1
|
Perez DM. Current Developments on the Role of α 1-Adrenergic Receptors in Cognition, Cardioprotection, and Metabolism. Front Cell Dev Biol 2021; 9:652152. [PMID: 34113612 PMCID: PMC8185284 DOI: 10.3389/fcell.2021.652152] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 04/29/2021] [Indexed: 12/13/2022] Open
Abstract
The α1-adrenergic receptors (ARs) are G-protein coupled receptors that bind the endogenous catecholamines, norepinephrine, and epinephrine. They play a key role in the regulation of the sympathetic nervous system along with β and α2-AR family members. While all of the adrenergic receptors bind with similar affinity to the catecholamines, they can regulate different physiologies and pathophysiologies in the body because they couple to different G-proteins and signal transduction pathways, commonly in opposition to one another. While α1-AR subtypes (α1A, α1B, α1C) have long been known to be primary regulators of vascular smooth muscle contraction, blood pressure, and cardiac hypertrophy, their role in neurotransmission, improving cognition, protecting the heart during ischemia and failure, and regulating whole body and organ metabolism are not well known and are more recent developments. These advancements have been made possible through the development of transgenic and knockout mouse models and more selective ligands to advance their research. Here, we will review the recent literature to provide new insights into these physiological functions and possible use as a therapeutic target.
Collapse
Affiliation(s)
- Dianne M Perez
- The Lerner Research Institute, The Cleveland Clinic Foundation, Cleveland, OH, United States
| |
Collapse
|
2
|
Wijaya LK, Stumbles PA, Drummond PD. A positive feedback loop between alpha 1-adrenoceptors and inflammatory cytokines in keratinocytes. Exp Cell Res 2020; 391:112008. [PMID: 32304706 DOI: 10.1016/j.yexcr.2020.112008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 04/02/2020] [Accepted: 04/12/2020] [Indexed: 01/11/2023]
Abstract
A positive feedback loop between inflammatory cytokines and alpha1-adrenoceptors (α1-AR) (a target of the sympathetic nervous system neurotransmitter norepinephrine) influences inflammatory responses in immune cells. This cross-talk between the sympathetic nervous system and immune system may play a role in promoting chronic inflammation. Emerging evidence shows that α1-AR interact with inflammatory cytokines in keratinocytes, and this epidermal adrenergic signalling may contribute to skin inflammatory responses following injury, disease or stress. In this study, utilizing an in vitro approach, we hypothesized that α1-AR interact in a positive feedback loop with inflammatory mediators in keratinocytes. The pro-inflammatory cytokine tumor necrosis factor α (TNFα) was used to induce an inflammatory state in cultured keratinocytes. TNFα increased interleukin (IL)-1β, IL-6, IL-8 and nerve growth factor (NGF) production and gene expression levels of α1-AR subtype B (α1B-AR). Additional stimulation of α1-AR further increased IL-6 levels, while maintaining high levels of IL-8 and decreasing levels of IL-1β and NGF. Our results suggest that reciprocal influences between α1-ARs and inflammatory cytokines may play a role in normal inflammatory responses. However, if unchecked, this cycle could contribute to pathology (e.g. chronic inflammatory diseases, chronic pain conditions, and stress-induced cancer progression).
Collapse
Affiliation(s)
- Linda K Wijaya
- College of Science, Health, Engineering and Education, Murdoch University, Perth, Western Australia, Australia
| | - Philip A Stumbles
- College of Science, Health, Engineering and Education, Murdoch University, Perth, Western Australia, Australia; Telethon Kids Institute, Perth, Western Australia, Australia
| | - Peter D Drummond
- College of Science, Health, Engineering and Education, Murdoch University, Perth, Western Australia, Australia.
| |
Collapse
|
3
|
Chowdhury MA, Sholl HK, Sharrett MS, Haller ST, Cooper CC, Gupta R, Liu LC. Exercise and Cardioprotection: A Natural Defense Against Lethal Myocardial Ischemia-Reperfusion Injury and Potential Guide to Cardiovascular Prophylaxis. J Cardiovasc Pharmacol Ther 2019; 24:18-30. [PMID: 30041547 PMCID: PMC7236859 DOI: 10.1177/1074248418788575] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Similar to ischemic preconditioning, high-intensity exercise has been shown to decrease infarct size following myocardial infarction. In this article, we review the literature on beneficial effects of exercise, exercise requirements for cardioprotection, common methods utilized in laboratories to study this phenomenon, and discuss possible mechanisms for exercise-mediated cardioprotection.
Collapse
Affiliation(s)
- Mohammed Andaleeb Chowdhury
- 1 Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
- * Mohammed Andaleeb Chowdhury, Haden K. Sholl, and Megan S. Sharrett contributed equally to this work
| | - Haden K Sholl
- 1 Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
- * Mohammed Andaleeb Chowdhury, Haden K. Sholl, and Megan S. Sharrett contributed equally to this work
| | - Megan S Sharrett
- 1 Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
| | - Steven T Haller
- 1 Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
| | - Christopher C Cooper
- 1 Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
| | - Rajesh Gupta
- 1 Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
| | - Lijun C Liu
- 1 Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
| |
Collapse
|
4
|
Zhu C, Rodda AE, Truong VX, Shi Y, Zhou K, Haynes JM, Wang B, Cook WD, Forsythe JS. Increased Cardiomyocyte Alignment and Intracellular Calcium Transients Using Micropatterned and Drug-Releasing Poly(Glycerol Sebacate) Elastomers. ACS Biomater Sci Eng 2018; 4:2494-2504. [PMID: 33435113 DOI: 10.1021/acsbiomaterials.8b00084] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Myocardial tissue engineering is a promising therapy for myocardial infarction recovery. The success of myocardial tissue engineering is likely to rely on the combination of cardiomyocytes, prosurvival regulatory signals, and a flexible biomaterial structure that can deliver them. In this study, poly(glycerol sebacate) (PGS), which exhibits stable elasticity under repeated tensile loading, was engineered to provide physical features that aligned cardiomyocytes in a similar manner to that seen in native cardiac tissue. In addition, a small molecule mimetic of brain derived neurotrophic factor (BDNF) was polymerized into the PGS to achieve a continuous and steady release. Micropatterning of PGS elastomers increased cell alignment, calcium transient homogeneity, and cell connectivity. The intensity of the calcium transients in cardiomyocytes was enhanced when cultured on PGS which released a small molecule BDNF mimetic. This study demonstrates that robust micropatterned elastomer films are a potential candidate for the delivery of functional cardiomyocytes and factors to the injured or dysfunctional myocardium, as well as providing novel in vitro platforms to study cardiomyocyte physiology.
Collapse
Affiliation(s)
- Chenghao Zhu
- Department of Materials Science and Engineering, Monash Institute of Medical Engineering, Monash University, Clayton, Victoria 3800, Australia
| | - Andrew E Rodda
- Department of Materials Science and Engineering, Monash Institute of Medical Engineering, Monash University, Clayton, Victoria 3800, Australia
| | - Vinh X Truong
- Department of Materials Science and Engineering, Monash Institute of Medical Engineering, Monash University, Clayton, Victoria 3800, Australia
| | - Yue Shi
- Department of Materials Science and Engineering, Monash Institute of Medical Engineering, Monash University, Clayton, Victoria 3800, Australia
| | - Kun Zhou
- Department of Materials Science and Engineering, Monash Institute of Medical Engineering, Monash University, Clayton, Victoria 3800, Australia
| | - John M Haynes
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Bing Wang
- Centre of Cardiovascular Research and Education in Therapeutics, School of Public Health and Preventive Medicine, Monash University, The Alfred Centre, 99 Commercial Road, Melbourne, Victoria 3004, Australia
| | - Wayne D Cook
- Department of Materials Science and Engineering, Monash Institute of Medical Engineering, Monash University, Clayton, Victoria 3800, Australia
| | - John S Forsythe
- Department of Materials Science and Engineering, Monash Institute of Medical Engineering, Monash University, Clayton, Victoria 3800, Australia
| |
Collapse
|
5
|
Sato M, Evans BA, Sandström AL, Chia LY, Mukaida S, Thai BS, Nguyen A, Lim L, Tan CYR, Baltos JA, White PJ, May LT, Hutchinson DS, Summers RJ, Bengtsson T. α 1A-Adrenoceptors activate mTOR signalling and glucose uptake in cardiomyocytes. Biochem Pharmacol 2017; 148:27-40. [PMID: 29175420 DOI: 10.1016/j.bcp.2017.11.016] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 11/22/2017] [Indexed: 12/20/2022]
Abstract
The capacity of G protein-coupled receptors to modulate mechanistic target of rapamycin (mTOR) activity is a newly emerging paradigm with the potential to link cell surface receptors with cell survival. Cardiomyocyte viability is linked to signalling pathways involving Akt and mTOR, as well as increased glucose uptake and utilization. Our aim was to determine whether the α1A-adrenoceptor (AR) couples to these protective pathways, and increased glucose uptake. We characterised α1A-AR signalling in CHO-K1 cells co-expressing the human α1A-AR and GLUT4 (CHOα1AGLUT4myc) and in neonatal rat ventricular cardiomyocytes (NRVM), and measured glucose uptake, intracellular Ca2+ mobilization, and phosphorylation of mTOR, Akt, 5' adenosine monophosphate-activated kinase (AMPK) and S6 ribosomal protein (S6rp). In both systems, noradrenaline and the α1A-AR selective agonist A61603 stimulated glucose uptake by parallel pathways involving mTOR and AMPK, whereas another α1-AR agonist oxymetazoline increased glucose uptake predominantly by mTOR. All agonists promoted phosphorylation of mTOR at Ser2448 and Ser2481, indicating activation of both mTORC1 and mTORC2, but did not increase Akt phosphorylation. In CHOα1AGLUT4myc cells, siRNA directed against rictor but not raptor suppressed α1A-AR mediated glucose uptake. We have thus identified mTORC2 as a key component in glucose uptake stimulated by α1A-AR agonists. Our findings identify a novel link between the α1A-AR, mTORC2 and glucose uptake, that have been implicated separately in cardiomyocyte survival. Our studies provide an improved framework for examining the utility of α1A-AR selective agonists as tools in the treatment of cardiac dysfunction.
Collapse
Affiliation(s)
- Masaaki Sato
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE-106 91 Stockholm, Sweden; Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, Victoria 3052, Australia
| | - Bronwyn A Evans
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, Victoria 3052, Australia
| | - Anna L Sandström
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE-106 91 Stockholm, Sweden
| | - Ling Yeong Chia
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, Victoria 3052, Australia
| | - Saori Mukaida
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, Victoria 3052, Australia
| | - Bui San Thai
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, Victoria 3052, Australia
| | - Anh Nguyen
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, Victoria 3052, Australia
| | - Linzi Lim
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, Victoria 3052, Australia
| | - Christina Y R Tan
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, Victoria 3052, Australia
| | - Jo-Anne Baltos
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, Victoria 3052, Australia
| | - Paul J White
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, Victoria 3052, Australia
| | - Lauren T May
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, Victoria 3052, Australia
| | - Dana S Hutchinson
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, Victoria 3052, Australia
| | - Roger J Summers
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, Victoria 3052, Australia
| | - Tore Bengtsson
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE-106 91 Stockholm, Sweden.
| |
Collapse
|
6
|
Vecchio EA, Chuo CH, Baltos JA, Ford L, Scammells PJ, Wang BH, Christopoulos A, White PJ, May LT. The hybrid molecule, VCP746, is a potent adenosine A2B receptor agonist that stimulates anti-fibrotic signalling. Biochem Pharmacol 2016; 117:46-56. [PMID: 27520486 DOI: 10.1016/j.bcp.2016.08.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 08/08/2016] [Indexed: 01/12/2023]
Abstract
We have recently described the rationally-designed adenosine receptor agonist, 4-(5-amino-4-benzoyl-3-(3-(trifluoromethyl)phenyl)thiophen-2-yl)-N-(6-(9-((2R,3R,4S,5R)-3,4-dihydroxy-5-(hydroxylmethyl)tetrahydro-furan-2-yl)-9H-purin-6-ylamino)hexyl)benzamide (VCP746), a hybrid molecule consisting of an adenosine moiety linked to an adenosine A1 receptor (A1AR) allosteric modulator moiety. At the A1AR, VCP746 mediated cardioprotection in the absence of haemodynamic side effects such as bradycardia. The current study has now identified VCP746 as an important pharmacological tool for the adenosine A2B receptor (A2BAR). The binding and function of VCP746 at the A2BAR was rigorously characterised in a heterologous expression system, in addition to examination of its anti-fibrotic signalling in cardiac- and renal-derived cells. In FlpInCHO cells stably expressing the human A2BAR, VCP746 was a high affinity, high potency A2BAR agonist that stimulated Gs- and Gq-mediated signal transduction, with an apparent lack of system bias relative to prototypical A2BAR agonists. The distinct agonist profile may result from an atypical binding mode of VCP746 at the A2BAR, which was consistent with a bivalent mechanism of receptor interaction. In isolated neonatal rat cardiac fibroblasts (NCF), VCP746 stimulated potent inhibition of both TGF-β1- and angiotensin II-mediated collagen synthesis. Similar attenuation of TGF-β1-mediated collagen synthesis was observed in renal mesangial cells (RMC). The anti-fibrotic signalling mediated by VCP746 in NCF and RMC was selectively reversed in the presence of an A2BAR antagonist. Thus, we believe, VCP746 represents an important tool to further investigate the role of the A2BAR in cardiac (patho)physiology.
Collapse
Affiliation(s)
- Elizabeth A Vecchio
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences & Department of Pharmacology, Monash University, Parkville, VIC 3052, Australia.
| | - Chung Hui Chuo
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences & Department of Pharmacology, Monash University, Parkville, VIC 3052, Australia.
| | - Jo-Anne Baltos
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences & Department of Pharmacology, Monash University, Parkville, VIC 3052, Australia.
| | - Leigh Ford
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia.
| | - Peter J Scammells
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia.
| | - Bing H Wang
- Centre of Cardiovascular Research and Education in Therapeutics, School of Public Health and Preventive Medicine, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC 3004, Australia.
| | - Arthur Christopoulos
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences & Department of Pharmacology, Monash University, Parkville, VIC 3052, Australia.
| | - Paul J White
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences & Department of Pharmacology, Monash University, Parkville, VIC 3052, Australia.
| | - Lauren T May
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences & Department of Pharmacology, Monash University, Parkville, VIC 3052, Australia.
| |
Collapse
|
7
|
Morris DP, Lei B, Longo LD, Bomsztyk K, Schwinn DA, Michelotti GA. Temporal Dissection of Rate Limiting Transcriptional Events Using Pol II ChIP and RNA Analysis of Adrenergic Stress Gene Activation. PLoS One 2015; 10:e0134442. [PMID: 26244980 PMCID: PMC4526373 DOI: 10.1371/journal.pone.0134442] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 07/10/2015] [Indexed: 12/13/2022] Open
Abstract
In mammals, increasing evidence supports mechanisms of co-transcriptional gene regulation and the generality of genetic control subsequent to RNA polymerase II (Pol II) recruitment. In this report, we use Pol II Chromatin Immunoprecipitation to investigate relationships between the mechanistic events controlling immediate early gene (IEG) activation following stimulation of the α1a-Adrenergic Receptor expressed in rat-1 fibroblasts. We validate our Pol II ChIP assay by comparison to major transcriptional events assessable by microarray and PCR analysis of precursor and mature mRNA. Temporal analysis of Pol II density suggests that reduced proximal pausing often enhances gene expression and was essential for Nr4a3 expression. Nevertheless, for Nr4a3 and several other genes, proximal pausing delayed the time required for initiation of productive elongation, consistent with a role in ensuring transcriptional fidelity. Arrival of Pol II at the 3’ cleavage site usually correlated with increased polyadenylated mRNA; however, for Nfil3 and probably Gprc5a expression was delayed and accompanied by apparent pre-mRNA degradation. Intragenic pausing not associated with polyadenylation was also found to regulate and delay Gprc5a expression. Temporal analysis of Nr4a3, Dusp5 and Nfil3 shows that transcription of native IEG genes can proceed at velocities of 3.5 to 4 kilobases/min immediately after activation. Of note, all of the genes studied here also used increased Pol II recruitment as an important regulator of expression. Nevertheless, the generality of co-transcriptional regulation during IEG activation suggests temporal and integrated analysis will often be necessary to distinguish causative from potential rate limiting mechanisms.
Collapse
Affiliation(s)
- Daniel P. Morris
- Center for Perinatal Biology, Loma Linda University, Loma Linda, California, United States of America
- * E-mail:
| | - Beilei Lei
- Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Lawrence D. Longo
- Center for Perinatal Biology, Loma Linda University, Loma Linda, California, United States of America
| | - Karol Bomsztyk
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Debra A. Schwinn
- Department of Anesthesiology, University of Iowa Carver College of Medicine, Iowa City, Iowa, United States of America
- Department of Pharmacology, University of Iowa Carver College of Medicine, Iowa City, Iowa, United States of America
- Department of Biochemistry, University of Iowa Carver College of Medicine, Iowa City, Iowa, United States of America
| | - Gregory A. Michelotti
- Department of Medicine, Division of Gastroenterology, Duke University Medical Center, Durham, North Carolina, United States of America
| |
Collapse
|
8
|
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: 3.6] [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
|
9
|
Alpha1a-adrenoceptor genetic variant induces cardiomyoblast-to-fibroblast-like cell transition via distinct signaling pathways. Cell Signal 2014; 26:1985-97. [PMID: 24835978 DOI: 10.1016/j.cellsig.2014.05.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Revised: 05/08/2014] [Accepted: 05/09/2014] [Indexed: 12/21/2022]
Abstract
The role of naturally occurring human α1a-Adrenergic Receptor (α1aAR) genetic variants associated with cardiovascular disorders is poorly understood. Here, we present the novel findings that expression of human α1aAR-247R (247R) genetic variant in cardiomyoblasts leads to transition of cardiomyoblasts into a fibroblast-like phenotype, evidenced by morphology and distinct de novo expression of characteristic genes. These fibroblast-like cells exhibit constitutive, high proliferative capacity and agonist-induced hypertrophy compared with cells prior to transition. We demonstrate that constitutive, synergistic activation of EGFR, Src and ERK kinases is the potential molecular mechanism of this transition. We also demonstrate that 247R triggers two distinct EGFR transactivation-dependent signaling pathways: 1) constitutive Gq-independent β-arrestin-1/Src/MMP/EGFR/ERK-dependent hyperproliferation and 2) agonist-induced Gq- and EGFR/STAT-dependent hypertrophy. Interestingly, in cardiomyoblasts agonist-independent hyperproliferation is MMP-dependent, but in fibroblast-like cells it is MMP-independent, suggesting that expression of α1aAR genetic variant in cardiomyocytes may trigger extracellular matrix remodeling. Thus, these novel findings demonstrate that EGFR transactivation by α1aAR-247R leads to hyperproliferation, hypertrophy and alterations in cardiomyoblasts, suggesting that these unique genetically-mediated alterations in signaling pathways and cellular function may lead to myocardial fibrosis. Such extracellular matrix remodeling may contribute to the genesis of arrhythmias in certain types of heart failure.
Collapse
|
10
|
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 PMCID: PMC3880467 DOI: 10.1124/pr.112.007203] [Citation(s) in RCA: 140] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [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
|
11
|
Schwinn D, Kleine-Brueggeney M, Oganesian A. Genomic Medicine. Anesth Analg 2013. [DOI: 10.1213/ane.0b013e31829ec0c7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
12
|
Lei B, Schwinn DA, Morris DP. Stimulation of α1a adrenergic receptors induces cellular proliferation or antiproliferative hypertrophy dependent solely on agonist concentration. PLoS One 2013; 8:e72430. [PMID: 23991110 PMCID: PMC3749976 DOI: 10.1371/journal.pone.0072430] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Accepted: 07/08/2013] [Indexed: 12/05/2022] Open
Abstract
Stimulation of α1aAdrenergic Receptors (ARs) is known to have anti-proliferative and hypertrophic effects; however, some studies also suggests this receptor can increase cell proliferation. Surprisingly, we find the α1aAR expressed in rat-1 fibroblasts can produce either phenotype, depending exclusively on agonist concentration. Stimulation of the α1aAR by high dose phenylephrine (>10−7 M) induces an antiproliferative, hypertrophic response accompanied by robust and extended p38 activation. Inhibition of p38 with SB203580 prevented the antiproliferative response, while inhibition of Erk or Jnk had no effect. In stark contrast, stimulation of the α1aAR with low dose phenylephrine (∼10−8 M) induced an Erk-dependent increase in cellular proliferation. Agonist-induced Erk phosphorylation was preceded by rapid FGFR and EGFR transactivation; however, only EGFR inhibition blocked Erk activation and proliferation. The general matrix metalloprotease inhibitor, GM6001, blocked agonist induced Erk activation within seconds, strongly suggesting EGFR activation involved extracellular triple membrane pass signaling. Erk activation required little Ca2+ release and was blocked by PLCβ or PKC inhibition but not by intracellular Ca2+ chelation, suggesting Ca2+ independent activation of novel PKC isoforms. In contrast, Ca2+ release was essential for PI3K/Akt activation, which was acutely maximal at non-proliferative doses of agonist. Remarkably, our data suggests EGFR transactivation leading to Erk induced proliferation has the lowest activation threshold of any α1aAR response. The ability of α1aARs to induce proliferation are discussed in light of evidence suggesting antagonistic growth responses reflect native α1aAR function.
Collapse
Affiliation(s)
- Beilei Lei
- Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Debra A. Schwinn
- Departments of Anesthesiology, Pharmacology, Biochemistry, University of Iowa Carver College of Medicine, Iowa City, Iowa, United States of America
| | - Daniel P. Morris
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
13
|
Papay RS, Shi T, Piascik MT, Naga Prasad SV, Perez DM. α₁A-adrenergic receptors regulate cardiac hypertrophy in vivo through interleukin-6 secretion. Mol Pharmacol 2013; 83:939-48. [PMID: 23404509 PMCID: PMC3629827 DOI: 10.1124/mol.112.084483] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Accepted: 02/12/2013] [Indexed: 01/17/2023] Open
Abstract
The role of α₁-adrenergic receptors (ARs) in the regulation of cardiac hypertrophy is still unclear, because transgenic mice demonstrated hypertrophy or the lack of it despite high receptor overexpression. To further address the role of the α₁-ARs in cardiac hypertrophy, we analyzed unique transgenic mice that overexpress constitutively active mutation (CAM) α₁A-ARs or CAM α₁B-ARs under the regulation of large fragments of their native promoters. These constitutively active receptors are expressed in all tissues that endogenously express their wild-type counterparts as opposed to only myocyte-targeted transgenic mice. In this study, we discovered that CAM α₁A-AR mice in vivo have cardiac hypertrophy independent of changes in blood pressure, corroborating earlier studies, but in contrast to myocyte-targeted α₁A-AR mice. We also found cardiac hypertrophy in CAM α₁B-AR mice, in agreement with previous studies, but hypertrophy only developed in older mice. We also discovered unique α₁-AR-mediated hypertrophic signaling that was AR subtype-specific with CAM α₁A-AR mice secreting atrial naturietic factor and interleukin-6 (IL-6), whereas CAM α₁B-AR mice expressed activated nuclear factor-κB (NF-κB). These particular hypertrophic signals were blocked when the other AR subtype was coactivated. We also discovered that crossbreeding the two CAM models (double CAM α₁A/B-AR) inhibited the development of hypertrophy and was reversible with single receptor activation, suggesting that coactivation of the receptors can lead to novel antagonistic signal transduction. This was confirmed by demonstrating antagonistic signals that were even lower than normal controls in the double CAM α₁A/B-AR mice for p38, NF-κB, and the IL-6/glycoprotein 130/signal transducer and activator of transcription 3 pathway. Because α₁A/B double knockout mice fail to develop hypertrophy in response to IL-6, our results suggest that IL-6 is a major mediator of α₁A-AR cardiac hypertrophy.
Collapse
Affiliation(s)
- Robert S Papay
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio 44195, USA
| | | | | | | | | |
Collapse
|
14
|
Abstract
Adrenoceptors and dopamine receptors are grouped together under the name 'catecholamine receptors.' Catecholamines and catecholaminergic drugs act on catecholamine receptors located on or near the cardiovascular system. The physiological effects of catecholamine receptor stimulation are only partly understood. The catecholaminergic drugs used in critical care medicine today are not selective, or are, at best, in part selective for the various catecholamine receptor subtypes. Many patients, however, depend on them. A variety of animal models has been developed to unravel catecholamine distribution and function. However, the identification of species heterogeneity makes it imperative to determine catecholamine receptor distribution and function in humans. In addition, age-related alterations in catecholamine receptor distribution and function have been identified in human adults. This might have implications for our understanding of the effect of catecholamines in pediatric patients. This article will focus on the pediatric population and will review currently available in vitro data on the distribution and the function of catecholamine receptors in the cardiovascular system of fetuses and children. Also discussed are relevant young animal models and in vivo hemodynamic effects of cardiotonic drugs acting on the catecholamine receptor in children requiring major cardiac surgery. A better understanding of these topics might provide clues for new, receptor subtype-selective, therapeutic approaches in newborns and children with cardiac disease.
Collapse
|
15
|
A novel player in cellular hypertrophy: Giβγ/PI3K-dependent activation of the RacGEF TIAM-1 is required for α₁-adrenoceptor induced hypertrophy in neonatal rat cardiomyocytes. J Mol Cell Cardiol 2012; 53:165-75. [PMID: 22564263 DOI: 10.1016/j.yjmcc.2012.04.015] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2011] [Revised: 04/25/2012] [Accepted: 04/26/2012] [Indexed: 01/08/2023]
Abstract
Activation of α(1)-adrenoceptors (α(1)-AR) by high catecholamine levels, e.g. in heart failure, is thought to be a driving force of cardiac hypertrophy. In this context several downstream mediators and cascades have been identified to potentially play a role in cardiomyocyte hypertrophy. One of these proteins is the monomeric G protein Rac1. However, until now it is unclear how this essential G protein is activated by α(1)-AR agonists and what are the downstream targets inducing cellular growth. By using protein-based as well as pharmacological inhibitors and the shRNA technique, we demonstrate that in neonatal rat cardiomyocytes (NRCM) Rac1 is activated via a cascade involving the α(1A)-AR subtype, G(i)βγ, the phosphoinositide-3'-kinase and the guanine nucleotide exchange factor Tiam1. We further demonstrate that this signaling induces an increase in protein synthesis, cell size and atrial natriuretic peptide expression. We identified the p21-activated kinase 2 (PAK2) as a downstream effector of Rac1 and were able to link this cascade to the activation of the pro-hypertrophic kinases ERK1/2 and p90RSK. Our data thus reveal a prominent role of the α(1A)-AR/G(i)βγ/Tiam1-mediated activation of Rac1 and its effector PAK2 in the induction of hypertrophy in NRCM.
Collapse
|
16
|
Constitutive coupling of a naturally occurring human alpha1a-adrenergic receptor genetic variant to EGFR transactivation pathway. Proc Natl Acad Sci U S A 2011; 108:19796-801. [PMID: 22089237 DOI: 10.1073/pnas.1116271108] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
We previously identified a naturally occurring human SNP, G247R, in the third intracellular loop of the α(1a)-adrenergic receptor (α(1a)-247R) and demonstrated that constitutive expression of α(1a)-247R results in twofold increased cell proliferation compared with WT. In the present study we elucidate molecular mechanisms and signal transduction pathways responsible for increased cell proliferation unique to α(1a)-247R, but not α(1a)-WT, α(1b), or α(1d)AR subtypes. We show that elevated levels of matrix metalloproteinase-7 (MMP7) and a disintegrin and metalloproteinase-12 (ADAM12) in α(1a)-247R-expressing cells are responsible for EGF receptor (EGFR) transactivation, downstream ERK activation, and increased cell proliferation; this pathway is confirmed using MMP, EGFR, and ERK inhibitors. We demonstrate that EGFR transactivation and downstream ERK activation depends on increased shedding of heparin-binding EGF. Finally, we demonstrate that knockdown of MMP7 or β-arrestin1 by shRNAs results in attenuation of proliferation of cells expressing α(1a)-247R. Importantly, accelerated cell proliferation triggered by the α(1a)-247R is serum- and agonist-independent, providing unique evidence for constitutive active coupling to the β-arrestin1/MMP/EGFR transactivation pathway by any G protein-coupled receptor. These findings raise the possibility of a previously unexplored mechanism for sympathetically mediated human hypertension triggered by a naturally occurring human genetic variant.
Collapse
|
17
|
Perez DM, Doze VA. Cardiac and neuroprotection regulated by α(1)-adrenergic receptor subtypes. J Recept Signal Transduct Res 2011; 31:98-110. [PMID: 21338248 DOI: 10.3109/10799893.2010.550008] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Sympathetic nervous system regulation by the α(1)-adrenergic receptor (AR) subtypes (α(1A), α(1B), α(1D)) is complex, whereby chronic activity can be either detrimental or protective for both heart and brain function. This review will summarize the evidence that this dual regulation can be mediated through the different α(1)-AR subtypes in the context of cardiac hypertrophy, heart failure, apoptosis, ischemic preconditioning, neurogenesis, locomotion, neurodegeneration, cognition, neuroplasticity, depression, anxiety, epilepsy, and mental illness.
Collapse
Affiliation(s)
- Dianne M Perez
- Department of Molecular Cardiology, NB50, The Lerner Research Institute, The Cleveland Clinic Foundation, Cleveland, Ohio 44195, USA.
| | | |
Collapse
|
18
|
Relationship between chemical structure, binding affinity and selectivity towards α1-adrenoceptors in the group of substituted n-phenylpiperazines. Part 2*. compounds containing ethane-1,2-diyl connecting chain. ACTA FACULTATIS PHARMACEUTICAE UNIVERSITATIS COMENIANAE 2011. [DOI: 10.2478/v10219-011-0005-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
19
|
Altered adrenergic receptor signaling following traumatic brain injury contributes to working memory dysfunction. Neuroscience 2010; 172:293-302. [PMID: 20974230 DOI: 10.1016/j.neuroscience.2010.10.048] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2010] [Revised: 10/07/2010] [Accepted: 10/16/2010] [Indexed: 11/21/2022]
Abstract
The prefrontal cortex is highly vulnerable to traumatic brain injury (TBI) and its structural and/or functional alterations as a result of TBI can give rise to persistent working memory (WM) dysfunction. Using a rodent model of TBI, we have described profound WM deficits following TBI that are associated with increases in prefrontal catecholamine (both dopamine and norepinephrine) content. In this study, we examined if enhanced norepinephrine signaling contributes to TBI-associated WM dysfunction. We demonstrate that administration of α1 adrenoceptor antagonists, but not α2A agonist, at 14 days post-injury significantly improved WM performance. mRNA analysis revealed increased levels of α1A, but not α1B or α1D, adrenoceptor in the medial prefrontal cortex (mPFC) of brain-injured rats. As α1A and 1B adrenoceptor promoters contain putative cAMP response element (CRE) sequences, we therefore examined if CRE-binding protein (CREB) actively engages these sequences in order to increase receptor gene transcription following TBI. Our results show that the phosphorylation of CREB is enhanced in the mPFC at time points during which increased α1A mRNA expression was observed. Chromatin immunoprecipitation (ChIP) assays using mPFC tissue from injured animals indicated increased phospho-CREB binding to the CRE sites of α1A, but not α1B, promoter compared to that observed in uninjured controls. To address the translatability of our findings, we tested the efficacy of the FDA-approved α1 antagonist Prazosin and observed that this drug improves WM in injured animals. Taken together, these studies suggest that enhanced CREB-mediated expression of α1 adrenoceptor contributes to TBI-associated WM dysfunction, and therapies aimed at reducing α1 signaling may be useful in the treatment of TBI-associated WM deficits in humans.
Collapse
|
20
|
|
21
|
Barreto F, Rezende D, Scaramello C, Silva C, Cunha V, Caricati-Neto A, Jurkiewicz A, Noël F, Quintas L. Lack of evidence for regulation of cardiac P-type ATPases and MAP kinases in transgenic mice with cardiac-specific overexpression of constitutively active α1B-adrenoceptors. Braz J Med Biol Res 2010; 43:500-5. [DOI: 10.1590/s0100-879x2010007500028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2009] [Accepted: 04/01/2010] [Indexed: 11/21/2022] Open
Affiliation(s)
- F. Barreto
- Universidade Federal do Rio de Janeiro, Brasil
| | | | | | | | | | | | | | - F. Noël
- Universidade Federal do Rio de Janeiro, Brasil
| | | |
Collapse
|
22
|
Alpha(1D)-adrenergic receptor insensitivity is associated with alterations in its expression and distribution in cultured vascular myocytes. Acta Pharmacol Sin 2009; 30:1585-93. [PMID: 19960004 DOI: 10.1038/aps.2009.160] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
AIM It is unclear why alpha(1D)-adrenergic receptors (alpha(1D)-ARs) play a critical role in the mediation of peripheral vascular resistance and blood pressure in situ but function inefficiently when studied in vitro. The present study examined the causes for these inconsistencies in native alpha(1)-adrenergic functional performance between the vascular smooth muscle and myocytes. METHODS The alpha(1)-adrenergic mediated contraction, Ca(2+) signaling and the subcellular receptor distribution were evaluated using the Fluo-4, BODIPY-FL prazosin and subtype-specific antibodies. RESULTS Rat aortic rings and freshly dissociated myocytes displayed contractile and increased intracellular Ca(2+) responses to stimulation with phenylephrine (PE, 10 micromol), respectively. However, the PE-induced responses disappeared completely in cultured aortic myocytes, whereas PE-enhanced Ca(2+) transients were seen in cultured rat cardiac myocytes. Further studies indicated that alpha(1D)-ARs, the major receptor subtype responsible for the alpha(1)-adrenergic regulation of aortic contraction, were distributed both intracellularly and at the cell membrane in freshly dispersed aortic myocytes, similar to the alpha(1A)-AR subcellular localization in the cultured cardiomyocytes. In the cultured aortic myocytes, however, in addition to a marked decrease in their protein expression relative to the aorta, most labeling signals for alpha(1D)-ARs were found in the cytoplasm. Importantly, treating the culture medium with charcoal/dextran caused the reappearance of alpha(1D)-ARs at the cell surface and a partial restoration of the Ca(2+) signal response to PE in approximately 30% of the cultured cells. CONCLUSION Reduction in alpha(1D)-AR total protein expression and disappearance from the cell surface contribute to the insensitivity of cultured vascular smooth muscle cells to alpha(1)-adrenergic receptor activation.
Collapse
|
23
|
Han J, Zou Z, Zhu C, Deng J, Wang J, Ran X, Shi C, Ai G, Li R, Cheng T, Su Y. DNA synthesis of rat bone marrow mesenchymal stem cells through alpha1-adrenergic receptors. Arch Biochem Biophys 2009; 490:96-102. [PMID: 19695215 DOI: 10.1016/j.abb.2009.08.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2009] [Revised: 08/06/2009] [Accepted: 08/12/2009] [Indexed: 11/16/2022]
Abstract
Multipotential bone marrow mesenchymal stem cells (BMSCs) are important in maintaining the microenvironment of the bone marrow (BM). Sympathetic nerves histologically innervate the BM; however, their role remains unclear. In this study, the effects of norepinephrine on DNA synthesis and the related signaling molecules involved in rBMSCs were examined. mRNA levels of the alpha1-adrenergic receptor subtypes increased following norepinephrine stimulation (10(-5) M for 30 min). DNA synthesis increased in dose- and time-dependent manners as determined by [(3)H]thymidine incorporation. Intracellular Ca(2+) concentration and translocation of protein kinase C from the cytosol to the membrane were also found to be elevated in rBMSCs. Phentolamine was able to suppress translocation of PKC. Norepinephrine also induced phosphorylation of ERK1/2, which was prevented by staurosporine treatment. Pretreatment with PD98059 inhibited ERK1/2 phosphorylation and DNA synthesis in rBMSCs. These findings indicate that norepinephrine stimulates DNA synthesis via alpha1-adrenergic receptors and downstream Ca(2+)/PKC and ERK1/2 activation in rBMSCs.
Collapse
Affiliation(s)
- Jing Han
- Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Yuan K, Rhee KS, Park WH, Kim SW, Kim SH. Different response of ANP secretion to adrenoceptor stimulation in renal hypertensive rat atria. Peptides 2008; 29:1207-15. [PMID: 18378355 DOI: 10.1016/j.peptides.2008.02.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2008] [Accepted: 02/19/2008] [Indexed: 11/29/2022]
Abstract
Sympathetic nervous system and atrial natriuretic peptide (ANP) system play fundamental roles in the regulation of cardiovascular functions. Overactivity of sympathetic nervous system can lead into cardiovascular diseases such as heart failure and hypertension. The present study aimed to define which adrenergic receptors (ARs) affect atrial contractility and ANP release and to determine their modification in renal hypertensive rat atria. An alpha(1)-AR agonist, cirazoline increased ANP release with positive inotropism. These alpha(1)-AR agonist-mediated responses were attenuated by the alpha(1A)-AR antagonist, but not by the alpha(1B)- or alpha(1D)-AR antagonist. An alpha(2)-AR agonist, guanabenz and clonidine increased ANP release with negative inotropism and decreased cAMP level. The order of potency for the increased ANP release was cirazoline>>phenylephrine=guanabenz>>clonidine. In contrast, a beta-AR agonist, isoproterenol decreased ANP release with positive inotropism and these responses were blocked by the beta(1)-AR antagonist but not by the beta(2)-AR antagonist. The increased cAMP level by isoproterenol was suppressed by pretreatment with both beta(1)- and beta(2)-AR antagonists. In renal hypertensive rat atria, the effects of isoproterenol on atrial contractility, ANP release, and cAMP level were attenuated whereas the effect of cirazoline on ANP release was unaltered. Atrial beta(1)-AR mRNA level but not alpha(1A)-AR mRNA level was decreased in renal hypertensive rats. These findings suggest that alpha(1A)- and beta(1)-AR oppositely regulate atrial ANP release and that atrial beta(1)-AR expression/function is impaired in renal hypertensive rats.
Collapse
Affiliation(s)
- Kuichang Yuan
- Department of Physiology, Medical School, Chonbuk National University, Jeonju 561-180, Republic of Korea
| | | | | | | | | |
Collapse
|
25
|
Long KM, Kirby R. An update on cardiovascular adrenergic receptor physiology and potential pharmacological applications in veterinary critical care. J Vet Emerg Crit Care (San Antonio) 2008. [DOI: 10.1111/j.1476-4431.2007.00266.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
26
|
Amirahmadi F, Turnbull L, Du XJ, Graham RM, Woodcock EA. Heightened alpha1A-adrenergic receptor activity suppresses ischaemia/reperfusion-induced Ins(1,4,5)P3 generation in the mouse heart: a comparison with ischaemic preconditioning. Clin Sci (Lond) 2008; 114:157-64. [PMID: 17696883 DOI: 10.1042/cs20070110] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Reperfusion of ischaemic rat or mouse hearts causes NE [noradrenaline ('norepinephrine')] release, stimulation of alpha(1)-ARs (alpha(1)-adrenergic receptors), PLC (phospholipase C) activation, Ins(1,4,5)P(3) generation and the development of arrhythmias. In the present study, we examined the effect of increased alpha(1A)-AR drive on these responses. In hearts from non-transgenic mice (alpha(1A)-WT), Ins(1,4,5)P(3) generation was observed after 2 min of reperfusion following 30 min of zero-flow ischaemia. No Ins(1,4,5)P(3) response was observed in hearts from transgenic mice with 66-fold overexpression of alpha(1A)-AR (alpha(1A)-TG). This was despite the fact that alpha(1A)-TG hearts had 8-10-fold higher PLC responses to NE than alpha(1A)-WT under normoxic conditions. The immediate phospholipid precursor of Ins(1,4,5)P(3), PtdIns(4,5)P(2), responded to ischaemia and reperfusion similarly in alpha(1A)-WT and alpha(1A)-TG mice. Thus the lack of Ins(1,4,5)P(3) generation in alpha(1A)-TG mice is not caused by limited availability of PtdIns(4,5)P(2). Overall, alpha(1)-AR-mediated PLC activity was markedly enhanced in alpha(1A)-WT mice under reperfusion conditions, but responses in alpha(1A)-TG mice were not significantly different in normoxia and post-ischaemic reperfusion. Ischaemic preconditioning prevented Ins(1,4,5)P(3) generation after 30 min of ischaemic insult in alpha(1A)-WT mice. However, the precursor lipid PtdIns(4,5)P(2) was also reduced by preconditioning, whereas heightened alpha(1A)-AR activity did not influence PtdIns(4,5)P(2) responses in reperfusion. Thus preconditioning and alpha(1A)-AR overexpression have different effects on early signalling responses, even though both prevented Ins(1,4,5)P(3) generation. These studies demonstrate a selective inhibitory action of heightened alpha(1A)-AR activity on immediate post-receptor signalling responses in early post-ischaemic reperfusion.
Collapse
Affiliation(s)
- Fatemeh Amirahmadi
- Cellular Biochemistry Laboratory, Baker Heart Research Institute, PO Box 6492, St. Kilda Road Central, Melbourne, VIC 8008, Australia
| | | | | | | | | |
Collapse
|
27
|
Woodcock EA. Roles of alpha1A- and alpha1B-adrenoceptors in heart: insights from studies of genetically modified mice. Clin Exp Pharmacol Physiol 2007; 34:884-8. [PMID: 17645635 DOI: 10.1111/j.1440-1681.2007.04707.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
1. Several mouse strains have been prepared in which different subtypes of the alpha1-adrenoceptor (AR) are overexpressed or deleted. The phenotypes of the animals generated vary depending on whether the receptors are expressed specifically in heart or generally throughout the animal, but some overall conclusions can be drawn. 2. Heightened activity of alpha1B-AR by overexpressing the receptors leads to depressed contractile responses to beta-AR activation, which may be related to activation of the inhibitory G-protein Gi. In contrast, alpha1A-AR cause substantially heightened contractility when overexpressed in heart. 3. Overexpressed alpha1B-AR predispose hearts to hypertrophy and worsen heart failure caused by pressure overload, whereas increased alpha1A-AR expression does not influence hypertrophic responses and, furthermore, improves outcomes after pressure overload or myocardial infarction. 4. Alpha1A-adrenoceptors mediate a preconditioning action to improve functional recovery after acute ischaemic insult, whereas alpha1B-AR are ineffective. Both subtypes appear to protect from inositol 1,4,5-trisphosphate generation and arrhythmogenesis in early postischaemic reperfusion. 5. Although some of the protective effects of heightened alpha1A-AR drive may be related to the enhanced contractility, it is also possible that alpha1A-AR protect from cardiomyocyte apoptotic responses.
Collapse
Affiliation(s)
- Elizabeth A Woodcock
- Cellular Biochemistry Laboratory, Baker Heart Research Institute, Melbourne, Victoria, Australia.
| |
Collapse
|
28
|
McConell GK, Bradley SJ, Stephens TJ, Canny BJ, Kingwell BA, Lee-Young RS. Skeletal muscle nNOSμ protein content is increased by exercise training in humans. Am J Physiol Regul Integr Comp Physiol 2007; 293:R821-8. [PMID: 17459909 DOI: 10.1152/ajpregu.00796.2006] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The major isoform of nitric oxide synthase (NOS) in skeletal muscle is the splice variant of neuronal NOS, termed nNOSμ. Exercise training increases nNOSμ protein levels in rat skeletal muscle, but data in humans are conflicting. We performed two studies to determine 1) whether resting nNOSμ protein expression is greater in skeletal muscle of 10 endurance-trained athletes compared with 11 sedentary individuals ( study 1) and 2) whether intense short-term (10 days) exercise training increases resting nNOSμ protein (within whole muscle and also within types I, IIa, and IIx fibers) in eight sedentary individuals ( study 2). In study 1, nNOSμ protein was ∼60% higher ( P < 0.05) in endurance-trained athletes compared with the sedentary participants. In study 2, nNOSμ protein expression was similar in types I, IIa, and IIx fibers before training. Ten days of intense exercise training significantly ( P < 0.05) increased nNOSμ protein levels in types I, IIa, and IIx fibers, a finding that was validated by using whole muscle samples. Endothelial NOS and inducible NOS protein were barely detectable in the skeletal muscle samples. In conclusion, nNOSμ protein expression is greater in endurance-trained individuals when compared with sedentary individuals. Ten days of intense exercise is also sufficient to increase nNOSμ expression in untrained individuals, due to uniform increases of nNOSμ within types I, IIa, and IIx fibers.
Collapse
Affiliation(s)
- Glenn K McConell
- Department of Physiology, The University of Melbourne, Parkville, Victoria 3010, Australia.
| | | | | | | | | | | |
Collapse
|
29
|
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: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
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
|
30
|
Fang L, Moore XL, Gao XM, Dart AM, Lim YL, Du XJ. Down-regulation of mitofusin-2 expression in cardiac hypertrophy in vitro and in vivo. Life Sci 2007; 80:2154-2160. [PMID: 17499311 DOI: 10.1016/j.lfs.2007.04.003] [Citation(s) in RCA: 111] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2006] [Revised: 03/22/2007] [Accepted: 04/10/2007] [Indexed: 11/26/2022]
Abstract
Mitofusin-2 (Mfn2) suppresses smooth muscle cell proliferation through inhibition of the Ras-extracellular signal-regulated kinases (ERK1/2) pathway. Since the ERK1/2 pathway is implicated in mediating hypertrophic signaling, we studied the changes in Mfn2 in cardiac hypertrophy using in vitro and in vivo models. Phenylephrine was used to induce hypertrophy in neonatal rat ventricular myocytes (NRVMs). In vivo hypertrophy models included spontaneously hypertensive rats (SHR), pressure-overload hypertrophy by transverse aortic constriction (TAC), hypertrophy of non-infarcted myocardium following myocardial infarction (MI), and cardiomyopathy due to cardiac-restricted overexpression of beta(2)-adrenergic receptors (beta(2)-TG). We determined hypertrophic parameters and analysed expression of atrial natriuretic peptide (ANP) and Mfn2 by real-time PCR. Phosphorylated-ERK1/2 (phospho-ERK) was measured by Western blot. Mfn2 was downregulated in phenylephrine treated NRCMs (by approximately 40%), hypertrophied hearts from SHR (by approximately 80%), mice with TAC (at 1 and 3 weeks, by approximately 50%), and beta(2)-TG mice (by approximately 20%). However, Mfn2 was not downregulated in hypertrophied hearts with 15 weeks of TAC, nor in hypertrophied non-infarcted myocardium following MI. phospho-ERK1/2 was increased in hypertrophied myocardium at 1 week post-TAC, but not in non-infarcted myocardium after MI, indicating that downregulated Mfn2 may be accompanied by an increase of phospho-ERK1/2. This study shows, for the first time, downregulated Mfn2 expression in hypertrophied hearts, which depends on the etiology and time course of hypertrophy. Further study is required to examine the causal relationship between Mfn2 and cardiac hypertrophy.
Collapse
Affiliation(s)
- Lu Fang
- Baker Heart Research Institute and Alfred Heart Centre, Alfred Hospital, Melbourne, Australia; Western Hospital, Department of Medicine, University of Melbourne, Melbourne, Australia
| | - Xiao-Lei Moore
- Baker Heart Research Institute and Alfred Heart Centre, Alfred Hospital, Melbourne, Australia
| | - Xiao-Ming Gao
- Baker Heart Research Institute and Alfred Heart Centre, Alfred Hospital, Melbourne, Australia
| | - Anthony M Dart
- Baker Heart Research Institute and Alfred Heart Centre, Alfred Hospital, Melbourne, Australia
| | - Yean Leng Lim
- Western Hospital, Department of Medicine, University of Melbourne, Melbourne, Australia
| | - Xiao-Jun Du
- Baker Heart Research Institute and Alfred Heart Centre, Alfred Hospital, Melbourne, Australia.
| |
Collapse
|
31
|
Hein P, Michel MC. Signal transduction and regulation: are all alpha1-adrenergic receptor subtypes created equal? Biochem Pharmacol 2006; 73:1097-106. [PMID: 17141737 DOI: 10.1016/j.bcp.2006.11.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2006] [Revised: 10/30/2006] [Accepted: 11/01/2006] [Indexed: 02/06/2023]
Abstract
The current manuscript reviews the evidence whether and how subtypes of alpha(1)-adrenergic receptors, i.e. alpha(1A)-, alpha(1B)- and alpha(1D)-adrenergic receptors, differentially couple to signal transduction pathways and exhibit differential susceptibility to regulation. In both regards studies in tissues or cells natively expressing the subtypes are hampered because the relative expression of the subtypes is poorly controlled and the observed effects may be cell-type specific. An alternative approach, i.e. transfection of multiple subtypes into the same host cell line overcomes this limitation, but it often remains unclear whether results in such artificial systems are representative for the physiological situation. The overall evidence suggests that indeed subtype-intrinsic and cell type-specific factors interact to direct alpha(1)-adrenergic receptor signaling and regulation. This may explain why so many apparently controversial findings have been reported from various tissues and cells. One of the few consistent themes is that alpha(1D)-adrenergic receptors signal less effectively upon agonist stimulation than the other subtypes, most likely because they exhibit spontaneous internalization.
Collapse
Affiliation(s)
- Peter Hein
- Department of Pharmacology, University of Würzburg, Würzburg, Germany
| | | |
Collapse
|
32
|
Faber JE, Szymeczek CL, Salvi SS, Zhang H. Enhanced α1-adrenergic trophic activity in pulmonary artery of hypoxic pulmonary hypertensive rats. Am J Physiol Heart Circ Physiol 2006; 291:H2272-81. [PMID: 16798826 DOI: 10.1152/ajpheart.00404.2006] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Mechanisms that induce the excessive proliferation of vascular wall cells in hypoxic pulmonary hypertension (PH) are not fully understood. Alveolar hypoxia causes sympathoexcitation, and norepinephrine can stimulate α1-adrenoceptor (α1-AR)-dependent hypertrophy/hyperplasia of smooth muscle cells and adventitial fibroblasts. Adrenergic trophic activity is augmented in systemic arteries by injury and altered shear stress, which are key pathogenic stimuli in hypoxic PH, and contributes to neointimal formation and flow-mediated hypertrophic remodeling. Here we examined whether norepinephrine stimulates growth of the pulmonary artery (PA) and whether this is augmented in PH. PA from normoxic and hypoxic rats [9 days of 0.1 fraction of inspired O2 (FiO2)] was studied in organ culture, where wall tension, Po2, and Pco2 were maintained at values present in normal and hypoxic PH rats. Norepinephrine treatment for 72 h increased DNA and protein content modestly in normoxic PA (+10%, P < 0.05). In hypoxic PA, these effects were augmented threefold ( P < 0.05), and protein synthesis was increased 34-fold ( P < 0.05). Inferior thoracic vena cava from normoxic or hypoxic rats was unaffected. Norepinephrine-induced growth in hypoxic PA was dose dependent, had efficacy greater than or equal to endothelin-1, required the presence of wall tension, and was inhibited by α1A-AR antagonist. In hypoxic pulmonary vasculature, α1A-AR was downregulated the least among α1-AR subtypes. These data demonstrate that norepinephrine has trophic activity in the PA that is augmented by PH. If evident in vivo in the pulmonary vasculature, adrenergic-induced growth may contribute to the vascular hyperplasia that participates in hypoxic PH.
Collapse
MESH Headings
- Adrenergic alpha-1 Receptor Agonists
- Adrenergic alpha-1 Receptor Antagonists
- Animals
- DNA/analysis
- DNA/metabolism
- Dose-Response Relationship, Drug
- Endothelin-1/pharmacology
- Fibroblasts/drug effects
- Fibroblasts/metabolism
- Hypertension, Pulmonary/etiology
- Hypertension, Pulmonary/pathology
- Hypertension, Pulmonary/physiopathology
- Hypoxia/complications
- Male
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Norepinephrine/pharmacology
- Organ Culture Techniques
- Proteins/analysis
- Proteins/metabolism
- Pulmonary Artery/drug effects
- Pulmonary Artery/pathology
- Rats
- Rats, Sprague-Dawley
- Receptors, Adrenergic, alpha-1/physiology
- Time Factors
Collapse
Affiliation(s)
- James E Faber
- Dept. of Cell and Molecular Physiology, 6309 MBRB, Univ. of North Carolina, Chapel Hill, NC 27599-7545, USA.
| | | | | | | |
Collapse
|
33
|
Xu Q, Xu N, Zhang T, Zhang H, Li Z, Yin F, Lu Z, Han Q, Zhang Y. Mammalian tolloid alters subcellular localization, internalization, and signaling of alpha(1a)-adrenergic receptors. Mol Pharmacol 2006; 70:532-541. [PMID: 16690783 DOI: 10.1124/mol.105.016451] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
In the present study, we identified the CUB5 domain of mammalian Tolloid (mTLD) as a novel protein binding to alpha(1A)-adrenergic receptor (AR) using the yeast two-hybrid system. Whereas CUB5 did not couple to either alpha(1B)-AR or alpha(1D)-AR. It was determined that amino acids 322 to 359 of alpha(1A)-AR were the major binding region for CUB5. The direct interaction between alpha(1A)-AR cytoplasmic tail and CUB5 was discovered by glutathione S-transferase pull-down assay. We confirmed the interaction of mTLD with alpha(1A)-AR in human embryonic kidney (HEK) 293 cells by immunoprecipitation, immunofluorescence, and fluorescence resonance energy transfer. Although mTLD did not affect the density and affinity of receptors in crudely prepared membranes from HEK293 cells stably expressing alpha(1A)-AR, it significantly altered the subcellular localization of the receptors. Moreover, mTLD reduced the level of cell surface alpha(1A)-ARs, delayed the initial rate of agonist-induced receptor internalization, and facilitated agonist-induced calcium transient. We have demonstrated that mTLD interacts with alpha(1A)-AR directly, alters the subcellular localization of receptor, and influences agonist-induced alpha(1A)-AR internalization and calcium signaling.
Collapse
Affiliation(s)
- Qi Xu
- Institute of Vascular Medicine, Peking University Third Hospital, No.49 Huayuan North Road, Haidian District, Beijing, P.R. China 100083
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Tual L, Morel OE, Favret F, Fouillit M, Guernier C, Buvry A, Germain L, Dhonneur G, Bernaudin JF, Richalet JP. Carvedilol inhibits right ventricular hypertrophy induced by chronic hypobaric hypoxia. Pflugers Arch 2006; 452:371-9. [PMID: 16639551 DOI: 10.1007/s00424-006-0058-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2005] [Revised: 02/04/2006] [Accepted: 02/26/2006] [Indexed: 10/24/2022]
Abstract
Right ventricular hypertrophy induced by chronic hypoxia is mainly due to a mechanical stress upon the ventricular wall secondary to pulmonary arterial hypertension. However, the hypoxic chronic activation of the sympathetic nervous system can contribute to the development of right ventricular hypertrophy either via myocardial adrenergic receptors and/or a vasoconstriction and remodeling of pulmonary arteries. To highlight the specific role of the sympathetic nervous system on hypoxia-induced right ventricular hypertrophy and particularly the efficiency of carvedilol, our study compared physiological, myocardial, and pulmonary arterial morphometric data in rats treated by alpha-(prazosin), or beta-(propranolol) or alphabeta-(carvedilol) antagonist and exposed to chronic hypobaric hypoxia (2 weeks at 380 mmHg barometric pressure). In chronic hypoxia, both systolic right ventricular pressure and Fulton's ratio (right/(left+septum) ventricular weight) were lower in rats treated by prazosin (-16.7 and -13.6%), propranolol (-28.6 and -12.7%) and carvedilol (-15.9 and -14.3%) respectively when compared to glucose (p<0.05). Surprisingly, prazosin was unable to reduce right ventricular hypertrophy induced by chronic hypoxia, whereas, left ventricular weight increased. Wall thickness index of pulmonary arteries increased in chronic hypoxia and was reduced by carvedilol. In conclusion, the hypoxia-induced activation of the adrenergic system participates in the development of right ventricular hypertrophy. Carvedilol is effective in reducing hypoxia-induced right ventricular hypertrophy, pulmonary arterial hypertension, and muscularization of pulmonary arteries.
Collapse
Affiliation(s)
- L Tual
- Université Paris 13, Laboratoire , Résponses cellulaires et fonctionnelles à l'hypoxie, EA 2363, Association pour la Recherche en Physiologie de l'Environment, 74 rue Marcen Cachin, 93017, Bobigny, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Myslivecek J, Nováková M, Palkovits M, Krizanová O, Kvetnanský R. Distribution of mRNA and binding sites of adrenoceptors and muscarinic receptors in the rat heart. Life Sci 2006; 79:112-20. [PMID: 16427094 DOI: 10.1016/j.lfs.2005.12.026] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2005] [Revised: 12/08/2005] [Accepted: 12/15/2005] [Indexed: 11/26/2022]
Abstract
Since there exist some obscurities in the expression of mRNAs and their receptors in the heart, we have investigated the gene expression (mRNA levels) of adrenoceptors (alpha1A-, alpha1B-, beta1-, beta2-, beta3-) and muscarinic receptors (M2) and the density of receptor binding sites (alpha1A-, alpha1B-, beta1-, beta2-adrenoceptors, muscarinic receptors). Moreover, the heart regions consist of tissue rich in ganglion cells (that are of importance in heart neural circuits) and those virtually free of them (myocytes). Therefore, we have examined the differences in the distribution of mRNAs/receptor binding sites in the atrial samples of the heart rich in ganglion cells vs. those are virtually free of them. Binding sites and mRNAs of muscarinic receptors and alpha1B-adrenoceptors differ in their distribution in different heart regions. The mRNAs for beta1- and beta2-adrenoceptors were almost equally distributed herein, while the amount of beta-adrenoceptors significantly differs in the heart regions. The alpha1A- and beta3-adrenoceptors mRNAs were also found in all investigated heart regions, but at significantly lower level and have not shown region differences. This is a new finding, especially to beta3-adrenoceptors, as they were not regularly found in each heart regions. alpha1B-adrenoceptors have similar distribution of their mRNAs and binding sites in some heart parts. Thus, we can conclude that there are noticeable differences in the presence of receptors in heart regions that contain ganglion cells in comparison to those are virtually free of them.
Collapse
Affiliation(s)
- Jaromír Myslivecek
- Institute of Physiology, 1st Faculty of Medicine, Charles University, Prague, Czech Republic.
| | | | | | | | | |
Collapse
|
36
|
Pare GC, Bauman AL, McHenry M, Michel JJC, Dodge-Kafka KL, Kapiloff MS. The mAKAP complex participates in the induction of cardiac myocyte hypertrophy by adrenergic receptor signaling. J Cell Sci 2005; 118:5637-46. [PMID: 16306226 DOI: 10.1242/jcs.02675] [Citation(s) in RCA: 106] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Maladaptive cardiac hypertrophy can progress to congestive heart failure, a leading cause of morbidity and mortality in the United States. A better understanding of the intracellular signal transduction network that controls myocyte cell growth may suggest new therapeutic directions. mAKAP is a scaffold protein that has recently been shown to coordinate signal transduction enzymes important for cytokine-induced cardiac hypertrophy. We now extend this observation and show mAKAP is important for adrenergic-mediated hypertrophy. One function of the mAKAP complex is to facilitate cAMP-dependent protein kinase A-catalyzed phosphorylation of the ryanodine receptor Ca2+-release channel. Experiments utilizing inhibition of the ryanodine receptor, RNA interference of mAKAP expression and replacement of endogenous mAKAP with a mutant form that does not bind to protein kinase A demonstrate that the mAKAP complex contributes to pro-hypertrophic signaling. Further, we show that calcineurin Abeta associates with mAKAP and that the formation of the mAKAP complex is required for the full activation of the pro-hypertrophic transcription factor NFATc. These data reveal a novel function of the mAKAP complex involving the integration of cAMP and Ca2+ signals that promote myocyte hypertrophy.
Collapse
Affiliation(s)
- Genevieve C Pare
- Department of Pediatrics, Heart Research Center, Oregon Health and Science University, NRC5 3181 S.W. Sam Jackson Park Road, Portland, OR 97239, USA
| | | | | | | | | | | |
Collapse
|
37
|
Ma S, Morilak DA. Chronic intermittent cold stress sensitises the hypothalamic-pituitary-adrenal response to a novel acute stress by enhancing noradrenergic influence in the rat paraventricular nucleus. J Neuroendocrinol 2005; 17:761-9. [PMID: 16219005 DOI: 10.1111/j.1365-2826.2005.01372.x] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Chronic intermittent cold stress sensitises activation of the hypothalamic-pituitary-adrenal (HPA) axis by novel acute stress. We have shown that enhanced noradrenergic function in limbic forebrain contributes to HPA sensitisation. In the present study, we investigated whether chronic intermittent cold also induced changes in noradrenergic function in the paraventricular nucleus (PVN), the primary mediator of the HPA stress response. Rats were exposed to chronic intermittent cold (7 days, 6 h per day, 4 degrees C). On the day after final cold exposure, there were no differences in baseline plasma ACTH, but the peak ACTH response to 30 min of acute immobilisation stress was greater in cold-stressed rats compared to controls. Bilateral microinjection of the alpha(1)-adrenergic receptor antagonist benoxathian into the PVN reduced acute stress-induced adrenocorticotrophic hormone (ACTH) levels by approximately 25% in controls. Furthermore, in cold-stressed rats, all of the sensitisation of the ACTH response was blocked by benoxathian, to a level comparable to benoxathian-treated controls. In a second study using microdialysis to measure norepinephrine release in the PVN, there were no differences in either baseline or acute stress-induced increases in norepinephrine release in the PVN of cold-stressed rats compared to controls. Thus, in a third study, we tested potential alterations in postsynaptic alpha(1)-receptor sensitivity after chronic cold stress. Dose-dependent activation of ACTH secretion by microinjection of the alpha(1)-adrenergic receptor agonist, phenylephrine, into the PVN was significantly enhanced in cold-stressed rats compared to controls. Thus, the sensitised HPA response to acute stress after chronic intermittent cold exposure is at least partly attributable to an enhanced response to alpha1-adrenergic receptor activation in the PVN. Chronic stress-induced plasticity in the acute stress response may be important for stress adaptation, but may also contribute to pathophysiological conditions associated with stress. Thus, understanding the neural mechanisms underlying such adaptations may help us understand the aetiology of such disorders, and contribute to the future development of more effective treatment or prevention strategies.
Collapse
Affiliation(s)
- S Ma
- Department of Pharmacology and Center for Biomedical Neuroscience, University of Texas Health Science Center, San Antonio, TX 78229-3900, USA
| | | |
Collapse
|
38
|
Woodcock EA, Matkovich SJ. Ins(1,4,5)P3 receptors and inositol phosphates in the heart-evolutionary artefacts or active signal transducers? Pharmacol Ther 2005; 107:240-51. [PMID: 15908009 DOI: 10.1016/j.pharmthera.2005.04.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The generation of the second messenger inositol 1,4,5-trisphosphate (Ins(1,4,5)P(3)) and its associated release of Ca(2+) from internal stores is a highly conserved module in intracellular signaling from Drosophila to mammals. Many cell types, often nonexcitable cells, depend on this pathway to couple external signals to intracellular Ca(2+) release. However, despite the presence of the requisite Ins(1,4,5)P(3) signaling machinery, excitable cells such as cardiac myocytes employ a robust alternate system of intracellular Ca(2+) release, namely, a coupled system of Ca(2+) influx, followed by Ca(2+) release via the IP(3)R-related ryanodine receptors. In these systems, Ins(1,4,5)P(3) signaling pathways appear to be largely dormant. In this review, we consider the general features of inositol phosphate (InsP) responses in cardiac myocytes and the molecules mediating these responses. The spatial localization of Ins(1,4,5)P(3) generation and Ins(1,4,5)P(3) receptor (IP(3)Rs) is likely of key importance, and we examine the state of knowledge in atrial, ventricular, and Purkinje myocytes. Several studies have implicated Ins(1,4,5)P(3) generation in both arrhythmogenic and hypertrophic responses, and possible mechanisms involving Ins(1,4,5)P(3) are discussed. While Ins(1,4,5)P(3) is unlikely to be a key player in cardiac excitation-contraction (EC) coupling, its potential role in an alternate Ca(2+) release system to signal changes in gene transcription warrants further investigation. Such studies will help to determine whether cardiac Ins(1,4,5)P(3) generation represents a vestigial pathway or plays an active role in cardiac signaling.
Collapse
Affiliation(s)
- Elizabeth A Woodcock
- Cellular Biochemistry Laboratory, Baker Heart Research Institute, Commercial Road, Melbourne, Australia.
| | | |
Collapse
|
39
|
Lei B, Morris DP, Smith MP, Svetkey LP, Newman MF, Rotter JI, Buchanan TA, Beckstrom-Sternberg SM, Green ED, Schwinn DA. Novel human alpha1a-adrenoceptor single nucleotide polymorphisms alter receptor pharmacology and biological function. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2005; 371:229-39. [PMID: 15900517 PMCID: PMC2367253 DOI: 10.1007/s00210-005-1019-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2004] [Accepted: 12/21/2004] [Indexed: 11/26/2022]
Abstract
We identified nine naturally-occurring human single nucleotide polymorphisms (SNPs) in the alpha(1a)-adrenoceptor (alpha(1a)AR) coding region, seven of which result in amino acid change. Utilizing rat-1 fibroblasts stably expressing wild type alpha(1a)AR or each SNP at both high and low levels, we investigated the effect of these SNPs on receptor function. Compared with wild type, two SNPs (R166K, V311I) cause a decrease in binding affinity for agonists norepinephrine, epinephrine, and phenylephrine, and also shift the dose-response curve for norepinephrine stimulation of inositol phosphate (IP) production to the right (reduced potency) without altering maximal IP activity. In addition, SNP V311I and I200S display altered antagonist binding. Interestingly, a receptor with SNP G247R (located in the third intracellular loop) displays increased maximal receptor IP activity and stimulates cell growth. The increased receptor signaling for alpha(1a)AR G247R is not mediated by altered ligand binding or a deficiency in agonist-mediated desensitization, but appears to be related to enhanced receptor-G protein coupling. In conclusion, four naturally-occurring human alpha(1a)AR SNPs induce altered receptor pharmacology and/or biological activity. This finding has potentially important implications in many areas of medicine and can be used to guide alpha(1a)AR SNP choice for future clinical studies.
Collapse
MESH Headings
- Adrenergic alpha-Agonists/pharmacology
- Adrenergic alpha-Antagonists/pharmacology
- Amino Acid Sequence
- Amino Acid Substitution
- Animals
- Cell Division/genetics
- Cells, Cultured
- Dose-Response Relationship, Drug
- GTP-Binding Proteins/metabolism
- Humans
- Molecular Sequence Data
- Mutagenesis, Site-Directed
- Phosphoric Monoester Hydrolases/metabolism
- Polymorphism, Single Nucleotide
- Radioligand Assay
- Rats
- Receptors, Adrenergic, alpha-1/drug effects
- Receptors, Adrenergic, alpha-1/genetics
- Receptors, Adrenergic, alpha-1/metabolism
- Signal Transduction
- Transfection
Collapse
Affiliation(s)
- Beilei Lei
- Department of Anesthesiology, Duke University Medical Center, Box 3094 Durham, NC, 27710, USA
- Department of Pharmacology/Cancer Biology Duke University Medical Center, Durham, NC, USA
| | - Daniel P. Morris
- Department of Anesthesiology, Duke University Medical Center, Box 3094 Durham, NC, 27710, USA
- Department of Pharmacology/Cancer Biology Duke University Medical Center, Durham, NC, USA
| | - Michael P. Smith
- Department of Anesthesiology, Duke University Medical Center, Box 3094 Durham, NC, 27710, USA
- Department of Pharmacology/Cancer Biology Duke University Medical Center, Durham, NC, USA
| | - Laura P. Svetkey
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Mark F. Newman
- Department of Anesthesiology, Duke University Medical Center, Box 3094 Durham, NC, 27710, USA
| | - Jerome I. Rotter
- Department of Medicine, Cedars-Sinai Medical Center and the University of California, Los Angeles, CA, USA
- Department of Pediatrics, Cedars-Sinai Medical Center and the University of California, Los Angeles, CA, USA
- Department of Human Genetics, Cedars-Sinai Medical Center and the University of California, Los Angeles, CA, USA
| | - Thomas A. Buchanan
- Department of Medicine, University of Southern California, Los Angeles, CA, USA
| | | | - Eric D. Green
- Genome Technology Branch and NIH Intramural Sequencing Center, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Debra A. Schwinn
- Department of Anesthesiology, Duke University Medical Center, Box 3094 Durham, NC, 27710, USA, e-mail: , Tel.: +1-919-6814781, Fax: +1-919-6814776
- Department of Pharmacology/Cancer Biology Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
40
|
Zhang Y, Yan J, Chen K, Song Y, Lu Z, Chen M, Han C, Zhang Y. Different roles of alpha1-adrenoceptor subtypes in mediating cardiomyocyte protein synthesis in neonatal rats. Clin Exp Pharmacol Physiol 2005; 31:626-33. [PMID: 15479171 DOI: 10.1111/j.1440-1681.2004.04063.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
1. Three different alpha1-adrenoceptor subtypes, designated alpha1A, alpha1B and alpha1D, have been cloned and identified pharmacologically in cardiomyocytes. In vitro studies have suggested that alpha1-adrenoceptors play an important role in facilitating cardiac hypertrophy. However, it remains controversial as to which subtype of alpha1-adrenoceptors is involved in this response. In the present study, we investigated the different role of each alpha1-adrenoceptor subtype in mediating cardiomyocyte protein synthesis, which is a most important characteristic of cardiac hypertrophy in cultured neonatal rat cardiomyocytes. 2. Cardiomyocyte hypertrophy was monitored by the following characteristic phenotypic changes: (i) an increase in protein synthesis; (ii) an increase in total protein content; and (iii) an increase in cardiomyocyte size. 3. The role of each alpha1-adrenoceptor subtype in mediating cardiomyocyte protein synthesis was investigated by the effect of specific alpha1-adrenoceptor subtype-selective antagonists on noradrenaline-induced [3H]-leucine incorporation. In addition, pKB values for alpha1-adrenoceptor subtype-selective antagonists were calculated and compared with the corresponding pKi values to further identify their effects. 4. Activation of alpha1-adrenoceptors by phenylephrine or noradrenaline in the presence of propranolol significantly increased [3H]-leucine incorporation, protein content and cell size. 5. Pre-incubating cardiomyocytes with 5-methyl-urapidil, RS 17053 or WB 4101 significantly inhibited noradrenaline-induced [3H]-leucine incorporation. However, there was no effect when cardiomyocytes were pre-incubated with BMY 7378. The correlation coefficients between pKB values for alpha1-adrenoceptor subtype-selective antagonists and pKi values obtained from cloned alpha1A-, alpha1B- or alpha1D-adrenoceptors were 0.92 (P <0.01), 0.66 (P >0.05) and 0.24 (P >0.05), respectively. 6. Our results suggest that the alpha1-adrenoceptor is dominantly responsible for adrenergic hypertrophy of cultured cardiomyocytes in neonatal rats. The efficiency in mediating cardiomyocyte protein synthesis is alpha1A > alpha1B >> alpha1D.
Collapse
Affiliation(s)
- Yongzhen Zhang
- Institute of Vascular Medicine, Peking University Third Hospital and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, PR China.
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Morris DP, Price RR, Smith MP, Lei B, Schwinn DA. Cellular trafficking of human alpha1a-adrenergic receptors is continuous and primarily agonist-independent. Mol Pharmacol 2004; 66:843-54. [PMID: 15258254 DOI: 10.1124/mol.104.000430] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Alpha1a-adrenergic receptors (alpha1aARs) are present intracellularly and at the cell surface in cultured and natural cell models, where they are subject to agonist-mediated desensitization and internalization. To explore alpha1aAR trafficking, a hemagglutinin (HA)-tagged alpha1aAR/enhanced green fluorescent protein (EGFP) fusion protein was expressed in rat-1 fibroblasts and tracked by EGFP fluorescence and antibody labeling of surface receptors. Confocal analysis of antibody-labeled surface receptors revealed unexpected constitutive internalization in the absence of agonist stimulation. In partial agreement, the inverse agonist prazosin also caused a modest 20 +/- 2% increase in surface receptor levels, suggesting a partial block of constitutive internalization caused by decreased basal activation. However, prazosin was unable to prevent internalization of antibody-tagged surface receptors observed by confocal microscopy or cause obvious redistribution of intracellular receptor to the surface, suggesting that the alpha1aAR is internalizing even in a basal-inactive state. In contrast to the alpha1aAR, surface labeling of an HA-tagged alpha1b-EGFP fusion protein did not result in any apparent constitutive internalization. Constitutive internalization of the alpha1aAR seems to occur alongside reversible agonist-induced internalization, and both seem to involve clathrin-mediated endocytosis but not degradation in lysozymes. Surface receptor density must be maintained by recycling, because the protein synthesis inhibitor cycloheximide has no effect on total or surface receptor density in agonist-treated or untreated cells for 6 h. Constitutive agonist-independent trafficking of alpha1aARs may provide a novel mechanism by which an internal pool of alpha1aARs are maintained and recycled to allow continuous agonist-induced signaling.
Collapse
Affiliation(s)
- Daniel P Morris
- Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | | | |
Collapse
|
42
|
Tzanidis A, Hannan RD, Thomas WG, Onan D, Autelitano DJ, See F, Kelly DJ, Gilbert RE, Krum H. Direct actions of urotensin II on the heart: implications for cardiac fibrosis and hypertrophy. Circ Res 2003; 93:246-53. [PMID: 12842917 DOI: 10.1161/01.res.0000084382.64418.bc] [Citation(s) in RCA: 164] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Urotensin II (UII) is a somatostatin-like peptide recently identified as a potent vasoconstrictor. In this study, we examined whether UII promotes cardiac remodeling through nonhemodynamic effects on the myocardium. In a rat model of heart failure after myocardial infarction (MI), increased UII peptide and UII receptor protein expression was observed in both infarct and noninfarct regions of the left ventricle compared with sham. Moreover, post-MI remodeling was associated with a significant 75% increase in UII receptor gene expression in the heart (P<0.05 versus sham controls), with this increase noted in both regions of the left ventricle. In vitro, UII (10-7 mol/L) stimulation of neonatal cardiac fibroblasts increased the level of mRNA transcripts for procollagens alpha1(I), alpha1(III), and fibronectin by 139+/-15% (P<0.01), 59+/-5% (P<0.05), and 141+/-14% (P<0.01), respectively, with a concomitant 23+/-2% increase in collagen peptide synthesis as determined by 3H-proline incorporation (P<0.01). UII had no effect on cellular hypertrophy, as determined by changes in total protein content in isolated neonatal cardiomyocytes. However, expression of recombinant rat UII receptor in neonatal cardiomyocytes resulted in significant UII-dependent activation of hypertrophic signaling as demonstrated by increased total protein content (unstimulated, 122.4+/-4.0 microg/well; rat UII, 147.6+/-7.0 microg/well; P<0.01) and activation of the hypertrophic phenotype through Galpha(q)- and Ras-dependent pathways. These results indicate that, in addition to potent hemodynamic effects, UII may be implicated in myocardial fibrogenesis through increased collagen synthesis by cardiac fibroblasts and may also be an important determinant of pathological cardiac hypertrophy in conditions characterized by UII receptor upregulation.
Collapse
MESH Headings
- Adenoviridae/genetics
- Animals
- Animals, Newborn
- Cell Division/drug effects
- Cells, Cultured
- Collagen/biosynthesis
- Disease Models, Animal
- Fibroblasts/cytology
- Fibroblasts/drug effects
- Fibroblasts/metabolism
- Fibrosis/etiology
- Fibrosis/pathology
- Fibrosis/physiopathology
- Gene Expression Regulation
- Heart/drug effects
- Heart/physiopathology
- Heart Failure/etiology
- Heart Failure/pathology
- Heart Failure/physiopathology
- Heart Ventricles/drug effects
- Heart Ventricles/pathology
- Heart Ventricles/physiopathology
- Hypertrophy/etiology
- Hypertrophy/pathology
- Hypertrophy/physiopathology
- Immunohistochemistry
- Myocardial Infarction/complications
- Myocardium/metabolism
- Myocardium/pathology
- Myocytes, Cardiac/cytology
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- RNA, Messenger/metabolism
- Rats
- Rats, Sprague-Dawley
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Receptors, G-Protein-Coupled
- Transfection
- Urotensins/metabolism
- Urotensins/pharmacology
Collapse
Affiliation(s)
- Alex Tzanidis
- NHMRC Centre of Clinical Research Excellence in Therapeutics, Department of MedicineEpidemiology and Preventive Medicine, Monash University Medical School, Prahran, Victoria, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Brandenburger Y, Arthur JF, Woodcock EA, Du XJ, Gao XM, Autelitano DJ, Rothblum LI, Hannan RD. Cardiac hypertrophy in vivo is associated with increased expression of the ribosomal gene transcription factor UBF. FEBS Lett 2003; 548:79-84. [PMID: 12885411 DOI: 10.1016/s0014-5793(03)00744-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The ribosomal DNA transcription-specific factor, UBF, is a key target for the regulation of ribosomal RNA synthesis and hypertrophic growth of isolated neonatal cardiomyocytes. In this study, we have examined whether UBF expression is also an important determinant of cardiac growth rates in vivo. We show that rDNA transcription, rRNA synthesis and UBF expression in left ventricular myocytes isolated from mice 1-6 weeks following transverse aortic constriction were significantly increased (2.5-3.5-fold) compared to the levels in myocytes from the left ventricle of sham-operated mice.
Collapse
Affiliation(s)
- Yves Brandenburger
- Molecular Cardiology, Baker Heart Research Institute, P.O. Box 6492, St Kilda Road Central, Melbourne, Vic., 8008, Australia
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Pönicke K, Heinroth-Hoffmann I, Brodde OE. Role of beta 1- and beta 2-adrenoceptors in hypertrophic and apoptotic effects of noradrenaline and adrenaline in adult rat ventricular cardiomyocytes. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2003; 367:592-9. [PMID: 12750877 DOI: 10.1007/s00210-003-0754-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2002] [Accepted: 03/18/2003] [Indexed: 12/20/2022]
Abstract
In adult rat ventricular cardiomyocytes alpha1-adrenoceptor (AR) stimulation causes increases in protein synthesis. On the other hand beta1-AR stimulation inhibits protein synthesis, and evokes apoptotic cell death. We studied, in adult rat ventricular cardiomyocytes, effects of noradrenaline (NA), adrenaline (ADR) and phenylephrine (PE) on protein synthesis (assessed by [3H]-phenylalanine incorporation into the cardiomyocytes) in relation to effects on early apoptosis (measured by Annexin V/propidium iodide staining). PE (10(-9)-10(-5) M) induced protein synthesis was not affected by the beta1-AR blocker CGP 20712A (CGP, 300 nM) or beta2-AR blocker ICI 118,551 (ICI, 55 nM). ADR (10(-9)-10(-5) M) induced protein synthesis was enhanced by CGP and decreased by ICI. Pretreatment of the cardiomyocytes with pertussis toxin (PTX) decreased NA- and ADR- induced protein synthesis, but did not affect PE-effects. NA (10(-5) M) and ADR (10(-5) M) caused a significant increase in the number of apoptotic cells; these effects were enhanced by PTX-treatment, abolished by CGP, but not significantly affected by ICI. Furthermore, there was a significant negative correlation between catecholamine-evoked apoptosis and catecholamine-induced hypertrophic effects. We conclude that, in ventricular cardiomyocytes of adult rats, growth-promoting effects of NA and ADR are composed of alpha1A-AR mediated increase in protein synthesis and beta1-AR mediated apoptosis that counteracts increases in protein synthesis. The role of beta2-adrenoceptor appears to be a balance of antiapoptotic effects via a PTX-sensitive pathway and proapoptotic effects via a GS-adenylyl cyclase pathway.
Collapse
Affiliation(s)
- Klaus Pönicke
- Institute of Pharmacology, University of Halle, 06097 Halle (Saale), Germany
| | | | | |
Collapse
|
45
|
Michelotti GA, Bauman MJ, Smith MP, Schwinn DA. Cloning and characterization of the rat alpha 1a-adrenergic receptor gene promoter. Demonstration of cell specificity and regulation by hypoxia. J Biol Chem 2003; 278:8693-705. [PMID: 12471020 DOI: 10.1074/jbc.m211986200] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Recent studies reveal important and distinct roles for cardiac alpha(1a) adrenergic receptors (alpha(1a)ARs). Surprisingly, given their importance in myocardial ischemia/reperfusion, hypoxia, and hypertrophy as well as frequent use of rat cardiomyocyte model systems, the rat alpha(1a)AR gene promoter has never been characterized. Therefore, we isolated 3.9 kb of rat alpha(1a)AR 5'-untranslated region and 5'-regulatory sequences and identified multiple transcription initiation sites. One proximal (P1) and several clustered upstream distal promoters (P2, P3, and P4) were delineated. Sequences surrounding both proximal and distal promoters lack typical TATA or CCAAT boxes but contain cis-elements for multiple myocardium-relevant nuclear regulators including Sp1, GATA, and CREB, findings consistent with enhanced cardiac basal alpha(1a)AR expression seen in Northern blots and reporter constructs. Promoter analysis using deletion reporter constructs reveals, in addition to a powerful upstream enhancer, a key region (-558/-542) important in regulating all alpha(1a)AR promoters with hypoxic stress. Gel shift analysis of this 14-bp region confirms a hypoxia-induced shift independent of direct hypoxia-inducible factor binding. Mutational analysis of this sequence identifies a novel 9-bp hypoxia response element, the loss of which severely attenuates hypoxia-mediated repression of alpha(1a)AR transcription. These findings for the alpha(1a) gene should facilitate elucidation of alpha(1)AR-mediated mechanisms involved in distinct myocardial pathologies.
Collapse
MESH Headings
- 5' Untranslated Regions
- Animals
- Base Sequence
- Blotting, Northern
- Cell Hypoxia
- Cells, Cultured
- Cloning, Molecular
- DNA
- Electrophoretic Mobility Shift Assay
- Gene Expression Regulation
- Promoter Regions, Genetic
- RNA, Messenger/genetics
- Rats
- Receptors, Adrenergic, alpha-1/chemistry
- Receptors, Adrenergic, alpha-1/genetics
- Receptors, Adrenergic, alpha-1/metabolism
Collapse
Affiliation(s)
- Gregory A Michelotti
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina 27710, USA
| | | | | | | |
Collapse
|
46
|
Kim DJ, Park SH, Lim CS, Chun JS, Kim JK, Song WK. Cellular localization of integrin isoforms in phenylephrine-induced hypertrophic cardiac myocytes. Cell Biochem Funct 2003; 21:41-8. [PMID: 12579520 DOI: 10.1002/cbf.988] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Cardiac hypertrophy is characterized by remodeling of the extracellular matrix (ECM). Integrins are cell-surface molecules that link the ECM to the cellular cytoskeleton where they play roles as signaling molecules and transducers of mechanical force. To clarify the possible roles of integrins in cardiac myocyte hypertrophy, we investigated the cellular localization and expression of ECM proteins and integrins in both normal cardiac myocytes and phenylephrine-induced hypertrophic myocytes. Addition of phenylephrine (PE) to cultured neonatal cardiac myocytes induced sarcomeric organization, increase in cell size, and synthesis of the hypertrophic marker, atrial natriuretic factor (ANF). In particular, fibronectin and collagen underwent dramatic localization changes during PE-induced cardiac hypertrophy. Significant changes were noted in the cellular localization of the respective collagen and fibronectin receptors, integrin alpha1 and alpha5, from diffuse to a sarcomeric banding pattern. Expression levels of integrins were also increased during hypertrophy. Treatment with okadaic acid (OA), an inhibitor of protein phosphatase 2A (PP2A), resulted in inhibition of hypertrophic response. These results suggest that dephosphorylation of integrin beta1 may be important in the induction of cardiac hypertrophy.
Collapse
Affiliation(s)
- Dae Joong Kim
- Department of Life Science, Kwangju Institute of Science and Technology, Kwangju 500-712, Korea
| | | | | | | | | | | |
Collapse
|
47
|
Steinberg SF. Alpha(1)-adrenergic receptor subtype function in cardiomyocytes: lessons from genetic models in mice. J Mol Cell Cardiol 2002; 34:1141-5. [PMID: 12392888 DOI: 10.1006/jmcc.2002.2057] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
48
|
Woodcock EA, Wang BH, Arthur JF, Lennard A, Matkovich SJ, Du XJ, Brown JH, Hannan RD. Inositol polyphosphate 1-phosphatase is a novel antihypertrophic factor. J Biol Chem 2002; 277:22734-42. [PMID: 11932254 DOI: 10.1074/jbc.m110405200] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Activation of G(q)-coupled alpha(1)-adrenergic receptors leads to hypertrophic growth of neonatal rat ventricular cardiomyocytes that is associated with increased expression of hypertrophy-related genes, including atrial natriuretic peptide (ANP) and myosin light chain-2 (MLC), as well as increased ribosome synthesis. The role of inositol phosphates in signaling pathways involved in these changes in gene expression was examined by overexpressing inositol phosphate-metabolizing enzymes and determining effects on ANP, MLC, and 45 S ribosomal gene expression following co-transfection of appropriate reporter gene constructs. Overexpression of enzymes that metabolize inositol 1,4,5-trisphosphate did not reduce ANP or MLC responses, but overexpression of the enzyme primarily responsible for metabolism of inositol 4,5-bisphosphate (Ins(1,4)P(2)), inositol polyphosphate 1-phosphatase (INPP), reduced ANP and MLC responses associated with alpha(1)-adrenergic receptor-mediated hypertrophy. Similarly overexpressed INPP reduced ANP and MLC responses associated with contraction-induced hypertrophy. In addition, overexpression of INPP reduced the increase in ribosomal DNA transcription associated with both hypertrophic models. Hypertrophied cells from both cell models as well as ventricular tissue from mouse hearts hypertrophied by pressure overload in vivo contained heightened levels of Ins(1,4)P(2), suggesting reduced INPP activity in three different models of hypertrophy. These studies provide evidence for an involvement of Ins(1,4)P(2) in hypertrophic signaling pathways in ventricular myocytes.
Collapse
MESH Headings
- Animals
- Atrial Natriuretic Factor/metabolism
- Blotting, Western
- CHO Cells
- Cardiac Myosins/metabolism
- Cells, Cultured
- Cloning, Molecular
- Cricetinae
- DNA, Complementary/metabolism
- DNA, Ribosomal/metabolism
- Electrophoresis, Polyacrylamide Gel
- Gene Library
- Genes, Reporter
- Humans
- Hypertrophy/drug therapy
- Inositol Phosphates/metabolism
- Mice
- Mice, Inbred C57BL
- Models, Chemical
- Myocardium/metabolism
- Myocardium/pathology
- Myosin Light Chains/metabolism
- Phosphoric Monoester Hydrolases/chemistry
- Phosphoric Monoester Hydrolases/pharmacology
- Promoter Regions, Genetic
- Protein Binding
- Protein Kinase C/metabolism
- Rats
- Signal Transduction
- Time Factors
- Transcription, Genetic
- Transfection
Collapse
Affiliation(s)
- Elizabeth A Woodcock
- Cellular Biochemistry Laboratory, Baker Medical Research Institute, PO Box 6492, St. Kilda Road Central, Melbourne, 8008, Victoria, Australia.
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Price RR, Morris DP, Biswas G, Smith MP, Schwinn DA. Acute agonist-mediated desensitization of the human alpha 1a-adrenergic receptor is primarily independent of carboxyl terminus regulation: implications for regulation of alpha 1aAR splice variants. J Biol Chem 2002; 277:9570-9. [PMID: 11781325 DOI: 10.1074/jbc.m111762200] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Despite important roles in myocardial hypertrophy and benign prostatic hyperplasia, little is known about acute effects of agonist stimulation on alpha(1a)-adrenergic receptor (alpha(1a)AR) signaling and function. Regulatory mechanisms are likely complex since 12 distinct human alpha(1a)AR carboxyl-terminal splice variants have been isolated. After determining the predominance of the alpha(1a-1)AR isoform in human heart and prostate, we stably expressed an epitope-tagged alpha(1a-1)AR cDNA in rat-1 fibroblasts and subsequently examined regulation of signaling, phosphorylation, and internalization of the receptor. Human alpha(1a)AR-mediated inositol phosphate signaling is acutely desensitized in response to both agonist and phorbol 12-myristate 13-acetate (PMA) exposure. Concurrent with desensitization, alpha(1a)ARs in (32)P(i)-labeled cells are rapidly phosphorylated in response to both NE and PMA stimulation. Despite the ability of PKC to desensitize alpha(1a)ARs when directly activated with PMA, inhibitors of PKC have no effect on agonist-mediated desensitization. In contrast, involvement of GRK kinases is suggested by the ability of GRK2 to desensitize alpha(1a)ARs. Internalization of cell surface alpha(1a)ARs also occurs in response to agonist stimulation (but not PKC activation), but is initiated more slowly than receptor desensitization. Significantly, deletion of the alpha(1a)AR carboxyl terminus has no effect on receptor internalization or either agonist-induced or GRK-mediated receptor desensitization. Because mechanisms underlying acute agonist-mediated regulation of human alpha(1a)ARs are primarily independent of the carboxyl terminus, they may be common to all functional alpha(1a)AR isoforms.
Collapse
Affiliation(s)
- R Reyn Price
- Department of Pharmacology, Duke University Medical Center, Durham, North Carolina 27710, USA
| | | | | | | | | |
Collapse
|
50
|
Thomas WG, Brandenburger Y, Autelitano DJ, Pham T, Qian H, Hannan RD. Adenoviral-directed expression of the type 1A angiotensin receptor promotes cardiomyocyte hypertrophy via transactivation of the epidermal growth factor receptor. Circ Res 2002; 90:135-42. [PMID: 11834705 DOI: 10.1161/hh0202.104109] [Citation(s) in RCA: 120] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Angiotensin II (Ang II) may cause cardiac hypertrophy via type 1 Ang II receptors (AT(1)) on cardiomyocytes and through growth factors released from cardiac fibroblasts. Whereas cardiomyocyte-specific AT(1) receptor expression produces cardiac hypertrophy and remodeling in vivo, delineation of the signals that mediate growth to Ang II is challenging because the prevailing in vitro model (cultured neonatal cardiomyocytes) expresses low levels of AT(1) receptor and responds inconsistently to Ang II. In this study, when AT(1A) receptors were expressed using adenovirus in cultured neonatal cardiomyocytes, Ang II stimulated a robust hypertrophy that was not secondary to the release of cardiac fibroblast-derived factors, specifically endothelin-1. Hypertrophy was accompanied by the induction of the immediate-early response genes, c-fos and c-jun, and reexpression of atrial natriuretic peptide (ANP). Ang II-induced activation of an ANP promoter-reporter was inhibited by the dominant/negative mutants, GalphaqI and N17Ras, indicating that hypertrophic signaling by the AT(1A) receptor is via heterotrimeric G protein coupling and downstream Ras pathways. AT(1A)-mediated cardiomyocyte hypertrophy and mitogen-activated protein kinase (MAPK) activation were inhibited by the MAPK kinase inhibitor, PD98059, and the epidermal growth factor (EGF) receptor kinase antagonist, AG1478, but not by PKC inhibitor, bisindolylmaleimide-1. Moreover, Ang II-induced MAPK activation was prevented by treatment with a matrix metalloproteinase inhibitor, consistent with the tyrosine phosphorylation of the EGF receptor in response to AT(1A) receptor activation. These data unequivocally demonstrate that Ang II can directly promote cardiac myocyte growth via AT(1A) receptors expressed on these cells and reveal for the first time the important contribution of EGF receptor-transactivated MAPK signaling to this process.
Collapse
Affiliation(s)
- Walter G Thomas
- Molecular Endocrinology, Baker Medical Research Institute, Melbourne, Australia
| | | | | | | | | | | |
Collapse
|