California (S.W.S.); Immundiagnostik AG, Bensheim, Germany (T.B.D.); and Charité-University Medicine Berlin, Campus Mitte, Medical Clinic for Cardiology and Angiology, Berlin, Germany (T.B.D.)
| | - Roger J Summers
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, Australia (M.L.H., R.J.S.); Neuropeptides Division, Florey Institute of Neuroscience and Mental Health and Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, Victoria, Australia (R.A.D.B.); Neuroscience Drug Discovery, Janssen Research & Development, LLC, San Diego, California (S.W.S.); Immundiagnostik AG, Bensheim, Germany (T.B.D.); and Charité-University Medicine Berlin, Campus Mitte, Medical Clinic for Cardiology and Angiology, Berlin, Germany (T.B.D.)
| |
Collapse
|
16
|
Woo AYH, Song Y, Xiao RP, Zhu W. Biased β2-adrenoceptor signalling in heart failure: pathophysiology and drug discovery. Br J Pharmacol 2014; 172:5444-56. [PMID: 25298054 DOI: 10.1111/bph.12965] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Revised: 08/27/2014] [Accepted: 09/28/2014] [Indexed: 12/27/2022] Open
Abstract
The body is constantly faced with a dynamic requirement for blood flow. The heart is able to respond to these changing needs by adjusting cardiac output based on cues emitted by circulating catecholamine levels. Cardiac β-adrenoceptors transduce the signal produced by catecholamine stimulation via Gs proteins to their downstream effectors to increase heart contractility. During heart failure, cardiac output is insufficient to meet the needs of the body; catecholamine levels are high and β-adrenoceptors become hyperstimulated. The hyperstimulated β1-adrenoceptors induce a cardiotoxic effect, which could be counteracted by the cardioprotective effect of β2-adrenoceptor-mediated Gi signalling. However, β2-adrenoceptor-Gi signalling negates the stimulatory effect of the Gs signalling on cardiomyocyte contraction and further exacerbates cardiodepression. Here, further to the localization of β1- and β2-adrenoceptors and β2-adrenoceptor-mediated β-arrestin signalling in cardiomyocytes, we discuss features of the dysregulation of β-adrenoceptor subtype signalling in the failing heart, and conclude that Gi-biased β2-adrenoceptor signalling is a pathogenic pathway in heart failure that plays a crucial role in cardiac remodelling. In contrast, β2-adrenoceptor-Gs signalling increases cardiomyocyte contractility without causing cardiotoxicity. Finally, we discuss a novel therapeutic approach for heart failure using a Gs-biased β2-adrenoceptor agonist and a β1-adrenoceptor antagonist in combination. This combination treatment normalizes the β-adrenoceptor subtype signalling in the failing heart and produces therapeutic effects that outperform traditional heart failure therapies in animal models. The present review illustrates how the concept of biased signalling can be applied to increase our understanding of the pathophysiology of diseases and in the development of novel therapies.
Collapse
Affiliation(s)
- Anthony Yiu-Ho Woo
- Institute of Molecular Medicine, Centre for Life Sciences, Peking University, Beijing, China.,Key Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Ying Song
- Institute of Molecular Medicine, Centre for Life Sciences, Peking University, Beijing, China
| | - Rui-Ping Xiao
- Institute of Molecular Medicine, Centre for Life Sciences, Peking University, Beijing, China.,Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, China
| | - Weizhong Zhu
- Department of Pharmacology, Nantong University School of Pharmacy, Nantong, China
| |
Collapse
|
17
|
Baker AJ. Adrenergic signaling in heart failure: a balance of toxic and protective effects. Pflugers Arch 2014; 466:1139-50. [PMID: 24623099 DOI: 10.1007/s00424-014-1491-5] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Revised: 02/24/2014] [Accepted: 02/26/2014] [Indexed: 10/25/2022]
Abstract
Heart failure with reduced ejection fraction involves activation of the sympathetic nervous system and chronic hyperactivation of the sympatho-adrenergic receptors (ARs) β-ARs and α1-ARs, which are thought to be cardiotoxic and worsen pathological remodeling and function. Concurrently, the failing heart manifests significant decreases in sympathetic nerve terminal density, decreased cardiac norepinephrine levels, and marked downregulation of β-AR abundance and signaling. Thus, a state of both feast and famine coexist with respect to the adrenergic state in heart failure. For the failing heart, the hyperadrenergic state is toxic. However, the role of hypoadrenergic mechanisms in the pathophysiology of heart failure is less clear. Cardiotoxic effects are known to arise from the β1-AR subtype, and use of β-AR blockers is a cornerstone of current heart failure therapy. However, cardioprotective effects arise from the β2-AR subtype that counteract hyperactive β1-AR signaling, but unfortunately, β2-AR cardioprotective signaling in heart failure is inhibited by β-AR blocker therapy. In contrast to current dogma, recent research shows β1-AR signaling can also be cardioprotective. Moreover, for some forms of heart failure, β2-AR signaling is cardiotoxic. Thus for both β-AR subtypes, there is a balance between cardiotoxic versus cardioprotective effects. In heart failure, stimulation of α1-ARs is widely thought to be cardiotoxic. However, also contrary to current dogma, recent research shows that α1-AR signaling is cardioprotective. Taken together, recent research identifies cardioprotective signaling arising from β1-AR, β2-AR, and α1-ARs. A goal for future therapies will to harness the protective effects of AR signaling while minimizing cardiotoxic effects. The trajectory of heart failure therapy changed radically from the previous and intuitive use of sympathetic agonists, which unfortunately resulted in greater mortality, to the current use of β-AR blockers, which initially seemed counterintuitive. As a cautionary note, if the slow adoption of beta-blocker therapy in heart failure is any guide, then new treatment strategies, especially counterintuitive therapies involving stimulating β-AR and α1-AR signaling, may take considerable time to develop and gain acceptance.
Collapse
Affiliation(s)
- Anthony J Baker
- Veterans Affairs Medical Center, San Francisco and Department of Medicine, University of California, Cardiology Division (111C), 4150 Clement St, San Francisco, CA, 94121, USA,
| |
Collapse
|
18
|
Competition for Gβγ dimers mediates a specific cross-talk between stimulatory and inhibitory G protein α subunits of the adenylyl cyclase in cardiomyocytes. Naunyn Schmiedebergs Arch Pharmacol 2013; 386:459-69. [PMID: 23615874 DOI: 10.1007/s00210-013-0876-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2013] [Accepted: 04/17/2013] [Indexed: 12/20/2022]
Abstract
Heterotrimeric G proteins are key regulators of signaling pathways in mammalian cells. Beyond G protein-coupled receptors, the amount and mutual ratio of specific G protein α, β, and γ subunits determine the G protein signaling. However, little is known about mechanisms that regulate the concentration and composition of G protein subunits at the plasma membrane. Here, we show a novel cross-talk between stimulatory and inhibitory G protein α subunits (Gα) that is mediated by G protein βγ dimers and controls the abundance of specific Gα subunits at the plasma membrane. Firstly, we observed in heart tissue from constitutively Gαi2- and Gαi3-deficient mice that the loss of Gαi2 and Gαi3 was accompanied by a slight increase in the protein content of the nontargeted Gαi isoform. Therefore, we analyzed whether overexpression of selected Gα subunits conversely impairs endogenous G protein α and β subunit levels in cardiomyocytes. Integration of overexpressed Gαi2 subunits into heterotrimeric G proteins was verified by co-immunoprecipitation. Adenoviral expression of increasing amounts of Gαi2 led to a reduction of Gαi3 (up to 90 %) and Gαs (up to 75 %) protein levels. Likewise, increasing amounts of adenovirally expressed Gαs resulted in a linear 75 % decrease in both Gαi2 and Gαi3 protein levels. In contrast, overexpression of either Gαi or Gαs isoform did not influence the amount of Gαo and Gαq, both of which are not involved in the regulation of adenylyl cyclase activity. The mRNA expression of the disappearing endogenous Gα subunits was not affected, indicating a posttranslational mechanism. Interestingly, the amount of endogenous G protein βγ dimers was not altered by any Gα overexpression. However, the increase of Gβγ level by adenoviral expression prevented the loss of endogenous Gαs and Gαi3 in Gαi2 overexpressing cardiomyocytes. Thus, our results provide evidence for a novel mechanism cross-regulating adenylyl cyclase-modulating Gαi isoforms and Gαs proteins. The Gα subunits apparently compete for a limited amount of Gβγ dimers, which are required for G protein heterotrimer formation at the plasma membrane.
Collapse
|
19
|
Bathgate RAD, Halls ML, van der Westhuizen ET, Callander GE, Kocan M, Summers RJ. Relaxin family peptides and their receptors. Physiol Rev 2013; 93:405-80. [PMID: 23303914 DOI: 10.1152/physrev.00001.2012] [Citation(s) in RCA: 399] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
There are seven relaxin family peptides that are all structurally related to insulin. Relaxin has many roles in female and male reproduction, as a neuropeptide in the central nervous system, as a vasodilator and cardiac stimulant in the cardiovascular system, and as an antifibrotic agent. Insulin-like peptide-3 (INSL3) has clearly defined specialist roles in male and female reproduction, relaxin-3 is primarily a neuropeptide involved in stress and metabolic control, and INSL5 is widely distributed particularly in the gastrointestinal tract. Although they are structurally related to insulin, the relaxin family peptides produce their physiological effects by activating a group of four G protein-coupled receptors (GPCRs), relaxin family peptide receptors 1-4 (RXFP1-4). Relaxin and INSL3 are the cognate ligands for RXFP1 and RXFP2, respectively, that are leucine-rich repeat containing GPCRs. RXFP1 activates a wide spectrum of signaling pathways to generate second messengers that include cAMP and nitric oxide, whereas RXFP2 activates a subset of these pathways. Relaxin-3 and INSL5 are the cognate ligands for RXFP3 and RXFP4 that are closely related to small peptide receptors that when activated inhibit cAMP production and activate MAP kinases. Although there are still many unanswered questions regarding the mode of action of relaxin family peptides, it is clear that they have important physiological roles that could be exploited for therapeutic benefit.
Collapse
Affiliation(s)
- R A D Bathgate
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences & Department of Pharmacology, Monash University, Victoria, Australia
| | | | | | | | | | | |
Collapse
|
20
|
Hussain RI, Aronsen JM, Afzal F, Sjaastad I, Osnes JB, Skomedal T, Levy FO, Krobert KA. The functional activity of inhibitory G protein (G(i)) is not increased in failing heart ventricle. J Mol Cell Cardiol 2012; 56:129-38. [PMID: 23220156 DOI: 10.1016/j.yjmcc.2012.11.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2012] [Revised: 11/22/2012] [Accepted: 11/24/2012] [Indexed: 11/16/2022]
Abstract
Beta-adrenergic receptor (βAR) inotropic effects are attenuated and muscarinic receptor-mediated inhibition thereof is enhanced in heart failure. We investigated if increased G(i) activity contributes to attenuated βAR-inotropic effects and potentiates muscarinic accentuated antagonism in failing rat ventricle. Contractility was measured in ventricular strips and adenylyl cyclase (AC) activity in ventricular membranes from rats with post-infarction heart failure (HF) or Sham-operated controls (Sham). The maximal βAR-mediated inotropic effect of isoproterenol was reduced by ~70% and basal, βAR- & forskolin-stimulated AC activity was significantly lower in HF vs. Sham. Carbachol-evoked antagonism of the βAR-mediated inotropic response was complete only in HF despite a ~40% reduction in the ability of carbachol to inhibit βAR-stimulated AC. However, neither the relative efficacy (contractility decreased by ~46%) nor the potency of carbachol to inhibit the βAR inotropic response differed between Sham and HF ventricle. Pertussis toxin (PTX) inactivation of G(i) did not increase the maximal βAR inotropic effect or the attenuated basal, βAR- & forskolin-stimulated AC activity in HF, but increased the potency of isoproterenol only in Sham (~0.5 log unit). In HF ventricle pretreated with PTX, simultaneous inhibition of phosphodiesterases 3,4 (PDE3,4) alone produced a larger inotropic response than isoproterenol in ventricle untreated with PTX (84% and 48% above basal respectively). In the absence of PTX, PDE3,4 inhibition evoked negligible inotropic effects in HF. These data are not consistent with the hypothesis that increased G(i) activity contributes to the reduced βAR-mediated inotropic response and AC activity in failing ventricle. The data, however, support the hypothesis that G(i), through chronic receptor independent inhibition of AC, together with PDE3,4 activity, is necessary to maintain a low basal level of contractility.
Collapse
Affiliation(s)
- R I Hussain
- Department of Pharmacology, Faculty of Medicine, University of Oslo, Oslo, Norway
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Halls ML. Constitutive formation of an RXFP1-signalosome: a novel paradigm in GPCR function and regulation. Br J Pharmacol 2012; 165:1644-1658. [PMID: 21557732 DOI: 10.1111/j.1476-5381.2011.01470.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The classical second messenger cAMP is important in diverse physiological processes, where its spatial and temporal compartmentalization allows precise control over multiple cellular events. Within this context, G-protein-coupled receptors (GPCRs) govern specialized pools of cAMP, which are functionally specific for the unique cellular effects attributed to a particular system. The relaxin receptor, RXFP1, is a GPCR that exerts pleiotropic physiological effects including a potent anti-fibrotic response, increased cancer metastases, and has efficacy as a vasodilator in heart failure. On a cellular level, relaxin stimulation of RXFP1 results in the activation of multiple G-protein pathways affecting cAMP accumulation. Specificity and diversity in the cAMP signal generated by RXFP1 is controlled by differential G-protein coupling dependent upon the background of cellular expression, and cAMP compartmentalization. Further complexity in cAMP signalling results from the constitutive assembly of an RXFP1-signalosome, which specifically responds to low concentrations of relaxin, and activates a distinct cAMP pathway. The RXFP1-signalosome is a higher-order protein complex that facilitates receptor sensitivity to attomolar concentration of peptide, exhibits constitutive activity and dual coupling to G-proteins and β-arrestins and reveals a concentration-biased agonism mediated by relaxin. The specific and directed formation of GPCR-centered signalosomes allows an even greater spatial and temporal control of cAMP, thus rationalizing the considerable physiological scope of this ubiquitous second messenger.
Collapse
Affiliation(s)
- Michelle L Halls
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| |
Collapse
|
22
|
Abstract
β-adrenergic receptor (βAR) stimulation by the sympathetic nervous system or circulating catecholamines is broadly involved in peripheral blood circulation, metabolic regulation, muscle contraction, and central neural activities. In the heart, acute βAR stimulation serves as the most powerful means to regulate cardiac output in response to a fight-or-flight situation, whereas chronic βAR stimulation plays an important role in physiological and pathological cardiac remodeling.There are three βAR subtypes, β(1)AR, β(2)AR and β(3)AR, in cardiac myocytes. Over the past two decades, we systematically investigated the molecular and cellular mechanisms underlying the different even opposite functional roles of β(1)AR and β(2)AR subtypes in regulating cardiac structure and function, with keen interest in the development of novel therapies based on our discoveries. We have made three major discoveries, including (1) dual coupling of β(2)AR to G(s) and G(i) proteins in cardiomyocytes, (2) cardioprotection by β(2)AR signaling in improving cardiac function and myocyte viability, and (3) PKA-independent, CaMKII-mediated β(1)AR apoptotic and maladaptive remodeling signaling in the heart. Based on these discoveries and salutary effects of β(1)AR blockade on patients with heart failure, we envision that activation of β(2)AR in combination with clinically used β(1)AR blockade should provide a safer and more effective therapy for the treatment of heart failure.
Collapse
|
23
|
Sun J, Fu L, Tang X, Han Y, Ma D, Cao J, Kang N, Ji H. Testosterone modulation of cardiac β-adrenergic signals in a rat model of heart failure. Gen Comp Endocrinol 2011; 172:518-25. [PMID: 21549119 DOI: 10.1016/j.ygcen.2011.04.019] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2010] [Revised: 03/16/2011] [Accepted: 04/20/2011] [Indexed: 01/08/2023]
Abstract
In this study, we examined the effects of castration and testosterone replacement on β-adrenoceptor and G protein expression in rats subjected to doxorubicin-induced heart failure. Five groups were included in this report: control, sham-castration with heart failure, castration with heart failure, castration+testosterone replacement with heart failure and castration+testosterone replacement and flutamide with heart failure. At 4 weeks post-treatment, echocardiography, hemodynamics and histopathology were assessed. Castration led to a further deterioration in myocardial performance, apoptosis and fibrosis, while testosterone replacement ameliorated these effects. Data obtained from Western blots revealed that testosterone upregulated the expression of β(2)-adrenoceptor, Gs, Gi(2) and bcl2 levels, downregulated the expression of β(3)-adrenoceptor, Gi(3) and GRK2 levels, and did not modify the expression of β(1)-adrenoceptor levels in the hearts of castrated rats subjected to doxorubicin-induced heart failure. Analyses of serum 17β-estradiol concentrations test confirmed that these effects of testosterone were exerted through the androgen pathway. Thus our findings suggest that testosterone may have beneficial effects for male heart failure patients with androgen deficiency and this protection involves modulation of the cardiac β-adrenergic system.
Collapse
Affiliation(s)
- Junfeng Sun
- Department of Cardiovascular Medicine, First Affiliated Hospital, Harbin Medical University, Harbin, China
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Chakir K, Zhu W, Tsang S, Woo AYH, Yang D, Wang X, Zeng X, Rhee MH, Mende U, Koitabashi N, Takimoto E, Blumer KJ, Lakatta EG, Kass DA, Xiao RP. RGS2 is a primary terminator of β₂-adrenergic receptor-mediated G(i) signaling. J Mol Cell Cardiol 2011; 50:1000-7. [PMID: 21291891 DOI: 10.1016/j.yjmcc.2011.01.015] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2010] [Revised: 01/23/2011] [Accepted: 01/24/2011] [Indexed: 01/08/2023]
Abstract
Two major β-adrenergic receptor (βAR) subtypes, β(1)AR and β(2)AR, are expressed in mammalian heart with β(1)AR coupling to G(s) and β(2)AR dually coupling to G(s) and G(i) proteins. In many types of chronic heart failure, myocardial contractile response to both β(1)AR and β(2)AR stimulation is severely impaired. The dysfunction of βAR signaling in failing hearts is largely attributable to an increase in G(i) signaling, because disruption of the G(i) signaling restores myocardial contractile response to β(1)AR as well as β(2)AR stimulation. However, the mechanism terminating the β(2)AR-G(i) signaling remains elusive, while it has been shown activation of the G(i) signaling is dependent on agonist stimulation and subsequent PKA-mediated phosphorylation of the receptor. Here we demonstrate that regulator of G protein signaling 2 (RGS2) is a primary terminator of the β(2)AR-G(i) signaling. Specifically, prolonged absence of agonist stimulation for 24h impairs the β(2)AR-G(i) signaling, resulting in enhanced β(2)AR- but not β(1)AR-mediated contractile response in cultured adult mouse cardiomyocytes. Increased β(2)AR contractile response is accompanied by a selective upregulation of RGS2 in the absence of alterations in other major cardiac RGS proteins (RGS3-5) or G(s), G(i) or βAR subtypes. Administration of a βAR agonist, isoproterenol (ISO, 1.0 nM), prevents RGS2 upregulation and restores the β(2)AR-G(i) signaling in cultured cells. Furthermore, RGS2 ablation, similar to βAR agonist stimulation, sustains the β(2)AR-G(i) signaling in cultured cells, whereas adenoviral overexpression of RGS2 suppresses agonist-activated β(2)AR-G(i) signaling in cardiomyocytes and HEK293 cells. These findings not only define RGS2 as a novel negative regulator of the β(2)AR-G(i) signaling but also provide a potential novel target for the treatment of chronic heart failure.
Collapse
Affiliation(s)
- Khalid Chakir
- Laboratory of Cardiovascular Science, Gerontology Research Center, National Institute on Aging, NIH, Baltimore, MD 21224, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Adameova A, Abdellatif Y, Dhalla NS. Role of the excessive amounts of circulating catecholamines and glucocorticoids in stress-induced heart disease. Can J Physiol Pharmacol 2010; 87:493-514. [PMID: 19767873 DOI: 10.1139/y09-042] [Citation(s) in RCA: 109] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Various stressful stimuli are known to activate the sympathetic nervous system to release catecholamines and the hypothalamic-pituitary-adrenal axis to release glucocorticoids in the circulation. Although initial actions of both catecholamines and glucocorticoids are beneficial for the function of the cardiovascular system, their delayed effects on the heart are deleterious. Glucocorticoids not only increase plasma levels of catecholamines by inhibiting their extraneuronal uptake, but they have also been shown to induce supersensitivity to catecholamines in the heart by upregulating different components of the betta-adrenoceptor signal transduction system. Low concentrations of catecholamines stimulate the heart by promoting Ca2+ movements, whereas excessive amounts of catecholamines produce cardiac dysfunction by inducing intracellular Ca2+ overload in cardiomyocytes. Several studies have shown, however, that under stressful conditions high concentrations of catecholamines become oxidized to form aminolutins and generate oxyradicals. These oxidation products of catecholamines have been demonstrated to produce coronary spasm, arrhythmias, and cardiac dysfunction by inducing Ca2+-handling abnormalities in both sarcolemmal and sarcoplasmic reticulum, defects in energy production by mitochondria, and myocardial cell damage. In this article we have focused the discussion to highlight the interrelationship between catecholamines and glucocorticoids and to emphasize the role of oxidation products of catecholamines in the development of stress-induced heart disease.
Collapse
Affiliation(s)
- Adriana Adameova
- Institute of Cardiovascular Sciences, St. Boniface General Hospital Research Centre, and Department of Physiology, Faculty of Medicine, University of Manitoba, 351 Tache Avenue, Winnipeg, MB R2H 2A6, Canada
| | | | | |
Collapse
|
26
|
Hamdani N, Paulus WJ, van Heerebeek L, Borbély A, Boontje NM, Zuidwijk MJ, Bronzwaer JGF, Simonides WS, Niessen HWM, Stienen GJM, van der Velden J. Distinct myocardial effects of beta-blocker therapy in heart failure with normal and reduced left ventricular ejection fraction. Eur Heart J 2009; 30:1863-72. [PMID: 19487234 DOI: 10.1093/eurheartj/ehp189] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
AIMS Left ventricular (LV) myocardial structure and function differ in heart failure (HF) with normal (N) and reduced (R) LV ejection fraction (EF). This difference could underlie an unequal outcome of trials with beta-blockers in heart failure with normal LVEF (HFNEF) and heart failure with reduced LVEF (HFREF) with mixed results observed in HFNEF and positive results in HFREF. To investigate whether beta-blockers have distinct myocardial effects in HFNEF and HFREF, myocardial structure, cardiomyocyte function, and myocardial protein composition were compared in HFNEF and HFREF patients without or with beta-blockers. METHODS AND RESULTS Patients, free of coronary artery disease, were divided into beta-(HFNEF) (n = 16), beta+(HFNEF) (n = 16), beta-(HFREF) (n = 17), and beta+(HFREF) (n = 22) groups. Using LV endomyocardial biopsies, we assessed collagen volume fraction (CVF) and cardiomyocyte diameter (MyD) by histomorphometry, phosphorylation of myofilamentary proteins by ProQ-Diamond phosphostained 1D-gels, and expression of beta-adrenergic signalling and calcium handling proteins by western immunoblotting. Cardiomyocytes were also isolated from the biopsies to measure active force (F(active)), resting force (F(passive)), and calcium sensitivity (pCa(50)). Myocardial effects of beta-blocker therapy were either shared by HFNEF and HFREF, unique to HFNEF or unique to HFREF. Higher F(active), higher pCa(50), lower phosphorylation of troponin I and myosin-binding protein C, and lower beta(2) adrenergic receptor expression were shared. Higher F(passive), lower CVF, lower MyD, and lower expression of stimulatory G protein were unique to HFNEF and lower expression of inhibitory G protein was unique to HFREF. CONCLUSION Myocardial effects unique to either HFNEF or HFREF could contribute to the dissimilar outcome of beta-blocker therapy in both HF phenotypes.
Collapse
Affiliation(s)
- Nazha Hamdani
- Laboratory for Physiology, Institute for Cardiovascular Research, VU University Medical Center, van der Boechorststraat 7, 1081 BT Amsterdam, the Netherlands
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Woo AYH, Wang TB, Zeng X, Zhu W, Abernethy DR, Wainer IW, Xiao RP. Stereochemistry of an agonist determines coupling preference of beta2-adrenoceptor to different G proteins in cardiomyocytes. Mol Pharmacol 2009; 75:158-65. [PMID: 18838481 PMCID: PMC2654765 DOI: 10.1124/mol.108.051078] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2008] [Accepted: 09/18/2008] [Indexed: 12/13/2022] Open
Abstract
A fundamental question regarding receptor-G protein interaction is whether different agonists can lead a receptor to different intracellular signaling pathways. Our previous studies have demonstrated that although most beta(2)-adrenoceptor agonists activate both G(s) and G(i) proteins, fenoterol, a full agonist of beta(2)-adrenoceptor, selectively activates G(s) protein. Fenoterol contains two chiral centers and may exist as four stereoisomers. We have synthesized a series of stereoisomers of fenoterol and its derivatives and characterized their receptor binding and pharmacological properties. We tested the hypothesis that the stereochemistry of an agonist determines selectivity of receptor coupling to different G protein(s). We found that the R,R isomers of fenoterol and methoxyfenoterol exhibited more potent effects to increase cardiomyocyte contraction than their S,R isomers. It is noteworthy that although (R,R)-fenoterol and (R,R)-methoxyfenoterol preferentially activate G(s) signaling, their S,R isomers were able to activate both G(s) and G(i) proteins as evidenced by the robust pertussis toxin sensitivities of their effects on cardiomyocyte contraction and on phosphorylation of extracellular signal-regulated kinase 1/2. The differential G protein selectivities of the fenoterol stereoisomers were further confirmed by photoaffinity labeling studies on G(s),G(i2), and G(i3) proteins. The inefficient G(i) signaling with the R,R isomers is not caused by the inability of the R,R isomers to trigger the protein kinase A (PKA)-mediated phosphorylation of the beta(2)-adrenoceptor, because the R,R isomers also markedly increased phosphorylation of the receptor at serine 262 by PKA. We conclude that in addition to receptor subtype and phosphorylation status, the stereochemistry of a given agonist plays an important role in determining receptor-G protein selectivity and downstream signaling events.
Collapse
Affiliation(s)
- Anthony Yiu-Ho Woo
- Laboratory of Cardiovascular Science, Gerontology Research Center, NIA, National Institutes of Health, 5600 Nathan Shock Drive, Baltimore, MD 21224, USA.
| | | | | | | | | | | | | |
Collapse
|
28
|
|
29
|
Osadchii OE. Myocardial phosphodiesterases and regulation of cardiac contractility in health and cardiac disease. Cardiovasc Drugs Ther 2007; 21:171-94. [PMID: 17373584 DOI: 10.1007/s10557-007-6014-6] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2006] [Accepted: 02/21/2007] [Indexed: 01/14/2023]
Abstract
Phosphodiesterase (PDE) inhibitors are potent cardiotonic agents used for parenteral inotropic support in heart failure. Contractile effects of these agents are mediated through cAMP-protein kinase A-induced stimulation of I (Ca2+) which ultimately results in increased Ca(2+)-induced sarcoplasmic reticulum Ca(2+) release. A number of additional effects such as increases in sarcoplasmic reticulum Ca(2+) stores, stimulation of reverse mode Na(+)-Ca(2+) exchange, direct or cAMP-mediated effects on sarcoplasmic reticulum ryanodine receptor, stimulation of the voltage-sensitive sarcoplasmic reticulum Ca(2+) release mechanism, as well as A(1) adenosine receptor blockade could contribute to positive inotropic responses to PDE inhibitors. Moreover, some PDE inhibitors exhibit Ca(2+) sensitizer properties as they could increase the affinity of troponin C Ca(2+)-binding sites as well as reduce Ca(2+) threshold for thin myofilament sliding and facilitate cross-bridge cycling. Inotropic responses to PDE inhibitors are significantly reduced in cardiac disease, an effect largely attributed to downregulation of cAMP-mediated signalling due to sustained sympathetic activation. Four PDE isoenzymes (PDE1, PDE2, PDE3 and PDE4) are present in myocardial tissue of various mammalian species, of which PDE3 and PDE4 are particularly involved in regulation of cardiac myocyte contraction. PDE cAMP-hydrolysing activity is preserved in compensated cardiac hypertrophy but significantly reduced in animal models of heart failure. However, clinical studies have not revealed any changes in distribution profile as well as kinetic and regulatory properties of myocardial PDEs in failing human hearts. A reduction of PDE inhibitors-induced contractile responses in heart failure has therefore been ascribed to reduced cAMP synthesis due to uncoupling of adenylyl cyclase from beta-adrenoreceptor. In cardiac myocytes, PDEs are targeted to distinct subcellular compartments by scaffolding proteins such as myomegalin, mAKAP and beta-arrestins. Over subcellular microdomains, cAMP hydrolysis by PDE3 and PDE4 allows to control the activity of local pools of protein kinase A and therefore the extent of protein kinase A-mediated phosphorylation of cellular proteins.
Collapse
Affiliation(s)
- Oleg E Osadchii
- Cardiology Group, School of Clinical Sciences, University Clinical Departments, University of Liverpool, The Duncan Building, Liverpool, UK.
| |
Collapse
|
30
|
Abstract
Most modern theories about stress recognize that although stress is not a disease, it may be the trigger for the majority of diseases when allostatic overload has been generated. During stress, the glucocorticoids and catecholamines play a key role in the regulation of physiological parameters and homeostasis during stress. In the heart, positive chronotropic, inotropic, and lusitropic responses to catecholamines are mediated by various subtypes of adrenergic receptors (beta-ARs), mainly beta1- and beta2-adrenergic receptors. beta-ARs also control cardiomyocyte growth and death, thus contributing to cardiac remodelling. The structural basis of each beta-AR subtype, as well as their signalling pathways, and adaptive responses to stress are discussed. The participation of beta3- and putative beta4-ARs in the control of cardiac function is also discussed, with emphasis on low affinity beta-AR isoforms and the role they play in the response to the catecholamines under stress. The changes in beta-AR signalling under pathogenic conditions as well as under stress are reviewed.
Collapse
Affiliation(s)
- Iraídes N Santos
- Institute of Biology, State University of Campinas (UNICAMP), Department of Physiology and Biophysics, Campinas, SP, Brazil
| | | |
Collapse
|
31
|
Zheng M, Zhu W, Han Q, Xiao RP. Emerging concepts and therapeutic implications of β-adrenergic receptor subtype signaling. Pharmacol Ther 2005; 108:257-68. [PMID: 15979723 DOI: 10.1016/j.pharmthera.2005.04.006] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2005] [Accepted: 04/20/2005] [Indexed: 12/31/2022]
Abstract
The stimulation of beta-adrenergic receptor (betaAR) plays a pivotal role in regulating myocardial function and morphology in the normal and failing heart. Three genetically and pharmacologically distinct betaAR subtypes, beta1AR, beta2AR, and beta3AR, are identified in various types of cells. While both beta1AR and beta2AR, the predominant betaAR subtypes expressed in the heart of many mammalian species including human, are coupled to the Gs-adenylyl cyclase-cAMP-PKA pathway, beta2AR dually activates pertussis toxin-sensitive Gi proteins. During acute stimulation, beta2AR-Gi coupling partially inhibits the Gs-mediated positive contractile and relaxant effects via a Gi-Gbetagamma-phosphoinositide 3-kinase (PI3K)-dependent mechanism in adult rodent cardiomyocytes. More importantly, persistent beta1AR stimulation evokes a multitude of cardiac toxic effects, including myocyte apoptosis and hypertrophy, via a calmodulin-dependent protein kinase II (CaMKII)-, rather than cAMP-PKA-, dependent mechanism in rodent heart in vivo and cultured cardiomyocytes. In contrast, persistent beta2AR activation protects myocardium by a cell survival pathway involving Gi, PI3K, and Akt. In this review, we attempt to highlight the distinct functionalities and signaling mechanisms of these betaAR subtypes and discuss how these subtype-specific properties of betaARs might affect the pathogenesis of congestive heart failure (CHF) and the therapeutic effectiveness of certain beta-blockers in the treatment of congestive heart failure.
Collapse
Affiliation(s)
- Ming Zheng
- Institute of Cardiovascular Sciences, Peking University, Beijing 100083, People's Republic of China
| | | | | | | |
Collapse
|
32
|
Xiao RP, Zhu W, Zheng M, Chakir K, Bond R, Lakatta EG, Cheng H. Subtype-specific beta-adrenoceptor signaling pathways in the heart and their potential clinical implications. Trends Pharmacol Sci 2004; 25:358-65. [PMID: 15219978 DOI: 10.1016/j.tips.2004.05.007] [Citation(s) in RCA: 113] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Rui-Ping Xiao
- Laboratory of Cardiovascular Science, National Institute on Aging/NIH, Baltimore, MD 21224, USA.
| | | | | | | | | | | | | |
Collapse
|
33
|
Xiao RP, Zhang SJ, Chakir K, Avdonin P, Zhu W, Bond RA, Balke CW, Lakatta EG, Cheng H. Enhanced G(i) signaling selectively negates beta2-adrenergic receptor (AR)--but not beta1-AR-mediated positive inotropic effect in myocytes from failing rat hearts. Circulation 2003; 108:1633-9. [PMID: 12975249 DOI: 10.1161/01.cir.0000087595.17277.73] [Citation(s) in RCA: 98] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND Myocardial contractile response to beta1- and beta2-adrenergic receptor (AR) stimulation is severely impaired in chronic heart failure, in which G(i) signaling and the ratio of beta2/beta1 are often increased. Because beta2-AR but not beta1-AR couples to G(s) and G(i) with the G(i) coupling negating the G(s)-mediated contractile response, we determined whether the heart failure-associated augmentation of G(i) signaling contributes differentially to the defects of these beta-AR subtypes and, if so, whether inhibition of G(i) or selective activation of beta2-AR/G(s) by ligands restores beta2-AR contractile response in the failing heart. METHODS AND RESULTS Cardiomyocytes were isolated from 18- to 24-month-old failing spontaneously hypertensive (SHR) or age-matched Wistar-Kyoto (WKY) rat hearts. In SHR cardiomyocytes, either beta-AR subtype-mediated inotropic effect was markedly diminished, whereas G(i) proteins and the beta2/beta1 ratio were increased. Disruption of G(i) signaling by pertussis toxin (PTX) enabled beta2- but not beta1-AR to induce a full positive inotropic response in SHR myocytes. Furthermore, screening of a panel of beta2-AR ligands revealed that the contractile response mediated by most beta2-AR agonists, including zinterol, salbutamol, and procaterol, was potentiated by PTX, indicating concurrent G(s) and G(i) activation. In contrast, fenoterol, another beta2-AR agonist, induced a full positive inotropic effect in SHR myocytes even in the absence of PTX. CONCLUSIONS We conclude that enhanced G(i) signaling is selectively involved in the dysfunction of beta2- but not beta1-AR in failing SHR hearts and that disruption of G(i) signaling by PTX or selective activation of beta2-AR/G(s) signaling by fenoterol restores the blunted beta2-AR contractile response in the failing heart.
Collapse
MESH Headings
- Adrenergic beta-1 Receptor Antagonists
- Adrenergic beta-2 Receptor Agonists
- Adrenergic beta-Agonists/pharmacology
- Animals
- Cardiac Output, Low/etiology
- Cardiac Output, Low/metabolism
- Cardiac Output, Low/physiopathology
- Cardiotonic Agents/pharmacology
- Cells, Cultured
- Chronic Disease
- Fenoterol/pharmacology
- GTP-Binding Protein alpha Subunits, Gi-Go/antagonists & inhibitors
- GTP-Binding Protein alpha Subunits, Gi-Go/metabolism
- Heterotrimeric GTP-Binding Proteins/analysis
- Ligands
- Myocardial Contraction/drug effects
- Myocardium/chemistry
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/physiology
- Pertussis Toxin/pharmacology
- Rats
- Rats, Inbred SHR
- Rats, Inbred WKY
- Receptors, Adrenergic, beta-1/analysis
- Receptors, Adrenergic, beta-1/metabolism
- Receptors, Adrenergic, beta-2/analysis
- Receptors, Adrenergic, beta-2/metabolism
- Signal Transduction/drug effects
Collapse
Affiliation(s)
- Rui-Ping Xiao
- the Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, Md 21224, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Moniotte S, Balligand JL. Potential use of beta(3)-adrenoceptor antagonists in heart failure therapy. CARDIOVASCULAR DRUG REVIEWS 2002; 20:19-26. [PMID: 12070531 DOI: 10.1111/j.1527-3466.2002.tb00079.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Recently, a functional, negatively inotropic, beta(3)-adrenoceptor was characterized in the human heart. Several studies now suggest that this receptor might play an important role in the pathophysiology of heart failure, by counterbalancing the effects of a beta(1)- and beta(2)-stimulation. Therefore, this review summarizes the rationale and effects of beta-adrenergic blockade in chronic heart failure and specifically addresses the question of the potential use of beta(3)-adrenoceptor antagonists in the treatment of heart failure and other pathophysiological conditions associated with a decreased cardiac contractility.
Collapse
Affiliation(s)
- Stéphane Moniotte
- Department of Pediatrics, University of Louvain Medical School, Brussels, Belgium.
| | | |
Collapse
|
35
|
Eckhart AD, Fentzke RC, Lepore J, Lang R, Lin H, Lefkowitz RJ, Koch WJ, Leiden JM. Inhibition of betaARK1 restores impaired biochemical beta-adrenergic receptor responsiveness but does not rescue CREB(A133) induced cardiomyopathy. J Mol Cell Cardiol 2002; 34:669-77. [PMID: 12054854 DOI: 10.1006/jmcc.2002.2007] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The myocardial beta-adrenergic receptor (betaAR) system plays a key role in dysfunctional signaling and physiology of the failing heart. Recently we described a murine model of dilated cardiomyopathy (DCM) produced by cardiac-specific expression of a dominant negative form of the CREB transcription factor (CREB(A133) mice). CREB(A133) mice display abnormalities within the betaAR signaling system including loss of inotropic reserve. Rapid desensitization of betaARs is mediated by the betaAR kinase (betaARK1), which is upregulated during heart failure. Inhibition of betaARK1 activity in the heart via expression of a peptide inhibitor (betaARKct) has been shown to enhance myocardial function and to "rescue" several animal models of heart failure. To determine the role of betaAR dysfunction in the progression of DCM in the CREB(A133) mice, we interbred them with mice expressing the betaARKct. Concurrent expression of the betaARKct peptide and CREB(A133) in mouse hearts resulted in the normalization of elevated betaARK1 levels. This biochemical change resulted in partial restoration of isoproterenol-stimulated adenylate cyclase activity as well as improvement in fractional shortening in response to betaAR stimulation. Interestingly, the progression of DCM and premature mortality was not altered. Therefore, the pathogenesis of DCM in CREB(A133) mice does not appear to involve abnormal betaAR signaling as a key element in its pathological progression and accordingly, the restoration of betaAR signaling is not sufficient to prevent the development and progression of all forms of heart failure.
Collapse
Affiliation(s)
- Andrea D Eckhart
- Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Armoundas AA, Wu R, Juang G, Marbán E, Tomaselli GF. Electrical and structural remodeling of the failing ventricle. Pharmacol Ther 2001; 92:213-30. [PMID: 11916538 DOI: 10.1016/s0163-7258(01)00171-1] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Heart failure (HF) is a complex disease that presents a major public health challenge to Western society. The prevalence of HF increases with age in the elderly population, and the societal disease burden will increase with prolongation of life expectancy. HF is initially characterized by an adaptive increase of neurohumoral activation to compensate for reduction of cardiac output. This leads to a combination of neurohumoral activation and mechanical stress in the failing heart that trigger a cascade of maladaptive electrical and structural events that impair both the systolic and diastolic function of the heart.
Collapse
Affiliation(s)
- A A Armoundas
- Division of Molecular Cardiobiology, Johns Hopkins University, Ross 844, 720 Rutland Avenue, Baltimore, MD 21205, USA.
| | | | | | | | | |
Collapse
|
37
|
Xiao RP. Beta-adrenergic signaling in the heart: dual coupling of the beta2-adrenergic receptor to G(s) and G(i) proteins. SCIENCE'S STKE : SIGNAL TRANSDUCTION KNOWLEDGE ENVIRONMENT 2001; 2001:re15. [PMID: 11604549 DOI: 10.1126/stke.2001.104.re15] [Citation(s) in RCA: 135] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Beta-adrenergic receptor (AR) subtypes are archetypical members of the G protein-coupled receptor (GPCR) superfamily. Whereas both beta1AR and beta2AR stimulate the classic G(s)-adenylyl cyclase-3',5'-adenosine monophosphate (cAMP)-protein kinase A (PKA) signaling cascade, beta2AR couples to both G(s) and G(i) proteins, activating bifurcated signaling pathways. In the heart, dual coupling of the beta2AR to G(s) and G(i) results in compartmentalization of the G(s)-stimulated cAMP signal, thus selectively affecting plasma membrane effectors (such as L-type Ca(2+) channels) and bypassing cytoplasmic target proteins (such as phospholamban and myofilament contractile proteins). More important, the beta2AR-to-G(i) branch delivers a powerful cell survival signal that counters apoptosis induced by the concurrent G(s)-mediated signal or by a wide range of assaulting factors. This survival pathway sequentially involves G(i), G(beta)(gamma), phosphoinositide 3-kinase, and Akt. Furthermore, cardiac-specific transgenic overexpression of betaAR subtypes in mice results in distinctly different phenotypes in terms of the likelihood of cardiac hypertrophy and heart failure. These findings indicate that stimulation of the two betaAR subtypes activates overlapping, but different, sets of signal transduction mechanisms, and fulfills distinct or even opposing physiological and pathophysiological roles. Because of these differences, selective activation of cardiac beta2AR may provide catecholamine-dependent inotropic support without cardiotoxic consequences, which might have beneficial effects in the failing heart.
Collapse
Affiliation(s)
- R P Xiao
- Laboratory of Cardiovascular Science, Gerontology Research Center, National Institute on Aging, Baltimore, MD 21224, USA.
| |
Collapse
|
38
|
Xiao RP. -Adrenergic Signaling in the Heart: Dual Coupling of the 2-Adrenergic Receptor to Gs and Gi Proteins. Sci Signal 2001. [DOI: 10.1126/scisignal.1042001re15] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
|
39
|
Moniotte S, Kobzik L, Feron O, Trochu JN, Gauthier C, Balligand JL. Upregulation of beta(3)-adrenoceptors and altered contractile response to inotropic amines in human failing myocardium. Circulation 2001; 103:1649-55. [PMID: 11273992 DOI: 10.1161/01.cir.103.12.1649] [Citation(s) in RCA: 231] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND Contrary to beta(1)- and beta(2)-adrenoceptors, beta(3)-adrenoceptors mediate a negative inotropic effect in human ventricular muscle. To assess their functional role in heart failure, our purpose was to compare the expression and contractile effect of beta(3)-adrenoceptors in nonfailing and failing human hearts. METHODS AND RESULTS We analyzed left ventricular samples from 29 failing (16 ischemic and 13 dilated cardiomyopathic) hearts (ejection fraction 18.6+/-2%) and 25 nonfailing (including 12 innervated) explanted hearts (ejection fraction 64.2+/-3%). beta(3)-Adrenoceptor proteins were identified by immunohistochemistry in ventricular cardiomyocytes from nonfailing and failing hearts. Contrary to beta(1)-adrenoceptor mRNA, Western blot analysis of beta(3)-adrenoceptor proteins showed a 2- to 3-fold increase in failing compared with nonfailing hearts. A similar increase was observed for Galpha(i-2) proteins that couple beta(3)-adrenoceptors to their negative inotropic effect. Contractile tension was measured in electrically stimulated myocardial samples ex vivo. In failing hearts, the positive inotropic effect of the nonspecific amine isoprenaline was reduced by 75% compared with that observed in nonfailing hearts. By contrast, the negative inotropic effect of beta(3)-preferential agonists was only mildly reduced. CONCLUSIONS Opposite changes occur in beta(1)- and beta(3)-adrenoceptor abundance in the failing left ventricle, with an imbalance between their inotropic influences that may underlie the functional degradation of the human failing heart.
Collapse
Affiliation(s)
- S Moniotte
- Department of Medicine, Unit of Pharmacology and Therapeutics, University of Louvain Medical School, Brussels, Belgium
| | | | | | | | | | | |
Collapse
|
40
|
Abstract
The acute contractile function of the heart is controlled by the effects of released nonepinephrine (NE) on cardiac adrenergic receptors. NE can also act in a more chronic fashion to induce cardiomyocyte growth, characterized by cell enlargement (hypertrophy), increased protein synthesis, alterations in gene expression and addition of sarcomeres. These responses enhance cardiomyocyte contractile function and thus allow the heart to compensate for increased stress. The hypertrophic effects of NE are mediated through Gq-coupled alpha(1)-adrenergic receptors and are mimicked by the actions of other neurohormones (endothelin, prostaglandin F(2alpha) angiotensin II) that also act on Gq-coupled receptors. Activation of phospholipase C by Gq is necessary for these responses, and protein kinase C and MAP kinases have also been implicated. Gq stimulated cardiac hypertrophy is also evident in transgenic mouse models. In contrast, stimulation of G(s)-coupled beta-adrenergic receptors or G(i)-coupled receptors do not directly effect cardiomyocyte hypertrophy. Apoptosis is also induced by G-protein-coupled receptor stimulation in cardiomyocytes. Sustained or excessive activation of either Gq- or Gs-signaling pathways results in apoptotic loss of cardiomyocytes both in vitro and in vivo. Apoptosis is associated with decreased ventricular function in the failing heart. Cardiomyocytes provide an ideal model system for understanding the basis for G-protein mediated hypertrophy and apoptosis, and the mechanisms responsible for the transition from compensatory to deleterious levels of signaling. This information may prove critical for designing interventions that prevent the pathophysiological consequences of heart failure.
Collapse
Affiliation(s)
- J W Adams
- University of California, San Diego, Department of Pharmacology, 9500 Gilman Drive, 0636, La Jolla, CA 92093-0636, USA
| | | |
Collapse
|
41
|
Toufektsian MC, Boucher FR, Tanguy S, Morel S, de Leiris JG. Cardiac toxicity of singlet oxygen: implication in reperfusion injury. Antioxid Redox Signal 2001; 3:63-9. [PMID: 11291599 DOI: 10.1089/152308601750100506] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Oxygen-derived free radicals (O2.-, H2O2, and .OH) that are produced during postischemic reperfusion are currently suspected to be involved in the pathogenesis of tissue injury. Another reactive oxygen species, the electronically excited molecular oxygen (1O2), is of increasing interest in the area of experimental research in cardiology. In this review are discussed the main potential sources of singlet oxygen in the organism, particularly in the myocardium, the various cardiovascular cytotoxic effects induced by this reactive oxygen intermediate, and the growing evidence of its involvement in ischemia/reperfusion injury.
Collapse
Affiliation(s)
- M C Toufektsian
- Laboratoire Stress Cardiovasculaires et Pathologies Associées, Université Joseph Fourier, Bâtiment Jean Roget, Domaine de la Merci, Grenoble, France
| | | | | | | | | |
Collapse
|
42
|
Abstract
Understanding of the pathophysiology of chronic systolic heart failure evolved from a purely mechanical model to one in which a cascade of neurohormones and biologically active molecules are thought to be critical in the development, maintenance, and progression of the disease. Two important neurohormonal systems are the sympathetic nervous and renin-angiotensin-aldosterone systems. Initially, increases in norepinephrine concentrations from the sympathetic nervous system and in angiotensin II and aldosterone are beneficial in the short term to maintain cardiac output after an insult to the myocardium. However, long-term exposure to these neurohormones causes alterations of myocytes and interstitial make-up of the heart. These alterations in myocardium lead to progression of heart failure and, eventually, death.
Collapse
Affiliation(s)
- B E Bleske
- University of Michigan College of Pharmacy, University of Michigan Health Systems, Ann Arbor 48109-1065, USA
| |
Collapse
|
43
|
Wang X, Dhalla NS. Modification of beta-adrenoceptor signal transduction pathway by genetic manipulation and heart failure. Mol Cell Biochem 2000; 214:131-55. [PMID: 11195784 DOI: 10.1023/a:1007131925048] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The beta-adrenoceptor (beta-AR) mediated signal transduction pathway in cardiomyocytes is known to involve beta1- and beta2-ARs, stimulatory (Gs) and inhibitory (Gi) guanine nucleotide binding proteins, adenylyl cyclase (AC) and cAMP-dependent protein kinase (PKA). The activation of beta1- and beta2-ARs has been shown to increase heart function by increasing Ca2+ -movements across the sarcolemmal membrane and sarcoplasmic reticulum through the stimulation of Gs-proteins, activation of AC and PKA enzymes and phosphorylation of the target sites. The activation of PKA has also been reported to increase phosphorylation of some myofibrillar proteins (for promoting cardiac relaxation) and nuclear proteins (for cardiac hypertrophy). The activation of beta2-AR has also been shown to affect Gi-proteins, stimulate mitogen activated protein kinase and increase protein synthesis by enhancing gene expression. Beta1- and beta2-ARs as well as AC are considered to be regulated by PKA- and protein kinase C (PKC)-mediated phosphorylations directly; both PKA and PKC also regulate beta-AR indirectly through the involvement of beta-AR kinase (betaARK), beta-arrestins and Gbeta gamma-protein subunits. Genetic manipulation of different components and regulators of beta-AR signal transduction pathway by employing transgenic and knockout mouse models has provided insight into their functional and regulatory characteristics in cardiomyocytes. The genetic studies have also helped in understanding the pathophysiological role of PARK in heart dysfunction and therapeutic role of betaARK inhibitors in the treatment of heart failure. Varying degrees of defects in the beta-AR signal transduction system have been identified in different types of heart failure to explain the attenuated response of the failing heart to sympathetic stimulation or catecholamine infusion. A decrease in beta1-AR density, an increase in the level of G1-proteins and overexpression of betaARK are usually associated with heart failure; however, these attenuations have been shown to be dependent upon the type and stage of heart failure as well as region of the heart. Both local and circulating renin-angiotensin systems, sympathetic nervous system and endothelial cell function appears to regulate the status of beta-AR signal transduction pathway in the failing heart. Thus different components and regulators of the beta-AR signal transduction pathway appears to represent important targets for the development of therapeutic interventions for the treatment of heart failure.
Collapse
Affiliation(s)
- X Wang
- Institute of Cardiovascular Sciences, Department of Physiology, Faculty of Medicine University of Manitoba, Winnipeg, Canada
| | | |
Collapse
|
44
|
Abstract
beta-Adrenergic-blocking agents underwent extensive research over the past 2 decades and emerged as a proven state-of-art therapy for the failing human heart. Through blockade of chronically elevated cardiac adrenergic stimulation, selective and nonselective agents with vasodilating properties prevent progression of myocardial dysfunction and cardiac remodeling. Most important, beta-adrenergic blockers added to conventional therapy of vasodilators and diuretics significantly increase survival to a 5-year rate similar to that of cardiac transplantation. The agents also significantly reduce hospitalizations, improve quality of life, and are well tolerated in clinical trials. The challenge in treating heart failure is to ensure that every eligible patient receives these life-saving drugs.
Collapse
Affiliation(s)
- M A Munger
- Utah Affiliated Hospitals Heart Failure Prevention and Treatment Program, University of Utah, Salt Lake City 84112, USA
| | | |
Collapse
|
45
|
Kilts JD, Gerhardt MA, Richardson MD, Sreeram G, Mackensen GB, Grocott HP, White WD, Davis RD, Newman MF, Reves JG, Schwinn DA, Kwatra MM. Beta(2)-adrenergic and several other G protein-coupled receptors in human atrial membranes activate both G(s) and G(i). Circ Res 2000; 87:705-9. [PMID: 11029407 DOI: 10.1161/01.res.87.8.705] [Citation(s) in RCA: 106] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Cardiac G protein-coupled receptors that couple to Galpha(s) and stimulate cAMP formation (eg, beta-adrenergic, histamine, serotonin, and glucagon receptors) play a key role in cardiac inotropy. Recent studies in rodent cardiac myocytes and transfected cells have revealed that one of these receptors, the beta(2)-adrenergic receptor (AR), also couples to the inhibitory G protein Galpha(i) (activation of which inhibits cAMP formation). If beta(2)ARs could be shown to couple to Galpha(i) in the human heart, it would have important ramifications, because levels of Galpha(i) increase with age and in failing human heart. Therefore, we investigated whether beta(2)ARs in the human heart activate Galpha(i). By photoaffinity labeling human atrial membranes with [(32)P]azidoanilido-GTP, followed by immunoprecipitation with antibodies specific for Galpha(i), we found that Galpha(i) is activated by stimulation of beta(2)ARs but not of beta(1)ARs. In addition, we found that other Galpha(s)-coupled receptors also couple to Galpha(i), including histamine, serotonin, and glucagon. When coupling of these receptors to Galpha(i) is disrupted by pertussis toxin, their ability to stimulate adenylyl cyclase is enhanced. These data provide the first evidence that beta(2)AR and many other Galpha(s)-coupled receptors in human atrium also couple to Galpha(i) and that abolishing the coupling of these receptors to Galpha(i) increases the receptor-mediated adenylyl cyclase activity.
Collapse
MESH Headings
- Adenylate Cyclase Toxin
- Adenylyl Cyclases/metabolism
- Adrenergic beta-1 Receptor Antagonists
- Adrenergic beta-2 Receptor Antagonists
- Adrenergic beta-Agonists/pharmacology
- Aged
- Atrial Appendage/chemistry
- Atrial Appendage/metabolism
- Cell Membrane/chemistry
- Dobutamine/pharmacology
- Ethanolamines/pharmacology
- GTP-Binding Protein alpha Subunits, Gi-Go/metabolism
- GTP-Binding Protein alpha Subunits, Gs/metabolism
- Humans
- Isoproterenol/pharmacology
- Middle Aged
- Myocardial Contraction/physiology
- Pertussis Toxin
- Photoaffinity Labels
- Precipitin Tests
- Receptors, Adrenergic, beta-1/analysis
- Receptors, Adrenergic, beta-1/metabolism
- Receptors, Adrenergic, beta-2/analysis
- Receptors, Adrenergic, beta-2/metabolism
- Receptors, Cell Surface/analysis
- Receptors, Cell Surface/metabolism
- Receptors, Glucagon/metabolism
- Receptors, Histamine/metabolism
- Receptors, Serotonin/metabolism
- Signal Transduction/drug effects
- Signal Transduction/physiology
- Virulence Factors, Bordetella/pharmacology
Collapse
Affiliation(s)
- J D Kilts
- Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
|
47
|
Abstract
Because of safety issues, components of the beta-adrenergic signaling pathway cannot currently be viewed as attractive targets for human gene therapy. Rather, the balance of evidence supports strategies that will target gene products specifically and directly at diastolic regulation. Augmenting the activity of the SR Ca2+ ATPase by AAV-mediated delivery of the SERCA2a gene, directed by a cardiac-specific promoter with a tightly regulable on-off switch is perhaps the most attractive strategy. PLB and cTnI also are attractive targets but only if molecular techniques can be devised to modulate their activity specifically and conditionally. Such techniques may involve modifying the phosphorylation sites in vitro and replacing wild type proteins in the failing heart with the modified forms, again using regulated AAV vectors for gene delivery.
Collapse
Affiliation(s)
- K A Webster
- Department of Molecular and Cellular Pharmacology, University of Miami Medical Center, Florida, USA.
| | | |
Collapse
|
48
|
Kouchi I, Zolk O, Jockenhövel F, Itter G, Linz W, Cremers B, Böhm M. Increase in G(i alpha) protein accompanies progression of post-infarction remodeling in hypertensive cardiomyopathy. Hypertension 2000; 36:42-7. [PMID: 10904010 DOI: 10.1161/01.hyp.36.1.42] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Hypertensive cardiac hypertrophy and myocardial infarction (MI) are clinically relevant risk factors for heart failure. There is no specific information addressing signaling alterations in the sequence of hypertrophy and post-MI remodeling. To investigate alterations in beta-adrenergic receptor G-protein signaling in ventricular remodeling with pre-existing hypertrophy, MI was induced by coronary artery ligation in Wistar-Kyoto rats (WKY) and spontaneously hypertensive rats (SHR). Ten weeks after the induction of MI, the progression of left ventricular dysfunction and increases in plasma atrial natriuretic peptide (ANP) and cardiac ANP mRNA were more pronounced in SHR than WKY. In addition, the impaired contractile response to beta-adrenergic stimulation was observed in the noninfarcted papillary muscle isolated from SHR. Immunochemical G(s alpha) protein and beta-adrenoceptor density were not significantly altered by MI in both strains. However, immunochemical G(i alpha) was increased (1.5-fold) in the noninfarcted left ventricle of the SHR in which infarction had been induced when compared with that in SHR that underwent sham operation. This increase was observed especially in rats with a high plasma ANP level. Furthermore, there was a positive correlation between G(i alpha) and the extent of post-MI remodeling in WKY. A similar correlation between G(i alpha) and the extent of hypertensive hypertrophy was observed in SHR. In conclusion, the vulnerability of hypertrophied hearts to ischemic damage is greater than that of normotensive hearts. An increase in G(i alpha) could be one mechanism involved in the transition from cardiac hypertrophy to cardiac failure when chronic pressure overload and loss of contractile mass from ischemic heart disease coexist.
Collapse
Affiliation(s)
- I Kouchi
- Klinik III für Innere Medizin, Universität zu Köln, Germany
| | | | | | | | | | | | | |
Collapse
|
49
|
Zolk O, Kouchi I, Schnabel P, Böhm M. Heterotrimeric G proteins in heart disease. Can J Physiol Pharmacol 2000. [DOI: 10.1139/y99-132] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Guanine nucleotide binding proteins (G proteins) are largely grouped into three classes: heterotrimeric G proteins, ras-like or small molecular weight GTP binding proteins, and others like Gh. In the heart G proteins transduce signals from a variety of membrane receptors to generate diverse effects on contractility, heart rate, and myocyte growth. This central position of G proteins forming a switchboard between extracellular signals and intracellular effectors makes them candidates possibly involved in the pathogenesis of cardiac hypertrophy, heart failure, and arrhythmia. This review focuses primarily on discoveries of heterotrimeric G protein alterations in heart diseases that help us to understand the pathogenesis and pathophysiology. We also discuss the underlying molecular mechanisms of heterotrimeric G protein signalling.Key words: G proteins, signal transduction, adrenergic system, heart failure, hypertrophy.
Collapse
|
50
|
Abstract
Beta-adrenergic blocking agents are now standard treatment for mild to moderate chronic heart failure (CHF). However, although many subjects improve on beta blockade, others do not, and some may even deteriorate. Even when subjects improve on beta blockade, they may subsequently decompensate and need acute treatment with a positive inotropic agent. In the presence of full beta blockade, a beta agonist such as dobutamine may have to be administered at very high (> 10 micrograms/kg/min) doses to increase cardiac output, and these doses may increase afterload. In contrast, phosphodiesterase inhibitors (PDEIs) such as milrinone or enoximone retain their full hemodynamic effects in the face of beta blockade. This is because the site of PDEI action is beyond the beta-adrenergic receptor, and because beta blockade reverses receptor pathway desensitization changes, which are detrimental to PDEI response. Moreover, when the combination of a PDEI and a beta-blocking agent is administered long term in CHF, their respective efficacies are additive and their adverse effects subtractive. The PDEI is administered first to increase the tolerability of beta-blocker initiation by counteracting the myocardial depressant effect of adrenergic withdrawal. With this combination, the signature effects of beta blockade (a substantial decrease in heart rate and an increase in left ventricular ejection fraction) are observed, the hemodynamic support conferred by the PDEI appears to be sustained, and clinical results are promising. However, large-scale placebo-controlled studies with PDEIs and beta blockers are needed to confirm these results.
Collapse
Affiliation(s)
- B D Lowes
- Division of Cardiology, University of Colorado Health Sciences Center, Denver 80262, USA
| | | | | | | |
Collapse
|