1
|
Gural B, Kirkland L, Hockett A, Sandroni P, Zhang J, Rosa-Garrido M, Swift SK, Chapski D, Flinn MA, O'Meara CC, Vondriska TM, Patterson M, Jensen BC, Rau CD. Novel Insights into Post-Myocardial Infarction Cardiac Remodeling through Algorithmic Detection of Cell-Type Composition Shifts. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.09.607400. [PMID: 39149394 PMCID: PMC11326268 DOI: 10.1101/2024.08.09.607400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Background Recent advances in single cell sequencing have led to an increased focus on the role of cell-type composition in phenotypic presentation and disease progression. Cell-type composition research in the heart is challenging due to large, frequently multinucleated cardiomyocytes that preclude most single cell approaches from obtaining accurate measurements of cell composition. Our in silico studies reveal that ignoring cell type composition when calculating differentially expressed genes (DEGs) can have significant consequences. For example, a relatively small change in cell abundance of only 10% can result in over 25% of DEGs being false positives. Methods We have implemented an algorithmic approach that uses snRNAseq datasets as a reference to accurately calculate cell type compositions from bulk RNAseq datasets through robust data cleaning, gene selection, and multi-sample cross-subject and cross-cell-type deconvolution. We applied our approach to cardiomyocyte-specific α1A adrenergic receptor (CM-α1A-AR) knockout mice. 8-12 week-old mice (either WT or CM-α1A-KO) were subjected to permanent left coronary artery (LCA) ligation or sham surgery (n=4 per group). Transcriptomes from the infarct border zones were collected 3 days later and analyzed using our algorithm to determine cell-type abundances, corrected differential expression calculations using DESeq2, and validated these findings using RNAscope. Results Uncorrected DEGs for the CM-α1A-KO X LCA interaction term featured many cell-type specific genes such as Timp4 (fibroblasts) and Aplnr (cardiomyocytes) and overall GO enrichment for terms pertaining to cardiomyocyte differentiation (P=3.1E-4). Using our algorithm, we observe a striking loss of cardiomyocytes and gain in fibroblasts in the α1A-KO + LCA mice that was not recapitulated in WT + LCA animals, although we did observe a similar increase in macrophage abundance in both conditions. This recapitulates prior results that showed a much more severe heart failure phenotype in CM-α1A-KO + LCA mice. Following correction for cell-type, our DEGs now highlight a novel set of genes enriched for GO terms such as cardiac contraction (P=3.7E-5) and actin filament organization (P=6.3E-5). Conclusions Our algorithm identifies and corrects for cell-type abundance in bulk RNAseq datasets opening new avenues for research on novel genes and pathways as well as an improved understanding of the role of cardiac cell types in cardiovascular disease.
Collapse
Affiliation(s)
- Brian Gural
- Department of Genetics and Computational Medicine Program, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Logan Kirkland
- McAllister Heart Institute, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Medicine, Division of Cardiology, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Abbey Hockett
- Department of Genetics and Computational Medicine Program, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Peyton Sandroni
- Department of Pharmacology, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jiandong Zhang
- McAllister Heart Institute, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Medicine, Division of Cardiology, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Manuel Rosa-Garrido
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Samantha K Swift
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Douglas Chapski
- Departments of Anesthesiology & Perioperative Medicine, Medicine/Cardiology, and Physiology, David Geffen School of Medicine; Molecular Biology Institute; University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Michael A Flinn
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Caitlin C O'Meara
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Thomas M Vondriska
- Departments of Anesthesiology & Perioperative Medicine, Medicine/Cardiology, and Physiology, David Geffen School of Medicine; Molecular Biology Institute; University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Michaela Patterson
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Brian C Jensen
- McAllister Heart Institute, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Medicine, Division of Cardiology, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Pharmacology, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Christoph D Rau
- Department of Genetics and Computational Medicine Program, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
2
|
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: 1.0] [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
|
3
|
Diaz-Falcon N, Clark-Price S, Holland M, Johnson J, Lascola K. Ultrasound dilution cardiac output and echocardiography findings in anesthetized mature alpacas (Vicugna pacos) during normotension, hypotension and hypertension. PLoS One 2023; 18:e0284299. [PMID: 37036882 PMCID: PMC10085030 DOI: 10.1371/journal.pone.0284299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 03/28/2023] [Indexed: 04/11/2023] Open
Abstract
Alpacas (Vicugna pacos) have physiologic adaptations to live at high altitude. These adaptations may result in unexpected responses to changes in cardiac performance and blood pressure during general anesthesia. There are few studies evaluating cardiovascular variables in anesthetized alpacas. The purpose of this study was to report cardiovascular performance in anesthetized mature alpacas during normotension, hypotension, and hypertension using ultrasound dilution and echocardiography. Six adult alpacas, 3 females and 3 castrated males, weighing 62.6 to 88.7 kg were anesthetized and maintained with isoflurane and placed in right lateral recumbency. Each alpaca underwent ultrasound dilution and echocardiography measurements during three cardiovascular phases, normotension, hypotension via increased isoflurane concentration, and hypertension via phenylephrine infusion. Variables were analyzed with a Friedman test and a post hoc Dunn's test when significant. A p < 0.05 was used for significance. Cardiac output, cardiac index, systemic vascular resistance, stroke volume, total ejection fraction, left ventricular internal diameter during diastole, and total stroke volume indexed to body weight were greater for hypertension compared to hypotension. Total ejection fraction, stroke volume, and left ventricular ejection time were greater for hypertions compared to normotension. There was no difference between ultrasound dilution and echocardiography determined cardiac output measurements within each cardiovascular phase. Phenylephrine appeared to have increased ventricular performance and/or increased preload in anesthetized, mature alpacas. For detecting change in cardiovascular status in anesthetized alpacas, ultrasound dilution and echocardiography may be useful.
Collapse
Affiliation(s)
- Noelia Diaz-Falcon
- Department of Clinical Sciences, College of Veterinary Medicine, Auburn University, Auburn, Alabama, United States of America
| | - Stuart Clark-Price
- Department of Clinical Sciences, College of Veterinary Medicine, Auburn University, Auburn, Alabama, United States of America
| | - Merrilee Holland
- Department of Clinical Sciences, College of Veterinary Medicine, Auburn University, Auburn, Alabama, United States of America
| | - Jacob Johnson
- Department of Clinical Sciences, College of Veterinary Medicine, Auburn University, Auburn, Alabama, United States of America
| | - Kara Lascola
- Department of Clinical Sciences, College of Veterinary Medicine, Auburn University, Auburn, Alabama, United States of America
| |
Collapse
|
4
|
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: 2.5] [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
|
5
|
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: 6] [Impact Index Per Article: 3.0] [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
|
6
|
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: 13] [Impact Index Per Article: 4.3] [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
|
7
|
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: 20] [Impact Index Per Article: 6.7] [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
|
8
|
Joyce W, Scholman KT, Jensen B, Wang T, Boukens BJ. α 1-adrenergic stimulation increases ventricular action potential duration in the intact mouse heart. Facets (Ott) 2021. [DOI: 10.1139/facets-2020-0081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The role of α1-adrenergic receptors (α-ARs) in the regulation of myocardial function is less well-understood than that of β-ARs. Previous reports in the mouse heart have described that α1-adrenergic stimulation shortens action potential duration in isolated cells or tissues, in contrast to prolongation of the action potential reported in most other mammalian hearts. It has since become appreciated, however, that the mouse heart exhibits marked variation in inotropic response to α1-adrenergic stimulation between ventricles and even individual cardiomyocytes. We investigated the effects of α1-adrenergic stimulation on action potential duration at 80% of repolarization in the right and left ventricles of Langendorff-perfused mouse hearts using optical mapping. In hearts under β-adrenergic blockade (propranolol), phenylephrine or noradrenaline perfusion both increased action potential duration in both ventricles. The increased action potential duration was partially reversed by subsequent perfusion with the α-adrenergic antagonist phentolamine (1 μmol L−1). These data show that α1-receptor stimulation may lead to a prolonging of action potential in the mouse heart and thereby refine our understanding of how action potential duration adjusts during sympathetic stimulation.
Collapse
Affiliation(s)
- William Joyce
- Department of Biology—Zoophysiology, Aarhus University, DK-8000 Aarhus C, Denmark
- Department of Biology, University of Ottawa, 30 Marie Curie, Ottawa, ON K1N 6N5, Canada
| | - Koen T. Scholman
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, 11005 AZ Amsterdam, the Netherlands
| | - Bjarke Jensen
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, 11005 AZ Amsterdam, the Netherlands
| | - Tobias Wang
- Department of Biology—Zoophysiology, Aarhus University, DK-8000 Aarhus C, Denmark
| | - Bastiaan J. Boukens
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, 11005 AZ Amsterdam, the Netherlands
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, 1100 DD Amsterdam, the Netherlands
| |
Collapse
|
9
|
Zhang J, Simpson PC, Jensen BC. Cardiac α1A-adrenergic receptors: emerging protective roles in cardiovascular diseases. Am J Physiol Heart Circ Physiol 2020; 320:H725-H733. [PMID: 33275531 DOI: 10.1152/ajpheart.00621.2020] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
α1-Adrenergic receptors (ARs) are catecholamine-activated G protein-coupled receptors (GPCRs) that are expressed in mouse and human myocardium and vasculature, and play essential roles in the regulation of cardiovascular physiology. Though α1-ARs are less abundant in the heart than β1-ARs, activation of cardiac α1-ARs results in important biologic processes such as hypertrophy, positive inotropy, ischemic preconditioning, and protection from cell death. Data from the Antihypertensive and Lipid-Lowering Treatment to Prevent Heart Attack Trial (ALLHAT) indicate that nonselectively blocking α1-ARs is associated with a twofold increase in adverse cardiac events, including heart failure and angina, suggesting that α1-AR activation might also be cardioprotective in humans. Mounting evidence implicates the α1A-AR subtype in these adaptive effects, including prevention and reversal of heart failure in animal models by α1A agonists. In this review, we summarize recent advances in our understanding of cardiac α1A-ARs.
Collapse
Affiliation(s)
- Jiandong Zhang
- McAllister Heart Institute, University of North Carolina, School of Medicine, Chapel Hill, North Carolina
| | - Paul C Simpson
- Department of Medicine and Research Service, San Francisco Veterans Affairs Medical Center and Cardiovascular Research Institute, University of California, San Francisco, California
| | - Brian C Jensen
- McAllister Heart Institute, University of North Carolina, School of Medicine, Chapel Hill, North Carolina
| |
Collapse
|
10
|
Kaykı-Mutlu G, Papazisi O, Palmen M, Danser AHJ, Michel MC, Arioglu-Inan E. Cardiac and Vascular α 1-Adrenoceptors in Congestive Heart Failure: A Systematic Review. Cells 2020; 9:E2412. [PMID: 33158106 PMCID: PMC7694190 DOI: 10.3390/cells9112412] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/29/2020] [Accepted: 11/02/2020] [Indexed: 02/07/2023] Open
Abstract
As heart failure (HF) is a devastating health problem worldwide, a better understanding and the development of more effective therapeutic approaches are required. HF is characterized by sympathetic system activation which stimulates α- and β-adrenoceptors (ARs). The exposure of the cardiovascular system to the increased locally released and circulating levels of catecholamines leads to a well-described downregulation and desensitization of β-ARs. However, information on the role of α-AR is limited. We have performed a systematic literature review examining the role of both cardiac and vascular α1-ARs in HF using 5 databases for our search. All three α1-AR subtypes (α1A, α1B and α1D) are expressed in human and animal hearts and blood vessels in a tissue-dependent manner. We summarize the changes observed in HF regarding the density, signaling and responses of α1-ARs. Conflicting findings arise from different studies concerning the influence that HF has on α1-AR expression and function; in contrast to β-ARs there is no consistent evidence for down-regulation or desensitization of cardiac or vascular α1-ARs. Whether α1-ARs are a therapeutic target in HF remains a matter of debate.
Collapse
Affiliation(s)
- Gizem Kaykı-Mutlu
- Department of Pharmacology, Faculty of Pharmacy, Ankara University, 06560 Ankara, Turkey; (G.K.-M.); (E.A.-I.)
| | - Olga Papazisi
- Department of Cardiothoracic Surgery, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (O.P.); (M.P.)
| | - Meindert Palmen
- Department of Cardiothoracic Surgery, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (O.P.); (M.P.)
| | - A. H. Jan Danser
- Department of Internal Medicine, Division of Pharmacology, Erasmus Medical Center, 3000 CA Rotterdam, The Netherlands;
| | - Martin C. Michel
- Department of Pharmacology, Johannes Gutenberg University, 55131 Mainz, Germany
| | - Ebru Arioglu-Inan
- Department of Pharmacology, Faculty of Pharmacy, Ankara University, 06560 Ankara, Turkey; (G.K.-M.); (E.A.-I.)
| |
Collapse
|
11
|
Carbajal-García A, Reyes-García J, Montaño LM. Androgen Effects on the Adrenergic System of the Vascular, Airway, and Cardiac Myocytes and Their Relevance in Pathological Processes. Int J Endocrinol 2020; 2020:8849641. [PMID: 33273918 PMCID: PMC7676939 DOI: 10.1155/2020/8849641] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 09/17/2020] [Accepted: 10/20/2020] [Indexed: 02/06/2023] Open
Abstract
INTRODUCTION Androgen signaling comprises nongenomic and genomic pathways. Nongenomic actions are not related to the binding of the androgen receptor (AR) and occur rapidly. The genomic effects implicate the binding to a cytosolic AR, leading to protein synthesis. Both events are independent of each other. Genomic effects have been associated with different pathologies such as vascular ischemia, hypertension, asthma, and cardiovascular diseases. Catecholamines play a crucial role in regulating vascular smooth muscle (VSM), airway smooth muscle (ASM), and cardiac muscle (CM) function and tone. OBJECTIVE The aim of this review is an updated analysis of the role of androgens in the adrenergic system of vascular, airway, and cardiac myocytes. Body. Testosterone (T) favors vasoconstriction, and its concentration fluctuation during life stages can affect the vascular tone and might contribute to the development of hypertension. In the VSM, T increases α1-adrenergic receptors (α 1-ARs) and decreases adenylyl cyclase expression, favoring high blood pressure and hypertension. Androgens have also been associated with asthma. During puberty, girls are more susceptible to present asthma symptoms than boys because of the increment in the plasmatic concentrations of T in young men. In the ASM, β 2-ARs are responsible for the bronchodilator effect, and T augments the expression of β 2-ARs evoking an increase in the relaxing response to salbutamol. The levels of T are also associated with an increment in atherosclerosis and cardiovascular risk. In the CM, activation of α 1A-ARs and β 2-ARs increases the ionotropic activity, leading to the development of contraction, and T upregulates the expression of both receptors and improves the myocardial performance. CONCLUSIONS Androgens play an essential role in the adrenergic system of vascular, airway, and cardiac myocytes, favoring either a state of health or disease. While the use of androgens as a therapeutic tool for treating asthma symptoms or heart disease is proposed, the vascular system is warmly affected.
Collapse
Affiliation(s)
- Abril Carbajal-García
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, CDMX, Mexico
| | - Jorge Reyes-García
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, CDMX, Mexico
| | - Luis M. Montaño
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, CDMX, Mexico
| |
Collapse
|
12
|
Janssen PM, Canan BD, Kilic A, Whitson BA, Baker AJ. Human Myocardium Has a Robust α1A-Subtype Adrenergic Receptor Inotropic Response. J Cardiovasc Pharmacol 2018; 72:136-142. [PMID: 29923888 PMCID: PMC6126952 DOI: 10.1097/fjc.0000000000000604] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Recent studies report that a single subtype of α1-adrenergic receptor (α1-AR), the α1A-subtype, mediates robust cardioprotective effects in multiple experimental models of heart failure, suggesting that the α1A-subtype is a potential therapeutic target for an agonist to treat heart failure. Moreover, we recently found that the α1A-subtype is present in human heart. The goal of this study was to assess the inotropic response mediated by the α1A-subtype in human myocardium, and to determine whether the response is downregulated in myocardium from failing human heart. We measured in vitro contractile responses of cardiac muscle preparations (trabeculae) isolated from the right ventricle from nonfailing and failing human hearts. Addition of the α1A-subtype agonist A61603 (100 nM) resulted in a large positive inotropic response (force increased ≈ 2-fold). This response represented ≈70% of the response mediated by the β-adrenergic receptor agonist isoproterenol (1 μM). Moreover, in myocardium from failing hearts, α1A-subtype responses remained robust, and only slightly reduced relative to nonfailing hearts. We conclude that α1A-subtype-mediated inotropy could represent a significant source of inotropic support in the human heart. Furthermore, the α1A-subtype remains functional in myocardium from failing human hearts and thus, might be a therapeutic target to support cardioprotective effects in patients with heart failure.
Collapse
Affiliation(s)
- Paul M.L. Janssen
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, Ohio
| | - Benjamin D. Canan
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, Ohio
| | - Ahmet Kilic
- Department of Surgery, The Ohio State University, Columbus, Ohio
| | - Bryan A Whitson
- Department of Surgery, The Ohio State University, Columbus, Ohio
| | - Anthony J. Baker
- Veterans Affairs Medical Center, San Francisco, and Department of Medicine, Univ. Calif. San Francisco, San Francisco
| |
Collapse
|
13
|
Carreón-Garcidueñas M, Godínez-Hernández D, Alvarado-Gómez N, Ortega-Varela LF, Cervantes-Durán C, Gauthereau-Torres MY. Participation of voltage-gated sodium and calcium channels in the acute cardiac effects of toluene. Toxicol Mech Methods 2018; 28:670-677. [PMID: 29925288 DOI: 10.1080/15376516.2018.1491664] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Inhaling solvents can lead to occurrence of cardiac arrhythmias and sudden sniffing death. Mechanisms related to this phenomenon are not fully understood. The purpose of this study was to investigate the effect of acute toluene exposure on heart reactivity to epinephrine and the participation of voltage-gated sodium and calcium channels. We found that acute toluene exposure increased perfusion pressure, left ventricular developed pressure, and heart rate. These actions were inhibited by lidocaine and nifedipine. Our results suggest that acute toluene exposure modify voltage-gated sodium and calcium channel function and expression likely due to a cardiac adrenergic mechanism and these effects could be participating, at least in part, in the presence of cardiac arrhythmias. To our best knowledge, this is the first report to establish a direct participation of voltage-gated Na+ and Ca2+ channels, toluene and epinephrine on cardiac function in rats.
Collapse
Affiliation(s)
- M Carreón-Garcidueñas
- a Instituto de Investigaciones Químico Biológicas , Universidad Michoacana de San Nicolás de Hidalgo, Francisco J. Mújica s/n, Edif. B-3, Ciudad Universitaria , Morelia , Mexico
| | - D Godínez-Hernández
- a Instituto de Investigaciones Químico Biológicas , Universidad Michoacana de San Nicolás de Hidalgo, Francisco J. Mújica s/n, Edif. B-3, Ciudad Universitaria , Morelia , Mexico
| | - N Alvarado-Gómez
- a Instituto de Investigaciones Químico Biológicas , Universidad Michoacana de San Nicolás de Hidalgo, Francisco J. Mújica s/n, Edif. B-3, Ciudad Universitaria , Morelia , Mexico
| | - L F Ortega-Varela
- b Escuela de Enfermería y Salud Pública , Universidad Michoacana de San Nicolás de Hidalgo , Morelia , Mexico
| | - C Cervantes-Durán
- c Facultad de Ciencias Médicas y Biológicas "Dr. Ignacio Chávez" , Universidad Michoacana de San Nicolás de Hidalgo, Dr. Rafael Carrillo esquina Dr. Salvador González Herrejón, Col. Bosque Cuauhtémoc , Morelia , Mexico
| | - M Y Gauthereau-Torres
- c Facultad de Ciencias Médicas y Biológicas "Dr. Ignacio Chávez" , Universidad Michoacana de San Nicolás de Hidalgo, Dr. Rafael Carrillo esquina Dr. Salvador González Herrejón, Col. Bosque Cuauhtémoc , Morelia , Mexico
| |
Collapse
|
14
|
Abstract
There are 2 α1-ARs on cardiac myocytes: α1A and α1B. α1A adrenergic receptors serve important cardioprotective roles and do not mediate cardiac hypertrophy. Dabuzalgron, an oral α1A-AR agonist developed for the treatment of urinary incontinence and tolerated well in Phase 2 clinical trials, protects against doxorubicin-induced cardiotoxicity in vivo. Dabuzalgron enhances contractile function, regulates transcription of genes related to energy production and mitochondrial function, and preserves myocardial ATP content after doxorubicin. Activation of α1A-ARs on cardiomyocytes protects against doxorubicin cytotoxicity and enhances mitochondrial function in vitro. These cytoprotective effects likely are mediated by activation of ERK 1/2. Future studies will explore whether dabuzalgron, a well-tolerated oral α1A-AR agonist, might be repurposed to treat heart failure.
Alpha-1 adrenergic receptors (α1-ARs) play adaptive and protective roles in the heart. Dabuzalgron is an oral selective α1A-AR agonist that was well tolerated in multiple clinical trials of treatment for urinary incontinence, but has never been used to treat heart disease in humans or animal models. In this study, the authors administered dabuzalgron to mice treated with doxorubicin (DOX), a widely used chemotherapeutic agent with dose-limiting cardiotoxicity that can lead to heart failure (HF). Dabuzalgron protected against DOX-induced cardiotoxicity, likely by preserving mitochondrial function. These results suggest that activating cardiac α1A-ARs with dabuzalgron, a well-tolerated oral agent, might represent a novel approach to treating HF.
Collapse
|
15
|
Willis MS, Ilaiwy A, Montgomery MD, Simpson PC, Jensen BC. The alpha-1A adrenergic receptor agonist A61603 reduces cardiac polyunsaturated fatty acid and endocannabinoid metabolites associated with inflammation in vivo. Metabolomics 2016; 12:155. [PMID: 28533737 PMCID: PMC5437747 DOI: 10.1007/s11306-016-1097-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
INTRODUCTION Alpha-1-adrenergic receptors (α1-ARs) are G-protein coupled receptors (GPCRs) with three highly homologous subtypes (α1A, α1B, and α1D). Of these three subtypes, only the α1A and α1B are expressed in the heart. Multiple pre-clinical models of heart injury demonstrate cardioprotective roles for the α1A. Non-selective α1-AR activation promotes glycolysis in the heart, but the functional α1-AR subtype and broader metabolic effects have not been studied. OBJECTIVES Given the high metabolic demands of the heart and previous evidence indicating benefit from α1A activation, we chose to investigate the effects of α1A activation on the cardiac metabolome in vivo. METHODS Mice were treated for one week with a low, subpressor dose of A61603, a highly selective and potent α1A agonist. Cardiac tissue and serum were analyzed using a non-targeted metabolomics approach. RESULTS We identified previously unrecognized metabolic responses to α1A activation, most notably broad reduction in the abundance of polyunsaturated fatty acids (PUFAs) and endocannabinoids (ECs). CONCLUSION Given the well characterized roles of PUFAs and ECs in inflammatory pathways, these findings suggest a possible role for cardiac α1A-ARs in the regulation of inflammation and may offer novel insight into the mechanisms underlying the cardioprotective benefit of selective pharmacologic α1A activation.
Collapse
Affiliation(s)
- Monte S. Willis
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC USA
- Department of Pathology & Laboratory Medicine, University of North Carolina, Chapel Hill, NC USA
- Department of Pharmacology, University of North Carolina, Chapel Hill, NC USA
| | - Amro Ilaiwy
- Sarah W. Stedman Nutrition and Metabolism Center, Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA
- Division of Endocrinology, Metabolism, and Nutrition, Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | | | - Paul C. Simpson
- VA Medical Center and University of California, San Francisco, CA, USA
| | - Brian C. Jensen
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC USA
- Department of Internal Medicine, Division of Cardiology University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
16
|
Abstract
Although convention dictates that G protein-coupled receptors localize to and signal at the plasma membrane, accumulating evidence suggests that G protein-coupled receptors localize to and signal at intracellular membranes, most notably the nucleus. In fact, there is now significant evidence indicating that endogenous alpha-1 adrenergic receptors (α1-ARs) localize to and signal at the nuclei in adult cardiac myocytes. Cumulatively, the data suggest that α1-ARs localize to the inner nuclear membrane, activate intranuclear signaling, and regulate physiologic function in adult cardiac myocytes. Although α1-ARs signal through Gαq, unlike other Gq-coupled receptors, α1-ARs mediate important cardioprotective functions including adaptive/physiologic hypertrophy, protection from cell death (survival signaling), positive inotropy, and preconditioning. Also unlike other Gq-coupled receptors, most, if not all, functional α1-ARs localize to the nuclei in adult cardiac myocytes, as opposed to the sarcolemma. Together, α1-AR nuclear localization and cardioprotection might suggest a novel model for compartmentalization of Gq-coupled receptor signaling in which nuclear Gq-coupled receptor signaling is cardioprotective.
Collapse
|
17
|
Cotecchia S, Del Vescovo CD, Colella M, Caso S, Diviani D. The alpha1-adrenergic receptors in cardiac hypertrophy: signaling mechanisms and functional implications. Cell Signal 2015; 27:1984-93. [PMID: 26169957 DOI: 10.1016/j.cellsig.2015.06.009] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 06/22/2015] [Accepted: 06/30/2015] [Indexed: 01/05/2023]
Abstract
Cardiac hypertrophy is a complex remodeling process of the heart induced by physiological or pathological stimuli resulting in increased cardiomyocyte size and myocardial mass. Whereas cardiac hypertrophy can be an adaptive mechanism to stressful conditions of the heart, prolonged hypertrophy can lead to heart failure which represents the primary cause of human morbidity and mortality. Among G protein-coupled receptors, the α1-adrenergic receptors (α1-ARs) play an important role in the development of cardiac hypertrophy as demonstrated by numerous studies in the past decades, both in primary cardiomyocyte cultures and genetically modified mice. The results of these studies have provided evidence of a large variety of α1-AR-induced signaling events contributing to the defining molecular and cellular features of cardiac hypertrophy. Recently, novel signaling mechanisms have been identified and new hypotheses have emerged concerning the functional role of the α1-adrenergic receptors in the heart. This review will summarize the main signaling pathways activated by the α1-AR in the heart and their functional implications in cardiac hypertrophy.
Collapse
Affiliation(s)
- Susanna Cotecchia
- Dipartimento di Bioscienze, Biotecnologie e Biofarmaceutica, Università di Bari, Via Orabona 4, 70125 Bari, Italy.
| | - Cosmo Damiano Del Vescovo
- Department de Pharmacologie et de de Toxicologie, Université de Lausanne, Rue du Bugnon 27, 1005, Lausanne, Switzerland
| | - Matilde Colella
- Dipartimento di Bioscienze, Biotecnologie e Biofarmaceutica, Università di Bari, Via Orabona 4, 70125 Bari, Italy
| | - Stefania Caso
- Dipartimento di Bioscienze, Biotecnologie e Biofarmaceutica, Università di Bari, Via Orabona 4, 70125 Bari, Italy; Department de Pharmacologie et de de Toxicologie, Université de Lausanne, Rue du Bugnon 27, 1005, Lausanne, Switzerland
| | - Dario Diviani
- Department de Pharmacologie et de de Toxicologie, Université de Lausanne, Rue du Bugnon 27, 1005, Lausanne, Switzerland
| |
Collapse
|
18
|
Alpha-1-adrenergic receptors in heart failure: the adaptive arm of the cardiac response to chronic catecholamine stimulation. J Cardiovasc Pharmacol 2014; 63:291-301. [PMID: 24145181 DOI: 10.1097/fjc.0000000000000032] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Alpha-1-adrenergic receptors (ARs) are G protein-coupled receptors activated by catecholamines. The alpha-1A and alpha-1B subtypes are expressed in mouse and human myocardium, whereas the alpha-1D protein is found only in coronary arteries. There are far fewer alpha-1-ARs than beta-ARs in the nonfailing heart, but their abundance is maintained or increased in the setting of heart failure, which is characterized by pronounced chronic elevation of catecholamines and beta-AR dysfunction. Decades of evidence from gain and loss-of-function studies in isolated cardiac myocytes and numerous animal models demonstrate important adaptive functions for cardiac alpha-1-ARs to include physiological hypertrophy, positive inotropy, ischemic preconditioning, and protection from cell death. Clinical trial data indicate that blocking alpha-1-ARs is associated with incident heart failure in patients with hypertension. Collectively, these findings suggest that alpha-1-AR activation might mitigate the well-recognized toxic effects of beta-ARs in the hyperadrenergic setting of chronic heart failure. Thus, exogenous cardioselective activation of alpha-1-ARs might represent a novel and viable approach to the treatment of heart failure.
Collapse
|
19
|
Wu SC, Dahl EF, Wright CD, Cypher AL, Healy CL, O'Connell TD. Nuclear localization of a1A-adrenergic receptors is required for signaling in cardiac myocytes: an “inside-out” a1-AR signaling pathway. J Am Heart Assoc 2014; 3:e000145. [PMID: 24772522 PMCID: PMC4187477 DOI: 10.1161/jaha.113.000145] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Background Recent studies indicate that α1‐adrenergic receptors (α1‐ARs) are cardioprotective by preventing cardiac myocyte death and augmenting contractility in heart failure. Although G‐protein‐coupled receptors are assumed to localize to and signal at the plasma membrane, we previously demonstrated that endogenous α1‐ARs localize to the nuclei in adult cardiac myocytes. However, the functional consequence of this nuclear localization remains unclear. Here, we attempted to reconcile nuclear localization of α1‐ARs with their physiologic function by examining α1‐AR‐induced contractility in adult cardiac myocytes. Methods and Results By measuring shortening in unloaded, cultured adult cardiac myocytes, we found that the α1A‐subtype regulated contractility through phosphorylation of cardiac troponin I (cTnI) at the protein kinase C (PKC) site, threonine 144. Reconstitution of an α1A‐subtype nuclear localization mutant in cardiac myocytes lacking α1‐ARs failed to rescue nuclear α1A‐mediated phosphorylation of cTnI and myocyte contractility. Leptomycin B, the nuclear export inhibitor, also blocked α1A‐mediated phosphorylation of cTnI. These data indicate that α1‐AR signaling originates in the nucleus. Consistent with these observations, we localized the α1A‐subtype to the inner nuclear membrane, identified PKCα, δ, and ε in the nucleus, and found that α1‐ARs activate PKCδ in nuclei isolated from adult cardiac myocytes. Finally, we found that a PKCδ nuclear localization mutant blunted α1‐induced phosphorylation of cTnI. Conclusions Together, our data identify a novel, “inside‐out” nuclear α1A‐subtype/PKCδ/cTnI‐signaling pathway that regulates contractile function in adult cardiac myocytes. Importantly, these data help resolve the discrepancy between nuclear localization of α1‐ARs and α1‐AR‐mediated physiologic function.
Collapse
Affiliation(s)
- Steven C. Wu
- Department of Integrative Biology and Physiology, The University of Minnesota, Minneapolis, MN (S.C.W., E.F.D., C.D.W., A.L.C., C.L.H., T.D.C.)
| | - Erika F. Dahl
- Department of Integrative Biology and Physiology, The University of Minnesota, Minneapolis, MN (S.C.W., E.F.D., C.D.W., A.L.C., C.L.H., T.D.C.)
| | - Casey D. Wright
- Department of Integrative Biology and Physiology, The University of Minnesota, Minneapolis, MN (S.C.W., E.F.D., C.D.W., A.L.C., C.L.H., T.D.C.)
- Novartis Animal Health US, Inc, 1447140th St, Larchwood, IA 51241
| | - Andrew L. Cypher
- Department of Integrative Biology and Physiology, The University of Minnesota, Minneapolis, MN (S.C.W., E.F.D., C.D.W., A.L.C., C.L.H., T.D.C.)
- Novartis Animal Health US, Inc, 1447140th St, Larchwood, IA 51241
| | - Chastity L. Healy
- Department of Integrative Biology and Physiology, The University of Minnesota, Minneapolis, MN (S.C.W., E.F.D., C.D.W., A.L.C., C.L.H., T.D.C.)
| | - Timothy D. O'Connell
- Department of Integrative Biology and Physiology, The University of Minnesota, Minneapolis, MN (S.C.W., E.F.D., C.D.W., A.L.C., C.L.H., T.D.C.)
| |
Collapse
|
20
|
Baker AJ. Adrenergic signaling in heart failure: a balance of toxic and protective effects. Pflugers Arch 2014; 466:1139-50. [PMID: 24623099 DOI: 10.1007/s00424-014-1491-5] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Revised: 02/24/2014] [Accepted: 02/26/2014] [Indexed: 10/25/2022]
Abstract
Heart failure with reduced ejection fraction involves activation of the sympathetic nervous system and chronic hyperactivation of the sympatho-adrenergic receptors (ARs) β-ARs and α1-ARs, which are thought to be cardiotoxic and worsen pathological remodeling and function. Concurrently, the failing heart manifests significant decreases in sympathetic nerve terminal density, decreased cardiac norepinephrine levels, and marked downregulation of β-AR abundance and signaling. Thus, a state of both feast and famine coexist with respect to the adrenergic state in heart failure. For the failing heart, the hyperadrenergic state is toxic. However, the role of hypoadrenergic mechanisms in the pathophysiology of heart failure is less clear. Cardiotoxic effects are known to arise from the β1-AR subtype, and use of β-AR blockers is a cornerstone of current heart failure therapy. However, cardioprotective effects arise from the β2-AR subtype that counteract hyperactive β1-AR signaling, but unfortunately, β2-AR cardioprotective signaling in heart failure is inhibited by β-AR blocker therapy. In contrast to current dogma, recent research shows β1-AR signaling can also be cardioprotective. Moreover, for some forms of heart failure, β2-AR signaling is cardiotoxic. Thus for both β-AR subtypes, there is a balance between cardiotoxic versus cardioprotective effects. In heart failure, stimulation of α1-ARs is widely thought to be cardiotoxic. However, also contrary to current dogma, recent research shows that α1-AR signaling is cardioprotective. Taken together, recent research identifies cardioprotective signaling arising from β1-AR, β2-AR, and α1-ARs. A goal for future therapies will to harness the protective effects of AR signaling while minimizing cardiotoxic effects. The trajectory of heart failure therapy changed radically from the previous and intuitive use of sympathetic agonists, which unfortunately resulted in greater mortality, to the current use of β-AR blockers, which initially seemed counterintuitive. As a cautionary note, if the slow adoption of beta-blocker therapy in heart failure is any guide, then new treatment strategies, especially counterintuitive therapies involving stimulating β-AR and α1-AR signaling, may take considerable time to develop and gain acceptance.
Collapse
Affiliation(s)
- Anthony J Baker
- Veterans Affairs Medical Center, San Francisco and Department of Medicine, University of California, Cardiology Division (111C), 4150 Clement St, San Francisco, CA, 94121, USA,
| |
Collapse
|
21
|
O'Connell TD, Jensen BC, Baker AJ, Simpson PC. Cardiac alpha1-adrenergic receptors: novel aspects of expression, signaling mechanisms, physiologic function, and clinical importance. Pharmacol Rev 2013; 66:308-33. [PMID: 24368739 DOI: 10.1124/pr.112.007203] [Citation(s) in RCA: 133] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Adrenergic receptors (AR) are G-protein-coupled receptors (GPCRs) that have a crucial role in cardiac physiology in health and disease. Alpha1-ARs signal through Gαq, and signaling through Gq, for example, by endothelin and angiotensin receptors, is thought to be detrimental to the heart. In contrast, cardiac alpha1-ARs mediate important protective and adaptive functions in the heart, although alpha1-ARs are only a minor fraction of total cardiac ARs. Cardiac alpha1-ARs activate pleiotropic downstream signaling to prevent pathologic remodeling in heart failure. Mechanisms defined in animal and cell models include activation of adaptive hypertrophy, prevention of cardiac myocyte death, augmentation of contractility, and induction of ischemic preconditioning. Surprisingly, at the molecular level, alpha1-ARs localize to and signal at the nucleus in cardiac myocytes, and, unlike most GPCRs, activate "inside-out" signaling to cause cardioprotection. Contrary to past opinion, human cardiac alpha1-AR expression is similar to that in the mouse, where alpha1-AR effects are seen most convincingly in knockout models. Human clinical studies show that alpha1-blockade worsens heart failure in hypertension and does not improve outcomes in heart failure, implying a cardioprotective role for human alpha1-ARs. In summary, these findings identify novel functional and mechanistic aspects of cardiac alpha1-AR function and suggest that activation of cardiac alpha1-AR might be a viable therapeutic strategy in heart failure.
Collapse
Affiliation(s)
- Timothy D O'Connell
- VA Medical Center (111-C-8), 4150 Clement St., San Francisco, CA 94121. ; or Dr. Timothy D. O'Connell, E-mail:
| | | | | | | |
Collapse
|
22
|
Ziyatdinova NI, Dementieva RE, Fashutdinov LI, Zefirov TL. Blockade of different subtypes of α(1)-adrenoceptors produces opposite effect on heart chronotropy in newborn rats. Bull Exp Biol Med 2013; 154:184-5. [PMID: 23330119 DOI: 10.1007/s10517-012-1906-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
We compared the effects of blockade of α(1A)-, α(1B)-, and α(1D)-subtypes of α(1)-adrenoceptors on the cardiac rhythm in newborn rats. Different responses of the heart were observed after blockade of several subtypes of α(1)-adrenoceptors. Administration of WB 4101, a selective blocker of α(1A)-adrenoceptors, increased heart rate, while blockade of α(1AD)-adrenoceptors with BMY 7378 decelerated of heart rhythm. Blockade of α(1B)-adrenoceptors with chloroethylclonidine produced no significant effects on heart chronotropy.
Collapse
Affiliation(s)
- N I Ziyatdinova
- Department of Anatomy, Physiology, and Human Health Protection, Kazan (Privolzhskii) Federal University, Russia
| | | | | | | |
Collapse
|
23
|
|
24
|
Jensen BC, O'Connell TD, Simpson PC. Alpha-1-adrenergic receptors: targets for agonist drugs to treat heart failure. J Mol Cell Cardiol 2010; 51:518-28. [PMID: 21118696 DOI: 10.1016/j.yjmcc.2010.11.014] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2010] [Accepted: 11/12/2010] [Indexed: 12/19/2022]
Abstract
Evidence from cell, animal, and human studies demonstrates that α1-adrenergic receptors mediate adaptive and protective effects in the heart. These effects may be particularly important in chronic heart failure, when catecholamine levels are elevated and β-adrenergic receptors are down-regulated and dysfunctional. This review summarizes these data and proposes that selectively activating α1-adrenergic receptors in the heart might represent a novel and effective way to treat heart failure. This article is part of a special issue entitled "Key Signaling Molecules in Hypertrophy and Heart Failure."
Collapse
Affiliation(s)
- Brian C Jensen
- Cardiology Division, VA Medical Center, San Francisco, CA, USA.
| | | | | |
Collapse
|
25
|
Jensen BC, Swigart PM, Montgomery MD, Simpson PC. Functional alpha-1B adrenergic receptors on human epicardial coronary artery endothelial cells. Naunyn Schmiedebergs Arch Pharmacol 2010; 382:475-82. [PMID: 20857090 PMCID: PMC2991196 DOI: 10.1007/s00210-010-0558-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2010] [Accepted: 08/30/2010] [Indexed: 11/25/2022]
Abstract
Alpha-1-adrenergic receptors (α1-ARs) regulate coronary arterial blood flow by binding catecholamines, norepinephrine (NE), and epinephrine (EPI), causing vasoconstriction when the endothelium is disrupted. Among the three α1-AR subtypes (α1A, α1B, and α1D), the α1D subtype predominates in human epicardial coronary arteries and is functional in human coronary smooth muscle cells (SMCs). However, the presence or function of α1-ARs on human coronary endothelial cells (ECs) is unknown. Here we tested the hypothesis that human epicardial coronary ECs express functional α1-ARs. Cultured human epicardial coronary artery ECs were studied using quantitative real-time reverse transcription polymerase chain reaction, radioligand binding, immunoblot, and (3)H-thymidine incorporation. The α1B-subtype messenger ribonucleic acid (mRNA) was predominant in cultured human epicardial coronary ECs (90-95% of total α1-AR mRNA), and total α1-AR binding density in ECs was twice that in coronary SMCs. Functionally, NE and EPI through the α1B subtype activated extracellular signal-regulated kinase (ERK) in ECs, stimulated phosphorylation of EC endothelial nitric oxide synthase (eNOS), and increased deoxyribonucleic acid (DNA) synthesis. These results are the first to demonstrate α1-ARs on human coronary ECs and indicate that the α1B subtype is predominant. Our findings provide another potential mechanism for adverse cardiac effects of drug antagonists that nonselectively inhibit all three α1-AR subtypes.
Collapse
Affiliation(s)
- Brian C Jensen
- Cardiology Division, VA Medical Center, University of California, San Francisco, San Francisco, CA, USA
| | | | | | | |
Collapse
|
26
|
Wu X, Lin S, Zhu C, Yue Z, Yu Y, Zhao F, Liu B, Dai J, Shi J. Homo- and heptanor-sterols and tremulane sesquiterpenes from cultures of Phellinus igniarius. JOURNAL OF NATURAL PRODUCTS 2010; 73:1294-300. [PMID: 20583752 DOI: 10.1021/np100216k] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Four steroids, a homopregnene (1) and three heptanorergosterane derivatives (2-4), nine tremulane sesquiterpenes (5-13), and 18 known compounds have been isolated from cultures of the fungus Phellinus igniarius. Their structures and absolute configurations were elucidated by spectroscopic data analysis. In preliminary in vitro assays, at 10(-5) M, compounds 8, 9, 13, and 3beta-hydroxy-11,12-O-isopropyldrimene (14) showed significant vascular-relaxing activities against phenylephrine-induced vasoconstriction with relaxing rates of 35.7%, 45.4%, 46.6%, and 32.1%, respectively, as compared with the blank control.
Collapse
Affiliation(s)
- Xiuli Wu
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College (Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education), Beijing, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Yeh CC, Li H, Malhotra D, Turcato S, Nicholas S, Tu R, Zhu BQ, Cha J, Swigart PM, Myagmar BE, Baker AJ, Simpson PC, Mann MJ. Distinctive ERK and p38 signaling in remote and infarcted myocardium during post-MI remodeling in the mouse. J Cell Biochem 2010; 109:1185-91. [PMID: 20186881 DOI: 10.1002/jcb.22498] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Global activation of MAP kinases has been reported in both human and experimental heart failure. Chronic remodeling of the surviving ventricular wall after myocardial infarction (MI) involves both myocyte loss and fibrosis; we hypothesized that this cardiomyopathy involves differential shifts in pro- and anti-apoptotic MAP kinase signaling in cardiac myocyte (CM) and non-myocyte. Cardiomyopathy after coronary artery ligation in mice was characterized by echocardiography, ex vivo Langendorff preparation, histologic analysis and measurements of apoptosis. Phosphorylation (activation) of signaling molecules was analyzed by Western blot, ELISA and immunohistochemistry. Post-MI remodeling involved dramatic changes in the phosphorylation of both stress-activated MAP (SAP) kinase p38 as well as ERK, a known mediator of cell survival, but not of SAP kinase JNK or the anti-apoptotic mediator of PI3K, Akt. Phosphorylation of p38 rose early after MI in the infarct, whereas a more gradual rise in the remote myocardium accompanied a rise in apoptosis in that region. In both areas, ERK phosphorylation was lowest early after MI and rose steadily thereafter, though infarct phosphorylation was consistently higher. Immunostaining of p-ERK localized to fibrotic areas populated primarily by non-myocytes, whereas staining of p38 phosphorylation was stronger in areas of progressive CM apoptosis. Relative segregation of CMs and non-myocytes in different regions of the post-MI myocardium revealed signaling patterns that imply cell type-specific changes in pro- and anti-apoptotic MAP kinase signaling. Prevention of myocyte loss and of LV remodeling after MI may therefore require cell type-specific manipulation of p38 and ERK activation.
Collapse
Affiliation(s)
- Che-Chung Yeh
- Division of Cardiothoracic Surgery, University of California, San Francisco VA Medical Center, San Francisco, CA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Methven L, Simpson PC, McGrath JC. Alpha1A/B-knockout mice explain the native alpha1D-adrenoceptor's role in vasoconstriction and show that its location is independent of the other alpha1-subtypes. Br J Pharmacol 2010; 158:1663-75. [PMID: 19888965 DOI: 10.1111/j.1476-5381.2009.00462.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
BACKGROUND AND PURPOSE Theoretically, three alpha(1)-adrenoceptor subtypes can interact at the signalling level to alter vascular contraction or at the molecular level to alter each other's cellular location. The alpha(1A/B)-adrenoceptor knockout mouse (alpha(1A/B)-KO) was used to study the isolated alpha(1D)-adrenoceptor to consider these potential interactions in native tissue. EXPERIMENTAL APPROACH Pharmacological analysis of carotid and mesenteric arteries employed wire myography and fluorescent ligand binding (alpha(1)-adrenoceptor ligand BODIPY FL-prazosin, QAPB). KEY RESULTS alpha(1A/B)-KO carotid had clear alpha(1D)-adrenoceptor-induced contractions. In WT carotid alpha(1D)-adrenoceptor dominated but all three alpha(1)-subtypes participated. alpha(1A/B)-KO mesenteric had alpha(1D)-adrenoceptor responses with high sensitivity and small maximum, explaining how alpha(1D)-adrenoceptor could determine agonist sensitivity in WT. In both arteries alpha(1A/B)-KO fluorescence levels were reduced but pharmacologically more consistent with 'pure'alpha(1D)-adrenoceptors. alpha(1D)-Adrenoceptor binding in alpha(1A/B)-KO was observed on the cell surface and intracellularly and was present in a high proportion of smooth-muscle cells in both strains, regardless of artery type. CONCLUSIONS AND IMPLICATIONS 'Pure'alpha(1D)-adrenoceptor pharmacology in alpha(1A/B)-KO provides a quantitative standard. Functionally, the alpha(1D)- and alpha(1A)-adrenoceptors produce additive responses and do not significantly compensate for each other. alpha(1D)-Adrenoceptor contributes to sensitivity even in resistance arteries. In alpha(1A/B)-KO, the loss of alpha(1A)- and alpha(1B)-adrenoceptors is reflected by a general decrease in fluorescence, but similar binding distribution to WT indicates that the alpha(1D)-adrenoceptor location in native smooth-muscle cells is not influenced by other alpha(1)-adrenoceptors. Equivalent levels of receptors did not correspond to equivalent responses. In conclusion, alpha(1)-subtypes do not interact but provide independent alternative signals for vascular regulation.
Collapse
Affiliation(s)
- L Methven
- Integrative and Systems Biology, Faculty of Biomedical and Life Sciences, University of Glasgow, Glasgow, UK.
| | | | | |
Collapse
|
29
|
Reply. J Am Coll Cardiol 2010. [DOI: 10.1016/j.jacc.2009.10.050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
30
|
Jensen BC, Swigart PM, Laden ME, DeMarco T, Hoopes C, Simpson PC. The alpha-1D Is the predominant alpha-1-adrenergic receptor subtype in human epicardial coronary arteries. J Am Coll Cardiol 2009; 54:1137-45. [PMID: 19761933 DOI: 10.1016/j.jacc.2009.05.056] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2008] [Revised: 05/13/2009] [Accepted: 05/19/2009] [Indexed: 11/15/2022]
Abstract
OBJECTIVES The goal was to identify alpha-1-adrenergic receptor (AR) subtypes in human coronary arteries. BACKGROUND The alpha1-ARs regulate human coronary blood flow. The alpha1-ARs exist as 3 molecular subtypes, alpha1A, alpha1B, and alpha1D, and the alpha1D subtype mediates coronary vasoconstriction in the mouse. However, the alpha1A is thought to be the only subtype in human coronary arteries. METHODS We obtained human epicardial coronary arteries and left ventricular (LV) myocardium from 19 transplant recipients and 6 unused donors (age 19 to 70 years; 68% male; 32% with coronary artery disease). We cultured coronary rings and human coronary smooth muscle cells. We assayed alpha1- and beta-AR subtype messenger ribonucleic acid (mRNA) by quantitative real-time reverse transcription polymerase chain reaction and subtype proteins by radioligand binding and extracellular signal-regulated kinase (ERK) activation. RESULTS The alpha1D subtype was 85% of total coronary alpha1-AR mRNA and 75% of total alpha1-AR protein, and alpha1D stimulation activated ERK. In contrast, the alpha1D was low in LV myocardium. Total coronary alpha1-AR levels were one-third of beta-ARs, which were 99% the beta2 subtype. CONCLUSIONS The alpha1D subtype is predominant and functional in human epicardial coronary arteries, whereas the alpha1A and alpha1B are present at very low levels. This distribution is similar to the mouse, where myocardial alpha1A- and alpha1B-ARs mediate beneficial functional responses and coronary alpha1Ds mediate vasoconstriction. Thus, alpha1D-selective antagonists might mediate coronary vasodilation, without the negative cardiac effects of nonselective alpha1-AR antagonists in current use. Furthermore, it could be possible to selectively activate beneficial myocardial alpha1A- and/or alpha1B-AR signaling without causing coronary vasoconstriction.
Collapse
|
31
|
Methven L, McBride M, Wallace GA, McGrath JC. The alpha 1B/D-adrenoceptor knockout mouse permits isolation of the vascular alpha 1A-adrenoceptor and elucidates its relationship to the other subtypes. Br J Pharmacol 2009; 158:209-24. [PMID: 19572943 PMCID: PMC2795267 DOI: 10.1111/j.1476-5381.2009.00269.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2008] [Revised: 01/30/2009] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND AND PURPOSE Mesenteric and carotid arteries from the alpha(1B/D)-adrenoceptor knockout (alpha(1B/D)-KO) were employed to isolate alpha(1A)-adrenoceptor pharmacology and location and to reveal these features in the wild-type (WT) mouse. EXPERIMENTAL APPROACH Functional pharmacology by wire myography and receptor localization by confocal microscopy, using the fluorescent alpha(1)-adrenoceptor ligand BODIPY FL-Prazosin (QAPB), on mesenteric (an 'alpha(1A)-adrenoceptor' tissue) and carotid (an 'alpha(1D)-adrenoceptor' tissue) arteries. KEY RESULTS Alpha(1B/D)-KO mesenteric arteries showed straightforward alpha(1A)-adrenoceptor agonist/antagonist pharmacology. WT had complex pharmacology with alpha(1A)- and alpha(1D)-adrenoceptor components. alpha(1B/D)-KO had a larger alpha(1A)-adrenoceptor response suggesting compensatory up-regulation: no increase in fluorescent ligand binding suggests up-regulation of signalling. alpha(1B/D)-KO carotid arteries had low efficacy alpha(1A)-adrenoceptor responses. WT had complex pharmacology consistent with co-activation of all three subtypes. Fluorescent binding had straightforward alpha(1A)-adrenoceptor characteristics in both arteries of alpha(1B/D)-KO. Fluorescent binding varied between cells in relative intracellular and surface distribution. Total fluorescence was reduced in the alpha(1B/D)-KO due to fewer smooth muscle cells showing fluorescent binding. WT binding was greater and sensitive to alpha(1A)- and alpha(1D)-adrenoceptor antagonists. CONCLUSIONS AND IMPLICATIONS The straightforward pharmacology and fluorescent binding in the alpha(1B/D)-KO was used to interpret the properties of the alpha(1A)-adrenoceptor in the WT. Reduced total fluorescence in alpha(1B/D)-KO arteries, despite a clear difference in the functionally dominant subtype, indicates that measurement of receptor protein is unlikely to correlate with function. Fewer cells bound QAPB in the alpha(1B/D)-KO suggesting different cellular phenotypes of alpha(1A)-adrenoceptor exist. The alpha(1B/D)-KO provides robust assays for the alpha(1A)-adrenoceptor and takes us closer to understanding multi-receptor subtype interactions.
Collapse
MESH Headings
- Adrenergic Agonists/pharmacology
- Adrenergic Antagonists/pharmacology
- Adrenergic alpha-1 Receptor Agonists
- Animals
- Carotid Arteries/drug effects
- Carotid Arteries/physiology
- Male
- Mesenteric Arteries/drug effects
- Mesenteric Arteries/physiology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/physiology
- Protein Subunits/classification
- Protein Subunits/physiology
- Receptors, Adrenergic, alpha-1/classification
- Receptors, Adrenergic, alpha-1/deficiency
- Receptors, Adrenergic, alpha-1/physiology
Collapse
Affiliation(s)
- L Methven
- Integrative and Systems Biology, Faculty of Biomedical and Life Sciences, University of Glasgow, Glasgow, UK.
| | | | | | | |
Collapse
|
32
|
Jensen BC, Swigart PM, De Marco T, Hoopes C, Simpson PC. {alpha}1-Adrenergic receptor subtypes in nonfailing and failing human myocardium. Circ Heart Fail 2009; 2:654-63. [PMID: 19919991 DOI: 10.1161/circheartfailure.108.846212] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND alpha1-adrenergic receptors (alpha1-ARs) play adaptive roles in the heart and protect against the development of heart failure. The 3 alpha1-AR subtypes, alpha1A, alpha1B, and alpha1D, have distinct physiological roles in mouse heart, but very little is known about alpha1 subtypes in human heart. Here, we test the hypothesis that the alpha1A and alpha1B subtypes are present in human myocardium, similar to the mouse, and are not downregulated in heart failure. METHODS AND RESULTS Hearts from transplant recipients and unused donors were failing (n=12; mean ejection fraction, 24%) or nonfailing (n=9; mean ejection fraction, 59%) and similar in age ( approximately 44 years) and sex ( approximately 70% male). We measured the alpha1-AR subtypes in multiple regions of both ventricles by quantitative real-time reverse-transcription polymerase chain reaction and radioligand binding. All 3 alpha1-AR subtype mRNAs were present, and alpha1A mRNA was most abundant ( approximately 65% of total alpha1-AR mRNA). However, only alpha1A and alpha1B binding were present, and the alpha1B was most abundant (60% of total). In failing hearts, alpha1A and alpha1B binding was not downregulated, in contrast with beta1-ARs. CONCLUSIONS Our data show for the first time that the alpha1A and alpha1B subtypes are both present in human myocardium, but alpha1D binding is not, and the alpha1 subtypes are not downregulated in heart failure. Because alpha1 subtypes in the human heart are similar to those in the mouse, where adaptive and protective effects of alpha1 subtypes are most convincing, it might become feasible to treat heart failure with a drug targeting the alpha1A and/or alpha1B.
Collapse
Affiliation(s)
- Brian C Jensen
- Cardiology Section and Research Service, San Francisco VA Medical Center, San Francisco, Calif, USA
| | | | | | | | | |
Collapse
|
33
|
Cao XJ, Li YF. Alteration of messenger RNA and protein levels of cardiac alpha(1)-adrenergic receptor and angiotensin II receptor subtypes during aging in rats. Can J Cardiol 2009; 25:415-20. [PMID: 19584972 DOI: 10.1016/s0828-282x(09)70509-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
BACKGROUND Structural and functional alterations in the senescent heart have been associated with an activated sympathetic nervous system and a regional cardiac renin-angiotensin system. To date, however, limited information related to their expression alteration during the whole procress of growth and development has been reported. OBJECTIVES To examine the expression of alpha(1)-adrenergic receptor (alpha(1)-AR) and angiotensin II receptor (ATR) subtypes in the left ventricle of hearts from young adult, middle-aged, presenescent and senescent rats. METHODS Semiquantitative reverse transcriptase polymerase chain reaction and Western blot were used to quantitate the messenger RNA and protein of alpha(1)-AR and ATR subtypes, respectively, in the left ventricles of three- (young adult), 12- (middle age), 18- (presenescent) and 24-month-old (senescent) Wistar rats. RESULTS AND CONCLUSIONS alpha(1A)-AR expression decreased gradually with age, and alpha(1D)-AR expression was repressed in middle age and presenescence, while the expression of alpha(1B)-AR remained unchanged during senescence. AT(1)R expression was unaffected by aging from young adulthood to presenescence, but exhibited a remarkable upregulation in senescence. There were no significant discrepancies of cardiac AT(2)R expression among the four age groups, but both messenger RNA and protein had a tendency to upregulate during aging. The results suggest that there are considerable changes of expression of cardiac alpha(1)-AR and ATR subtypes during growth and development. The change of cardiac alpha(1)-AR and ATR expression during aging is a protective response to senescence by keeping normal myocardial contractility, while the upregulation of AT(1)R and AT(2)R promotes age-related myocardium hypertrophy and cardiac remodelling.
Collapse
Affiliation(s)
- Xiao-jing Cao
- The Cardiology Department, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | | |
Collapse
|
34
|
Wright CD, Chen Q, Baye NL, Huang Y, Healy CL, Kasinathan S, O'Connell TD. Nuclear alpha1-adrenergic receptors signal activated ERK localization to caveolae in adult cardiac myocytes. Circ Res 2008; 103:992-1000. [PMID: 18802028 DOI: 10.1161/circresaha.108.176024] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
We previously identified an alpha1-AR-ERK (alpha1A-adrenergic receptor-extracellular signal-regulated kinase) survival signaling pathway in adult cardiac myocytes. Here, we investigated localization of alpha1-AR subtypes (alpha1A and alpha1B) and how their localization influences alpha1-AR signaling in cardiac myocytes. Using binding assays on myocyte subcellular fractions or a fluorescent alpha1-AR antagonist, we localized endogenous alpha1-ARs to the nucleus in wild-type adult cardiac myocytes. To clarify alpha1 subtype localization, we reconstituted alpha1 signaling in cultured alpha1A- and alpha1B-AR double knockout cardiac myocytes using alpha1-AR-green fluorescent protein (GFP) fusion proteins. Similar to endogenous alpha1-ARs and alpha1A- and alpha1B-GFP colocalized with LAP2 at the nuclear membrane. alpha1-AR nuclear localization was confirmed in vivo using alpha1-AR-GFP transgenic mice. The alpha1-signaling partners Galphaq and phospholipase Cbeta1 also colocalized with alpha1-ARs only at the nuclear membrane. Furthermore, we observed rapid catecholamine uptake mediated by norepinephrine-uptake-2 and found that alpha1-mediated activation of ERK was not inhibited by a membrane impermeant alpha1-blocker, suggesting alpha1 signaling is initiated at the nucleus. Contrary to prior studies, we did not observe alpha1-AR localization to caveolae, but we found that alpha1-AR signaling initiated at the nucleus led to activated ERK localized to caveolae. In summary, our results show that nuclear alpha1-ARs transduce signals to caveolae at the plasma membrane in cardiac myocytes.
Collapse
Affiliation(s)
- Casey D Wright
- Cardiovascular Research Center, Sanford Research/USD, Sioux Falls, SD 57105, USA
| | | | | | | | | | | | | |
Collapse
|
35
|
McCloskey DT, Turcato S, Wang GY, Turnbull L, Zhu BQ, Bambino T, Nguyen AP, Lovett DH, Nissenson RA, Karliner JS, Baker AJ. Expression of a Gi-coupled receptor in the heart causes impaired Ca2+ handling, myofilament injury, and dilated cardiomyopathy. Am J Physiol Heart Circ Physiol 2007; 294:H205-12. [PMID: 17965283 DOI: 10.1152/ajpheart.00829.2007] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Increased signaling by G(i)-coupled receptors has been implicated in dilated cardiomyopathy. To investigate the mechanisms, we used transgenic mice that develop dilated cardiomyopathy after conditional expression of a cardiac-targeted G(i)-coupled receptor (Ro1). Activation of G(i) signaling by the Ro1 agonist spiradoline caused decreased cellular cAMP levels and bradycardia in Langendorff-perfused hearts. However, acute termination of Ro1 signaling with the antagonist nor-binaltorphimine did not reverse the Ro1-induced contractile dysfunction, indicating that Ro1 cardiomyopathy was not due to acute effects of receptor signaling. Early after initiation of Ro1 expression, there was a 40% reduction in the abundance of the sarcoplasmic reticulum Ca(2+)-ATPase (P < 0.05); thereafter, there was progressive impairment of both Ca(2+) handling and force development assessed with ventricular trabeculae. Six weeks after initiation of Ro1 expression, systolic Ca(2+) concentration was reduced to 0.61 +/- 0.08 vs. 0.91 +/- 0.07 microM for control (n = 6-8; P < 0.05), diastolic Ca(2+) concentration was elevated to 0.41 +/- 0.07 vs. 0.23 +/- 0.06 microM for control (n = 6-8; P < 0.01), and the decline phase of the Ca(2+) transient (time from peak to 50% decline) was slowed to 0.25 +/- 0.02 s vs. 0.13 +/- 0.02 s for control (n = 6-8; P < 0.01). Early after initiation of Ro1 expression, there was a ninefold elevation of matrix metalloproteinase-2 (P < 0.01), which is known to cause myofilament injury. Consistent with this, 6 wk after initiation of Ro1 expression, Ca(2+)-saturated myofilament force in skinned trabeculae was reduced to 21 +/- 2 vs. 38 +/- 0.1 mN/mm(2) for controls (n = 3; P < 0.01). Furthermore, electron micrographs revealed extensive myofilament damage. These findings may have implications for some forms of human heart failure in which increased activity of G(i)-coupled receptors leads to impaired Ca(2+) handling and myofilament injury, contributing to impaired ventricular pump function and heart failure.
Collapse
Affiliation(s)
- Diana T McCloskey
- VA Medical Center, Cardiology Division (111C ), University of California-San Francisco, 4150 Clement St., San Francisco, CA 94121, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Luo DL, Gao J, Fan LL, Tang Y, Zhang YY, Han QD. Receptor subtype involved in alpha 1-adrenergic receptor-mediated Ca2+ signaling in cardiomyocytes. Acta Pharmacol Sin 2007; 28:968-74. [PMID: 17588332 DOI: 10.1111/j.1745-7254.2007.00605.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
AIM The enhancement of intracellular Ca2+ signaling in response to alpha 1-adrenergic receptor (alpha 1-AR) stimulation is an essential signal transduction event in the regulation of cardiac functions, such as cardiac growth, cardiac contraction, and cardiac adaptation to various situations. The present study was intended to determine the role(s) of the alpha 1-AR subtype(s) in mediating this response. METHODS We evaluated the effects of subtype-specific agonists and antagonists of the alpha 1- AR on the intracellular Ca2+ signaling of neonatal rat ventricular myocytes using a confocal microscope. RESULTS After being cultured for 48 h, the myocytes exhibited spontaneous local Ca2+ release, sparks, and global Ca2+ transients. The activation of the alpha 1-AR with phenylephrine, a selective agonist of the alpha 1-AR, dose-dependently increased the frequency of Ca2+ transients with an EC50 value of 2.3 micromol/L. Blocking the alpha 1A-AR subtype with 5-methylurapidil (5-Mu) inhibited the stimulatory effect of phenylephrine with an IC(50) value of 6.7 nmol/L. In contrast, blockade of the alpha 1B-AR and alpha 1D-AR subtypes with chloroethylclonidine and BMY 7378, respectively, did not affect the phenylephrine effect. Similarly, the local Ca2+ spark numbers were also increased by the activation of the alpha 1-AR, and this effect could be abolished selectively by 5-Mu. More importantly, A61603, a novel selective alpha 1A-AR agonist, mimicked the effects of phenylephrine, but with more potency (EC(50) value =6.9 nmol/L) in the potentiation of Ca2+ transients, and blockade of the alpha 1A-AR by 5-Mu caused abolishment of its effects. CONCLUSION These results indicate that alpha 1-adrenergic stimulation of intracellular Ca2+ activity is mediated selectively by the alpha 1A-AR.
Collapse
Affiliation(s)
- Da-li Luo
- Department of Pharmacology, School of Chemical Biology and Pharmaceutical Sciences, Capital University of Medical Sciences, Beijing 100069, China.
| | | | | | | | | | | |
Collapse
|
37
|
Brum PC, Rolim NPL, Bacurau AVN, Medeiros A. Neurohumoral activation in heart failure: the role of adrenergic receptors. AN ACAD BRAS CIENC 2007; 78:485-503. [PMID: 16936938 DOI: 10.1590/s0001-37652006000300009] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2005] [Accepted: 11/04/2005] [Indexed: 01/08/2023] Open
Abstract
Heart failure (HF) is a common endpoint for many forms of cardiovascular disease and a significant cause of morbidity and mortality. The development of end-stage HF often involves an initial insult to the myocardium that reduces cardiac output and leads to a compensatory increase in sympathetic nervous system activity. Acutely, the sympathetic hyperactivity through the activation of beta-adrenergic receptors increases heart rate and cardiac contractility, which compensate for decreased cardiac output. However, chronic exposure of the heart to elevated levels of catecholamines released from sympathetic nerve terminals and the adrenal gland may lead to further pathologic changes in the heart, resulting in continued elevation of sympathetic tone and a progressive deterioration in cardiac function. On a molecular level, altered beta-adrenergic receptor signaling plays a pivotal role in the genesis and progression of HF. beta-adrenergic receptor number and function are decreased, and downstream mechanisms are altered. In this review we will present an overview of the normal beta-adrenergic receptor pathway in the heart and the consequences of sustained adrenergic activation in HF. The myopathic potential of individual components of the adrenergic signaling will be discussed through the results of research performed in genetic modified animals. Finally, we will discuss the potential clinical impact of beta-adrenergic receptor gene polymorphisms for better understanding the progression of HF.
Collapse
MESH Headings
- Animals
- Cardiac Output, Low/physiopathology
- Disease Models, Animal
- Disease Progression
- Humans
- Mice
- Polymorphism, Genetic
- Receptors, Adrenergic, beta-1/genetics
- Receptors, Adrenergic, beta-1/physiology
- Receptors, Adrenergic, beta-2/genetics
- Receptors, Adrenergic, beta-2/physiology
- Signal Transduction/physiology
Collapse
Affiliation(s)
- Patricia C Brum
- Laboratório de Fisiologia Cel. e Mol. do Exercício, Departamento de Biodinâmica do Movimento Humano, Escola de Educação Física e Esporte, Universidade de São Paulo, São Paulo, SP, Brasil.
| | | | | | | |
Collapse
|
38
|
Wang GY, McCloskey DT, Turcato S, Swigart PM, Simpson PC, Baker AJ. Contrasting inotropic responses to α1-adrenergic receptor stimulation in left versus right ventricular myocardium. Am J Physiol Heart Circ Physiol 2006; 291:H2013-7. [PMID: 16731650 DOI: 10.1152/ajpheart.00167.2006] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The left ventricle (LV) and right ventricle (RV) have differing hemodynamics and embryological origins, but it is unclear whether they are regulated differently. In particular, no previous studies have directly compared the LV versus RV myocardial inotropic responses to α1-adrenergic receptor (α1-AR) stimulation. We compared α1-AR inotropy of cardiac trabeculae from the LV versus RV of adult mouse hearts. As previously reported, for mouse RV trabeculae, α1-AR stimulation with phenylephrine (PE) caused a triphasic contractile response with overall negative inotropy. In marked contrast, LV trabeculae had an overall positive inotropic response to PE. Stimulation of a single subtype (α1A-AR) with A-61603 also mediated contrasting LV/RV inotropy, suggesting differential activation of multiple α1-AR-subtypes was not involved. Contrasting LV/RV α1-AR inotropy was not abolished by inhibiting protein kinase C, suggesting differential activation of PKC isoforms was not involved. However, contrasting LV/RV α1-AR inotropic responses did involve different effects on myofilament Ca2+ sensitivity: submaximal force of skinned trabeculae was increased by PE pretreatment for LV but was decreased by PE for RV. For LV myocardium, α1-AR-induced net positive inotropy was abolished by the myosin light chain kinase inhibitor ML-9. This study suggests that LV and RV myocardium have fundamentally different inotropic responses to α1-AR stimulation, involving different effects on myofilament function and myosin light chain phosphorylation.
Collapse
Affiliation(s)
- Guan-Ying Wang
- VA Medical Center, Cardiology Division (111C 4150 Clement St., San Francisco, CA 94121, USA
| | | | | | | | | | | |
Collapse
|
39
|
Grimm M, Mahnecke N, Soja F, El-Armouche A, Haas P, Treede H, Reichenspurner H, Eschenhagen T. The MLCK-mediated alpha1-adrenergic inotropic effect in atrial myocardium is negatively modulated by PKCepsilon signaling. Br J Pharmacol 2006; 148:991-1000. [PMID: 16783412 PMCID: PMC1751924 DOI: 10.1038/sj.bjp.0706803] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
The present study examined the role of myosin light chain kinase (MLCK), PKC isozymes, and inositol 1,4,5-trisphosphate (IP(3)) receptor in the positive inotropic effect of alpha(1)-adrenergic stimulation in atrial myocardium. We measured inotropic effects of phenylephrine (0.3-300 microM) in isolated left atrial preparations (1 Hz, 37 degrees C, 1.8 mM Ca(2+), 0.3 microM nadolol) from male 8-week FVB mice (n=200). Phenylephrine concentration-dependently increased force of contraction from 1.5+/-0.1 to 2.8+/-0.1 mN (mean+/-s.e.m., n=42), which was associated with increased MLC-2a phosphorylation at serine 21 and 22 by 67% and translocation of PKCepsilon but not PKCalpha to membrane (+30%) and myofilament (+50%) fractions.MLCK inhibition using ML-7 or wortmannin right-shifted the concentration-response curve of phenylephrine, reducing its inotropic effect at 10 microM by 73% and 81%, respectively. The compound KIE1-1 (500 nM), an intracellularly acting PKCepsilon translocation inhibitor peptide, prevented PKCepsilon translocation and augmented the maximal inotropic effect of phenylephrine by 40%. In contrast, inhibition of Ca(2+)-dependent PKC translocation (KIC1-1, 500 nM) had no effect. Chelerythrine, a PKC inhibitor, decreased basal force without changing the inotropic effect of phenylephrine. The IP(3) receptor blocker 2-APB (2 and 20 microM) concentration-dependently decreased basal force, but did not affect the concentration-response curve of phenylephrine. These results indicate that activation of MLCK is required for the positive inotropic effect of alpha(1)-adrenergic stimulation, that the Ca(2+)-independent PKCepsilon negatively modulates this effect, and that PKCalpha and IP(3) receptor activation is not involved.
Collapse
Affiliation(s)
- Michael Grimm
- Institute of Experimental and Clinical Pharmacology and Toxicology, University Medical Center, Hamburg, Germany
| | - Nina Mahnecke
- Institute of Experimental and Clinical Pharmacology and Toxicology, University Medical Center, Hamburg, Germany
| | - Friederike Soja
- Institute of Experimental and Clinical Pharmacology and Toxicology, University Medical Center, Hamburg, Germany
| | - Ali El-Armouche
- Institute of Experimental and Clinical Pharmacology and Toxicology, University Medical Center, Hamburg, Germany
| | - Pascal Haas
- Institute of Experimental and Clinical Pharmacology and Toxicology, University Medical Center, Hamburg, Germany
| | - Hendrik Treede
- Department of Cardiovascular Surgery, University Medical Center, Hamburg, Germany
| | | | - Thomas Eschenhagen
- Institute of Experimental and Clinical Pharmacology and Toxicology, University Medical Center, Hamburg, Germany
- Author for correspondence:
| |
Collapse
|
40
|
O’Connell TD, Swigart PM, Rodrigo M, Ishizaka S, Joho S, Turnbull L, Tecott LH, Baker AJ, Foster E, Grossman W, Simpson PC. Alpha1-adrenergic receptors prevent a maladaptive cardiac response to pressure overload. J Clin Invest 2006; 116:1005-15. [PMID: 16585965 PMCID: PMC1421341 DOI: 10.1172/jci22811] [Citation(s) in RCA: 116] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2004] [Accepted: 01/10/2006] [Indexed: 01/06/2023] Open
Abstract
An alpha1-adrenergic receptor (alpha1-AR) antagonist increased heart failure in the Antihypertensive and Lipid-Lowering Treatment to Prevent Heart Attack Trial (ALLHAT), but it is unknown whether this adverse result was due to alpha1-AR inhibition or a nonspecific drug effect. We studied cardiac pressure overload in mice with double KO of the 2 main alpha1-AR subtypes in the heart, alpha 1A (Adra1a) and alpha 1B (Adra1b). At 2 weeks after transverse aortic constriction (TAC), KO mouse survival was only 60% of WT, and surviving KO mice had lower ejection fractions and larger end-diastolic volumes than WT mice. Mechanistically, final heart weight and myocyte cross-sectional area were the same after TAC in KO and WT mice. However, KO hearts after TAC had increased interstitial fibrosis, increased apoptosis, and failed induction of the fetal hypertrophic genes. Before TAC, isolated KO myocytes were more susceptible to apoptosis after oxidative and beta-AR stimulation, and beta-ARs were desensitized. Thus, alpha1-AR deletion worsens dilated cardiomyopathy after pressure overload, by multiple mechanisms, indicating that alpha1-signaling is required for cardiac adaptation. These results suggest that the adverse cardiac effects of alpha1-antagonists in clinical trials are due to loss of alpha1-signaling in myocytes, emphasizing concern about clinical use of alpha1-antagonists, and point to a revised perspective on sympathetic activation in heart failure.
Collapse
Affiliation(s)
- Timothy D. O’Connell
- Cardiology Division, San Francisco Veterans Affairs Medical Center, San Francisco, California, USA.
Cardiovascular Research Institute and Department of Medicine, UCSF, San Francisco, California, USA.
Cardiology Division, Department of Medicine, UCSF, San Francisco, California, USA.
Department of Radiology, UCSF, San Francisco, California, USA.
Department of Psychiatry, UCSF, San Francisco, California, USA
| | - Philip M. Swigart
- Cardiology Division, San Francisco Veterans Affairs Medical Center, San Francisco, California, USA.
Cardiovascular Research Institute and Department of Medicine, UCSF, San Francisco, California, USA.
Cardiology Division, Department of Medicine, UCSF, San Francisco, California, USA.
Department of Radiology, UCSF, San Francisco, California, USA.
Department of Psychiatry, UCSF, San Francisco, California, USA
| | - M.C. Rodrigo
- Cardiology Division, San Francisco Veterans Affairs Medical Center, San Francisco, California, USA.
Cardiovascular Research Institute and Department of Medicine, UCSF, San Francisco, California, USA.
Cardiology Division, Department of Medicine, UCSF, San Francisco, California, USA.
Department of Radiology, UCSF, San Francisco, California, USA.
Department of Psychiatry, UCSF, San Francisco, California, USA
| | - Shinji Ishizaka
- Cardiology Division, San Francisco Veterans Affairs Medical Center, San Francisco, California, USA.
Cardiovascular Research Institute and Department of Medicine, UCSF, San Francisco, California, USA.
Cardiology Division, Department of Medicine, UCSF, San Francisco, California, USA.
Department of Radiology, UCSF, San Francisco, California, USA.
Department of Psychiatry, UCSF, San Francisco, California, USA
| | - Shuji Joho
- Cardiology Division, San Francisco Veterans Affairs Medical Center, San Francisco, California, USA.
Cardiovascular Research Institute and Department of Medicine, UCSF, San Francisco, California, USA.
Cardiology Division, Department of Medicine, UCSF, San Francisco, California, USA.
Department of Radiology, UCSF, San Francisco, California, USA.
Department of Psychiatry, UCSF, San Francisco, California, USA
| | - Lynne Turnbull
- Cardiology Division, San Francisco Veterans Affairs Medical Center, San Francisco, California, USA.
Cardiovascular Research Institute and Department of Medicine, UCSF, San Francisco, California, USA.
Cardiology Division, Department of Medicine, UCSF, San Francisco, California, USA.
Department of Radiology, UCSF, San Francisco, California, USA.
Department of Psychiatry, UCSF, San Francisco, California, USA
| | - Laurence H. Tecott
- Cardiology Division, San Francisco Veterans Affairs Medical Center, San Francisco, California, USA.
Cardiovascular Research Institute and Department of Medicine, UCSF, San Francisco, California, USA.
Cardiology Division, Department of Medicine, UCSF, San Francisco, California, USA.
Department of Radiology, UCSF, San Francisco, California, USA.
Department of Psychiatry, UCSF, San Francisco, California, USA
| | - Anthony J. Baker
- Cardiology Division, San Francisco Veterans Affairs Medical Center, San Francisco, California, USA.
Cardiovascular Research Institute and Department of Medicine, UCSF, San Francisco, California, USA.
Cardiology Division, Department of Medicine, UCSF, San Francisco, California, USA.
Department of Radiology, UCSF, San Francisco, California, USA.
Department of Psychiatry, UCSF, San Francisco, California, USA
| | - Elyse Foster
- Cardiology Division, San Francisco Veterans Affairs Medical Center, San Francisco, California, USA.
Cardiovascular Research Institute and Department of Medicine, UCSF, San Francisco, California, USA.
Cardiology Division, Department of Medicine, UCSF, San Francisco, California, USA.
Department of Radiology, UCSF, San Francisco, California, USA.
Department of Psychiatry, UCSF, San Francisco, California, USA
| | - William Grossman
- Cardiology Division, San Francisco Veterans Affairs Medical Center, San Francisco, California, USA.
Cardiovascular Research Institute and Department of Medicine, UCSF, San Francisco, California, USA.
Cardiology Division, Department of Medicine, UCSF, San Francisco, California, USA.
Department of Radiology, UCSF, San Francisco, California, USA.
Department of Psychiatry, UCSF, San Francisco, California, USA
| | - Paul C. Simpson
- Cardiology Division, San Francisco Veterans Affairs Medical Center, San Francisco, California, USA.
Cardiovascular Research Institute and Department of Medicine, UCSF, San Francisco, California, USA.
Cardiology Division, Department of Medicine, UCSF, San Francisco, California, USA.
Department of Radiology, UCSF, San Francisco, California, USA.
Department of Psychiatry, UCSF, San Francisco, California, USA
| |
Collapse
|
41
|
Turnbull L, Zhou HZ, Swigart PM, Turcato S, Karliner JS, Conklin BR, Simpson PC, Baker AJ. Sustained preconditioning induced by cardiac transgenesis with the tetracycline transactivator. Am J Physiol Heart Circ Physiol 2006; 290:H1103-9. [PMID: 16243914 DOI: 10.1152/ajpheart.00732.2005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Preconditioning protocols that protect the heart from ischemic injury may aid in the development of new therapies. However, the temporal window of cardioprotection is limited to a few days after the preconditioning stimulus. Here we report a sustained cardioprotected phenotype in mice expressing a tetracycline transactivator (tTA) transcription factor under the control of the α-myosin heavy chain (αMHC) promoter. αMHC-tTA mice were originally designed for tetracycline-regulated gene expression in the heart (Tet system). However, we found that after 45 min of global ischemia at 37°C, left ventricular developed pressure (LVDP) of Langendorff-perfused αMHC-tTA mouse hearts rapidly recovered in 5 min to 60% of initial levels, whereas LVDP of wild-type (WT) littermates recovered to only 10% of the initial level. Improved postischemic recovery of function for αMHC-tTA hearts was associated with a 50% decrease of infarct size and a significantly smaller release of lactate dehydrogenase to the coronary effluent. Improved postischemic recovery was not attributable to differences in coronary flow that was similar for WT- and αMHC-tTA hearts during recovery. Moreover, improved postischemic recovery of αMHC-tTA hearts was not abolished by inhibitors of classical cardioprotective effectors (mitochondrial ATP-sensitive K+ channels, PKC, or adenosine receptors), suggesting a novel mechanism. Finally, the tetracycline analog doxycycline, which inhibits binding of tTA to DNA, did not abolish improved recovery for αMHC-tTA hearts. The sustained cardioprotected phenotype of αMHC-tTA hearts may have implications for developing new therapies to minimize cardiac ischemic injury. Furthermore, investigations of cardioprotection using the Tet system may be aberrantly influenced by sustained preconditioning induced by cardiac transgenesis with tTA.
Collapse
Affiliation(s)
- Lynne Turnbull
- Department of Radiology, University of California, San Francisco, California 94121, USA
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Turcato S, Turnbull L, Wang GY, Honbo N, Simpson PC, Karliner JS, Baker AJ. Ischemic preconditioning depends on age and gender. Basic Res Cardiol 2006; 101:235-43. [PMID: 16450074 DOI: 10.1007/s00395-006-0585-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2005] [Revised: 12/01/2005] [Accepted: 12/22/2005] [Indexed: 10/25/2022]
Abstract
UNLABELLED The goal of this study was to determine if an ischemic preconditioning (IPC) protocol improved post-ischemic functional recovery of female mouse hearts. A previous study found that IPC did not occur in hearts from 10-week-old females. We studied Langendorff-perfused hearts from both 10- and 18-week-old mice (males and females). Hearts were subjected to 45 min ischemia and 45 reperfusion (I/R); IPC involved pretreatment with 3 min ischemia. We measured hemodynamics, infarct size and levels of the phosphorylated prosurvival kinase Akt (p-Akt). Similar to a previous study, for 10- week-old mice we found that the IPC protocol appreciably improved recovery of LV developed pressure (LVDP) for hearts from males but not females. However, for 18-week-old mice we found that the IPC protocol doubled the recovery of LVDP for both males and females. For both ages, hearts from females had greater recovery of LVDP and higher levels of p-Akt compared to males. CONCLUSIONS These findings are consistent with growing evidence that preconditioning induced by ischemia or other interventions can occur in hearts from females. However, for hearts from females, preconditioning depends on age. Moreover, consistent with previous studies, hearts from females have greater inherent resistance to ischemic injury, possibly involving increased signaling via p-Akt.
Collapse
Affiliation(s)
- S Turcato
- University of California, San Francisco, VA Medical Center, Cardiology Division 111C, 4150 Clement St, San Francisco, CA 94121, USA
| | | | | | | | | | | | | |
Collapse
|
43
|
McCloskey DT, Turnbull L, Swigart PM, Zambon AC, Turcato S, Joho S, Grossman W, Conklin BR, Simpson PC, Baker AJ. Cardiac transgenesis with the tetracycline transactivator changes myocardial function and gene expression. Physiol Genomics 2005; 22:118-26. [PMID: 15797971 DOI: 10.1152/physiolgenomics.00016.2005] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The cardiac-specific tetracycline-regulated gene expression system (tet-system) is a powerful tool using double-transgenic mice. The cardiac alpha-myosin heavy chain promoter (alphaMHC) drives lifetime expression of a tetracycline-inhibited transcription activator (tTA). Crossing alphaMHC-tTA mice with mice containing a tTA-responsive promoter linked to a target gene yields double-transgenic mice having tetracycline-repressed expression of the target gene in the heart. Using the tet-system, some studies use nontransgenic mice for the control group, whereas others use single-transgenic alphaMHC-tTA mice. However, previous studies found that high-level expression of a modified activator protein caused cardiomyopathy. Therefore, we tested whether cardiac expression of tTA was associated with altered function of alphaMHC-tTA mice compared with wild-type (WT) littermates. We monitored in vivo and in vitro function and gene expression profiles for myocardium from WT and alphaMHC-tTA mice. Compared with WT littermates, alphaMHC-tTA mice had a greater heart-to-body weight ratio (approximately 10%), ventricular dilation, and decreased ejection fraction, suggesting mild cardiomyopathy. In vitro, submaximal contractions were greater compared with WT and were associated with greater myofilament Ca2+ sensitivity. Gene expression profiling revealed that the expression of 153 genes was significantly changed by >20% when comparing alphaMHC-tTA with WT myocardium. These findings demonstrate that introduction of the alphaMHC-tTA construct causes significant effects on myocardial gene expression and major functional abnormalities in vivo and in vitro. For studies using the tet-system, these results suggest caution in the use of controls, since alphaMHC-tTA myocardium differs appreciably from WT. Furthermore, the results raise the possibility that the phenotype conferred by a target gene may be influenced by the modified genetic background of alphaMHC-tTA myocardium.
Collapse
Affiliation(s)
- Diana T McCloskey
- Department of Radiology, University of California, San Francisco, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Uberti MA, Hague C, Oller H, Minneman KP, Hall RA. Heterodimerization with β2-Adrenergic Receptors Promotes Surface Expression and Functional Activity of α1D-Adrenergic Receptors. J Pharmacol Exp Ther 2004; 313:16-23. [PMID: 15615865 DOI: 10.1124/jpet.104.079541] [Citation(s) in RCA: 113] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The alpha1D-adrenergic receptor (alpha1D-AR) is a G protein-coupled receptor (GPCR) that is poorly trafficked to the cell surface and largely nonfunctional when heterologously expressed by itself in a variety of cell types. We screened a library of approximately 30 other group I GPCRs in a quantitative luminometer assay for the ability to promote alpha1D-AR cell surface expression. Strikingly, these screens revealed only two receptors capable of inducing robust increases in the amount of alpha1D-AR at the cell surface: alpha1B-AR and beta2-AR. Confocal imaging confirmed that coexpression with beta2-AR resulted in translocation of alpha1D-AR from intracellular sites to the plasma membrane. Additionally, coimmunoprecipitation studies demonstrated that alpha1D-AR and beta2-AR specifically interact to form heterodimers when coexpressed in HEK-293 cells. Ligand binding studies revealed an increase in total alpha1D-AR binding sites upon coexpression with beta2-AR, but no apparent effect on the pharmacological properties of the receptors. In functional studies, coexpression with beta2-AR significantly enhanced the coupling of alpha1D-AR to norepinephrine-stimulated Ca2+ mobilization. Heterodimerization of beta2-AR with alpha1D-AR also conferred the ability of alpha1D-AR to cointernalize upon beta2-AR agonist stimulation, revealing a novel mechanism by which these different adrenergic receptor subtypes may regulate each other's activity. These findings demonstrate that the selective association of alpha1D-AR with other receptors is crucial for receptor surface expression and function and also shed light on a novel mechanism of cross talk between alpha1- and beta2-ARs that is mediated through heterodimerization and cross-internalization.
Collapse
MESH Headings
- Adrenergic beta-Agonists/pharmacology
- Albuterol/pharmacology
- Blotting, Western
- Calcium/metabolism
- Cell Line
- Humans
- Immunoprecipitation
- Microscopy, Confocal
- Plasmids/genetics
- Radioligand Assay
- Receptors, Adrenergic, alpha-1/biosynthesis
- Receptors, Adrenergic, alpha-1/genetics
- Receptors, Adrenergic, alpha-1/physiology
- Receptors, Adrenergic, beta-2/genetics
- Receptors, Adrenergic, beta-2/physiology
- Receptors, Cell Surface/metabolism
- Receptors, G-Protein-Coupled/metabolism
Collapse
Affiliation(s)
- Michelle A Uberti
- Department of Pharmacology, Emory University School of Medicine, 5113 Rollins Research Center, 1510 Clifton Rd., Atlanta, GA 30322, USA
| | | | | | | | | |
Collapse
|
45
|
Abstract
The biological effects of epinephrine and norepinephrine are mediated via 9 different adrenergic receptor subtypes, which all belong to the superfamily of G protein-coupled receptors. Although pharmacological ligands for adrenergic receptors have an important place in medical therapy, the full therapeutic potential of the 9 adrenergic receptor subtypes has not been explored yet. To dissect the physiological relevance of adrenergic receptor subtype diversity, gene-targeted mouse models carrying deletions in these receptor genes ("knockout mice") have been generated. This review gives an overview of the phenotypes observed in mice deficient in adrenergic receptors and discusses the therapeutic relevance of subtype-specific drug therapy.
Collapse
MESH Headings
- Animals
- Mice
- Mice, Knockout
- Models, Animal
- Receptors, Adrenergic, alpha/genetics
- Receptors, Adrenergic, alpha/physiology
- Receptors, Adrenergic, beta/genetics
- Receptors, Adrenergic, beta/physiology
- Signal Transduction/physiology
Collapse
Affiliation(s)
- Melanie Philipp
- Institute of Pharmacology and Toxicology, University of Würzburg, Versbacher Strasse 9, D-97078, Würzburg, Germany
| | | |
Collapse
|
46
|
Hague C, Chen Z, Uberti M, Minneman KP. α1-Adrenergic receptor subtypes: non-identical triplets with different dancing partners? Life Sci 2003; 74:411-8. [PMID: 14609720 DOI: 10.1016/j.lfs.2003.07.008] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Alpha(1)-adrenergic receptors are one of the three subfamilies of G protein coupled receptors activated by epinephrine and norepinephrine to control important functions in many target organs. Three human subtypes (alpha(1A), alpha(1B), alpha(1D)) are derived from separate genes and are highly homologous in their transmembrane domains but not in their amino or carboxyl termini. Recent advances in our understanding of these "non-identical triplets" include development of knockout mice lacking single or multiple subtypes, new insights into subcellular localization and trafficking, identification of allosteric modulators, and increasing evidence for an important role in brain function. Although all three subtypes activate the same G(q/11) signaling pathway, they also appear to interact with different protein binding partners. Recent evidence suggests they may also form dimers, and may initiate independent signals through pathways yet to be clearly elucidated. Thus, this subfamily represents a common phenomenon of a group of similar but non-identical receptor subtypes activated by the same neurotransmitter, whose individual functional roles remain to be clearly established.
Collapse
Affiliation(s)
- Chris Hague
- Department of Pharmacology, Emory University, Atlanta, GA 30322, USA.
| | | | | | | |
Collapse
|