1
|
Pyle WG. From signal to action: the roles of β 1-adrenergic receptors in cardiac function and disease. Am J Physiol Heart Circ Physiol 2025; 328:H830-H831. [PMID: 40062933 DOI: 10.1152/ajpheart.00151.2025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 03/03/2025] [Accepted: 03/03/2025] [Indexed: 03/27/2025]
Affiliation(s)
- W Glen Pyle
- IMPART Team Canada Investigator Network, Dalhousie Medicine, Dalhousie University, Saint John, New Brunswick, Canada
- Women's Health Research Institute, B.C. Women's Hospital Health Centre, Vancouver, British Columbia, Canada
| |
Collapse
|
2
|
Fan Q, Wang Y, An Q, Ling Y. Right ventricular dysfunction following surgical repair of tetralogy of Fallot: Molecular pathways and therapeutic prospects. Biomed Pharmacother 2025; 184:117924. [PMID: 39983432 DOI: 10.1016/j.biopha.2025.117924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 02/11/2025] [Accepted: 02/15/2025] [Indexed: 02/23/2025] Open
Abstract
Tetralogy of Fallot (TOF) is the most common cyanotic congenital heart disease (CHD). Although surgical correction of TOF is possible, patients often face challenges related to right ventricle dysfunction (RVD) post-surgery, which can significantly impact their long-term survival. The causes of RVD in TOF patients are complex, involving both the unique structural characteristics of the TOF heart and damage resulting from surgical interventions. Residual anatomical issues following TOF repair are often unavoidable, placing the RV under stress and leading to the activation of multiple molecular pathways. This review comprehensively outlines the causes of RVD in patients after TOF surgery, particularly focusing the molecular pathways that contribute to RVD, including established signaling pathways as well as emerging pathways identified through transcriptomic analysis of RV myocardium in TOF patients. We also highlight the features of these molecular pathways concerning RVD, as well as the influence of gender disparities on these molecular pathways. By interpreting the causes and molecular mechanisms underlying RVD after TOF surgery, this review provides new insights for managing RVD in repaired TOF, potentially paving the way for targeted therapies aimed at improving long-term outcomes for those affected by RVD.
Collapse
Affiliation(s)
- Qiang Fan
- Department of Cardiovascular Surgery, West China Hospital of Sichuan University, No. 37 GuoXue Xiang, Chengdu, Sichuan 610041, China.
| | - Yabo Wang
- Department of Cardiovascular Surgery, West China Hospital of Sichuan University, No. 37 GuoXue Xiang, Chengdu, Sichuan 610041, China.
| | - Qi An
- Department of Cardiovascular Surgery, West China Hospital of Sichuan University, No. 37 GuoXue Xiang, Chengdu, Sichuan 610041, China.
| | - Yunfei Ling
- Department of Cardiovascular Surgery, West China Hospital of Sichuan University, No. 37 GuoXue Xiang, Chengdu, Sichuan 610041, China.
| |
Collapse
|
3
|
Kaplan A, El‐Samadi L, Zahreddine R, Amin G, Booz GW, Zouein FA. Canonical or non-canonical, all aspects of G protein-coupled receptor kinase 2 in heart failure. Acta Physiol (Oxf) 2025; 241:e70010. [PMID: 39960030 PMCID: PMC11831727 DOI: 10.1111/apha.70010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 01/12/2025] [Accepted: 01/20/2025] [Indexed: 02/20/2025]
Abstract
G protein-coupled receptor kinase 2 (GRK2) with its multidomain structure performs various crucial cellular functions under both normal and pathological conditions. Overexpression of GRK2 is linked to cardiovascular diseases, and its inhibition or deletion has been shown to be protective. The functions of GRK2 extend beyond G protein-coupled receptor (GPCR) signaling, influencing non-GPCR substrates as well. Increased GRK2 in heart failure (HF) initially may be protective but ultimately leads to maladaptive effects such as GPCR desensitization, insulin resistance, and apoptosis. The multifunctional nature of GRK2, including its action in hypertrophic gene expression, insulin signaling, and cardiac fibrosis, highlights its complex role in HF pathogenesis. Additionally, GRK2 is involved in mitochondrial biogenesis and lipid metabolism. GRK2 also regulates epinephrine secretion from the adrenal gland and its increase in circulating lymphocytes can be used to monitor HF status. Overall, GRK2 is a multifaceted protein with significant implications for HF and the regulation of GRK2 is crucial for understanding and treating cardiovascular diseases.
Collapse
Affiliation(s)
- Abdullah Kaplan
- Department of Pharmacology and ToxicologyAmerican University of Beirut Faculty of MedicineBeirutLebanon
- The Cardiovascular, Renal, and Metabolic Diseases Research Center of ExcellenceAmerican University of Beirut Medical CenterBeirutLebanon
- Cardiology ClinicKemer Public HospitalAntalyaTurkey
| | - Lana El‐Samadi
- Department of Pharmacology and ToxicologyAmerican University of Beirut Faculty of MedicineBeirutLebanon
- The Cardiovascular, Renal, and Metabolic Diseases Research Center of ExcellenceAmerican University of Beirut Medical CenterBeirutLebanon
| | - Rana Zahreddine
- Department of Pharmacology and ToxicologyAmerican University of Beirut Faculty of MedicineBeirutLebanon
- The Cardiovascular, Renal, and Metabolic Diseases Research Center of ExcellenceAmerican University of Beirut Medical CenterBeirutLebanon
| | - Ghadir Amin
- Department of Pharmacology and ToxicologyAmerican University of Beirut Faculty of MedicineBeirutLebanon
- The Cardiovascular, Renal, and Metabolic Diseases Research Center of ExcellenceAmerican University of Beirut Medical CenterBeirutLebanon
- Department of Pharmacology and Toxicology, School of MedicineUniversity of Mississippi Medical CenterJacksonMississippiUSA
| | - George W. Booz
- Department of Pharmacology and Toxicology, School of MedicineUniversity of Mississippi Medical CenterJacksonMississippiUSA
| | - Fouad A. Zouein
- Department of Pharmacology and ToxicologyAmerican University of Beirut Faculty of MedicineBeirutLebanon
- The Cardiovascular, Renal, and Metabolic Diseases Research Center of ExcellenceAmerican University of Beirut Medical CenterBeirutLebanon
- Department of Pharmacology and Toxicology, School of MedicineUniversity of Mississippi Medical CenterJacksonMississippiUSA
| |
Collapse
|
4
|
Kimball TH, Gromova T, Gehred ND, Chapski DJ, Wang K, Vaseghi M, Fischer MA, Lefer DJ, Vondriska TM. Rapid onset fibrotic remodeling and ventricular dysfunction induced by phenylephrine involve targeted reprogramming of myocyte and fibroblast transcriptomes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.11.617933. [PMID: 39464022 PMCID: PMC11507669 DOI: 10.1101/2024.10.11.617933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Catecholamine dysregulation is a common feature of multiple acute and chronic cardiac conditions, including heart failure. To investigate the role of altered α-adrenergic stimulation on cardiac function, we developed a short-term exposure model, administering phenylephrine subcutaneously to mice for one week. Compared to vehicle-injected controls, phenylephrine-treated animals exhibited increased ejection fraction, decreased chamber size, diastolic dysfunction and ventricular hypertrophy in the absence of hypertension. Remarkably, these animals developed extensive fibrotic remodeling of the tissue that plateaued at 24 hours and myocyte hypertrophy localized to regions of fibrotic deposition after 3 days of treatment. Transcriptome analyses of purified myocyte and fibroblast populations from these hearts revealed an unexpected role for myocytes in the production of extracellular matrix. Comparison with other models of cardiac stress, including pressure overload hypertrophy and cytokine activation of fibroblasts, identified stimulus-specific transcriptional circuits associated with cardiac pathology. Given the rapid, robust fibrotic response that preceded myocyte hypertrophy, intercellular communication analyses were conducted to investigate fibroblast to myocyte signaling, identifying potential crosstalk between these cells. These studies thoroughly describe and phenotypically characterize a new model of short-term catecholamine stress and provide an atlas of transcriptional remodeling in myocytes and fibroblasts.
Collapse
Affiliation(s)
- Todd H. Kimball
- Departments of Anesthesiology & Perioperative Medicine, David Geffen School of Medicine
| | - Tatiana Gromova
- Departments of Anesthesiology & Perioperative Medicine, David Geffen School of Medicine
| | - Natalie D. Gehred
- Departments of Anesthesiology & Perioperative Medicine, David Geffen School of Medicine
| | - Douglas J. Chapski
- Departments of Anesthesiology & Perioperative Medicine, David Geffen School of Medicine
| | - Ke Wang
- Department of Cardiac Surgery, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles
| | - Marmar Vaseghi
- Cardiac Arrhythmia Center, Division of Cardiology, Department of Medicine, University of California, Los Angeles
| | - Matthew A. Fischer
- Departments of Anesthesiology & Perioperative Medicine, David Geffen School of Medicine
| | - David J. Lefer
- Department of Cardiac Surgery, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles
| | - Thomas M. Vondriska
- Departments of Anesthesiology & Perioperative Medicine, David Geffen School of Medicine
- Physiology, David Geffen School of Medicine
- Medicine, David Geffen School of Medicine
- Molecular Biology Institute, University of California, Los Angeles
| |
Collapse
|
5
|
Parichatikanond W, Duangrat R, Kurose H, Mangmool S. Regulation of β-Adrenergic Receptors in the Heart: A Review on Emerging Therapeutic Strategies for Heart Failure. Cells 2024; 13:1674. [PMID: 39451192 PMCID: PMC11506672 DOI: 10.3390/cells13201674] [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: 09/05/2024] [Revised: 09/24/2024] [Accepted: 10/08/2024] [Indexed: 10/26/2024] Open
Abstract
The prolonged overstimulation of β-adrenergic receptors (β-ARs), a member of the G protein-coupled receptor (GPCR) family, causes abnormalities in the density and functionality of the receptor and contributes to cardiac dysfunctions, leading to the development and progression of heart diseases, especially heart failure (HF). Despite recent advancements in HF therapy, mortality and morbidity rates continue to be high. Treatment with β-AR antagonists (β-blockers) has improved clinical outcomes and reduced overall hospitalization and mortality rates. However, several barriers in the management of HF remain, providing opportunities to develop new strategies that focus on the functions and signal transduction of β-ARs involved in the pathogenesis of HF. As β-AR can signal through multiple pathways influenced by different receptor subtypes, expression levels, and signaling components such as G proteins, G protein-coupled receptor kinases (GRKs), β-arrestins, and downstream effectors, it presents a complex mechanism that could be targeted in HF management. In this narrative review, we focus on the regulation of β-ARs at the receptor, G protein, and effector loci, as well as their signal transductions in the physiology and pathophysiology of the heart. The discovery of potential ligands for β-AR that activate cardioprotective pathways while limiting off-target signaling is promising for the treatment of HF. However, applying findings from preclinical animal models to human patients faces several challenges, including species differences, the genetic variability of β-ARs, and the complexity and heterogeneity of humans. In this review, we also summarize recent updates and future research on the regulation of β-ARs in the molecular basis of HF and highlight potential therapeutic strategies for HF.
Collapse
Affiliation(s)
| | - Ratchanee Duangrat
- Department of Pharmacology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand;
| | - Hitoshi Kurose
- Pharmacology for Life Sciences, Graduate School of Pharmaceutical Sciences, Tokushima University, Tokushima 770-8505, Japan;
- Pharmacology for Life Sciences, Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8505, Japan
| | - Supachoke Mangmool
- Department of Pharmaceutical Care, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
6
|
Schanche T, Han YS, Jensen CW, Arteaga GM, Tveita T, Sieck GC. β-adrenergic stimulation after rewarming does not mitigate hypothermia-induced contractile dysfunction in rat cardiomyocytes. Cryobiology 2024; 116:104927. [PMID: 38857777 DOI: 10.1016/j.cryobiol.2024.104927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 06/06/2024] [Accepted: 06/06/2024] [Indexed: 06/12/2024]
Abstract
Victims of severe accidental hypothermia are frequently treated with catecholamines to counteract the hemodynamic instability associated with hypothermia-induced cardiac contractile dysfunction. However, we previously reported that the inotropic effects of epinephrine are diminished after hypothermia and rewarming (H/R) in an intact animal model. Thus, the goal of this study was to investigate the effects of Epi treatment on excitation-contraction coupling in isolated rat cardiomyocytes after H/R. In adult male rats, cardiomyocytes isolated from the left ventricle were electrically stimulated at 0.5 Hz and evoked cytosolic [Ca2+] and contractile responses (sarcomere length shortening) were measured. In initial experiments, the effects of varying concentrations of epinephrine on evoked cytosolic [Ca2+] and contractile responses at 37 °C were measured. In a second series of experiments, cardiomyocytes were cooled from 37 °C to 15 °C, maintained at 15 °C for 2 h, then rewarmed to 37 °C (H/R protocol). Immediately after rewarming, the effects of epinephrine treatment on evoked cytosolic [Ca2+] and contractile responses of cardiomyocytes were determined. At 37 °C, epinephrine treatment increased both cytosolic [Ca2+] and contractile responses of cardiomyocytes in a concentration-dependent manner peaking at 25-50 nM. The evoked contractile response of cardiomyocytes after H/R was reduced while the cytosolic [Ca2+] response was slightly elevated. The diminished contractile response of cardiomyocytes after H/R was not mitigated by epinephrine (25 nM) and epinephrine treatment reduced the exponential time decay constant (Tau), but did not increase the cytosolic [Ca2+] response. We conclude that epinephrine treatment does not mitigate H/R-induced contractile dysfunction in cardiomyocytes.
Collapse
Affiliation(s)
- Torstein Schanche
- Department of Physiology & Biomedical Engineering, Mayo Clinic Rochester, MN, USA; Anesthesia and Critical Care Research Group, Department of Clinical Medicine, UiT, The Arctic University of Norway, 9037, Tromsø, Norway
| | - Young Soo Han
- Department of Physiology & Biomedical Engineering, Mayo Clinic Rochester, MN, USA
| | - Cole W Jensen
- Department of Physiology & Biomedical Engineering, Mayo Clinic Rochester, MN, USA
| | - Grace M Arteaga
- Department of Physiology & Biomedical Engineering, Mayo Clinic Rochester, MN, USA
| | - Torkjel Tveita
- Department of Physiology & Biomedical Engineering, Mayo Clinic Rochester, MN, USA; Anesthesia and Critical Care Research Group, Department of Clinical Medicine, UiT, The Arctic University of Norway, 9037, Tromsø, Norway; Division of Surgical Medicine and Intensive Care, University Hospital of North Norway, 9038, Tromsø, Norway
| | - Gary C Sieck
- Department of Physiology & Biomedical Engineering, Mayo Clinic Rochester, MN, USA.
| |
Collapse
|
7
|
Underwood L, Jiang CS, Oh JY, Sato PY. Unheralded Adrenergic Receptor Signaling in Cellular Oxidative Stress and Death. CURRENT OPINION IN PHYSIOLOGY 2024; 40:100766. [PMID: 39070968 PMCID: PMC11271747 DOI: 10.1016/j.cophys.2024.100766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Catecholamines (CAs) bind and activate adrenergic receptors (ARs), thus exuding a key role in cardiac adaptations to global physiological queues. Prolonged exposure to high levels of CAs promotes deleterious effects on the cardiovascular system, leading to organ dysfunction and heart failure (HF). In addition to the prominent role of ARs in inotropic and chronotropic responses, recent studies have delved into elucidating mechanisms contributing to CA toxicity and cell death. Central to this process is understanding the involvement of α1AR and βAR in cardiac remodeling and mechanisms of cellular survival. Here, we highlight the complexity of AR signaling and the fundamental need for a better understanding of its contribution to oxidative stress and cell death. This crucial informational nexus remains a barrier to the development of new therapeutic strategies for cardiovascular diseases.
Collapse
Affiliation(s)
- Lilly Underwood
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL
| | - Chun-Sun Jiang
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL
| | - Joo-Yeun Oh
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL
| | - Priscila Y Sato
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
8
|
Liu S, Anderson PJ, Rajagopal S, Lefkowitz RJ, Rockman HA. G Protein-Coupled Receptors: A Century of Research and Discovery. Circ Res 2024; 135:174-197. [PMID: 38900852 PMCID: PMC11192237 DOI: 10.1161/circresaha.124.323067] [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] [Indexed: 06/22/2024]
Abstract
GPCRs (G protein-coupled receptors), also known as 7 transmembrane domain receptors, are the largest receptor family in the human genome, with ≈800 members. GPCRs regulate nearly every aspect of human physiology and disease, thus serving as important drug targets in cardiovascular disease. Sharing a conserved structure comprised of 7 transmembrane α-helices, GPCRs couple to heterotrimeric G-proteins, GPCR kinases, and β-arrestins, promoting downstream signaling through second messengers and other intracellular signaling pathways. GPCR drug development has led to important cardiovascular therapies, such as antagonists of β-adrenergic and angiotensin II receptors for heart failure and hypertension, and agonists of the glucagon-like peptide-1 receptor for reducing adverse cardiovascular events and other emerging indications. There continues to be a major interest in GPCR drug development in cardiovascular and cardiometabolic disease, driven by advances in GPCR mechanistic studies and structure-based drug design. This review recounts the rich history of GPCR research, including the current state of clinically used GPCR drugs, and highlights newly discovered aspects of GPCR biology and promising directions for future investigation. As additional mechanisms for regulating GPCR signaling are uncovered, new strategies for targeting these ubiquitous receptors hold tremendous promise for the field of cardiovascular medicine.
Collapse
Affiliation(s)
- Samuel Liu
- Department of Medicine, Duke University Medical
Center
| | - Preston J. Anderson
- Cell and Molecular Biology (CMB), Duke University, Durham,
NC, 27710, USA
- Duke Medical Scientist Training Program, Duke University,
Durham, NC, 27710, USA
| | - Sudarshan Rajagopal
- Department of Medicine, Duke University Medical
Center
- Cell and Molecular Biology (CMB), Duke University, Durham,
NC, 27710, USA
- Deparment of Biochemistry Duke University, Durham, NC,
27710, USA
| | - Robert J. Lefkowitz
- Department of Medicine, Duke University Medical
Center
- Deparment of Biochemistry Duke University, Durham, NC,
27710, USA
- Howard Hughes Medical Institute, Duke University Medical
Center, Durham, North Carolina 27710, USA
| | - Howard A. Rockman
- Department of Medicine, Duke University Medical
Center
- Cell and Molecular Biology (CMB), Duke University, Durham,
NC, 27710, USA
| |
Collapse
|
9
|
Yuan X, Yu T, Zhang Z, Li S. Non-invasive assessment of proarrhythmic risks associated with isoprenaline and the dietary supplement ingredient synephrine using human induced pluripotent stem cell-derived cardiomyocytes. Front Cardiovasc Med 2024; 11:1407138. [PMID: 38911513 PMCID: PMC11190318 DOI: 10.3389/fcvm.2024.1407138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 05/23/2024] [Indexed: 06/25/2024] Open
Abstract
Background There have been conflicting reports about the proarrhythmic risk of p-synephrine (SYN). To address this, human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) combined with the microelectrode array (MEA) system have been utilized to assess arrhythmia risks, particularly in the context of adrenomimetic drugs. Aim This study aims to determine whether MEA recordings from hiPSC-CMs could predict the proarrhythmic risk of adrenomimetic drugs and to investigate the cardiovascular effects and mechanisms of SYN. Materials and methods We employed MEA recordings to assess the electrophysiological properties of hiPSC-CMs and conducted concentration-response analyses to evaluate the effects of SYN and Isoprenaline (ISO) on beating rate and contractility. A risk scoring system for proarrhythmic risks was established based on hiPSC-CMs in this study. ISO, a classic beta-adrenergic drug, was also evaluated. Furthermore, the study evaluated the risk of SYN and recorded the concentration-response of beating rate, contractility and the change in the presence or absence of selective β1, β2 and β3 adrenergic blockers. Results Our results suggested that ISO carries a high risk of inducing arrhythmias, aligning with existing literature. SYN caused a 30% prolongation of the field potential duration (FPD) at a concentration of 206.326 μM, a change significantly different from baseline measurements and control treatments. The half maximal effective concentration (EC50) of SYN (3.31 μM) to affect hiPSC-CM beating rate is much higher than that of ISO (18.00 nM). The effect of SYN at an EC50 of 3.31 μM is about ten times more potent in hiPSC-CMs compared to neonatal rat cardiomyocytes (34.12 μM). SYN increased the contractility of cardiomyocytes by 29.97 ± 11.65%, compared to ISO's increase of 50.56 ± 24.15%. β1 receptor blockers almost eliminated the beating rate increase induced by both ISO and SYN, while neither β2 nor β3 blockers had a complete inhibitory effect. Conclusion The MEA and hiPSC-CM system could effectively predict the risk of adrenomimetic drugs. The study concludes that the proarrhythmia risk of SYN at conventional doses is low. SYN is more sensitive in increasing beating rate and contractility in human cardiomyocytes compared to rats, primarily activating β1 receptor.
Collapse
Affiliation(s)
| | | | | | - Sen Li
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
10
|
Bode C, Preissl S, Hein L, Lother A. Catecholamine treatment induces reversible heart injury and cardiomyocyte gene expression. Intensive Care Med Exp 2024; 12:48. [PMID: 38733526 PMCID: PMC11088585 DOI: 10.1186/s40635-024-00632-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 05/07/2024] [Indexed: 05/13/2024] Open
Abstract
BACKGROUND Catecholamines are commonly used as therapeutic drugs in intensive care medicine to maintain sufficient organ perfusion during shock. However, excessive or sustained adrenergic activation drives detrimental cardiac remodeling and may lead to heart failure. Whether catecholamine treatment in absence of heart failure causes persistent cardiac injury, is uncertain. In this experimental study, we assessed the course of cardiac remodeling and recovery during and after prolonged catecholamine treatment and investigated the molecular mechanisms involved. RESULTS C57BL/6N wild-type mice were assigned to 14 days catecholamine treatment with isoprenaline and phenylephrine (IsoPE), treatment with IsoPE and subsequent recovery, or healthy control groups. IsoPE improved left ventricular contractility but caused substantial cardiac fibrosis and hypertrophy. However, after discontinuation of catecholamine treatment, these alterations were largely reversible. To uncover the molecular mechanisms involved, we performed RNA sequencing from isolated cardiomyocyte nuclei. IsoPE treatment resulted in a transient upregulation of genes related to extracellular matrix formation and transforming growth factor signaling. While components of adrenergic receptor signaling were downregulated during catecholamine treatment, we observed an upregulation of endothelin-1 and its receptors in cardiomyocytes, indicating crosstalk between both signaling pathways. To follow this finding, we treated mice with endothelin-1. Compared to IsoPE, treatment with endothelin-1 induced minor but longer lasting changes in cardiomyocyte gene expression. DNA methylation-guided analysis of enhancer regions identified immediate early transcription factors such as AP-1 family members Jun and Fos as key drivers of pathological gene expression following catecholamine treatment. CONCLUSIONS The results from this study show that prolonged catecholamine exposure induces adverse cardiac remodeling and gene expression before the onset of left ventricular dysfunction which has implications for clinical practice. The observed changes depend on the type of stimulus and are largely reversible after discontinuation of catecholamine treatment. Crosstalk with endothelin signaling and the downstream transcription factors identified in this study provide new opportunities for more targeted therapeutic approaches that may help to separate desired from undesired effects of catecholamine treatment.
Collapse
Affiliation(s)
- Christine Bode
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Sebastian Preissl
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Lutz Hein
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- BIOSS Centre for Biological Signaling Studies, University of Freiburg, Freiburg, Germany
| | - Achim Lother
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
- Interdisciplinary Medical Intensive Care, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
11
|
Zhang J, Sandroni PB, Huang W, Gao X, Oswalt L, Schroder MA, Lee S, Shih YYI, Huang HYS, Swigart PM, Myagmar BE, Simpson PC, Rossi JS, Schisler JC, Jensen BC. Cardiomyocyte Alpha-1A Adrenergic Receptors Mitigate Postinfarct Remodeling and Mortality by Constraining Necroptosis. JACC Basic Transl Sci 2024; 9:78-96. [PMID: 38362342 PMCID: PMC10864988 DOI: 10.1016/j.jacbts.2023.08.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 08/26/2023] [Accepted: 08/29/2023] [Indexed: 02/17/2024]
Abstract
Clinical studies have shown that α1-adrenergic receptor antagonists (α-blockers) are associated with increased heart failure risk. The mechanism underlying that hazard and whether it arises from direct inhibition of cardiomyocyte α1-ARs or from systemic effects remain unclear. To address these issues, we created a mouse with cardiomyocyte-specific deletion of the α1A-AR subtype and found that it experienced 70% mortality within 7 days of myocardial infarction driven, in part, by excessive activation of necroptosis. We also found that patients taking α-blockers at our center were at increased risk of death after myocardial infarction, providing clinical correlation for our translational animal models.
Collapse
Affiliation(s)
- Jiandong Zhang
- Division of Cardiology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
- UNC McAllister Heart Institute, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Peyton B. Sandroni
- UNC McAllister Heart Institute, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
- Department of Medicine, University of California-San Francisco, San Francisco, California, USA
| | - Wei Huang
- UNC McAllister Heart Institute, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Xiaohua Gao
- Department of Epidemiology, University of North Carolina Gillings School of Public Health, Chapel Hill, North Carolina, USA
| | - Leah Oswalt
- UNC McAllister Heart Institute, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Melissa A. Schroder
- UNC McAllister Heart Institute, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - SungHo Lee
- Center for Animal MRI, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Yen-Yu I. Shih
- Center for Animal MRI, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Hsiao-Ying S. Huang
- Mechanical and Aerospace Engineering Department, North Carolina State University, Raleigh, North Carolina, USA
| | - Philip M. Swigart
- Department of Medicine, University of California-San Francisco, San Francisco, California, USA
- San Francisco VA Medical Center, San Francisco, California, USA
| | - Bat E. Myagmar
- Department of Medicine, University of California-San Francisco, San Francisco, California, USA
- San Francisco VA Medical Center, San Francisco, California, USA
| | - Paul C. Simpson
- Department of Medicine, University of California-San Francisco, San Francisco, California, USA
- San Francisco VA Medical Center, San Francisco, California, USA
| | - Joseph S. Rossi
- Division of Cardiology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Jonathan C. Schisler
- UNC McAllister Heart Institute, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
- Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Brian C. Jensen
- Division of Cardiology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
- UNC McAllister Heart Institute, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
- Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina, USA
| |
Collapse
|
12
|
Mastos C, Xu X, Keen AC, Halls ML. Signalling of Adrenoceptors: Canonical Pathways and New Paradigms. Handb Exp Pharmacol 2024; 285:147-184. [PMID: 38227198 DOI: 10.1007/164_2023_704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2024]
Abstract
The concept of G protein-coupled receptors initially arose from studies of the β-adrenoceptor, adenylyl cyclase, and cAMP signalling pathway. Since then both canonical G protein-coupled receptor signalling pathways and emerging paradigms in receptor signalling have been defined by experiments focused on adrenoceptors. Here, we discuss the evidence for G protein coupling specificity of the nine adrenoceptor subtypes. We summarise the ability of each of the adrenoceptors to activate proximal signalling mediators including cAMP, calcium, mitogen-activated protein kinases, and protein kinase C pathways. Finally, we highlight the importance of precise spatial and temporal control of adrenoceptor signalling that is controlled by the localisation of receptors at intracellular membranes and in larger protein complexes.
Collapse
Affiliation(s)
- Chantel Mastos
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Xiaomeng Xu
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Alastair C Keen
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Michelle L Halls
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia.
| |
Collapse
|
13
|
Leemann S, Schneider-Warme F, Kleinlogel S. Cardiac optogenetics: shining light on signaling pathways. Pflugers Arch 2023; 475:1421-1437. [PMID: 38097805 PMCID: PMC10730638 DOI: 10.1007/s00424-023-02892-y] [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] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 11/23/2023] [Accepted: 11/24/2023] [Indexed: 12/21/2023]
Abstract
In the early 2000s, the field of neuroscience experienced a groundbreaking transformation with the advent of optogenetics. This innovative technique harnesses the properties of naturally occurring and genetically engineered rhodopsins to confer light sensitivity upon target cells. The remarkable spatiotemporal precision offered by optogenetics has provided researchers with unprecedented opportunities to dissect cellular physiology, leading to an entirely new level of investigation. Initially revolutionizing neuroscience, optogenetics quickly piqued the interest of the wider scientific community, and optogenetic applications were expanded to cardiovascular research. Over the past decade, researchers have employed various optical tools to observe, regulate, and steer the membrane potential of excitable cells in the heart. Despite these advancements, achieving control over specific signaling pathways within the heart has remained an elusive goal. Here, we review the optogenetic tools suitable to control cardiac signaling pathways with a focus on GPCR signaling, and delineate potential applications for studying these pathways, both in healthy and diseased hearts. By shedding light on these exciting developments, we hope to contribute to the ongoing progress in basic cardiac research to facilitate the discovery of novel therapeutic possibilities for treating cardiovascular pathologies.
Collapse
Affiliation(s)
- Siri Leemann
- Institute of Physiology, University of Bern, Bern, Switzerland.
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg - Bad Krozingen, and Medical Faculty, University of Freiburg, Freiburg, Germany.
| | - Franziska Schneider-Warme
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg - Bad Krozingen, and Medical Faculty, University of Freiburg, Freiburg, Germany
| | - Sonja Kleinlogel
- Institute of Physiology, University of Bern, Bern, Switzerland
- F. Hoffmann-La Roche, Translational Medicine Neuroscience, Basel, Switzerland
| |
Collapse
|
14
|
Moric-Janiszewska E, Smolik S, Szydłowski L, Kapral M. Associations between Selected ADRB1 and CYP2D6 Gene Polymorphisms in Children with Ventricular and Supraventricular Arrhythmias. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:2057. [PMID: 38138160 PMCID: PMC10744405 DOI: 10.3390/medicina59122057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 11/13/2023] [Accepted: 11/17/2023] [Indexed: 12/24/2023]
Abstract
Background and Objectives: Tachycardia is a common cardiovascular disease. Drugs blocking β1-adrenergic receptors (ADRB1) are used in the therapy of arrhythmogenic heart diseases. Disease-related polymorphisms can be observed within the ADRB1 gene. The two most important are Ser49Gly and Arg389Gly, and they influence the treatment efficacy. The family of the cytochrome P450 system consists of the isoenzyme CYP2D6 (Debrisoquine 4-hydroxylase), which is involved in phase I metabolism of almost 25% of clinically important drugs, including antiarrhythmic drugs. A study was conducted to detect the ADRB1 and CYP2D6 gene polymorphisms. Materials and Methods: The material for the test was whole blood from 30 patients with ventricular and supraventricular tachycardia and 20 controls. The samples were obtained from the Department of Pediatric Cardiology. The first to be made was the extraction of DNA using a GeneMATRIX Quick Blood DNA Purification Kit from EURx. The selected ADRB1 and CYP2D6 gene polymorphisms were detected by high-resolution melting polymerase chain reaction (HRM-PCR) analysis. Results: Based on the analysis of melt profile data for each PCR product, the identification of polymorphisms was carried out. Heterozygotes and homozygotes were found in the examined alleles. Conclusions: The frequency of the Arg389Gly polymorphism differs statistically significantly between the control group and patients with supraventricular and ventricular arrhythmias, as well as between these two groups of patients. Moreover, the Arg389Gly polymorphism was statistically more prevalent in the group of girls with SVT arrhythmia compared to girls with VT. A few carriers of homozygous and heterozygous systems of the S49G polymorphism were detected among patients with arrhythmias, as well as control group. The percentage of individuals carrying the CYP2D6 4 allele as either homozygous or heterozygous was observed in the study and control groups. The high prevalence of the CYP2D6*4 allele carriers in both groups prompts the optimization of beta-1 blocker therapy.
Collapse
Affiliation(s)
- Ewa Moric-Janiszewska
- Department of Biochemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, Jedności 8B, 41-200 Sosnowiec, Poland
| | - Sławomir Smolik
- Department of Biochemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, Jedności 8B, 41-200 Sosnowiec, Poland
| | - Lesław Szydłowski
- Department of Pediatric Cardiology, Faculty of Medical Sciences in Katowice, Medical University of Silesia in Katowice, Medyków 16, 40-752 Katowice, Poland
| | - Małgorzata Kapral
- Department of Biochemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, Jedności 8B, 41-200 Sosnowiec, Poland
| |
Collapse
|
15
|
Dang W, Cao N, Zhang Y, Li W, Li H. Association among β2-adrenergic receptor autoantibodies and proximal left anterior descending artery lesions in patients with initial ST-segment elevation myocardial infarction. Clin Cardiol 2023; 46:1371-1379. [PMID: 37587904 PMCID: PMC10642316 DOI: 10.1002/clc.24129] [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: 01/30/2023] [Revised: 07/24/2023] [Accepted: 08/08/2023] [Indexed: 08/18/2023] Open
Abstract
BACKGROUND β2 -adrenergic receptor autoantibody (β2 -AA) are widely present in patients with many different types of cardiovascular diseases. Proximal left anterior descending (LAD) artery lesions are associated with adverse prognostic events in patients with ST-segment elevation myocardial infarction (STEMI). HYPOTHESIS β2 -AA is associated with the presence of proximal LAD lesions in patients with STEMI. METHODS A cohort of 153 patients with STEMI who underwent primary percutaneous coronary intervention (PPCI) was enrolled in the study. Baseline characteristics were compared between the proximal LAD group (n = 62) and the nonproximal LAD group (n = 91). Admission serum of patients was collected to detect the level of β2 -AA. Data for echocardiogram within 24 hours after PPCI and at the 6-month follow-up were recorded. RESULTS The optical density values and positive rates of β2 -AA in the proximal LAD group were higher than those in the nonproximal LAD group (p < 0.05). β2 -AA positively correlated with high sensitivity C-reactive protein and peak N-terminal pro-B type natriuretic peptide levels in the proximal LAD group, but those were not relevant in the nonproximal LAD group. Multivariate logistic regression analysis revealed that high β2 -AA levels was independently associated with the presence of proximal LAD lesions in patients with STEMI. Furthermore, a receiver operating characteristic curve was used to show the efficiency of β2 -AA levels to detect proximal LAD lesions, and the AUC of the β2-AA OD value was 0.658 (95% confidence interval 0.568-0.749; p = 0.001). CONCLUSIONS The STEMI patients with high β2 -AA levels had a greater possibility having proximal LAD lesions.
Collapse
Affiliation(s)
- Wenxi Dang
- Department of Cardiology, Cardiovascular Center, Beijing Friendship HospitalCapital Medical UniversityBeijingChina
- Beijing Key Laboratory of Metabolic Disorder Related Cardiovascular DiseaseBeijingChina
| | - Ning Cao
- Department of Cardiology, Cardiovascular Center, Beijing Friendship HospitalCapital Medical UniversityBeijingChina
- Beijing Key Laboratory of Metabolic Disorder Related Cardiovascular DiseaseBeijingChina
- Laboratory of Clinical MedicineCapital Medical UniversityBeijingChina
| | - Yue Zhang
- Department of Cardiology, Cardiovascular Center, Beijing Friendship HospitalCapital Medical UniversityBeijingChina
| | - Weiping Li
- Department of Cardiology, Cardiovascular Center, Beijing Friendship HospitalCapital Medical UniversityBeijingChina
- Beijing Key Laboratory of Metabolic Disorder Related Cardiovascular DiseaseBeijingChina
- Laboratory of Clinical MedicineCapital Medical UniversityBeijingChina
| | - Hongwei Li
- Department of Cardiology, Cardiovascular Center, Beijing Friendship HospitalCapital Medical UniversityBeijingChina
- Beijing Key Laboratory of Metabolic Disorder Related Cardiovascular DiseaseBeijingChina
- Laboratory of Clinical MedicineCapital Medical UniversityBeijingChina
| |
Collapse
|
16
|
Joyce W, Warwicker J, Shiels HA, Perry SF. Evolution and divergence of teleost adrenergic receptors: why sometimes 'the drugs don't work' in fish. J Exp Biol 2023; 226:jeb245859. [PMID: 37823524 DOI: 10.1242/jeb.245859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/13/2023]
Abstract
Adrenaline and noradrenaline, released as hormones and/or neurotransmitters, exert diverse physiological functions in vertebrates, and teleost fishes are widely used as model organisms to study adrenergic regulation; however, such investigations often rely on receptor subtype-specific pharmacological agents (agonists and antagonists; see Glossary) developed and validated in mammals. Meanwhile, evolutionary (phylogenetic and comparative genomic) studies have begun to unravel the diversification of adrenergic receptors (ARs) and reveal that whole-genome duplications and pseudogenization events in fishes results in notable distinctions from mammals in their genomic repertoire of ARs, while lineage-specific gene losses within teleosts have generated significant interspecific variability. In this Review, we visit the evolutionary history of ARs (including α1-, α2- and β-ARs) to highlight the prominent interspecific differences in teleosts, as well as between teleosts and other vertebrates. We also show that structural modelling of teleost ARs predicts differences in ligand binding affinity compared with mammalian orthologs. To emphasize the difficulty of studying the roles of different AR subtypes in fish, we collate examples from the literature of fish ARs behaving atypically compared with standard mammalian pharmacology. Thereafter, we focus on specific case studies of the liver, heart and red blood cells, where our understanding of AR expression has benefited from combining pharmacological approaches with molecular genetics. Finally, we briefly discuss the ongoing advances in 'omics' technologies that, alongside classical pharmacology, will provide abundant opportunities to further explore adrenergic signalling in teleosts.
Collapse
Affiliation(s)
- William Joyce
- Department of Biology - Zoophysiology, Aarhus University, 8000 Aarhus C, Denmark
| | - Jim Warwicker
- Division of Molecular and Cellular Function, Faculty of Biology, Medicine and Health, Manchester Institute of Biotechnology, The University of Manchester, Manchester, M1 7DN, UK
| | - Holly A Shiels
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PL, UK
| | - Steve F Perry
- Department of Biology, University of Ottawa, 30 Marie Curie, Ottawa, ON, Canada, K1N 6N5
| |
Collapse
|
17
|
Dhalla NS, Bhullar SK, Adameova A, Mota KO, de Vasconcelos CML. Status of β 1-Adrenoceptor Signal Transduction System in Cardiac Hypertrophy and Heart Failure. Rev Cardiovasc Med 2023; 24:264. [PMID: 39076390 PMCID: PMC11270071 DOI: 10.31083/j.rcm2409264] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 05/30/2023] [Accepted: 06/15/2023] [Indexed: 07/31/2024] Open
Abstract
Although β 1-adrenoceptor ( β 1-AR) signal transduction, which maintains cardiac function, is downregulated in failing hearts, the mechanisms for such a defect in heart failure are not fully understood. Since cardiac hypertrophy is invariably associated with heart failure, it is possible that the loss of β 1-AR mechanisms in failing heart occurs due to hypertrophic process. In this regard, we have reviewed the information from a rat model of adaptive cardiac hypertrophy and maladaptive hypertrophy at 4 and 24 weeks after inducing pressure overload as well as adaptive cardiac hypertrophy and heart failure at 4 and 24 weeks after inducing volume overload, respectively. Varying degrees of alterations in β 1-AR density as well as isoproterenol-induced increases in cardiac function, intracellular Ca 2 + -concentration in cardiomyocytes and adenylyl cyclase activity in crude membranes have been reported under these hypertrophic conditions. Adaptive hypertrophy at 4 weeks of pressure or volume overload showed unaltered or augmented increases in the activities of different components of β 1-AR signaling. On the other hand, maladaptive hypertrophy due to pressure overload and heart failure due to volume overload at 24 weeks revealed depressions in the activities of β 1-AR signal transduction pathway. These observations provide evidence that β 1-AR signal system is either unaltered or upregulated in adaptive cardiac hypertrophy and downregulated in maladaptive cardiac hypertrophy or heart failure. Furthermore, the information presented in this article supports the concept that downregulation of β 1-AR mechanisms in heart failure or maladaptive cardiac hypertrophy is not due to hypertrophic process per se. It is suggested that a complex mechanism involving the autonomic imbalance may be of a critical importance in determining differential alterations in non-failing and failing hearts.
Collapse
Affiliation(s)
- Naranjan S. Dhalla
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, and Department of Physiology and Pathophysiology, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R2H 2A6, Canada
| | - Sukhwinder K. Bhullar
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, and Department of Physiology and Pathophysiology, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R2H 2A6, Canada
| | - Adriana Adameova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University and Centre of Experimental Medicine, Institute for Heart Research, Slovak Academy of Sciences, 811 03 Bratislava, Slovakia
| | - Karina Oliveira Mota
- Heart Biophysics Laboratory, Department of Physiology, Center for Biological and Health Sciences, Federal University of Sergipe, 73330 Sergipe, Brazil
| | - Carla Maria Lins de Vasconcelos
- Heart Biophysics Laboratory, Department of Physiology, Center for Biological and Health Sciences, Federal University of Sergipe, 73330 Sergipe, Brazil
| |
Collapse
|
18
|
Bourque K, Jones-Tabah J, Pétrin D, Martin RD, Tanny JC, Hébert TE. Comparing the signaling and transcriptome profiling landscapes of human iPSC-derived and primary rat neonatal cardiomyocytes. Sci Rep 2023; 13:12248. [PMID: 37507481 PMCID: PMC10382583 DOI: 10.1038/s41598-023-39525-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 07/26/2023] [Indexed: 07/30/2023] Open
Abstract
The inaccessibility of human cardiomyocytes significantly hindered years of cardiovascular research efforts. To overcome these limitations, non-human cell sources were used as proxies to study heart function and associated diseases. Rodent models became increasingly acceptable surrogates to model the human heart either in vivo or through in vitro cultures. More recently, due to concerns regarding animal to human translation, including cross-species differences, the use of human iPSC-derived cardiomyocytes presented a renewed opportunity. Here, we conducted a comparative study, assessing cellular signaling through cardiac G protein-coupled receptors (GPCRs) in rat neonatal cardiomyocytes (RNCMs) and human induced pluripotent stem cell-derived cardiomyocytes. Genetically encoded biosensors were used to explore GPCR-mediated nuclear protein kinase A (PKA) and extracellular signal-regulated kinase 1/ 2 (ERK1/2) activities in both cardiomyocyte populations. To increase data granularity, a single-cell analytical approach was conducted. Using automated high content microscopy, our analyses of nuclear PKA and ERK1/2 signaling revealed distinct response clusters in rat and human cardiomyocytes. In line with this, bulk RNA-seq revealed key differences in the expression patterns of GPCRs, G proteins and downstream effector expression levels. Our study demonstrates that human stem cell-derived models of the cardiomyocyte offer distinct advantages for understanding cellular signaling in the heart.
Collapse
Affiliation(s)
- Kyla Bourque
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, H3G 1Y6, Canada
| | - Jace Jones-Tabah
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, H3G 1Y6, Canada
| | - Darlaine Pétrin
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, H3G 1Y6, Canada
| | - Ryan D Martin
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, H3G 1Y6, Canada
| | - Jason C Tanny
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, H3G 1Y6, Canada
| | - Terence E Hébert
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, H3G 1Y6, Canada.
| |
Collapse
|
19
|
Neubert E, Rassler B, Hoschke A, Raffort C, Salameh A. Effects of Normobaric Hypoxia and Adrenergic Blockade over 72 h on Cardiac Function in Rats. Int J Mol Sci 2023; 24:11417. [PMID: 37511176 PMCID: PMC10379660 DOI: 10.3390/ijms241411417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 07/06/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
In rats, acute normobaric hypoxia depressed left ventricular (LV) inotropic function. After 24 h of hypoxic exposure, a slight recovery of LV function occurred. We speculated that prolonged hypoxia (72 h) would induce acclimatization and, hence, recovery of LV function. Moreover, we investigated biomarkers of nitrosative stress and apoptosis as possible causes of hypoxic LV depression. To elucidate the role of hypoxic sympathetic activation, we studied whether adrenergic blockade would further deteriorate the general state of the animals and their cardiac function. Ninety-four rats were exposed over 72 h either to normal room air (N) or to normobaric hypoxia (H). The rodents received infusion (0.1 mL/h) with 0.9% NaCl or with different adrenergic blockers. Despite clear signs of acclimatization to hypoxia, the LV depression continued persistently after 72 h of hypoxia. Immunohistochemical analyses revealed significant increases in markers of nitrosative stress, adenosine triphosphate deficiency and apoptosis in the myocardium, which could provide a possible explanation for the absence of LV function recovery. Adrenergic blockade had a slightly deteriorative effect on the hypoxic LV function compared to the hypoxic group with maintained sympathetic efficacy. These findings show that hypoxic sympathetic activation compensates, at least partially, for the compromised function in hypoxic conditions, therefore emphasizing its importance for hypoxia adaptation.
Collapse
Affiliation(s)
- Elias Neubert
- Carl-Ludwig-Institute of Physiology, University of Leipzig, 04103 Leipzig, Germany; (E.N.); (A.H.)
| | - Beate Rassler
- Carl-Ludwig-Institute of Physiology, University of Leipzig, 04103 Leipzig, Germany; (E.N.); (A.H.)
| | - Annekathrin Hoschke
- Carl-Ludwig-Institute of Physiology, University of Leipzig, 04103 Leipzig, Germany; (E.N.); (A.H.)
| | - Coralie Raffort
- Department of Pediatric Cardiology, Heart Centre, University of Leipzig, 04289 Leipzig, Germany; (C.R.); (A.S.)
| | - Aida Salameh
- Department of Pediatric Cardiology, Heart Centre, University of Leipzig, 04289 Leipzig, Germany; (C.R.); (A.S.)
| |
Collapse
|
20
|
Zhang D, Zhao MM, Wu JM, Wang R, Xue G, Xue YB, Shao JQ, Zhang YY, Dong ED, Li ZY, Xiao H. Dual-omics reveals temporal differences in acute sympathetic stress-induced cardiac inflammation following α 1 and β-adrenergic receptors activation. Acta Pharmacol Sin 2023; 44:1350-1365. [PMID: 36737635 PMCID: PMC10310713 DOI: 10.1038/s41401-022-01048-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 12/28/2022] [Indexed: 02/05/2023]
Abstract
Sympathetic stress is prevalent in cardiovascular diseases. Sympathetic overactivation under strong acute stresses triggers acute cardiovascular events including myocardial infarction (MI), sudden cardiac death, and stress cardiomyopathy. α1-ARs and β-ARs, two dominant subtypes of adrenergic receptors in the heart, play a significant role in the physiological and pathologic regulation of these processes. However, little is known about the functional similarities and differences between α1- and β-ARs activated temporal responses in stress-induced cardiac pathology. In this work, we systematically compared the cardiac temporal genome-wide profiles of acute α1-AR and β-AR activation in the mice model by integrating transcriptome and proteome. We found that α1- and β-AR activations induced sustained and transient inflammatory gene expression, respectively. Particularly, the overactivation of α1-AR but not β-AR led to neutrophil infiltration at one day, which was closely associated with the up-regulation of chemokines, activation of NF-κB pathway, and sustained inflammatory response. Furthermore, there are more metabolic disorders under α1-AR overactivation compared with β-AR overactivation. These findings provide a new therapeutic strategy that, besides using β-blocker as soon as possible, blocking α1-AR within one day should also be considered in the treatment of acute stress-associated cardiovascular diseases.
Collapse
Affiliation(s)
- Di Zhang
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Ming-Ming Zhao
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital; NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education; Beijing Key Laboratory of Cardiovascular Receptors Research; Haihe Laboratory of Cell Ecosystem, Beijing, 100191, China
| | - Ji-Min Wu
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital; NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education; Beijing Key Laboratory of Cardiovascular Receptors Research; Haihe Laboratory of Cell Ecosystem, Beijing, 100191, China
| | - Rui Wang
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital; NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education; Beijing Key Laboratory of Cardiovascular Receptors Research; Haihe Laboratory of Cell Ecosystem, Beijing, 100191, China
| | - Gang Xue
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Yan-Bo Xue
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Ji-Qi Shao
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - You-Yi Zhang
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital; NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education; Beijing Key Laboratory of Cardiovascular Receptors Research; Haihe Laboratory of Cell Ecosystem, Beijing, 100191, China
| | - Er-Dan Dong
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital; NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education; Beijing Key Laboratory of Cardiovascular Receptors Research; Haihe Laboratory of Cell Ecosystem, Beijing, 100191, China.
| | - Zhi-Yuan Li
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China.
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China.
| | - Han Xiao
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital; NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education; Beijing Key Laboratory of Cardiovascular Receptors Research; Haihe Laboratory of Cell Ecosystem, Beijing, 100191, China.
| |
Collapse
|
21
|
Joyce W. Evolutionary loss of the ß1-adrenergic receptor in salmonids. Gen Comp Endocrinol 2023; 338:114279. [PMID: 37019291 DOI: 10.1016/j.ygcen.2023.114279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 03/21/2023] [Accepted: 03/30/2023] [Indexed: 04/07/2023]
Abstract
Whole-genome duplications (WGDs) have been at the heart of the diversification of ß-adrenergic receptors (ß-ARs) in vertebrates. Non-teleost jawed vertebrates typically possess three ß-AR genes: adrb1 (ß1-AR), adrb2 (ß2-AR), and adrb3 (ß3-AR), originating from the ancient 2R (two rounds) WGDs. Teleost fishes, owing to the teleost-specific WGD, have five ancestral adrb paralogs (adrb1, adrb2a, adrb2b, adrb3a and adrb3b). Salmonids are particularly intriguing from an evolutionary perspective as they experienced an additional WGD after separating from other teleosts. Moreover, adrenergic regulation in salmonids, especially rainbow trout, has been intensively studied for decades. However, the repertoire of adrb genes in salmonids has not been yet characterized. An exhaustive genome survey of diverse salmonids, spanning five genera, complemented by phylogenetic sequence analysis, revealed each species has seven adrb paralogs: two adrb2a, two adrb2b, two adrb3a and one adrb3b. Surprisingly, salmonids emerge as the first known jawed vertebrate lineage to lack adrb1. adrb1 is nevertheless highly expressed in the hearts of non-salmonid teleosts, indicating that the wealth of data on adrenergic regulation in salmonids should be generalised to other teleost fishes with caution. It is hypothesised that the loss of adrb1 could have been viable because of the evolutionary radiation of adrb2 and adrb3 genes attributable to the salmonid WGD.
Collapse
Affiliation(s)
- William Joyce
- Department of Biology - Zoophysiology, Aarhus University, 8000 Aarhus C, Denmark; Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PL, United Kingdom.
| |
Collapse
|
22
|
de Moura AL, Brum PC, de Carvalho AETS, Spadari RC. Effect of stress on the chronotropic and inotropic responses to β-adrenergic agonists in isolated atria of KOβ2 mice. Life Sci 2023; 322:121644. [PMID: 37004731 DOI: 10.1016/j.lfs.2023.121644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 03/06/2023] [Accepted: 03/26/2023] [Indexed: 04/03/2023]
Abstract
Altered sensitivity to the chronotropic and inotropic effects of catecholamines and reduction in β1/β2-adrenoceptor (β1/β2-AR) ratio were reported in failing and in senescent human heart, as well as in isolated atria and ventricle of rats submitted to stress. This was due to downregulation of β1-AR with or without up-regulation of β2-AR. AIMS To investigate the stress-induced behavior of β1-AR in the heart of mice expressing a non-functional β2-AR subtype. The guiding hypothesis is that the absence of β2-AR signaling will not affect the behavior of β1-AR during stress and that those are independent processes. MATERIALS AND METHODS The chronotropic and inotropic responses to β-AR agonists in isolated atria of stressed mice expressing a non-functional β2-AR were analyzed. The mRNA and protein expressions of β1- and β2-AR were also determined. KEY FINDINGS No deaths were observed in mice under stress protocol. Atria of stressed mice displayed reduced sensitivity to isoprenaline compared to the controls, an effect that was abolished by the β2- and β1-AR antagonists 50 nM ICI118,551 and 300 nM CGP20712A, respectively. Sensitivity and maximum response to the β-agonists dobutamine and salbutamol were not altered by stress or ICI118,551. The responses to dobutamine and salbutamol were prevented by CGP20712A. The expression of β1-AR was reduced at protein levels. SIGNIFICANCE Collectively, our data provide evidence that the cardiac β2-AR is not essential for survival in a stressful situation and that the stress-induced reduction of β1-AR expression was independent of the β2-AR presence.
Collapse
|
23
|
Rupert C, López JE, Cortez-Toledo E, De la Cruz Cabrera O, Chesler NC, Simpson PC, Campbell SG, Baker AJ. Increased length-dependent activation of human engineered heart tissue after chronic α 1A-adrenergic agonist treatment: testing a novel heart failure therapy. Am J Physiol Heart Circ Physiol 2023; 324:H293-H304. [PMID: 36637971 PMCID: PMC9886349 DOI: 10.1152/ajpheart.00279.2022] [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: 06/06/2022] [Revised: 12/06/2022] [Accepted: 12/23/2022] [Indexed: 01/14/2023]
Abstract
Chronic stimulation of cardiac α1A-adrenergic receptors (α1A-ARs) improves symptoms in multiple preclinical models of heart failure. However, the translational significance remains unclear. Human engineered heart tissues (EHTs) provide a means of quantifying the effects of chronic α1A-AR stimulation on human cardiomyocyte physiology. EHTs were created from thin slices of decellularized pig myocardium seeded with human induced pluripotent stem cell (iPSC)-derived cardiomyocytes and fibroblasts. With a paired experimental design, EHTs were cultured for 3 wk, mechanically tested, cultured again for 2 wk with α1A-AR agonist A61603 (10 nM) or vehicle control, and retested after drug washout for 24 h. Separate control experiments determined the effects of EHT age (3-5 wk) or repeat mechanical testing. We found that chronic A61603 treatment caused a 25% increase of length-dependent activation (LDA) of contraction compared with vehicle treatment (n = 7/group, P = 0.035). EHT force was not increased after chronic A61603 treatment. However, after vehicle treatment, EHT force was increased by 35% relative to baseline testing (n = 7/group, P = 0.022), suggesting EHT maturation. Control experiments suggested that increased EHT force resulted from repeat mechanical testing, not from EHT aging. RNA-seq analysis confirmed that the α1A-AR is expressed in human EHTs and found chronic A61603 treatment affected gene expression in biological pathways known to be activated by α1A-ARs, including the MAP kinase signaling pathway. In conclusion, increased LDA in human EHT after chronic A61603 treatment raises the possibility that chronic stimulation of the α1A-AR might be beneficial for increasing LDA in human myocardium and might be beneficial for treating human heart failure by restoring LDA.NEW & NOTEWORTHY Chronic stimulation of α1A-adrenergic receptors (α1A-ARs) is known to mediate therapeutic effects in animal heart failure models. To investigate the effects of chronic α1A-AR stimulation in human cardiomyocytes, we tested engineered heart tissue (EHT) created with iPSC-derived cardiomyocytes. RNA-seq analysis confirmed human EHT expressed α1A-ARs. Chronic (2 wk) α1A-AR stimulation with A61603 (10 nM) increased length-dependent activation (LDA) of contraction. Chronic α1A-AR stimulation might be beneficial for treating human heart failure by restoring LDA.
Collapse
Affiliation(s)
- C. Rupert
- Propria LLC, Branford, Connecticut, United States
| | - J. E. López
- Division of Cardiovascular Medicine, Department of Internal Medicine,
University of California Davis, Davis, California, United States
| | - E. Cortez-Toledo
- Division of Cardiovascular Medicine, Department of Internal Medicine,
University of California Davis, Davis, California, United States
| | | | - N. C. Chesler
- Edwards Lifesciences Foundation Cardiovascular Innovation Research Center, Irvine, California, United States
- Department of Biomedical Engineering, University of California, Irvine, California, United States
| | - P. C. Simpson
- Cardiology Division, Veterans Affairs Medical Center, San Francisco, California, United States
- Department of Medicine, University of California, San Francisco, California, United States
| | - S. G. Campbell
- Departments of Biomedical Engineering and Cellular and Molecular Physiology, Yale University, New Haven, Connecticut, United States
| | - A. J. Baker
- Cardiology Division, Veterans Affairs Medical Center, San Francisco, California, United States
- Department of Medicine, University of California, San Francisco, California, United States
| |
Collapse
|
24
|
Grisanti LA. Radioligand Binding to Quantify Adrenergic Receptor Expression in the Heart. Curr Protoc 2023; 3:e649. [PMID: 36602296 PMCID: PMC9827508 DOI: 10.1002/cpz1.649] [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] [Indexed: 01/06/2023]
Abstract
β-adrenergic receptors regulate cardiac function in both the healthy and failing heart. Their expression is decreased in heart failure due to chronic overactivation of the sympathetic nervous system, contributing to declines in cardiac function and disease progression. Furthermore, therapies that prevent β-adrenergic receptor downregulation or restore β-adrenergic receptor levels are beneficial, making the determination of cardiac β-adrenergic receptor expression in the heart an important consideration. Although quantitative RT-PCR can provide an indication of β-adrenergic receptor density and subtype expression, mRNA levels do not always correlate with functional protein levels. Additionally, antibodies to β-adrenergic receptors lack specificity, making immunoblotting and other antibody-based techniques unreliable. Radioligand binding assays were developed over 50 years ago and remain the gold standard for quantifying β-adrenergic receptor densities in biological samples. This technique capitalizes on the binding of high-affinity, highly specific ligands to receptors and can give quantifiable levels of receptor expression. Furthermore, competition assays using subtype-selective antagonists generate binding profiles and can differentiate β-adrenergic receptor subtype expression in cardiac tissue. This article focuses on the quantification of β-adrenergic receptors in the heart using saturation and competition radioligand binding techniques to quantify β-adrenergic receptor density and ligand affinities in cardiac membranes. © 2023 Wiley Periodicals LLC. Basic Protocol: Radioligand binding to quantify adrenergic receptor expression in the heart.
Collapse
Affiliation(s)
- Laurel A. Grisanti
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA
| |
Collapse
|
25
|
Lam T, Mastos C, Sloan EK, Halls ML. Pathological changes in GPCR signal organisation: Opportunities for targeted therapies for triple negative breast cancer. Pharmacol Ther 2023; 241:108331. [PMID: 36513135 DOI: 10.1016/j.pharmthera.2022.108331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 12/05/2022] [Accepted: 12/07/2022] [Indexed: 12/14/2022]
Abstract
Triple negative breast cancer (TNBC) has the poorest prognosis compared to other breast cancer subtypes, due to a historical lack of targeted therapies and high rates of relapse. Greater insight into the components of signalling pathways in TNBC tumour cells has led to the clinical evaluation, and in some cases approval, of targeted therapies. In the last decade, G protein-coupled receptors, such as the β2-adrenoceptor, have emerged as potential new therapeutic targets. Here, we describe how the β2-adrenoceptor accelerates TNBC progression in response to stress, and the unique signalling pathway activated by the β2-adrenoceptor to drive the invasion of an aggressive TNBC tumour cell. We highlight evidence that supports an altered organisation of GPCRs in tumour cells, and suggests that activation of the same GPCR in a different cellular location can control unique cell responses. Finally, we speculate how the relocation of GPCRs to the "wrong" place in tumour cells presents opportunities to develop targeted anti-cancer GPCR drugs with greater efficacy and minimal adverse effects.
Collapse
Affiliation(s)
- Terrance Lam
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Chantel Mastos
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Erica K Sloan
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Michelle L Halls
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia.
| |
Collapse
|
26
|
The Role of Sympathetic Nerves in Osteoporosis: A Narrative Review. Biomedicines 2022; 11:biomedicines11010033. [PMID: 36672541 PMCID: PMC9855775 DOI: 10.3390/biomedicines11010033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/09/2022] [Accepted: 12/14/2022] [Indexed: 12/25/2022] Open
Abstract
Osteoporosis, a systemic bone disease, is characterized by decreased bone density due to various reasons, destructed bone microstructure, and increased bone fragility. The incidence of osteoporosis is very high among the elderly, and patients with osteoporosis are prone to suffer from spine fractures and hip fractures, which cause great harm to patients. Meanwhile, osteoporosis is mainly treated with anti-osteoporosis drugs that have side effects. Therefore, the development of new treatment modalities has a significant clinical impact. Sympathetic nerves play an important role in various physiological activities and the regulation of osteoporosis as well. Therefore, the role of sympathetic nerves in osteoporosis was reviewed, aiming to provide information for future targeting of sympathetic nerves in osteoporosis.
Collapse
|
27
|
Sandroni PB, Fisher-Wellman KH, Jensen BC. Adrenergic Receptor Regulation of Mitochondrial Function in Cardiomyocytes. J Cardiovasc Pharmacol 2022; 80:364-377. [PMID: 35170492 PMCID: PMC9365878 DOI: 10.1097/fjc.0000000000001241] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 02/01/2022] [Indexed: 01/31/2023]
Abstract
ABSTRACT Adrenergic receptors (ARs) are G protein-coupled receptors that are stimulated by catecholamines to induce a wide array of physiological effects across tissue types. Both α1- and β-ARs are found on cardiomyocytes and regulate cardiac contractility and hypertrophy through diverse molecular pathways. Acute activation of cardiomyocyte β-ARs increases heart rate and contractility as an adaptive stress response. However, chronic β-AR stimulation contributes to the pathobiology of heart failure. By contrast, mounting evidence suggests that α1-ARs serve protective functions that may mitigate the deleterious effects of chronic β-AR activation. Here, we will review recent studies demonstrating that α1- and β-ARs differentially regulate mitochondrial biogenesis and dynamics, mitochondrial calcium handling, and oxidative phosphorylation in cardiomyocytes. We will identify potential mechanisms of these actions and focus on the implications of these findings for the modulation of contractile function in the uninjured and failing heart. Collectively, we hope to elucidate important physiological processes through which these well-studied and clinically relevant receptors stimulate and fuel cardiac contraction to contribute to myocardial health and disease.
Collapse
Affiliation(s)
- Peyton B. Sandroni
- University of North Carolina School of Medicine, Department of Pharmacology
- University of North Carolina School of Medicine, McAllister Heart Institute
| | - Kelsey H. Fisher-Wellman
- East Carolina University Brody School of Medicine, Department of Physiology
- East Carolina University Diabetes and Obesity Institute
| | - Brian C. Jensen
- University of North Carolina School of Medicine, Department of Pharmacology
- University of North Carolina School of Medicine, McAllister Heart Institute
- University of North Carolina School of Medicine, Department of Medicine, Division of Cardiology
| |
Collapse
|
28
|
Muslimova E, Rebrova T, Kondratieva D, Korepanov V, Sonduev E, Kozlov B, Afanasiev S. Expression of the β1-adrenergic receptor (ADRB1) gene in the myocardium and β-adrenergic reactivity of the body in patients with a history of myocardium infraction. Gene 2022; 844:146820. [PMID: 35987510 DOI: 10.1016/j.gene.2022.146820] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 08/08/2022] [Accepted: 08/12/2022] [Indexed: 11/18/2022]
Abstract
β1-adrenergic receptors (β1-AR) directly affect on intracardiac hemodynamic and the ability of the heart to tolerate physical activity by regulating its inotropic and chronotropic functions. Severe hypersympathicotonia, specific to coronary artery disease (CAD) and chronic heart failure (HF), leads to impaired functioning of β1-AR. The aim of this research was to assess the expression level of the β1-AR ADRB1 gene in the myocardium, to evaluate the β-adrenergic reactivity of the membrane (β-ARM) of erythrocytes, and to analyze the association of these parameters with myocardial contractile dysfunction in patients with a myocardial infarction (MI) in the past and without it. The study included 126 patients with chronic CAD. Among the patients, 55.6 % had a history of MI at least 6 months ago. The expression of the ADRB1 gene was assessed using real-time polymerase chain reaction. With this purpose, we isolated RNA from the right atrial appendage, which was excised when a heart-lung machine was connected during a planned coronary bypass surgery. β-ARM was evaluated in 57 patients. This method is based on the fact of inhibition of hemolysis of erythrocytes, placed in a hyposmotic medium, in the presence of a β-blocker. Within the whole sample of patients, the expression of the ADRB1 gene is comparable in different functional classes of HF. There was no linear correlation between the expression of the ADRB1 gene and left ventricle ejection fraction (LVEF). In patients with a history of MI, the expression of the ADRB1 gene was elevated when compared to a group of patients without MI (p = 0.017). Patients with a history of MI had higher values of β-ARM than those without MI (p = 0.017). The reverse correlation between β-ARM and LVEF (r = -0,570, p = 0,002) was revealed in the group of patients without MI but not in the group of patients with a history of MI (r = -0,137, p = 0,479). In the sample of patients with chronic CAD, in the myocardium of subjects with a history of MI, the relative expression of ADRB1 gene was higher compared to the group of patients without MI. In patients with different functional classes (FC) of HF and with different ejection fraction, both with MI and without it, ADRB1 gene expression was comparable. In the group of patients with a history of MI, an increase in β-ARM was observed, i.e. decrease in the number or sensitivity of β-AR. Among patients without MI, an inverse correlation was found between β-ARM and LVEF.
Collapse
Affiliation(s)
- Elvira Muslimova
- Laboratory of Molecular and Cellular Pathology and Gene Diagnostics, Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia.
| | - Tatiana Rebrova
- Laboratory of Molecular and Cellular Pathology and Gene Diagnostics, Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Dina Kondratieva
- Laboratory of Molecular and Cellular Pathology and Gene Diagnostics, Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Viacheslav Korepanov
- Laboratory of Molecular and Cellular Pathology and Gene Diagnostics, Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Erdeni Sonduev
- Department of Cardiovascular Surgery, Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Boris Kozlov
- Department of Cardiovascular Surgery, Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Sergey Afanasiev
- Laboratory of Molecular and Cellular Pathology and Gene Diagnostics, Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| |
Collapse
|
29
|
Joyce W, Pan YK, Garvey K, Saxena V, Perry S. Regulation of heart rate following genetic deletion of the ß1 adrenergic receptor in larval zebrafish. Acta Physiol (Oxf) 2022; 235:e13849. [PMID: 35665450 PMCID: PMC9539580 DOI: 10.1111/apha.13849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 05/17/2022] [Accepted: 06/01/2022] [Indexed: 12/01/2022]
Abstract
Aim Although zebrafish are gaining popularity as biomedical models of cardiovascular disease, our understanding of their cardiac control mechanisms is fragmentary. Our goal was to clarify the controversial role of the ß1‐adrenergic receptor (AR) in the regulation of heart rate in zebrafish. Methods CRISPR‐Cas9 was used to delete the adrb1 gene in zebrafish allowing us to generate a stable adrb1−/− line. Larval heart rates were measured during pharmacological protocols and with exposure to hypercapnia. Expression of the five zebrafish adrb genes were measured in larval zebrafish hearts using qPCR. Results Compared with genetically matched wild‐types (adrb1+/+), adrb1−/− larvae exhibited ~20 beats min−1 lower heart rate, measured from 2 to 21 days post‐fertilization (dpf). Nevertheless, adrb1−/− larvae exhibited preserved positive chronotropic responses to pharmacological treatment with AR agonists (adrenaline, noradrenaline, isoproterenol), which were blocked by propranolol (general ß‐AR antagonist). Regardless of genotype, larvae exhibited similar increases in heart rate in response to hypercapnia (1% CO2) at 5 dpf, but tachycardia was blunted in adrb1−/− larvae at 6 dpf. adrb1 gene expression was abolished in the hearts of adrb1−/− larvae, confirming successful knockout. While gene expression of adrb2a and adrb3a was unchanged, adrb2b and adrb3b mRNA levels increased in adrb1−/− larval hearts. Conclusion Despite adrb1 contributing to the setting of resting heart rate in larvae, it is not strictly essential for zebrafish, as we generated a viable and breeding adrb1−/− line. The chronotropic effects of adrenergic stimulation persist in adrb1−/− zebrafish, likely due to the upregulation of other ß‐AR subtypes.
Collapse
Affiliation(s)
- William Joyce
- Department of Biology University of Ottawa Ottawa Ontario Canada
- Department of Biology – Zoophysiology Aarhus Universitet Aarhus C Denmark
| | - Yihang K. Pan
- Department of Biology University of Ottawa Ottawa Ontario Canada
| | - Kayla Garvey
- Department of Biology University of Ottawa Ottawa Ontario Canada
| | - Vishal Saxena
- Department of Biology University of Ottawa Ottawa Ontario Canada
| | - Steve F. Perry
- Department of Biology University of Ottawa Ottawa Ontario Canada
| |
Collapse
|
30
|
Bogush N, Tan L, Naqvi E, Calvert JW, Graham RM, Taylor WR, Naqvi N, Husain A. Remuscularization with triiodothyronine and β 1-blocker therapy reverses post-ischemic left ventricular dysfunction and adverse remodeling. Sci Rep 2022; 12:8852. [PMID: 35614155 PMCID: PMC9132945 DOI: 10.1038/s41598-022-12723-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 05/09/2022] [Indexed: 11/19/2022] Open
Abstract
Renewal of the myocardium by preexisting cardiomyocytes is a powerful strategy for restoring the architecture and function of hearts injured by myocardial infarction. To advance this strategy, we show that combining two clinically approved drugs, but neither alone, muscularizes the heart through cardiomyocyte proliferation. Specifically, in adult murine cardiomyocytes, metoprolol, a cardioselective β1-adrenergic receptor blocker, when given with triiodothyronine (T3, a thyroid hormone) accentuates the ability of T3 to stimulate ERK1/2 phosphorylation and proliferative signaling by inhibiting expression of the nuclear phospho-ERK1/2-specific phosphatase, dual-specificity phosphatase-5. While short-duration metoprolol plus T3 therapy generates new heart muscle in healthy mice, in mice with myocardial infarction-induced left ventricular dysfunction and pathological remodeling, it remuscularizes the heart, restores contractile function and reverses chamber dilatation; outcomes that are enduring. If the beneficial effects of metoprolol plus T3 are replicated in humans, this therapeutic strategy has the potential to definitively address ischemic heart failure.
Collapse
Affiliation(s)
- Nikolay Bogush
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, 3311 WMRB, 323 WMRB, 101 Woodruff Circle, Atlanta, GA, 30322, USA
| | - Lin Tan
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, 3311 WMRB, 323 WMRB, 101 Woodruff Circle, Atlanta, GA, 30322, USA
| | - Emmen Naqvi
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, 3311 WMRB, 323 WMRB, 101 Woodruff Circle, Atlanta, GA, 30322, USA
| | - John W Calvert
- Department of Surgery, Carlyle Fraser Heart Center, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Robert M Graham
- Victor Chang Cardiac Research Institute, Sydney, NSW, 2010, Australia
| | - W Robert Taylor
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, 3311 WMRB, 323 WMRB, 101 Woodruff Circle, Atlanta, GA, 30322, USA
- Cardiology Division, Atlanta Veterans Affairs Medical Center, Decatur, GA, 30033, USA
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA, 30322, USA
| | - Nawazish Naqvi
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, 3311 WMRB, 323 WMRB, 101 Woodruff Circle, Atlanta, GA, 30322, USA.
| | - Ahsan Husain
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, 3311 WMRB, 323 WMRB, 101 Woodruff Circle, Atlanta, GA, 30322, USA.
| |
Collapse
|
31
|
Qiao L, Fan X, Yang Z, El-Battrawy I, Zhou X, Akin I. Glucose Counteracts Isoprenaline Effects on Ion Channel Functions in Human-Induced Pluripotent Stem Cell-Derived Cardiomyocytes. J Cardiovasc Dev Dis 2022; 9:jcdd9030076. [PMID: 35323624 PMCID: PMC8955312 DOI: 10.3390/jcdd9030076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/01/2022] [Accepted: 03/02/2022] [Indexed: 02/01/2023] Open
Abstract
Recent studies indicate that the disorder of glucose metabolism in myocardial tissue is involved in the development of Takotsubo syndrome (TTS). This study investigated the effects of a high level of glucose on the pathogenesis of TTS, focusing on the electrophysiological mechanisms. Human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) were treated with toxic concentration of isoprenaline (Iso, 1 mM) and a high level of glucose (22 mM) to mimic the setting of TTS and diabetes mellitus (DM). Iso prolonged action potential duration (APD) through enhancing the late sodium channel current and suppressing the transient outward potassium current (Ito). However, a high level of glucose prevented the APD prolongation and the change in Ito. High-level glucose reduced the expression levels of PI3K/Akt, β1-adrenoceptors, Gs-protein, and PKA, suggesting their involvement in the protective effects of high-level glucose against toxic effects of catecholamine. High glucose level did not influence Iso-induced ROS-generation, suggesting that the protective effects of high-level glucose against Iso did not result from changes in ROS generation. High-level glucose may protect cardiomyocytes from the toxic effects of catecholamine excess through suppressing β1-adrenoceptor-Gs-PKA signaling. DM may reduce the risk for occurrence of arrhythmias due to QT prolongation in TTS patients.
Collapse
Affiliation(s)
- Lin Qiao
- Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care, Medical Faculty Mannheim, University Medical Centre Mannheim (UMM), University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany; (L.Q.); (X.F.); (Z.Y.); (I.E.-B.); (I.A.)
| | - Xuehui Fan
- Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care, Medical Faculty Mannheim, University Medical Centre Mannheim (UMM), University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany; (L.Q.); (X.F.); (Z.Y.); (I.E.-B.); (I.A.)
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention of Cardiovascular Diseases, Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, China
- European Center for AngioScience (ECAS) and German Center for Cardiovascular Research (DZHK) Partner Site Heidelberg/Mannheim, 68167 Mannheim, Germany
| | - Zhen Yang
- Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care, Medical Faculty Mannheim, University Medical Centre Mannheim (UMM), University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany; (L.Q.); (X.F.); (Z.Y.); (I.E.-B.); (I.A.)
| | - Ibrahim El-Battrawy
- Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care, Medical Faculty Mannheim, University Medical Centre Mannheim (UMM), University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany; (L.Q.); (X.F.); (Z.Y.); (I.E.-B.); (I.A.)
- European Center for AngioScience (ECAS) and German Center for Cardiovascular Research (DZHK) Partner Site Heidelberg/Mannheim, 68167 Mannheim, Germany
| | - Xiaobo Zhou
- Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care, Medical Faculty Mannheim, University Medical Centre Mannheim (UMM), University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany; (L.Q.); (X.F.); (Z.Y.); (I.E.-B.); (I.A.)
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention of Cardiovascular Diseases, Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, China
- European Center for AngioScience (ECAS) and German Center for Cardiovascular Research (DZHK) Partner Site Heidelberg/Mannheim, 68167 Mannheim, Germany
- Correspondence: ; Tel.: +49-621-383-1448; Fax: +49-621-383-1474
| | - Ibrahim Akin
- Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care, Medical Faculty Mannheim, University Medical Centre Mannheim (UMM), University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany; (L.Q.); (X.F.); (Z.Y.); (I.E.-B.); (I.A.)
- European Center for AngioScience (ECAS) and German Center for Cardiovascular Research (DZHK) Partner Site Heidelberg/Mannheim, 68167 Mannheim, Germany
| |
Collapse
|
32
|
Investigation of the Antiremodeling Effects of Losartan, Mirabegron and Their Combination on the Development of Doxorubicin-Induced Chronic Cardiotoxicity in a Rat Model. Int J Mol Sci 2022; 23:ijms23042201. [PMID: 35216317 PMCID: PMC8877618 DOI: 10.3390/ijms23042201] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 02/11/2022] [Accepted: 02/12/2022] [Indexed: 12/16/2022] Open
Abstract
Despite the effectiveness of doxorubicin (DOXO) as a chemotherapeutic agent, dose-dependent development of chronic cardiotoxicity limits its application. The angiotensin-II receptor blocker losartan is commonly used to treat cardiac remodeling of various etiologies. The beta-3 adrenergic receptor agonist mirabegron was reported to improve chronic heart failure. Here we investigated the effects of losartan, mirabegron and their combination on the development of DOXO-induced chronic cardiotoxicity. Male Wistar rats were divided into five groups: (i) control; (ii) DOXO-only; (iii) losartan-treated DOXO; (iv) mirabegron-treated DOXO; (v) losartan plus mirabegron-treated DOXO groups. The treatments started 5 weeks after DOXO administration. At week 8, echocardiography was performed. At week 9, left ventricles were prepared for histology, qRT-PCR, and Western blot measurements. Losartan improved diastolic but not systolic dysfunction and ameliorated SERCA2a repression in our DOXO-induced cardiotoxicity model. The DOXO-induced overexpression of Il1 and Il6 was markedly decreased by losartan and mirabegron. Mirabegron and the combination treatment improved systolic and diastolic dysfunction and significantly decreased overexpression of Smad2 and Smad3 in our DOXO-induced cardiotoxicity model. Only mirabegron reduced DOXO-induced cardiac fibrosis significantly. Mirabegron and its combination with losartan seem to be promising therapeutic tools against DOXO-induced chronic cardiotoxicity.
Collapse
|
33
|
Chronic isoprenaline/phenylephrine vs. exclusive isoprenaline stimulation in mice: critical contribution of alpha 1-adrenoceptors to early cardiac stress responses. Basic Res Cardiol 2022; 117:15. [PMID: 35286475 PMCID: PMC8921177 DOI: 10.1007/s00395-022-00920-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 02/04/2022] [Accepted: 02/07/2022] [Indexed: 01/31/2023]
Abstract
Hyperactivity of the sympathetic nervous system is a major driver of cardiac remodeling, exerting its effects through both α-, and β-adrenoceptors (α-, β-ARs). As the relative contribution of subtype α1-AR to cardiac stress responses remains poorly investigated, we subjected mice to either subcutaneous perfusion with the β-AR agonist isoprenaline (ISO, 30 mg/kg × day) or to a combination of ISO and the stable α1-AR agonist phenylephrine (ISO/PE, 30 mg/kg × day each). Telemetry analysis revealed similar hemodynamic responses under both ISO and ISO/PE treatment i.e., permanently increased heart rates and only transient decreases in mean blood pressure during the first 24 h. Echocardiography and single cell analysis after 1 week of exposure showed that ISO/PE-, but not ISO-treated animals established α1-AR-mediated inotropic responsiveness to acute adrenergic stimulation. Morphologically, additional PE perfusion limited concentric cardiomyocyte growth and enhanced cardiac collagen deposition during 7 days of treatment. Time-course analysis demonstrated a diverging development in transcriptional patterns at day 4 of treatment i.e., increased expression of selected marker genes Xirp2, Nppa, Tgfb1, Col1a1, Postn under chronic ISO/PE treatment which was either less pronounced or absent in the ISO group. Transcriptome analyses at day 4 via RNA sequencing demonstrated that additional PE treatment caused a marked upregulation of genes allocated to extracellular matrix and fiber organization along with a more pronounced downregulation of genes involved in metabolic processes, muscle adaptation and cardiac electrophysiology. Consistently, transcriptome changes under ISO/PE challenge more effectively recapitulated early transcriptional alterations in pressure overload-induced experimental heart failure and in human hypertrophic cardiomyopathy.
Collapse
|
34
|
Adu-Amankwaah J, Adzika GK, Adekunle AO, Ndzie Noah ML, Mprah R, Bushi A, Akhter N, Huang F, Xu Y, Adzraku SY, Nadeem I, Sun H. ADAM17, A Key Player of Cardiac Inflammation and Fibrosis in Heart Failure Development During Chronic Catecholamine Stress. Front Cell Dev Biol 2021; 9:732952. [PMID: 34966735 PMCID: PMC8710811 DOI: 10.3389/fcell.2021.732952] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 11/16/2021] [Indexed: 12/24/2022] Open
Abstract
Heart failure development is characterized by persistent inflammation and progressive fibrosis owing to chronic catecholamine stress. In a chronic stress state, elevated catecholamines result in the overstimulation of beta-adrenergic receptors (βARs), specifically β2-AR coupling with Gαi protein. Gαi signaling increases the activation of receptor-stimulated p38 mitogen-activated-protein-kinases (p38 MAPKs) and extracellular signal-regulated kinases (ERKs). Phosphorylation by these kinases is a common way to positively regulate the catalytic activity of A Disintegrin and Metalloprotease 17 (ADAM17), a metalloprotease that has grown much attention in recent years and has emerged as a chief regulatory hub in inflammation, fibrosis, and immunity due to its vital proteolytic activity. ADAM17 cleaves and activates proinflammatory cytokines and fibrotic factors that enhance cardiac dysfunction via inflammation and fibrosis. However, there is limited information on the cardiovascular aspect of ADAM17, especially in heart failure. Hence, this concise review provides a comprehensive insight into the structure of ADAM17, how it is activated and regulated during chronic catecholamine stress in heart failure development. This review highlights the inflammatory and fibrotic roles of ADAM17’s substrates; Tumor Necrosis Factor α (TNFα), soluble interleukin-6 receptor (sIL-6R), and amphiregulin (AREG). Finally, how ADAM17-induced chronic inflammation and progressive fibrosis aggravate cardiac dysfunction is discussed.
Collapse
Affiliation(s)
| | | | | | | | - Richard Mprah
- Department of Physiology, Xuzhou Medical University, Xuzhou, China
| | | | - Nazma Akhter
- Department of Physiology, Xuzhou Medical University, Xuzhou, China
| | - Fei Huang
- Department of Physiology, Xuzhou Medical University, Xuzhou, China
| | - Yaxin Xu
- Department of Physiology, Xuzhou Medical University, Xuzhou, China
| | - Seyram Yao Adzraku
- Key Laboratory of Bone Marrow Stem Cell, Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Iqra Nadeem
- Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, China
| | - Hong Sun
- Department of Physiology, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
35
|
Wang J, Pani B, Gokhan I, Xiong X, Kahsai AW, Jiang H, Ahn S, Lefkowitz RJ, Rockman HA. β-Arrestin-Biased Allosteric Modulator Potentiates Carvedilol-Stimulated β Adrenergic Receptor Cardioprotection. Mol Pharmacol 2021; 100:568-579. [PMID: 34561298 PMCID: PMC8626783 DOI: 10.1124/molpharm.121.000359] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 09/20/2021] [Indexed: 01/14/2023] Open
Abstract
β 1 adrenergic receptors (β 1ARs) are central regulators of cardiac function and a drug target for cardiac disease. As a member of the G protein-coupled receptor family, β 1ARs activate cellular signaling by primarily coupling to Gs proteins to activate adenylyl cyclase, cAMP-dependent pathways, and the multifunctional adaptor-transducer protein β-arrestin. Carvedilol, a traditional β-blocker widely used in treating high blood pressure and heart failure by blocking β adrenergic receptor-mediated G protein activation, can selectively stimulate Gs-independent β-arrestin signaling of β adrenergic receptors, a process known as β-arrestin-biased agonism. Recently, a DNA-encoded small-molecule library screen against agonist-occupied β 2 adrenergic receptors (β 2ARs) identified Compound-6 (Cmpd-6) to be a positive allosteric modulator for agonists on β 2ARs. Intriguingly, it was further discovered that Cmpd-6 is positively cooperative with the β-arrestin-biased ligand carvedilol at β 2ARs. Here we describe the surprising finding that at β 1ARs unlike β 2ARs, Cmpd-6 is cooperative only with carvedilol and not agonists. Cmpd-6 increases the binding affinity of carvedilol for β 1ARs and potentiates carvedilol-stimulated, β-arrestin-dependent β 1AR signaling, such as epidermal growth factor receptor transactivation and extracellular signal-regulated kinase activation, whereas it does not have an effect on Gs-mediated cAMP generation. In vivo, Cmpd-6 enhances the antiapoptotic, cardioprotective effect of carvedilol in response to myocardial ischemia/reperfusion injury. This antiapoptotic role of carvedilol is dependent on β-arrestins since it is lost in mice with myocyte-specific deletion of β-arrestins. Our findings demonstrate that Cmpd-6 is a selective β-arrestin-biased allosteric modulator of β 1ARs and highlight its potential clinical utility in enhancing carvedilol-mediated cardioprotection against ischemic injury. SIGNIFICANCE STATEMENT: This study demonstrates the positive cooperativity of Cmpd-6 on β1ARs as a β-arrestin-biased positive allosteric modulator. Cmpd-6 selectively enhances the affinity and cellular signaling of carvedilol, a known β-arrestin-biased β-blocker for β1ARs, whereas it has minimal effect on other ligands tested. Importantly, Cmpd-6 enhances the β-arrestin-dependent in vivo cardioprotective effect of carvedilol during ischemia/reperfusion injury-induced apoptosis. The data support the potential therapeutic application of Cmpd-6 to enhance the clinical benefits of carvedilol in the treatment of cardiac disease.
Collapse
Affiliation(s)
- Jialu Wang
- Departments of Medicine (J.W., B.P, I.G., X.X., A.W.K., H.J., S.A., R.J.L., H.A.R.), Biochemistry (R.J.W.), Howard Hughes Medical Institute (R.J.L.), and Cell Biology (H.A.R.), Duke University Medical Center, Durham, North Carolina
| | - Biswaranjan Pani
- Departments of Medicine (J.W., B.P, I.G., X.X., A.W.K., H.J., S.A., R.J.L., H.A.R.), Biochemistry (R.J.W.), Howard Hughes Medical Institute (R.J.L.), and Cell Biology (H.A.R.), Duke University Medical Center, Durham, North Carolina
| | - Ilhan Gokhan
- Departments of Medicine (J.W., B.P, I.G., X.X., A.W.K., H.J., S.A., R.J.L., H.A.R.), Biochemistry (R.J.W.), Howard Hughes Medical Institute (R.J.L.), and Cell Biology (H.A.R.), Duke University Medical Center, Durham, North Carolina
| | - Xinyu Xiong
- Departments of Medicine (J.W., B.P, I.G., X.X., A.W.K., H.J., S.A., R.J.L., H.A.R.), Biochemistry (R.J.W.), Howard Hughes Medical Institute (R.J.L.), and Cell Biology (H.A.R.), Duke University Medical Center, Durham, North Carolina
| | - Alem W Kahsai
- Departments of Medicine (J.W., B.P, I.G., X.X., A.W.K., H.J., S.A., R.J.L., H.A.R.), Biochemistry (R.J.W.), Howard Hughes Medical Institute (R.J.L.), and Cell Biology (H.A.R.), Duke University Medical Center, Durham, North Carolina
| | - Haoran Jiang
- Departments of Medicine (J.W., B.P, I.G., X.X., A.W.K., H.J., S.A., R.J.L., H.A.R.), Biochemistry (R.J.W.), Howard Hughes Medical Institute (R.J.L.), and Cell Biology (H.A.R.), Duke University Medical Center, Durham, North Carolina
| | - Seungkirl Ahn
- Departments of Medicine (J.W., B.P, I.G., X.X., A.W.K., H.J., S.A., R.J.L., H.A.R.), Biochemistry (R.J.W.), Howard Hughes Medical Institute (R.J.L.), and Cell Biology (H.A.R.), Duke University Medical Center, Durham, North Carolina
| | - Robert J Lefkowitz
- Departments of Medicine (J.W., B.P, I.G., X.X., A.W.K., H.J., S.A., R.J.L., H.A.R.), Biochemistry (R.J.W.), Howard Hughes Medical Institute (R.J.L.), and Cell Biology (H.A.R.), Duke University Medical Center, Durham, North Carolina
| | - Howard A Rockman
- Departments of Medicine (J.W., B.P, I.G., X.X., A.W.K., H.J., S.A., R.J.L., H.A.R.), Biochemistry (R.J.W.), Howard Hughes Medical Institute (R.J.L.), and Cell Biology (H.A.R.), Duke University Medical Center, Durham, North Carolina
| |
Collapse
|
36
|
Bruning R, Dykes H, Jones TW, Wayne NB, Sikora Newsome A. Beta-Adrenergic Blockade in Critical Illness. Front Pharmacol 2021; 12:735841. [PMID: 34721025 PMCID: PMC8554196 DOI: 10.3389/fphar.2021.735841] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Accepted: 09/27/2021] [Indexed: 12/31/2022] Open
Abstract
Catecholamine upregulation is a core pathophysiological feature in critical illness. Sustained catecholamine β-adrenergic induction produces adverse effects relevant to critical illness management. β-blockers (βB) have proposed roles in various critically ill disease states, including sepsis, trauma, burns, and cardiac arrest. Mounting evidence suggests βB improve hemodynamic and metabolic parameters culminating in decreased burn healing time, reduced mortality in traumatic brain injury, and improved neurologic outcomes following cardiac arrest. In sepsis, βB appear hemodynamically benign after acute resuscitation and may augment cardiac function. The emergence of ultra-rapid βB provides new territory for βB, and early data suggest significant improvements in mitigating atrial fibrillation in persistently tachycardic septic patients. This review summarizes the evidence regarding the pharmacotherapeutic role of βB on relevant pathophysiology and clinical outcomes in various types of critical illness.
Collapse
Affiliation(s)
- Rebecca Bruning
- Department of Clinical and Administrative Pharmacy, University of Georgia College of Pharmacy, Augusta, GA, United States
| | - Hannah Dykes
- Department of Clinical and Administrative Pharmacy, University of Georgia College of Pharmacy, Augusta, GA, United States
| | - Timothy W Jones
- Department of Clinical and Administrative Pharmacy, University of Georgia College of Pharmacy, Augusta, GA, United States
| | - Nathaniel B Wayne
- Department of Pharmacy, Augusta University Medical Center, Augusta, GA, United States
| | - Andrea Sikora Newsome
- Department of Clinical and Administrative Pharmacy, University of Georgia College of Pharmacy, Augusta, GA, United States
| |
Collapse
|
37
|
Gaitán-González P, Sánchez-Hernández R, Arias-Montaño JA, Rueda A. Tale of two kinases: Protein kinase A and Ca 2+/calmodulin-dependent protein kinase II in pre-diabetic cardiomyopathy. World J Diabetes 2021; 12:1704-1718. [PMID: 34754372 PMCID: PMC8554373 DOI: 10.4239/wjd.v12.i10.1704] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 07/28/2021] [Accepted: 08/30/2021] [Indexed: 02/06/2023] Open
Abstract
Metabolic syndrome is a pre-diabetic state characterized by several biochemical and physiological alterations, including insulin resistance, visceral fat accumulation, and dyslipidemias, which increase the risk for developing cardiovascular disease. Metabolic syndrome is associated with augmented sympathetic tone, which could account for the etiology of pre-diabetic cardiomyopathy. This review summarizes the current knowledge of the pathophysiological consequences of enhanced and sustained β-adrenergic response in pre-diabetes, focusing on cardiac dysfunction reported in diet-induced experimental models of pre-diabetic cardiomyopathy. The research reviewed indicates that both protein kinase A and Ca2+/calmodulin-dependent protein kinase II play important roles in functional responses mediated by β1-adrenoceptors; therefore, alterations in the expression or function of these kinases can be deleterious. This review also outlines recent information on the role of protein kinase A and Ca2+/calmodulin-dependent protein kinase II in abnormal Ca2+ handling by cardiomyocytes from diet-induced models of pre-diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Pamela Gaitán-González
- Department of Biochemistry, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México 07360, Mexico
| | - Rommel Sánchez-Hernández
- Department of Physiology, Biophysics and Neurosciences, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México 07360, Mexico
| | - José-Antonio Arias-Montaño
- Department of Physiology, Biophysics and Neurosciences, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México 07360, Mexico
| | - Angélica Rueda
- Department of Biochemistry, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México 07360, Mexico
| |
Collapse
|
38
|
Tanner MA, Maitz CA, Grisanti LA. Immune cell β 2-adrenergic receptors contribute to the development of heart failure. Am J Physiol Heart Circ Physiol 2021; 321:H633-H649. [PMID: 34415184 PMCID: PMC8816326 DOI: 10.1152/ajpheart.00243.2021] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 08/16/2021] [Accepted: 08/16/2021] [Indexed: 02/07/2023]
Abstract
β-Adrenergic receptors (βARs) regulate normal and pathophysiological heart function through their impact on contractility. βARs are also regulators of immune function where they play a unique role depending on the disease condition and immune cell type. Emerging evidence suggests an important role for the β2AR subtype in regulating remodeling in the pathological heart; however, the importance of these responses has never been examined. In heart failure, catecholamines are elevated, leading to chronic βAR activation and contributing to the detrimental effects in the heart. We hypothesized that immune cell β2AR plays a critical role in the development of heart failure in response to chronic catecholamine elevations through their regulation of immune cell infiltration. To test this, chimeric mice were generated by performing bone marrow transplant (BMT) experiments using wild-type (WT) or β2AR knockout (KO) donors. WT and β2ARKO BMT mice were chronically administered the βAR agonist isoproterenol. Immune cell recruitment to the heart was examined by histology and flow cytometry. Numerous changes in immune cell recruitment were observed with isoproterenol administration in WT BMT mice including proinflammatory myeloid populations and lymphocytes with macrophages made up the majority of immune cells in the heart and which were absent in β2ARKO BMT animal. β2ARKO BMT mice had decreased cardiomyocyte death, hypertrophy, and interstitial fibrosis following isoproterenol treatment, culminating in improved function. These findings demonstrate an important role for immune cell β2AR expression in the heart's response to chronically elevated catecholamines.NEW & NOTEWORTHY Immune cell β2-adrenergic receptors (β2ARs) are important for proinflammatory macrophage infiltration to the heart in a chronic isoproterenol administration model of heart failure. Mice lacking immune cell β2AR have decreased immune cell infiltration to their heart, primarily proinflammatory macrophage populations. This decrease culminated to decreased cardiac injury with lessened cardiomyocyte death, decreased interstitial fibrosis and hypertrophy, and improved function demonstrating that β2AR regulation of immune responses plays an important role in the heart's response to persistent βAR stimulation.
Collapse
Affiliation(s)
- Miles A Tanner
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, Missouri
| | - Charles A Maitz
- Department of Veterinary Medicine and Surgery, University of Missouri, College of Veterinary Medicine, Columbia, Missouri
| | - Laurel A Grisanti
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, Missouri
| |
Collapse
|
39
|
Grisanti LA. Cardiomyocyte Na +/K +-ATPase-α2 overexpression confers protection in ischemic heart failure. Am J Physiol Heart Circ Physiol 2021; 321:H736-H737. [PMID: 34533399 PMCID: PMC8803311 DOI: 10.1152/ajpheart.00505.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Laurel A Grisanti
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, Missouri
| |
Collapse
|
40
|
Kovács ZZA, Szűcs G, Freiwan M, Kovács MG, Márványkövi FM, Dinh H, Siska A, Farkas K, Kovács F, Kriston A, Horváth P, Kővári B, Cserni BG, Cserni G, Földesi I, Csont T, Sárközy M. Comparison of the antiremodeling effects of losartan and mirabegron in a rat model of uremic cardiomyopathy. Sci Rep 2021; 11:17495. [PMID: 34471171 PMCID: PMC8410807 DOI: 10.1038/s41598-021-96815-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Accepted: 08/17/2021] [Indexed: 02/07/2023] Open
Abstract
Uremic cardiomyopathy is characterized by diastolic dysfunction (DD), left ventricular hypertrophy (LVH), and fibrosis. Angiotensin-II plays a major role in the development of uremic cardiomyopathy via nitro-oxidative and inflammatory mechanisms. In heart failure, the beta-3 adrenergic receptor (β3-AR) is up-regulated and coupled to endothelial nitric oxide synthase (eNOS)-mediated pathways, exerting antiremodeling effects. We aimed to compare the antiremodeling effects of the angiotensin-II receptor blocker losartan and the β3-AR agonist mirabegron in uremic cardiomyopathy. Chronic kidney disease (CKD) was induced by 5/6th nephrectomy in male Wistar rats. Five weeks later, rats were randomized into four groups: (1) sham-operated, (2) CKD, (3) losartan-treated (10 mg/kg/day) CKD, and (4) mirabegron-treated (10 mg/kg/day) CKD groups. At week 13, echocardiographic, histologic, laboratory, qRT-PCR, and Western blot measurements proved the development of uremic cardiomyopathy with DD, LVH, fibrosis, inflammation, and reduced eNOS levels, which were significantly ameliorated by losartan. However, mirabegron showed a tendency to decrease DD and fibrosis; but eNOS expression remained reduced. In uremic cardiomyopathy, β3-AR, sarcoplasmic reticulum ATPase (SERCA), and phospholamban levels did not change irrespective of treatments. Mirabegron reduced the angiotensin-II receptor 1 expression in uremic cardiomyopathy that might explain its mild antiremodeling effects despite the unchanged expression of the β3-AR.
Collapse
Affiliation(s)
- Zsuzsanna Z A Kovács
- MEDICS Research Group, Department of Biochemistry, Interdisciplinary Center of Excellence, Albert Szent-Györgyi Medical School, University of Szeged, Dóm tér 9, Szeged, 6720, Hungary
| | - Gergő Szűcs
- MEDICS Research Group, Department of Biochemistry, Interdisciplinary Center of Excellence, Albert Szent-Györgyi Medical School, University of Szeged, Dóm tér 9, Szeged, 6720, Hungary
| | - Marah Freiwan
- MEDICS Research Group, Department of Biochemistry, Interdisciplinary Center of Excellence, Albert Szent-Györgyi Medical School, University of Szeged, Dóm tér 9, Szeged, 6720, Hungary
| | - Mónika G Kovács
- MEDICS Research Group, Department of Biochemistry, Interdisciplinary Center of Excellence, Albert Szent-Györgyi Medical School, University of Szeged, Dóm tér 9, Szeged, 6720, Hungary
| | - Fanni M Márványkövi
- MEDICS Research Group, Department of Biochemistry, Interdisciplinary Center of Excellence, Albert Szent-Györgyi Medical School, University of Szeged, Dóm tér 9, Szeged, 6720, Hungary
| | - Hoa Dinh
- MEDICS Research Group, Department of Biochemistry, Interdisciplinary Center of Excellence, Albert Szent-Györgyi Medical School, University of Szeged, Dóm tér 9, Szeged, 6720, Hungary
| | - Andrea Siska
- Department of Laboratory Medicine, Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis utca 6, Szeged, 6720, Hungary
| | - Katalin Farkas
- Department of Laboratory Medicine, Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis utca 6, Szeged, 6720, Hungary
| | - Ferenc Kovács
- Synthetic and Systems Biology Unit, Biological Research Centre, Eötvös Loránd Research Network, Temesvári krt. 62, Szeged, 6726, Hungary
- Single-Cell Technologies Ltd, Temesvári krt. 62, Szeged, 6726, Hungary
| | - András Kriston
- Synthetic and Systems Biology Unit, Biological Research Centre, Eötvös Loránd Research Network, Temesvári krt. 62, Szeged, 6726, Hungary
- Single-Cell Technologies Ltd, Temesvári krt. 62, Szeged, 6726, Hungary
| | - Péter Horváth
- Synthetic and Systems Biology Unit, Biological Research Centre, Eötvös Loránd Research Network, Temesvári krt. 62, Szeged, 6726, Hungary
- Single-Cell Technologies Ltd, Temesvári krt. 62, Szeged, 6726, Hungary
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, 00014, Helsinki, Finland
| | - Bence Kővári
- Department of Pathology, Albert Szent-Györgyi Medical School, University of Szeged, Állomás utca 1, Szeged, 6720, Hungary
| | - Bálint Gábor Cserni
- Department of Pathology, Albert Szent-Györgyi Medical School, University of Szeged, Állomás utca 1, Szeged, 6720, Hungary
| | - Gábor Cserni
- Department of Pathology, Albert Szent-Györgyi Medical School, University of Szeged, Állomás utca 1, Szeged, 6720, Hungary
| | - Imre Földesi
- Department of Laboratory Medicine, Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis utca 6, Szeged, 6720, Hungary
| | - Tamás Csont
- MEDICS Research Group, Department of Biochemistry, Interdisciplinary Center of Excellence, Albert Szent-Györgyi Medical School, University of Szeged, Dóm tér 9, Szeged, 6720, Hungary.
| | - Márta Sárközy
- MEDICS Research Group, Department of Biochemistry, Interdisciplinary Center of Excellence, Albert Szent-Györgyi Medical School, University of Szeged, Dóm tér 9, Szeged, 6720, Hungary.
| |
Collapse
|
41
|
Huang M, Fan X, Yang Z, Cyganek L, Li X, Yuecel G, Lan H, Li Y, Wendel A, Lang S, Bieback K, El-Battrawy I, Zhou X, Akin I, Borggrefe M. Alpha 1-adrenoceptor signalling contributes to toxic effects of catecholamine on electrical properties in cardiomyocytes. Europace 2021; 23:1137-1148. [PMID: 33604602 DOI: 10.1093/europace/euab008] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 01/07/2021] [Indexed: 12/18/2022] Open
Abstract
AIMS This study aimed to investigate possible roles and underlying mechanisms of alpha-adrenoceptor coupled signalling for the pathogenesis of Takotsubo syndrome (TTS). METHODS AND RESULTS Human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) were treated with a toxic concentration of epinephrine (Epi, 0.5 mM for 1 h) to mimic the setting of TTS. Patch-clamp technique, polymerase chain reaction (PCR) and Fluorescence-activated cell sorting (FACS) were employed for the study. High concentration Epi suppressed the depolarization velocity, prolonged duration of action potentials and induced arrhythmic events in hiPSC-CMs. The Epi effects were attenuated by an alpha-adrenoceptor blocker (phentolamine), suggesting involvement of alpha-adrenoceptor signalling in arrhythmogenesis related to QT interval prolongation in the setting of TTS. An alpha 1-adrenoceptor agonist (phenylephrine) but not an alpha 2-adrenoceptor agonist (clonidine) mimicked Epi effects. Epi enhanced ROS production, which could be attenuated by the alpha- adrenoceptor blocker. Treatment of cells with H2O2 (100 µM) mimicked the effects of Epi on action potentials and a reactive oxygen species (ROS)-blocker (N-acetyl-I-cysteine, 1 mM) prevented the Epi effects, indicating that the ROS signalling is involved in the alpha-adrenoceptor actions. Nicotinamide adenine dinucleotide phosphate hydrogen (NADPH) oxidases were involved in alpha 1-adrenoceptor signalling. A protein kinase C (PKC) blocker suppressed the effects of Epi, phenylephrine and ROS as well, implying that PKC participated in alpha 1-adrenoceptor signalling and acted as a downstream factor of ROS. The abnormal action potentials resulted from alpha 1-adrenoceptor activation-induced dysfunctions of ion channels including the voltage-dependent Na+ and L-type Ca2+ channels. CONCLUSIONS Alpha 1-adrenoceptor signalling plays important roles for arrhythmogenesis of TTS. Alpha-adrenoceptor blockers might be clinically helpful for treating arrhythmias in patients with TTS.
Collapse
Affiliation(s)
- Mengying Huang
- First Department of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany
| | - Xuehui Fan
- First Department of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany
| | - Zhen Yang
- First Department of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany
| | - Lukas Cyganek
- DZHK (German Center for Cardiovascular Research), Partner Sites Heidelberg-Mannheim and Göttingen, Germany.,Stem Cell Unit, Clinic for Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany
| | - Xin Li
- First Department of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany
| | - Goekhan Yuecel
- First Department of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany.,DZHK (German Center for Cardiovascular Research), Partner Sites Heidelberg-Mannheim and Göttingen, Germany
| | - Huan Lan
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China
| | - Yingrui Li
- First Department of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany
| | - Angela Wendel
- First Department of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany
| | - Siegfried Lang
- First Department of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany.,DZHK (German Center for Cardiovascular Research), Partner Sites Heidelberg-Mannheim and Göttingen, Germany
| | - Karen Bieback
- Institute of Transfusion Medicine and Immunology, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany
| | - Ibrahim El-Battrawy
- First Department of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany.,DZHK (German Center for Cardiovascular Research), Partner Sites Heidelberg-Mannheim and Göttingen, Germany
| | - Xiaobo Zhou
- First Department of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany.,DZHK (German Center for Cardiovascular Research), Partner Sites Heidelberg-Mannheim and Göttingen, Germany.,Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China
| | - Ibrahim Akin
- First Department of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany.,DZHK (German Center for Cardiovascular Research), Partner Sites Heidelberg-Mannheim and Göttingen, Germany
| | - Martin Borggrefe
- First Department of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany.,DZHK (German Center for Cardiovascular Research), Partner Sites Heidelberg-Mannheim and Göttingen, Germany
| |
Collapse
|
42
|
Ahern BM, Sebastian A, Levitan BM, Goh J, Andres DA, Satin J. L-type channel inactivation balances the increased peak calcium current due to absence of Rad in cardiomyocytes. J Gen Physiol 2021; 153:212476. [PMID: 34269819 PMCID: PMC8289690 DOI: 10.1085/jgp.202012854] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 06/25/2021] [Indexed: 12/15/2022] Open
Abstract
The L-type Ca2+ channel (LTCC) provides trigger calcium to initiate cardiac contraction in a graded fashion that is regulated by L-type calcium current (ICa,L) amplitude and kinetics. Inactivation of LTCC is controlled to fine-tune calcium flux and is governed by voltage-dependent inactivation (VDI) and calcium-dependent inactivation (CDI). Rad is a monomeric G protein that regulates ICa,L and has recently been shown to be critical to β-adrenergic receptor (β-AR) modulation of ICa,L. Our previous work showed that cardiomyocyte-specific Rad knockout (cRadKO) resulted in elevated systolic function, underpinned by an increase in peak ICa,L, but without pathological remodeling. Here, we sought to test whether Rad-depleted LTCC contributes to the fight-or-flight response independently of β-AR function, resulting in ICa,L kinetic modifications to homeostatically balance cardiomyocyte function. We recorded whole-cell ICa,L from ventricular cardiomyocytes from inducible cRadKO and control (CTRL) mice. The kinetics of ICa,L stimulated with isoproterenol in CTRL cardiomyocytes were indistinguishable from those of unstimulated cRadKO cardiomyocytes. CDI and VDI are both enhanced in cRadKO cardiomyocytes without differences in action potential duration or QT interval. To confirm that Rad loss modulates LTCC independently of β-AR stimulation, we crossed a β1,β2-AR double-knockout mouse with cRadKO, resulting in a Rad-inducible triple-knockout mouse. Deletion of Rad in cardiomyocytes that do not express β1,β2-AR still yielded modulated ICa,L and elevated basal heart function. Thus, in the absence of Rad, increased Ca2+ influx is homeostatically balanced by accelerated CDI and VDI. Our results indicate that the absence of Rad can modulate the LTCC without contribution of β1,β2-AR signaling and that Rad deletion supersedes β-AR signaling to the LTCC to enhance in vivo heart function.
Collapse
Affiliation(s)
- Brooke M Ahern
- Department of Physiology, University of Kentucky, Lexington, KY
| | | | - Bryana M Levitan
- Department of Physiology, University of Kentucky, Lexington, KY.,Gill Heart and Vascular Institute, University of Kentucky, Lexington, KY
| | - Jensen Goh
- Department of Physiology, University of Kentucky, Lexington, KY
| | - Douglas A Andres
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY
| | - Jonathan Satin
- Department of Physiology, University of Kentucky, Lexington, KY
| |
Collapse
|
43
|
Kilian LS, Frank D, Rangrez AY. RhoA Signaling in Immune Cell Response and Cardiac Disease. Cells 2021; 10:1681. [PMID: 34359851 PMCID: PMC8306393 DOI: 10.3390/cells10071681] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 07/01/2021] [Accepted: 07/01/2021] [Indexed: 11/25/2022] Open
Abstract
Chronic inflammation, the activation of immune cells and their cross-talk with cardiomyocytes in the pathogenesis and progression of heart diseases has long been overlooked. However, with the latest research developments, it is increasingly accepted that a vicious cycle exists where cardiomyocytes release cardiocrine signaling molecules that spiral down to immune cell activation and chronic state of low-level inflammation. For example, cardiocrine molecules released from injured or stressed cardiomyocytes can stimulate macrophages, dendritic cells, neutrophils and even T-cells, which then subsequently increase cardiac inflammation by co-stimulation and positive feedback loops. One of the key proteins involved in stress-mediated cardiomyocyte signal transduction is a small GTPase RhoA. Importantly, the regulation of RhoA activation is critical for effective immune cell response and is being considered as one of the potential therapeutic targets in many immune-cell-mediated inflammatory diseases. In this review we provide an update on the role of RhoA at the juncture of immune cell activation, inflammation and cardiac disease.
Collapse
Affiliation(s)
- Lucia Sophie Kilian
- Department of Internal Medicine III, Cardiology, Angiology, Intensive Care, University Medical Center Kiel, 24105 Kiel, Germany;
- DZHK, German Centre for Cardiovascular Research, Partner Site Hamburg/Kiel/Lübeck, 24105 Kiel, Germany
| | - Derk Frank
- Department of Internal Medicine III, Cardiology, Angiology, Intensive Care, University Medical Center Kiel, 24105 Kiel, Germany;
- DZHK, German Centre for Cardiovascular Research, Partner Site Hamburg/Kiel/Lübeck, 24105 Kiel, Germany
| | - Ashraf Yusuf Rangrez
- Department of Internal Medicine III, Cardiology, Angiology, Intensive Care, University Medical Center Kiel, 24105 Kiel, Germany;
- DZHK, German Centre for Cardiovascular Research, Partner Site Hamburg/Kiel/Lübeck, 24105 Kiel, Germany
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, 69120 Heidelberg, Germany
| |
Collapse
|
44
|
Odnoshivkina YG, Petrov AM. The Role of Neuro-Cardiac Junctions
in Sympathetic Regulation of the Heart. J EVOL BIOCHEM PHYS+ 2021. [DOI: 10.1134/s0022093021030078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
45
|
Perez DM. Targeting Adrenergic Receptors in Metabolic Therapies for Heart Failure. Int J Mol Sci 2021; 22:5783. [PMID: 34071350 PMCID: PMC8198887 DOI: 10.3390/ijms22115783] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 05/20/2021] [Accepted: 05/22/2021] [Indexed: 12/14/2022] Open
Abstract
The heart has a reduced capacity to generate sufficient energy when failing, resulting in an energy-starved condition with diminished functions. Studies have identified numerous changes in metabolic pathways in the failing heart that result in reduced oxidation of both glucose and fatty acid substrates, defects in mitochondrial functions and oxidative phosphorylation, and inefficient substrate utilization for the ATP that is produced. Recent early-phase clinical studies indicate that inhibitors of fatty acid oxidation and antioxidants that target the mitochondria may improve heart function during failure by increasing compensatory glucose oxidation. Adrenergic receptors (α1 and β) are a key sympathetic nervous system regulator that controls cardiac function. β-AR blockers are an established treatment for heart failure and α1A-AR agonists have potential therapeutic benefit. Besides regulating inotropy and chronotropy, α1- and β-adrenergic receptors also regulate metabolic functions in the heart that underlie many cardiac benefits. This review will highlight recent studies that describe how adrenergic receptor-mediated metabolic pathways may be able to restore cardiac energetics to non-failing levels that may offer promising therapeutic strategies.
Collapse
Affiliation(s)
- Dianne M Perez
- The Lerner Research Institute, The Cleveland Clinic Foundation, 9500 Euclid Ave, Cleveland, OH 44195, USA
| |
Collapse
|
46
|
Perez DM. Current Developments on the Role of α 1-Adrenergic Receptors in Cognition, Cardioprotection, and Metabolism. Front Cell Dev Biol 2021; 9:652152. [PMID: 34113612 PMCID: PMC8185284 DOI: 10.3389/fcell.2021.652152] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 04/29/2021] [Indexed: 12/13/2022] Open
Abstract
The α1-adrenergic receptors (ARs) are G-protein coupled receptors that bind the endogenous catecholamines, norepinephrine, and epinephrine. They play a key role in the regulation of the sympathetic nervous system along with β and α2-AR family members. While all of the adrenergic receptors bind with similar affinity to the catecholamines, they can regulate different physiologies and pathophysiologies in the body because they couple to different G-proteins and signal transduction pathways, commonly in opposition to one another. While α1-AR subtypes (α1A, α1B, α1C) have long been known to be primary regulators of vascular smooth muscle contraction, blood pressure, and cardiac hypertrophy, their role in neurotransmission, improving cognition, protecting the heart during ischemia and failure, and regulating whole body and organ metabolism are not well known and are more recent developments. These advancements have been made possible through the development of transgenic and knockout mouse models and more selective ligands to advance their research. Here, we will review the recent literature to provide new insights into these physiological functions and possible use as a therapeutic target.
Collapse
Affiliation(s)
- Dianne M Perez
- The Lerner Research Institute, The Cleveland Clinic Foundation, Cleveland, OH, United States
| |
Collapse
|
47
|
Peter AK, Walker CJ, Ceccato T, Trexler CL, Ozeroff CD, Lugo KR, Perry AR, Anseth KS, Leinwand LA. Cardiac Fibroblasts Mediate a Sexually Dimorphic Fibrotic Response to β-Adrenergic Stimulation. J Am Heart Assoc 2021; 10:e018876. [PMID: 33998248 PMCID: PMC8483546 DOI: 10.1161/jaha.120.018876] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background Biological sex is an important modifier of cardiovascular disease and women generally have better outcomes compared with men. However, the contribution of cardiac fibroblasts (CFs) to this sexual dimorphism is relatively unexplored. Methods and Results Isoproterenol (ISO) was administered to rats as a model for chronic β‐adrenergic receptor (β‐AR)‐mediated cardiovascular disease. ISO‐treated males had higher mortality than females and also developed fibrosis whereas females did not. Gonadectomy did not abrogate this sex difference. To determine the cellular contribution to this phenotype, CFs were studied. CFs from both sexes had increased proliferation in vivo in response to ISO, but CFs from female hearts proliferated more than male cells. In addition, male CFs were significantly more activated to myofibroblasts by ISO. To investigate potential regulatory mechanisms for the sexually dimorphic fibrotic response, β‐AR mRNA and PKA (protein kinase A) activity were measured. In response to ISO treatment, male CFs increased expression of β1‐ and β2‐ARs, whereas expression of both receptors decreased in female CFs. Moreover, ISO‐treated male CFs had higher PKA activity relative to vehicle controls, whereas ISO did not activate PKA in female CFs. Conclusions Chronic in vivo β‐AR stimulation causes fibrosis in male but not female rat hearts. Male CFs are more activated than female CFs, consistent with elevated fibrosis in male rat hearts and may be caused by higher β‐AR expression and PKA activation in male CFs. Taken together, our data suggest that CFs play a substantial role in mediating sex differences observed after cardiac injury.
Collapse
Affiliation(s)
- Angela K Peter
- BioFrontiers Institute University of Colorado Boulder CO.,Department of Molecular, Cellular and Developmental Biology University of Colorado Boulder CO
| | - Cierra J Walker
- BioFrontiers Institute University of Colorado Boulder CO.,Materials Science and Engineering Program University of Colorado Boulder Boulder CO
| | - Tova Ceccato
- BioFrontiers Institute University of Colorado Boulder CO.,Department of Molecular, Cellular and Developmental Biology University of Colorado Boulder CO
| | - Christa L Trexler
- BioFrontiers Institute University of Colorado Boulder CO.,Department of Molecular, Cellular and Developmental Biology University of Colorado Boulder CO
| | - Christopher D Ozeroff
- BioFrontiers Institute University of Colorado Boulder CO.,Department of Molecular, Cellular and Developmental Biology University of Colorado Boulder CO
| | | | - Amy R Perry
- BioFrontiers Institute University of Colorado Boulder CO
| | - Kristi S Anseth
- BioFrontiers Institute University of Colorado Boulder CO.,Department of Chemical and Biological Engineering University of Colorado Boulder CO
| | - Leslie A Leinwand
- BioFrontiers Institute University of Colorado Boulder CO.,Department of Molecular, Cellular and Developmental Biology University of Colorado Boulder CO
| |
Collapse
|
48
|
Walsh SK, Lipina C, Ang SY, Sato M, Chia LY, Kocan M, Hutchinson DS, Summers RJ, Wainwright CL. GPR55 regulates the responsiveness to, but does not dimerise with, α 1A-adrenoceptors. Biochem Pharmacol 2021; 188:114560. [PMID: 33844984 DOI: 10.1016/j.bcp.2021.114560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 03/26/2021] [Accepted: 04/06/2021] [Indexed: 11/15/2022]
Abstract
Emerging evidence suggests that G protein coupled receptor 55 (GPR55) may influence adrenoceptor function/activity in the cardiovascular system. Whether this reflects direct interaction (dimerization) between receptors or signalling crosstalk has not been investigated. This study explored the interaction between GPR55 and the alpha 1A-adrenoceptor (α1A-AR) in the cardiovascular system and the potential to influence function/signalling activities. GPR55 and α1A-AR mediated changes in both cardiac and vascular function was assessed in male wild-type (WT) and GPR55 homozygous knockout (GPR55-/-) mice by pressure volume loop analysis and isolated vessel myography, respectively. Dimerization of GPR55 with the α1A-AR was examined in transfected Chinese hamster ovary-K1 (CHO-K1) cells via Bioluminescence Resonance Energy Transfer (BRET). GPR55 and α1A-AR mediated signalling (extracellular signal-regulated kinase 1/2 (ERK1/2) phosphorylation) was investigated in neonatal rat ventricular cardiomyocytes using AlphaScreen proximity assays. GPR55-/- mice exhibited both enhanced pressor and inotropic responses to A61603 (α1A-AR agonist), while in isolated vessels, A61603 induced vasoconstriction was attenuated by a GPR55-dependent mechanism. Conversely, GPR55-mediated vasorelaxation was not altered by pharmacological blockade of α1A-ARs with tamsulosin. While cellular studies demonstrated that GPR55 and α1A-AR failed to dimerize, pharmacological blockade of GPR55 altered α1A-AR mediated signalling and reduced ERK1/2 phosphorylation. Taken together, this study provides evidence that GPR55 and α1A-AR do not dimerize to form heteromers, but do interact at the signalling level to modulate the function of α1A-AR in the cardiovascular system.
Collapse
Affiliation(s)
- Sarah K Walsh
- Cardiometabolic Health Research, School of Pharmacy and Life Sciences, Robert Gordon University, Sir Ian Wood Building, Aberdeen AB10 7GJ, UK.
| | - Christopher Lipina
- Division of Cell Signalling and Immunology, Sir James Black Centre, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Sheng Y Ang
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Masaaki Sato
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Ling Yeong Chia
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Martina Kocan
- The Florey Institute of Neuroscience and Mental Health and School of Biosciences, University of Melbourne, Parkville, VIC, Australia
| | - Dana S Hutchinson
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Roger J Summers
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Cherry L Wainwright
- Cardiometabolic Health Research, School of Pharmacy and Life Sciences, Robert Gordon University, Sir Ian Wood Building, Aberdeen AB10 7GJ, UK
| |
Collapse
|
49
|
Berisha F, Götz KR, Wegener JW, Brandenburg S, Subramanian H, Molina CE, Rüffer A, Petersen J, Bernhardt A, Girdauskas E, Jungen C, Pape U, Kraft AE, Warnke S, Lindner D, Westermann D, Blankenberg S, Meyer C, Hasenfuß G, Lehnart SE, Nikolaev VO. cAMP Imaging at Ryanodine Receptors Reveals β 2-Adrenoceptor Driven Arrhythmias. Circ Res 2021; 129:81-94. [PMID: 33902292 DOI: 10.1161/circresaha.120.318234] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Filip Berisha
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Germany (F.B., H.S., C.E.M., A.E.K., V.O.N.).,Department of Cardiology (F.B., C.J., U.P., S.W., D.L., D.W., S. Blankenberg, C.M.), University Heart and Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Germany (F.B., H.S., C.E.M., A.E., S.W., D.L., D.W., S. Blankenberg, V.O.N.)
| | - Konrad R Götz
- Department of Cardiology and Pulmonology, Heart Research Center Göttingen, Georg August University Medical Center, Germany (K.R.G., J.W.W., S. Brandenburg, G.H., S.E.L.)
| | - Jörg W Wegener
- Department of Cardiology and Pulmonology, Heart Research Center Göttingen, Georg August University Medical Center, Germany (K.R.G., J.W.W., S. Brandenburg, G.H., S.E.L.).,DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany (J.W.W., S. Brandenburg, G.H., S.E.L.)
| | - Sören Brandenburg
- Department of Cardiology and Pulmonology, Heart Research Center Göttingen, Georg August University Medical Center, Germany (K.R.G., J.W.W., S. Brandenburg, G.H., S.E.L.).,DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany (J.W.W., S. Brandenburg, G.H., S.E.L.)
| | - Hariharan Subramanian
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Germany (F.B., H.S., C.E.M., A.E.K., V.O.N.).,DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Germany (F.B., H.S., C.E.M., A.E., S.W., D.L., D.W., S. Blankenberg, V.O.N.)
| | - Cristina E Molina
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Germany (F.B., H.S., C.E.M., A.E.K., V.O.N.).,DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Germany (F.B., H.S., C.E.M., A.E., S.W., D.L., D.W., S. Blankenberg, V.O.N.)
| | - André Rüffer
- Department of Cardiovascular Surgery, University Heart and Vascular Center Hamburg, Germany (A.R., J.P., A.B., E.G.)
| | - Johannes Petersen
- Department of Cardiovascular Surgery, University Heart and Vascular Center Hamburg, Germany (A.R., J.P., A.B., E.G.)
| | - Alexander Bernhardt
- Department of Cardiovascular Surgery, University Heart and Vascular Center Hamburg, Germany (A.R., J.P., A.B., E.G.)
| | - Evaldas Girdauskas
- Department of Cardiovascular Surgery, University Heart and Vascular Center Hamburg, Germany (A.R., J.P., A.B., E.G.)
| | - Christiane Jungen
- Department of Cardiology (F.B., C.J., U.P., S.W., D.L., D.W., S. Blankenberg, C.M.), University Heart and Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Cardiology-Electrophysiology, cNEP (Cardiac Neuro- and Electrophysiology Research Group) (C.J., U.P., C.M.), University Heart and Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ulrike Pape
- Department of Cardiology (F.B., C.J., U.P., S.W., D.L., D.W., S. Blankenberg, C.M.), University Heart and Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Cardiology-Electrophysiology, cNEP (Cardiac Neuro- and Electrophysiology Research Group) (C.J., U.P., C.M.), University Heart and Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Axel E Kraft
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Germany (F.B., H.S., C.E.M., A.E.K., V.O.N.).,DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Germany (F.B., H.S., C.E.M., A.E., S.W., D.L., D.W., S. Blankenberg, V.O.N.)
| | - Svenja Warnke
- Department of Cardiology (F.B., C.J., U.P., S.W., D.L., D.W., S. Blankenberg, C.M.), University Heart and Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Germany (F.B., H.S., C.E.M., A.E., S.W., D.L., D.W., S. Blankenberg, V.O.N.)
| | - Diana Lindner
- Department of Cardiology (F.B., C.J., U.P., S.W., D.L., D.W., S. Blankenberg, C.M.), University Heart and Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Germany (F.B., H.S., C.E.M., A.E., S.W., D.L., D.W., S. Blankenberg, V.O.N.)
| | - Dirk Westermann
- Department of Cardiology (F.B., C.J., U.P., S.W., D.L., D.W., S. Blankenberg, C.M.), University Heart and Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Germany (F.B., H.S., C.E.M., A.E., S.W., D.L., D.W., S. Blankenberg, V.O.N.)
| | - Stefan Blankenberg
- Department of Cardiology (F.B., C.J., U.P., S.W., D.L., D.W., S. Blankenberg, C.M.), University Heart and Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Germany (F.B., H.S., C.E.M., A.E., S.W., D.L., D.W., S. Blankenberg, V.O.N.)
| | - Christian Meyer
- Department of Cardiology (F.B., C.J., U.P., S.W., D.L., D.W., S. Blankenberg, C.M.), University Heart and Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Cardiology-Electrophysiology, cNEP (Cardiac Neuro- and Electrophysiology Research Group) (C.J., U.P., C.M.), University Heart and Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Gerd Hasenfuß
- Department of Cardiology and Pulmonology, Heart Research Center Göttingen, Georg August University Medical Center, Germany (K.R.G., J.W.W., S. Brandenburg, G.H., S.E.L.).,DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany (J.W.W., S. Brandenburg, G.H., S.E.L.)
| | - Stephan E Lehnart
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany (J.W.W., S. Brandenburg, G.H., S.E.L.)
| | - Viacheslav O Nikolaev
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Germany (F.B., H.S., C.E.M., A.E.K., V.O.N.).,DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Germany (F.B., H.S., C.E.M., A.E., S.W., D.L., D.W., S. Blankenberg, V.O.N.)
| |
Collapse
|
50
|
Why Do We Not Assess Sympathetic Nervous System Activity in Heart Failure Management: Might GRK2 Serve as a New Biomarker? Cells 2021; 10:cells10020457. [PMID: 33669936 PMCID: PMC7924864 DOI: 10.3390/cells10020457] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 02/11/2021] [Accepted: 02/19/2021] [Indexed: 12/11/2022] Open
Abstract
Heart failure (HF) represents the end-stage condition of several structural and functional cardiovascular diseases, characterized by reduced myocardial pump function and increased pressure load. The dysregulation of neurohormonal systems, especially the hyperactivity of the cardiac adrenergic nervous system (ANS), constitutes a hallmark of HF and exerts a pivotal role in its progression. Indeed, it negatively affects patients’ prognosis, being associated with high morbidity and mortality rates, with a tremendous burden on global healthcare systems. To date, all the techniques proposed to assess the cardiac sympathetic nervous system are burdened by intrinsic limits that hinder their implementation in clinical practice. Several biomarkers related to ANS activity, which may potentially support the clinical management of such a complex syndrome, are slow to be implemented in the routine practice for several limitations due to their assessment and clinical impact. Lymphocyte G-protein-coupled Receptor Kinase 2 (GRK2) levels reflect myocardial β-adrenergic receptor function in HF and have been shown to add independent prognostic information related to ANS overdrive. In the present manuscript, we provide an overview of the techniques currently available to evaluate cardiac ANS in HF and future perspectives in this field of relevant scientific and clinical interest.
Collapse
|