1
|
Tian T, Yu Q, Yang D, Zhang X, Zhang C, Li J, Luo T, Zhang K, Lv X, Wang Y, Wang H, Li H. Endothelial α 1-adrenergic receptor activation improves cardiac function in septic mice via PKC-ERK/p38MAPK signaling pathway. Int Immunopharmacol 2024; 141:112937. [PMID: 39182270 DOI: 10.1016/j.intimp.2024.112937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 07/30/2024] [Accepted: 08/12/2024] [Indexed: 08/27/2024]
Abstract
Cardiomyopathy is particularly common in septic patients. Our previous studies have shown that activation of the alpha 1 adrenergic receptor (α1-AR) on cardiomyocytes inhibits sepsis-induced myocardial dysfunction. However, the role of cardiac endothelial α1-AR in septic cardiomyopathy has not been determined. Here, we identified α1-AR expression in mouse and human endothelial cells and showed that activation of α1-AR with phenylephrine (PE) improved cardiac function and survival by preventing cardiac endothelial injury in septic mice. Mechanistically, activating α1-AR with PE decreased the expression of ICAM-1, VCAM-1, iNOS, E-selectin, and p-p38MAPK, while promoting PKC and ERK1/2 phosphorylation in LPS-treated endothelial cells. These effects were abolished by a PKC inhibitor or α1-AR antagonist. PE also reduced p65 nuclear translocation, but this suppression is not blocked by PKC inhibition. Treatment with U0126 (a specific ERK1/2 inhibitor) reversed the effects of PE on p38MAPK phosphorylation. Our results demonstrate that cardiac endothelial α1-AR activation prevents sepsis-induced myocardial dysfunction in mice by inhibiting the endothelial injury via PKC-ERK/p38MAPK signaling pathway and a PKC-independent inhibition of p65 nuclear translocation. These findings offer a new perspective for septic patients with cardiac dysfunction by inhibiting cardiac endothelial cell injury through α1-AR activation.
Collapse
Affiliation(s)
- Tian Tian
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Qing Yu
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Duomeng Yang
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Xue Zhang
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Chanjuan Zhang
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Jianling Li
- Department of Anesthesiology, The First Affiliated Hospital, Jinan University, Guangzhou 510632, Guangdong, China
| | - Tao Luo
- Department of Pathophysiology, Zhuhai Campus of Zunyi Medical University, Zhuhai 519041, China
| | - Keke Zhang
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Xiuxiu Lv
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Yiyang Wang
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Huadong Wang
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Hongmei Li
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
2
|
Dong ZK, Wang YF, Li WP, Jin WL. Neurobiology of cancer: Adrenergic signaling and drug repurposing. Pharmacol Ther 2024:108750. [PMID: 39527999 DOI: 10.1016/j.pharmthera.2024.108750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 10/04/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Cancer neuroscience, as an emerging converging discipline, provides us with new perspectives on the interactions between the nervous system and cancer progression. As the sympathetic nervous system, in particular adrenergic signaling, plays an important role in the regulation of tumor activity at every hierarchical level of life, from the tumor cell to the tumor microenvironment, and to the tumor macroenvironment, it is highly desirable to dissect its effects. Considering the far-reaching implications of drug repurposing for antitumor drug development, such a large number of adrenergic receptor antagonists on the market has great potential as one of the means of antitumor therapy, either as primary or adjuvant therapy. Therefore, this review aims to summarize the impact of adrenergic signaling on cancer development and to assess the status and prospects of intervening in adrenergic signaling as a therapeutic tool against tumors.
Collapse
Affiliation(s)
- Zi-Kai Dong
- The First Clinical Medical College of Lanzhou University, Lanzhou 730000, PR China; Institute of Cancer Neuroscience, Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, The First Clinical Medical College of Lanzhou University, Lanzhou 730000, PR China
| | - Yong-Fei Wang
- The First Clinical Medical College of Lanzhou University, Lanzhou 730000, PR China; Institute of Cancer Neuroscience, Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, The First Clinical Medical College of Lanzhou University, Lanzhou 730000, PR China
| | - Wei-Ping Li
- The First Clinical Medical College of Lanzhou University, Lanzhou 730000, PR China; Department of Urology, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, PR China
| | - Wei-Lin Jin
- The First Clinical Medical College of Lanzhou University, Lanzhou 730000, PR China; Institute of Cancer Neuroscience, Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, The First Clinical Medical College of Lanzhou University, Lanzhou 730000, PR China.
| |
Collapse
|
3
|
Kimball TH, Gromova T, Gehred ND, Chapski DJ, Wang K, Vaseghi M, Fischer MA, Lefer DJ, Vondriska TM. Rapid onset fibrotic remodeling and ventricular dysfunction induced by phenylephrine involve targeted reprogramming of myocyte and fibroblast transcriptomes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.11.617933. [PMID: 39464022 PMCID: PMC11507669 DOI: 10.1101/2024.10.11.617933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Catecholamine dysregulation is a common feature of multiple acute and chronic cardiac conditions, including heart failure. To investigate the role of altered α-adrenergic stimulation on cardiac function, we developed a short-term exposure model, administering phenylephrine subcutaneously to mice for one week. Compared to vehicle-injected controls, phenylephrine-treated animals exhibited increased ejection fraction, decreased chamber size, diastolic dysfunction and ventricular hypertrophy in the absence of hypertension. Remarkably, these animals developed extensive fibrotic remodeling of the tissue that plateaued at 24 hours and myocyte hypertrophy localized to regions of fibrotic deposition after 3 days of treatment. Transcriptome analyses of purified myocyte and fibroblast populations from these hearts revealed an unexpected role for myocytes in the production of extracellular matrix. Comparison with other models of cardiac stress, including pressure overload hypertrophy and cytokine activation of fibroblasts, identified stimulus-specific transcriptional circuits associated with cardiac pathology. Given the rapid, robust fibrotic response that preceded myocyte hypertrophy, intercellular communication analyses were conducted to investigate fibroblast to myocyte signaling, identifying potential crosstalk between these cells. These studies thoroughly describe and phenotypically characterize a new model of short-term catecholamine stress and provide an atlas of transcriptional remodeling in myocytes and fibroblasts.
Collapse
Affiliation(s)
- Todd H. Kimball
- Departments of Anesthesiology & Perioperative Medicine, David Geffen School of Medicine
| | - Tatiana Gromova
- Departments of Anesthesiology & Perioperative Medicine, David Geffen School of Medicine
| | - Natalie D. Gehred
- Departments of Anesthesiology & Perioperative Medicine, David Geffen School of Medicine
| | - Douglas J. Chapski
- Departments of Anesthesiology & Perioperative Medicine, David Geffen School of Medicine
| | - Ke Wang
- Department of Cardiac Surgery, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles
| | - Marmar Vaseghi
- Cardiac Arrhythmia Center, Division of Cardiology, Department of Medicine, University of California, Los Angeles
| | - Matthew A. Fischer
- Departments of Anesthesiology & Perioperative Medicine, David Geffen School of Medicine
| | - David J. Lefer
- Department of Cardiac Surgery, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles
| | - Thomas M. Vondriska
- Departments of Anesthesiology & Perioperative Medicine, David Geffen School of Medicine
- Physiology, David Geffen School of Medicine
- Medicine, David Geffen School of Medicine
- Molecular Biology Institute, University of California, Los Angeles
| |
Collapse
|
4
|
Hamaguchi S, Agata N, Seki M, Namekata I, Tanaka H. Developmental Changes in the Excitation-Contraction Mechanisms of the Ventricular Myocardium and Their Sympathetic Regulation in Small Experimental Animals. J Cardiovasc Dev Dis 2024; 11:267. [PMID: 39330325 PMCID: PMC11432613 DOI: 10.3390/jcdd11090267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 08/02/2024] [Accepted: 08/11/2024] [Indexed: 09/28/2024] Open
Abstract
The developmental changes in the excitation-contraction mechanisms of the ventricular myocardium of small animals (guinea pig, rat, mouse) and their sympathetic regulation will be summarized. The action potential duration monotonically decreases during pre- and postnatal development in the rat and mouse, while in the guinea pig it decreases during the fetal stage but turns into an increase just before birth. Such changes can be attributed to changes in the repolarizing potassium currents. The T-tubule and the sarcoplasmic reticulum are scarcely present in the fetal cardiomyocyte, but increase during postnatal development. This causes a developmental shift in the Ca2+ handling from a sarcolemma-dependent mechanism to a sarcoplasmic reticulum-dependent mechanism. The sensitivity for beta-adrenoceptor-mediated positive inotropy decreases during early postnatal development, which parallels the increase in sympathetic nerve innervation. The alpha-adrenoceptor-mediated inotropy in the mouse changes from positive in the neonate to negative in the adult. This can be explained by the change in the excitation-contraction mechanism mentioned above. The shortening of the action potential duration enhances trans-sarcolemmal Ca2+ extrusion by the Na+-Ca2+ exchanger. The sarcoplasmic reticulum-dependent mechanism of contraction in the adult allows Na+-Ca2+ exchanger activity to cause negative inotropy, a mechanism not observed in neonatal myocardium. Such developmental studies would provide clues towards a more comprehensive understanding of cardiac function.
Collapse
Affiliation(s)
| | | | | | | | - Hikaru Tanaka
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Toho University, Funabashi 274-8510, Japan; (S.H.); (N.A.); (M.S.); (I.N.)
| |
Collapse
|
5
|
Zhang J, Sandroni PB, Huang W, Gao X, Oswalt L, Schroder MA, Lee S, Shih YYI, Huang HYS, Swigart PM, Myagmar BE, Simpson PC, Rossi JS, Schisler JC, Jensen BC. Cardiomyocyte Alpha-1A Adrenergic Receptors Mitigate Postinfarct Remodeling and Mortality by Constraining Necroptosis. JACC Basic Transl Sci 2024; 9:78-96. [PMID: 38362342 PMCID: PMC10864988 DOI: 10.1016/j.jacbts.2023.08.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 08/26/2023] [Accepted: 08/29/2023] [Indexed: 02/17/2024]
Abstract
Clinical studies have shown that α1-adrenergic receptor antagonists (α-blockers) are associated with increased heart failure risk. The mechanism underlying that hazard and whether it arises from direct inhibition of cardiomyocyte α1-ARs or from systemic effects remain unclear. To address these issues, we created a mouse with cardiomyocyte-specific deletion of the α1A-AR subtype and found that it experienced 70% mortality within 7 days of myocardial infarction driven, in part, by excessive activation of necroptosis. We also found that patients taking α-blockers at our center were at increased risk of death after myocardial infarction, providing clinical correlation for our translational animal models.
Collapse
Affiliation(s)
- Jiandong Zhang
- Division of Cardiology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
- UNC McAllister Heart Institute, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Peyton B. Sandroni
- UNC McAllister Heart Institute, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
- Department of Medicine, University of California-San Francisco, San Francisco, California, USA
| | - Wei Huang
- UNC McAllister Heart Institute, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Xiaohua Gao
- Department of Epidemiology, University of North Carolina Gillings School of Public Health, Chapel Hill, North Carolina, USA
| | - Leah Oswalt
- UNC McAllister Heart Institute, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Melissa A. Schroder
- UNC McAllister Heart Institute, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - SungHo Lee
- Center for Animal MRI, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Yen-Yu I. Shih
- Center for Animal MRI, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Hsiao-Ying S. Huang
- Mechanical and Aerospace Engineering Department, North Carolina State University, Raleigh, North Carolina, USA
| | - Philip M. Swigart
- Department of Medicine, University of California-San Francisco, San Francisco, California, USA
- San Francisco VA Medical Center, San Francisco, California, USA
| | - Bat E. Myagmar
- Department of Medicine, University of California-San Francisco, San Francisco, California, USA
- San Francisco VA Medical Center, San Francisco, California, USA
| | - Paul C. Simpson
- Department of Medicine, University of California-San Francisco, San Francisco, California, USA
- San Francisco VA Medical Center, San Francisco, California, USA
| | - Joseph S. Rossi
- Division of Cardiology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Jonathan C. Schisler
- UNC McAllister Heart Institute, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
- Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Brian C. Jensen
- Division of Cardiology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
- UNC McAllister Heart Institute, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
- Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina, USA
| |
Collapse
|
6
|
Roy R, Koch WJ. A (Alpha 1-Adrenergic Receptors), B (Blocking Alpha 1-Adrenergic Receptors), C (Catecholamines): On the Road to Heart Failure. JACC Basic Transl Sci 2024; 9:97-99. [PMID: 38362339 PMCID: PMC10864958 DOI: 10.1016/j.jacbts.2023.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/17/2024]
Abstract
Corresponding Author First Author
Collapse
Affiliation(s)
- Rajika Roy
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, USA
- Cardiovascular Research Center, Duke University School of Medicine, Durham, North Carolina, USA
| | - Walter J. Koch
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, USA
- Cardiovascular Research Center, Duke University School of Medicine, Durham, North Carolina, USA
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| |
Collapse
|
7
|
Baudier C, Fougerousse F, Asselbergs FW, Guedj M, Komajda M, Kotecha D, Thomas Lumbers R, Schmidt AF, Tyl B. Unraveling the relationships between alpha- and beta-adrenergic modulation and the risk of heart failure. Front Cardiovasc Med 2023; 10:1148931. [PMID: 37920183 PMCID: PMC10619754 DOI: 10.3389/fcvm.2023.1148931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 09/22/2023] [Indexed: 11/04/2023] Open
Abstract
Background The effects of α and ß adrenergic receptor modulation on the risk of developing heart failure (HF) remains uncertain due to a lack of randomized controlled trials. This study aimed to estimate the effects of α and ß adrenergic receptors modulation on the risk of HF and to provide proof of principle for genetic target validation studies in HF. Methods Genetic variants within the cis regions encoding the adrenergic receptors α1A, α2B, ß1, and ß2 associated with blood pressure in a 757,601-participant genome-wide association study (GWAS) were selected as instruments to perform a drug target Mendelian randomization study. Effects of these variants on HF risk were derived from the HERMES GWAS (542,362 controls; 40,805 HF cases). Results Lower α1A or ß1 activity was associated with reduced HF risk: odds ratio (OR) 0.83 (95% CI 0.74-0.93, P = 0.001) and 0.95 (95% CI 0.93-0.97, P = 8 × 10-6). Conversely, lower α2B activity was associated with increased HF risk: OR 1.09 (95% CI 1.05-1.12, P = 3 × 10-7). No evidence of an effect of lower ß2 activity on HF risk was found: OR 0.99 (95% CI 0.92-1.07, P = 0.95). Complementary analyses showed that these effects were consistent with those on left ventricular dimensions and acted independently of any potential effect on coronary artery disease. Conclusions This study provides genetic evidence that α1A or ß1 receptor inhibition will likely decrease HF risk, while lower α2B activity may increase this risk. Genetic variant analysis can assist with drug development for HF prevention.
Collapse
Affiliation(s)
- Claire Baudier
- Translational Medicine Division, Institut de Recherches Internationales Servier, Suresnes, France
| | - Françoise Fougerousse
- Center for Therapeutic Innovation Cardiovascular & Metabolic Disease, Institut de Recherches Internationales Servier, Suresnes, France
| | - Folkert W. Asselbergs
- Institute of Health Informatics, University College London, London, United Kingdom
- The National Institute for Health Research University College London Hospitals Biomedical Research Centre, University College London, London, United Kingdom
- Department of Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centre, University of Amsterdam, Amsterdam, Netherlands
| | - Mickael Guedj
- Translational Medicine Division, Institut de Recherches Internationales Servier, Suresnes, France
| | - Michel Komajda
- Department of Cardiology, Hospital Saint Joseph and Sorbonne University, Paris, France
| | - Dipak Kotecha
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom
- West Midlands NHS Secure Data Environment, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
- NIHR Birmingham Biomedical Research Centre, Birmingham, United Kingdom
| | - R. Thomas Lumbers
- Institute of Health Informatics, University College London, London, United Kingdom
- Health Data Research UK London, University College London, London, United Kingdom
- UCL British Heart Foundation Research Accelerator, London, United Kingdom
| | - Amand F. Schmidt
- Institute of Health Informatics, University College London, London, United Kingdom
- Department of Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centre, University of Amsterdam, Amsterdam, Netherlands
- UCL British Heart Foundation Research Accelerator, London, United Kingdom
- Institute of Cardiovascular Science, Faculty of Population Health Sciences, University College London, London, United Kingdom
- Department of Cardiology, Division Heart and Lungs, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Benoît Tyl
- Center for Therapeutic Innovation Cardiovascular & Metabolic Disease, Institut de Recherches Internationales Servier, Suresnes, France
| |
Collapse
|
8
|
Sung DJ, Park S, Noh HJ, Golpasandi S, Eun SH, Lee H, Kim B, Wie J, Seo MS, Park SW, Bae YM. Receptor-specific contributions of caveolae, PKC, and Src tyrosine kinase to serotonergic and adrenergic regulation of Kv channels and vasoconstriction. Life Sci 2023; 328:121903. [PMID: 37394095 DOI: 10.1016/j.lfs.2023.121903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 06/28/2023] [Accepted: 06/28/2023] [Indexed: 07/04/2023]
Abstract
AIMS Caveolae are invaginated, Ω-shaped membrane structures. They are now recognized as portals for signal transduction of multiple chemical and mechanical stimuli. Notably, the contribution of caveolae has been reported to be receptor-specific. However, details of how they differentially contribute to receptor signaling remain unclear. MAIN METHODS Using isometric tension measurements, patch-clamping, and western blotting, we examined the contribution of caveolae and their related signaling pathways to serotonergic (5-HT2A receptor-mediated) and adrenergic (α1-adrenoceptor-mediated) signaling in rat mesenteric arteries. KEY FINDINGS Disruption of caveolae by methyl-β-cyclodextrin effectively blocked vasoconstriction mediated by the 5-HT2A receptor (5-HT2AR), but not by the α1-adrenoceptor. Caveolar disruption selectively impaired 5-HT2AR-mediated voltage-dependent K+ channel (Kv) inhibition, but not α1-adrenoceptor-mediated Kv inhibition. In contrast, both serotonergic and α1-adrenergic effects on vasoconstriction, as well as Kv currents, were similarly blocked by the Src tyrosine kinase inhibitor PP2. However, inhibition of protein kinase C (PKC) by either GO6976 or chelerythrine selectively attenuated the effects mediated by the α1-adrenoceptor, but not by 5-HT2AR. Disruption of caveolae decreased 5-HT2AR-mediated Src phosphorylation, but not α1-adrenoceptor-mediated Src phosphorylation. Finally, the PKC inhibitor GO6976 blocked Src phosphorylation by the α1-adrenoceptor, but not by 5-HT2AR. SIGNIFICANCE 5-HT2AR-mediated Kv inhibition and vasoconstriction are dependent on caveolar integrity and Src tyrosine kinase, but not on PKC. In contrast, α1-adrenoceptor-mediated Kv inhibition and vasoconstriction are not dependent on caveolar integrity, but rather on PKC and Src tyrosine kinase. Caveolae-independent PKC is upstream of Src activation for α1-adrenoceptor-mediated Kv inhibition and vasoconstriction.
Collapse
Affiliation(s)
- Dong Jun Sung
- Department of Sport and Health Studies, College of Biomedical and Health Science, Konkuk University, Chungju 27478, Republic of Korea; Sports Convergence Institute, Konkuk University, Chungju 27478, Republic of Korea; Center for Metabolic Diseases, Konkuk University, Chungju 27478, Republic of Korea; Research Institute for Biomedical & Health Science, Chungju 27478, Republic of Korea
| | - Solah Park
- Department of Physiology, KU Open Innovation Center, Research Institute of Medical Science, Konkuk University School of Medicine, Chungju 27478, Republic of Korea
| | - Hyun Ju Noh
- Department of Physiology, KU Open Innovation Center, Research Institute of Medical Science, Konkuk University School of Medicine, Chungju 27478, Republic of Korea
| | - Shadi Golpasandi
- Department of Physiology, KU Open Innovation Center, Research Institute of Medical Science, Konkuk University School of Medicine, Chungju 27478, Republic of Korea
| | - Seo Hyeon Eun
- Department of Physiology, KU Open Innovation Center, Research Institute of Medical Science, Konkuk University School of Medicine, Chungju 27478, Republic of Korea
| | - Hyeryeong Lee
- Department of Physiology, KU Open Innovation Center, Research Institute of Medical Science, Konkuk University School of Medicine, Chungju 27478, Republic of Korea
| | - Bokyung Kim
- Department of Physiology, KU Open Innovation Center, Research Institute of Medical Science, Konkuk University School of Medicine, Chungju 27478, Republic of Korea
| | - Jinhong Wie
- Department of Physiology, KU Open Innovation Center, Research Institute of Medical Science, Konkuk University School of Medicine, Chungju 27478, Republic of Korea
| | - Mi Seon Seo
- Department of Physiology, KU Open Innovation Center, Research Institute of Medical Science, Konkuk University School of Medicine, Chungju 27478, Republic of Korea
| | - Sang Woong Park
- Department of Emergency Medical Services, Eulji University, Seongnam 13135, Republic of Korea.
| | - Young Min Bae
- Department of Physiology, KU Open Innovation Center, Research Institute of Medical Science, Konkuk University School of Medicine, Chungju 27478, Republic of Korea.
| |
Collapse
|
9
|
Zhang D, Zhao MM, Wu JM, Wang R, Xue G, Xue YB, Shao JQ, Zhang YY, Dong ED, Li ZY, Xiao H. Dual-omics reveals temporal differences in acute sympathetic stress-induced cardiac inflammation following α 1 and β-adrenergic receptors activation. Acta Pharmacol Sin 2023; 44:1350-1365. [PMID: 36737635 PMCID: PMC10310713 DOI: 10.1038/s41401-022-01048-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 12/28/2022] [Indexed: 02/05/2023] Open
Abstract
Sympathetic stress is prevalent in cardiovascular diseases. Sympathetic overactivation under strong acute stresses triggers acute cardiovascular events including myocardial infarction (MI), sudden cardiac death, and stress cardiomyopathy. α1-ARs and β-ARs, two dominant subtypes of adrenergic receptors in the heart, play a significant role in the physiological and pathologic regulation of these processes. However, little is known about the functional similarities and differences between α1- and β-ARs activated temporal responses in stress-induced cardiac pathology. In this work, we systematically compared the cardiac temporal genome-wide profiles of acute α1-AR and β-AR activation in the mice model by integrating transcriptome and proteome. We found that α1- and β-AR activations induced sustained and transient inflammatory gene expression, respectively. Particularly, the overactivation of α1-AR but not β-AR led to neutrophil infiltration at one day, which was closely associated with the up-regulation of chemokines, activation of NF-κB pathway, and sustained inflammatory response. Furthermore, there are more metabolic disorders under α1-AR overactivation compared with β-AR overactivation. These findings provide a new therapeutic strategy that, besides using β-blocker as soon as possible, blocking α1-AR within one day should also be considered in the treatment of acute stress-associated cardiovascular diseases.
Collapse
Affiliation(s)
- Di Zhang
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Ming-Ming Zhao
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital; NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education; Beijing Key Laboratory of Cardiovascular Receptors Research; Haihe Laboratory of Cell Ecosystem, Beijing, 100191, China
| | - Ji-Min Wu
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital; NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education; Beijing Key Laboratory of Cardiovascular Receptors Research; Haihe Laboratory of Cell Ecosystem, Beijing, 100191, China
| | - Rui Wang
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital; NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education; Beijing Key Laboratory of Cardiovascular Receptors Research; Haihe Laboratory of Cell Ecosystem, Beijing, 100191, China
| | - Gang Xue
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Yan-Bo Xue
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Ji-Qi Shao
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - You-Yi Zhang
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital; NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education; Beijing Key Laboratory of Cardiovascular Receptors Research; Haihe Laboratory of Cell Ecosystem, Beijing, 100191, China
| | - Er-Dan Dong
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital; NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education; Beijing Key Laboratory of Cardiovascular Receptors Research; Haihe Laboratory of Cell Ecosystem, Beijing, 100191, China.
| | - Zhi-Yuan Li
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China.
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China.
| | - Han Xiao
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital; NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education; Beijing Key Laboratory of Cardiovascular Receptors Research; Haihe Laboratory of Cell Ecosystem, Beijing, 100191, China.
| |
Collapse
|
10
|
Alrasheed NM, Alammari RB, Alshammari TK, Alamin MA, Alharbi AO, Alonazi AS, Bin Dayel AF, Alrasheed NM. α1A Adrenoreceptor blockade attenuates myocardial infarction by modulating the integrin-linked kinase/TGF-β/Smad signaling pathways. BMC Cardiovasc Disord 2023; 23:153. [PMID: 36964489 PMCID: PMC10037904 DOI: 10.1186/s12872-023-03188-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 03/16/2023] [Indexed: 03/26/2023] Open
Abstract
Background Myocardial infarction (MI) is considered a public health problem. According to the World Health Organization, MI is a leading cause of death and comorbidities worldwide. Activation of the α1A adrenergic receptor is a contributing factor to the development of MI. Tamsulosin, an α1A adrenergic blocker, has gained wide popularity as a medication for the treatment of benign prostatic hyperplasia. Limited evidence from previous studies has revealed the potential cardioprotective effects of tamsulosin, as its inhibitory effect on the α1A adrenoceptor protects the heart by acting on the smooth muscle of blood vessels, which results in hypotension; however, its effect on the infarcted heart is still unclear. The mechanisms of the expected cardioprotective effects mediated by tamsulosin are not yet understood. Transforming growth factor-beta (TGF-β), a mediator of fibrosis, is considered an attractive therapeutic target for remodeling after MI. The role of α1A adrenoceptor inhibition or its relationships with integrin-linked kinase (ILK) and TGF-β/small mothers against decapentaplegic (Smad) signaling pathways in attenuating MI are unclear. The present study was designed to investigate whether tamsulosin attenuates MI by modulating an ILK-related TGF-β/Smad pathway. Methods Twenty-four adult male Wistar rats were randomly divided into 4 groups: control, ISO, TAM, and ISO + TAM. ISO (150 mg/kg, intraperitoneally) was injected on Days 20 and 21 to induce MI. Tamsulosin (0.8 mg/kg, orally) was administered for 21 days, prior to ISO injection for 2 consecutive days. Heart-to-body weight ratios and cardiac and fibrotic biomarker levels were subsequently determined. ILK, TGF-β1, p-Smad2/3, and collagen III protein expression levels were determined using biomolecular methods. Results Tamsulosin significantly attenuated the relative heart-to-body weight index (p < 0.5) and creatine kinase-MB level (p < 0.01) compared with those in the ISO control group. While ISO resulted in superoxide anion production and enhanced oxidative damage, tamsulosin significantly prevented this damage through antioxidant defense mechanisms, increasing glutathione and superoxide dismutase levels (p < 0.05) and decreasing lipid peroxide oxidation levels (p < 0.01). The present data revealed that tamsulosin reduced TGF-β/p-Smad2/3 expression and enhanced ILK expression. Conclusion Tamsulosin may exert a cardioprotective effect by modulating the ILK-related TGF-β/Smad signaling pathway. Thus, tamsulosin may be a useful therapeutic approach for preventing MI. Supplementary Information The online version contains supplementary material available at 10.1186/s12872-023-03188-w.
Collapse
Affiliation(s)
- Nawal M. Alrasheed
- grid.56302.320000 0004 1773 5396Department of Pharmacology and Toxicology, College of Pharmacy , King Saud University, P.O. Box 70474, Riyadh, 11567 Saudi Arabia
| | - Raghad B. Alammari
- grid.56302.320000 0004 1773 5396Pharm D. Student, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Tahani K. Alshammari
- grid.56302.320000 0004 1773 5396Department of Pharmacology and Toxicology, College of Pharmacy , King Saud University, P.O. Box 70474, Riyadh, 11567 Saudi Arabia
| | - Maha A. Alamin
- grid.56302.320000 0004 1773 5396Department of Pharmacology and Toxicology, College of Pharmacy , King Saud University, P.O. Box 70474, Riyadh, 11567 Saudi Arabia
| | - Abeer O. Alharbi
- grid.56302.320000 0004 1773 5396Department of Pharmacology and Toxicology, College of Pharmacy , King Saud University, P.O. Box 70474, Riyadh, 11567 Saudi Arabia
| | - Asma S. Alonazi
- grid.56302.320000 0004 1773 5396Department of Pharmacology and Toxicology, College of Pharmacy , King Saud University, P.O. Box 70474, Riyadh, 11567 Saudi Arabia
| | - Anfal F. Bin Dayel
- grid.56302.320000 0004 1773 5396Department of Pharmacology and Toxicology, College of Pharmacy , King Saud University, P.O. Box 70474, Riyadh, 11567 Saudi Arabia
| | - Nouf M. Alrasheed
- grid.56302.320000 0004 1773 5396Department of Pharmacology and Toxicology, College of Pharmacy , King Saud University, P.O. Box 70474, Riyadh, 11567 Saudi Arabia
| |
Collapse
|
11
|
Kim JD, Kwon C, Nakamura K, Muromachi N, Mori H, Muroi SI, Yamada Y, Saito H, Nakagawa Y, Fukamizu A. Increased angiotensin II coupled with decreased Adra1a expression enhances cardiac hypertrophy in pregnancy-associated hypertensive mice. J Biol Chem 2023; 299:102964. [PMID: 36736425 PMCID: PMC10011504 DOI: 10.1016/j.jbc.2023.102964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 12/27/2022] [Accepted: 01/26/2023] [Indexed: 02/05/2023] Open
Abstract
Cardiac hypertrophy is a crucial risk factor for hypertensive disorders during pregnancy, but its progression during pregnancy remains unclear. We previously showed cardiac hypertrophy in a pregnancy-associated hypertensive (PAH) mouse model, in which an increase in angiotensin II (Ang II) levels was induced by human renin and human angiotensinogen, depending on pregnancy conditions. Here, to elucidate the factors involved in the progression of cardiac hypertrophy, we performed a comprehensive analysis of changes in gene expression in the hearts of PAH mice and compared them with those in control mice. We found that alpha-1A adrenergic receptor (Adra1a) mRNA levels in the heart were significantly reduced under PAH conditions, whereas the renin-angiotensin system was upregulated. Furthermore, we found that Adra1a-deficient PAH mice exhibited more severe cardiac hypertrophy than PAH mice. Our study suggests that Adra1a levels are regulated by renin-angiotensin system and that changes in Adra1a expression are involved in progressive cardiac hypertrophy in PAH mice.
Collapse
Affiliation(s)
- Jun-Dal Kim
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, Tsukuba, Ibaraki, Japan; Division of Complex Bioscience Research, Department of Research and Development, Institute of National Medicine, University of Toyama, Toyama, Japan; AMED-CREST, Japan Agency for Medical Research and Development, Chiyoda-ku, Tokyo, Japan.
| | - Chulwon Kwon
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Kanako Nakamura
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, Tsukuba, Ibaraki, Japan; Graduate School of Sciences and Technology, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Naoto Muromachi
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, Tsukuba, Ibaraki, Japan; Doctoral Program in Life and Agricultural Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Haruka Mori
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, Tsukuba, Ibaraki, Japan; Graduate School of Sciences and Technology, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Shin-Ichi Muroi
- Division of Complex Bioscience Research, Department of Research and Development, Institute of National Medicine, University of Toyama, Toyama, Japan; AMED-CREST, Japan Agency for Medical Research and Development, Chiyoda-ku, Tokyo, Japan
| | - Yasunari Yamada
- Division of Complex Bioscience Research, Department of Research and Development, Institute of National Medicine, University of Toyama, Toyama, Japan
| | - Hodaka Saito
- Division of Complex Bioscience Research, Department of Research and Development, Institute of National Medicine, University of Toyama, Toyama, Japan
| | - Yoshimi Nakagawa
- Division of Complex Bioscience Research, Department of Research and Development, Institute of National Medicine, University of Toyama, Toyama, Japan
| | - Akiyoshi Fukamizu
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, Tsukuba, Ibaraki, Japan; AMED-CREST, Japan Agency for Medical Research and Development, Chiyoda-ku, Tokyo, Japan; International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan.
| |
Collapse
|
12
|
Farraj AK, Martin BL, Schladweiler MC, Miller CN, Smoot J, Williams W, Fisher A, Oshiro W, Tennant A, Martin WK, Henriquez AR, Grindstaff R, Gavett SH, Gilmour MI, Kodavanti UP, Hazari MS, Dye JA. Mild allergic airways responses to an environmental mixture increase cardiovascular risk in rats. Toxicol Sci 2022; 191:106-122. [PMID: 36269214 PMCID: PMC9887678 DOI: 10.1093/toxsci/kfac112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Recent epidemiological findings link asthma to adverse cardiovascular responses. Yet, the precise cardiovascular impacts of asthma have been challenging to disentangle from the potential cardiovascular effects caused by asthma medication. The purpose of this study was to determine the impacts of allergic airways disease alone on cardiovascular function in an experimental model. Female Wistar rats were intranasally sensitized and then challenged once per week for 5 weeks with saline vehicle or a mixture of environmental allergens (ragweed, house dust mite, and Aspergillus fumigatus). Ventilatory and cardiovascular function, measured using double-chamber plethysmography and implantable blood pressure (BP) telemetry and cardiovascular ultrasound, respectively, were assessed before sensitization and after single and final allergen challenge. Responses to a single 0.5 ppm ozone exposure and to the cardiac arrhythmogenic agent aconitine were also assessed after final challenge. A single allergen challenge in sensitized rats increased tidal volume and specific airways resistance in response to provocation with methacholine and increased bronchoalveolar lavage fluid (BALF) eosinophils, neutrophils, lymphocytes, cytokines interleukin (IL)-4, IL-5, IL-10, IL-1β, tumor necrosis factor-α, and keratinocyte chemoattract-growth-related oncogene characteristic of allergic airways responses. Lung responses after final allergen challenge in sensitized rats were diminished, although ozone exposure increased BALF IL-6, IL-13, IL-1 β, and interferon-γ and modified ventilatory responses only in the allergen group. Final allergen challenge also increased systolic and mean arterial BP, stroke volume, cardiac output, end-diastolic volume, sensitivity to aconitine-induced cardiac arrhythmia, and cardiac gene expression with lesser effects after a single challenge. These findings demonstrate that allergic airways responses may increase cardiovascular risk in part by altering BP and myocardial function and by causing cardiac electrical instability.
Collapse
Affiliation(s)
- Aimen K Farraj
- To whom correspondence should be addressed at US Environmental Protection Agency, 109 T.W. Alexander Drive, Mail Code: B105-02, Research Triangle Park, NC 27709, USA. E-mail:
| | - Brandi L Martin
- Oak Ridge Institute for Science and Education, Oak Ridge, Tennessee 37830, USA
| | - Mette C Schladweiler
- Public Health and Integrated Toxicology Division, US Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA
| | - Colette N Miller
- Oak Ridge Institute for Science and Education, Oak Ridge, Tennessee 37830, USA
| | - Jacob Smoot
- Oak Ridge Institute for Science and Education, Oak Ridge, Tennessee 37830, USA
| | - Wanda Williams
- Public Health and Integrated Toxicology Division, US Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA
| | - Anna Fisher
- Public Health and Integrated Toxicology Division, US Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA
| | - Wendy Oshiro
- Public Health and Integrated Toxicology Division, US Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA
| | - Alan Tennant
- Biomolecular and Computational Toxicology Division, US Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA
| | - W Kyle Martin
- Curriculum of Toxicology and Environmental Medicine, UNC Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Andres R Henriquez
- Oak Ridge Institute for Science and Education, Oak Ridge, Tennessee 37830, USA
| | - Rachel Grindstaff
- Oak Ridge Institute for Science and Education, Oak Ridge, Tennessee 37830, USA
| | - Stephen H Gavett
- Public Health and Integrated Toxicology Division, US Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA
| | - M Ian Gilmour
- Public Health and Integrated Toxicology Division, US Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA
| | - Urmila P Kodavanti
- Public Health and Integrated Toxicology Division, US Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA
| | - Mehdi S Hazari
- Public Health and Integrated Toxicology Division, US Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA
| | - Janice A Dye
- Public Health and Integrated Toxicology Division, US Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA
| |
Collapse
|
13
|
Sandroni PB, Fisher-Wellman KH, Jensen BC. Adrenergic Receptor Regulation of Mitochondrial Function in Cardiomyocytes. J Cardiovasc Pharmacol 2022; 80:364-377. [PMID: 35170492 PMCID: PMC9365878 DOI: 10.1097/fjc.0000000000001241] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 02/01/2022] [Indexed: 01/31/2023]
Abstract
ABSTRACT Adrenergic receptors (ARs) are G protein-coupled receptors that are stimulated by catecholamines to induce a wide array of physiological effects across tissue types. Both α1- and β-ARs are found on cardiomyocytes and regulate cardiac contractility and hypertrophy through diverse molecular pathways. Acute activation of cardiomyocyte β-ARs increases heart rate and contractility as an adaptive stress response. However, chronic β-AR stimulation contributes to the pathobiology of heart failure. By contrast, mounting evidence suggests that α1-ARs serve protective functions that may mitigate the deleterious effects of chronic β-AR activation. Here, we will review recent studies demonstrating that α1- and β-ARs differentially regulate mitochondrial biogenesis and dynamics, mitochondrial calcium handling, and oxidative phosphorylation in cardiomyocytes. We will identify potential mechanisms of these actions and focus on the implications of these findings for the modulation of contractile function in the uninjured and failing heart. Collectively, we hope to elucidate important physiological processes through which these well-studied and clinically relevant receptors stimulate and fuel cardiac contraction to contribute to myocardial health and disease.
Collapse
Affiliation(s)
- Peyton B. Sandroni
- University of North Carolina School of Medicine, Department of Pharmacology
- University of North Carolina School of Medicine, McAllister Heart Institute
| | - Kelsey H. Fisher-Wellman
- East Carolina University Brody School of Medicine, Department of Physiology
- East Carolina University Diabetes and Obesity Institute
| | - Brian C. Jensen
- University of North Carolina School of Medicine, Department of Pharmacology
- University of North Carolina School of Medicine, McAllister Heart Institute
- University of North Carolina School of Medicine, Department of Medicine, Division of Cardiology
| |
Collapse
|
14
|
Shiu P, Grewal GS, Kozik TM. Midodrine to optimize heart failure therapy in patients with concurrent hypotension. SAGE Open Med Case Rep 2022; 10:2050313X221100400. [PMID: 35601610 PMCID: PMC9121496 DOI: 10.1177/2050313x221100400] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 04/22/2022] [Indexed: 11/19/2022] Open
Abstract
According to the Centers for Disease Control and Prevention statistics, about 6.2
million adults in the United States have heart failure. Guideline-Directed
Medical Therapy (GDMT) involving the use of renin-angiotensin-aldosterone system
inhibitors with or without a neprilysin inhibitor, β-blockers,
mineralocorticoid-receptor-antagonists, and sodium-glucose cotransporter-2
inhibitors serve as the backbone for heart failure with reduced ejection
fraction (HFrEF) therapy. However, in patients with refractory hypotension, the
initiation of GDMT may not be possible. We present four cases where the use of
midodrine, an alpha adrenergic agonist, serves as bridge therapy for the
initiation or continuation of GDMT with marked clinical improvement. These cases
illustrate how exacerbations of HFrEF may be ameliorated with outpatient
midodrine titration among patients with baseline, persistent hypotension such
that GDMT may be better tolerated.
Collapse
Affiliation(s)
- Paul Shiu
- Graduate Medical Education, St. Joseph’s Medical Center, Stockton, CA, USA
| | | | - Teri M Kozik
- Graduate Medical Education, St. Joseph’s Medical Center, Stockton, CA, USA
| |
Collapse
|
15
|
Murganti F, Derks W, Baniol M, Simonova I, Trus P, Neumann K, Khattak S, Guan K, Bergmann O. FUCCI-Based Live Imaging Platform Reveals Cell Cycle Dynamics and Identifies Pro-proliferative Compounds in Human iPSC-Derived Cardiomyocytes. Front Cardiovasc Med 2022; 9:840147. [PMID: 35548410 PMCID: PMC9081338 DOI: 10.3389/fcvm.2022.840147] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 03/16/2022] [Indexed: 01/23/2023] Open
Abstract
One of the major goals in cardiac regeneration research is to replace lost ventricular tissue with new cardiomyocytes. However, cardiomyocyte proliferation drops to low levels in neonatal hearts and is no longer efficient in compensating for the loss of functional myocardium in heart disease. We generated a human induced pluripotent stem cell (iPSC)-derived cardiomyocyte-specific cell cycle indicator system (TNNT2-FUCCI) to characterize regular and aberrant cardiomyocyte cycle dynamics. We visualized cell cycle progression in TNNT2-FUCCI and found G2 cycle arrest in endoreplicating cardiomyocytes. Moreover, we devised a live-cell compound screening platform to identify pro-proliferative drug candidates. We found that the alpha-adrenergic receptor agonist clonidine induced cardiomyocyte proliferation in vitro and increased cardiomyocyte cell cycle entry in neonatal mice. In conclusion, the TNNT2-FUCCI system is a versatile tool to characterize cardiomyocyte cell cycle dynamics and identify pro-proliferative candidates with regenerative potential in the mammalian heart.
Collapse
Affiliation(s)
| | - Wouter Derks
- Center for Regenerative Therapies Dresden, TU Dresden, Dresden, Germany
| | - Marion Baniol
- Karolinska Institute, Cell and Molecular Biology (CMB), Stockholm, Sweden
| | - Irina Simonova
- Center for Regenerative Therapies Dresden, TU Dresden, Dresden, Germany
| | - Palina Trus
- Center for Regenerative Therapies Dresden, TU Dresden, Dresden, Germany
| | - Katrin Neumann
- Center for Regenerative Therapies Dresden, TU Dresden, Dresden, Germany
| | - Shahryar Khattak
- Center for Regenerative Therapies Dresden, TU Dresden, Dresden, Germany
- Royal College of Surgeons Ireland (RCSI) in Bahrain, Adliya, Bahrain
| | - Kaomei Guan
- Institute of Pharmacology and Toxicology, TU Dresden, Dresden, Germany
| | - Olaf Bergmann
- Center for Regenerative Therapies Dresden, TU Dresden, Dresden, Germany
- Karolinska Institute, Cell and Molecular Biology (CMB), Stockholm, Sweden
- *Correspondence: Olaf Bergmann
| |
Collapse
|
16
|
Yang M, Tao L, Zhao CC, Wang ZL, Yu ZJ, Zhou W, Wen YL, Li LF, Tian Y, Sheng J. Antifatigue Effect of Panax Notoginseng Leaves Fermented With Microorganisms: In-vitro and In-vivo Evaluation. Front Nutr 2022; 9:824525. [PMID: 35273989 PMCID: PMC8904179 DOI: 10.3389/fnut.2022.824525] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 01/25/2022] [Indexed: 11/15/2022] Open
Abstract
Fatigue is a common physiological phenomenon caused by many complicated factors. Excessive fatigue will lead to a series of uncomfortable reactions and damage body health. Panax notoginseng leaves (PNL) is a new resource food that good for soothing nerves, nourishing the heart, and strengthening the spleen. Microbial fermentation could increase the content of bio-ingredients and produce new active ingredients. However, the effect of fermented P. notoginseng leaves (FPNL) on antifatigue and the molecular mechanisms remain to be elucidated. Thus, in this study, we evaluated the antifatigue effect of co-fermented P. notoginseng leaves by Saccharomyces cerevisiae and Bacillus subtilis in-vitro and in-vivo, and its mechanism was further elucidated. The results showed that FPNL exhibited higher saponins, organic phenolic acids content, and antioxidant activity than PNL. FPNL improved ISO-induced H9c2 myocardial cell damage by alleviating apoptosis (modulating Bax and Bcl-2 protein expression) and reducing antioxidant activity in-vitro. Moreover, in-vivo experiment showed that FPNL significantly prolonged the weight-loading swimming time of mice. After gavaged FPNL, the levels of liver glycogen (LG) and serum lactate dehydrogenase (LDH) activity were increased in mice. In contrast, the levels of blood urea nitrogen (BUN), lactate acid, and malondialdehyde (MDA) were decreased. In summary, our results indicated that FPNL showed a good antifatigue effect in-vivo and in-vitro.
Collapse
Affiliation(s)
- Min Yang
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, China
- Engineering Research Center of Development and Utilization of Food and Drug Homologous Resources, Ministry of Education, Yunnan Agricultural University, Kunming, China
| | - Liang Tao
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, China
- Engineering Research Center of Development and Utilization of Food and Drug Homologous Resources, Ministry of Education, Yunnan Agricultural University, Kunming, China
- National Research and Development Professional Center for Moringa Processing Technology, Yunnan Agricultural University, Kunming, China
| | - Cun-Chao Zhao
- Engineering Research Center of Development and Utilization of Food and Drug Homologous Resources, Ministry of Education, Yunnan Agricultural University, Kunming, China
- National Research and Development Professional Center for Moringa Processing Technology, Yunnan Agricultural University, Kunming, China
| | - Zi-Lin Wang
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, China
- Engineering Research Center of Development and Utilization of Food and Drug Homologous Resources, Ministry of Education, Yunnan Agricultural University, Kunming, China
| | - Zhi-Jin Yu
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, China
- National Research and Development Professional Center for Moringa Processing Technology, Yunnan Agricultural University, Kunming, China
| | - Wen Zhou
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Yan-Long Wen
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, China
- Engineering Research Center of Development and Utilization of Food and Drug Homologous Resources, Ministry of Education, Yunnan Agricultural University, Kunming, China
| | - Ling-Fei Li
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, China
- Engineering Research Center of Development and Utilization of Food and Drug Homologous Resources, Ministry of Education, Yunnan Agricultural University, Kunming, China
- *Correspondence: Ling-Fei Li
| | - Yang Tian
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, China
- Engineering Research Center of Development and Utilization of Food and Drug Homologous Resources, Ministry of Education, Yunnan Agricultural University, Kunming, China
- National Research and Development Professional Center for Moringa Processing Technology, Yunnan Agricultural University, Kunming, China
- Yang Tian
| | - Jun Sheng
- Engineering Research Center of Development and Utilization of Food and Drug Homologous Resources, Ministry of Education, Yunnan Agricultural University, Kunming, China
- Key Laboratory of Pu-erh Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming, China
- Jun Sheng
| |
Collapse
|
17
|
Letson HL, Biros E, Morris JL, Dobson GP. ALM Fluid Therapy Shifts Sympathetic Hyperactivity to Parasympathetic Dominance in the Rat Model of Non-Compressible Hemorrhagic Shock. Shock 2022; 57:264-273. [PMID: 34798632 DOI: 10.1097/shk.0000000000001886] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
ABSTRACT Excessive sympathetic outflow following trauma can lead to cardiac dysfunction, inflammation, coagulopathy, and poor outcomes. We previously reported that buprenorphine analgesia decreased survival after hemorrhagic trauma. Our aim is to examine the underlying mechanisms of mortality in a non-compressible hemorrhage rat model resuscitated with saline or adenosine, lidocaine, magnesium (ALM). Anesthetized adult male Sprague-Dawley rats were randomly assigned to Saline control group or ALM therapy group (both n = 10). Hemorrhage was induced by 50% liver resection. After 15 min, 0.7 mL/kg 3% NaCl ± ALM intravenous bolus was administered, and after 60 min, 0.9% NaCl ± ALM was infused for 4 h (0.5 mL/kg/h) with 72 h monitoring. Animals received 6-12-hourly buprenorphine for analgesia. Hemodynamics, heart rate variability, echocardiography, and adiponectin were measured. Cardiac tissue was analyzed for adrenergic/cholinergic receptor expression, inflammation, and histopathology. Four ALM animals and one Saline control survived to 72 h. Mortality was associated with up to 97% decreases in adrenergic (β-1, α-1A) and cholinergic (M2) receptor expression, cardiac inflammation, myocyte Ca2+ loading, and histopathology, indicating heart ischemia/failure. ALM survivors had higher cardiac output and stroke volume, a 30-fold increase in parasympathetic/sympathetic receptor expression ratio, and higher circulating adiponectin compared to Saline controls. Paradoxically, Saline cardiac adiponectin hormone levels were higher than ALM, with no change in receptor expression, indicating intra-cardiac synthesis. Mortality appears to be a "systems failure" associated with CNS dysregulation of cardiac function. Survival involves an increased parasympathetic dominance to support cardiac pump function with reduced myocardial inflammation. Increased cardiac α-1A adrenergic receptor in ALM survivors may be significant, as this receptor is highly protective during heart dysfunction/failure.
Collapse
Affiliation(s)
- Hayley L Letson
- Heart and Trauma Research Laboratory, College of Medicine and Dentistry, James Cook University, Queensland, Australia
| | | | | | | |
Collapse
|
18
|
Chronic isoprenaline/phenylephrine vs. exclusive isoprenaline stimulation in mice: critical contribution of alpha 1-adrenoceptors to early cardiac stress responses. Basic Res Cardiol 2022; 117:15. [PMID: 35286475 PMCID: PMC8921177 DOI: 10.1007/s00395-022-00920-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 02/04/2022] [Accepted: 02/07/2022] [Indexed: 01/31/2023]
Abstract
Hyperactivity of the sympathetic nervous system is a major driver of cardiac remodeling, exerting its effects through both α-, and β-adrenoceptors (α-, β-ARs). As the relative contribution of subtype α1-AR to cardiac stress responses remains poorly investigated, we subjected mice to either subcutaneous perfusion with the β-AR agonist isoprenaline (ISO, 30 mg/kg × day) or to a combination of ISO and the stable α1-AR agonist phenylephrine (ISO/PE, 30 mg/kg × day each). Telemetry analysis revealed similar hemodynamic responses under both ISO and ISO/PE treatment i.e., permanently increased heart rates and only transient decreases in mean blood pressure during the first 24 h. Echocardiography and single cell analysis after 1 week of exposure showed that ISO/PE-, but not ISO-treated animals established α1-AR-mediated inotropic responsiveness to acute adrenergic stimulation. Morphologically, additional PE perfusion limited concentric cardiomyocyte growth and enhanced cardiac collagen deposition during 7 days of treatment. Time-course analysis demonstrated a diverging development in transcriptional patterns at day 4 of treatment i.e., increased expression of selected marker genes Xirp2, Nppa, Tgfb1, Col1a1, Postn under chronic ISO/PE treatment which was either less pronounced or absent in the ISO group. Transcriptome analyses at day 4 via RNA sequencing demonstrated that additional PE treatment caused a marked upregulation of genes allocated to extracellular matrix and fiber organization along with a more pronounced downregulation of genes involved in metabolic processes, muscle adaptation and cardiac electrophysiology. Consistently, transcriptome changes under ISO/PE challenge more effectively recapitulated early transcriptional alterations in pressure overload-induced experimental heart failure and in human hypertrophic cardiomyopathy.
Collapse
|
19
|
Du X. Sympatho-adrenergic mechanisms in heart failure: new insights into pathophysiology. MEDICAL REVIEW (BERLIN, GERMANY) 2021; 1:47-77. [PMID: 37724075 PMCID: PMC10388789 DOI: 10.1515/mr-2021-0007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 06/02/2021] [Indexed: 09/20/2023]
Abstract
The sympathetic nervous system is activated in the setting of heart failure (HF) to compensate for hemodynamic instability. However, acute sympathetic surge or sustained high neuronal firing rates activates β-adrenergic receptor (βAR) signaling contributing to myocardial remodeling, dysfunction and electrical instability. Thus, sympatho-βAR activation is regarded as a hallmark of HF and forms pathophysiological basis for β-blocking therapy. Building upon earlier research findings, studies conducted in the recent decades have significantly advanced our understanding on the sympatho-adrenergic mechanism in HF, which forms the focus of this article. This review notes recent research progress regarding the roles of cardiac β2AR or α1AR in the failing heart, significance of β1AR-autoantibodies, and βAR signaling through G-protein independent signaling pathways. Sympatho-βAR regulation of immune cells or fibroblasts is specifically discussed. On the neuronal aspects, knowledge is assembled on the remodeling of sympathetic nerves of the failing heart, regulation by presynaptic α2AR of NE release, and findings on device-based neuromodulation of the sympathetic nervous system. The review ends with highlighting areas where significant knowledge gaps exist but hold promise for new breakthroughs.
Collapse
Affiliation(s)
- Xiaojun Du
- Faculty of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, 76 West Yanta Road, Xi’an710061, Shaanxi, China
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC3004, Australia
| |
Collapse
|
20
|
Li R, Qi Y, Yuan Q, Xu L, Gao M, Xu Y, Han X, Yin L, Liu C. Protective effects of dioscin against isoproterenol-induced cardiac hypertrophy via adjusting PKCε/ERK-mediated oxidative stress. Eur J Pharmacol 2021; 907:174277. [PMID: 34171391 DOI: 10.1016/j.ejphar.2021.174277] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/17/2021] [Accepted: 06/18/2021] [Indexed: 11/24/2022]
Abstract
Cardiac hypertrophy (CH) plays a central role in cardiac remodeling and is an independent risk factor for cardiac events. It is imperative to find drugs with protective effect on CH. Dioscin, one natural product, shows various pharmacological activities, and PKCepsilon (PKCε) plays an important role in the physiological hypertrophic responses. Thus, we aimed to investigate the possible protective effect of dioscin on CH through PKCε. In the present study, the isoproterenol (ISO)-induced H9C2 cells and primary cardiomyocytes models, and the ISO-induced rat model were established, and the pharmacodynamics and mechanism of dioscin were investigated. In vitro results prompted that, dioscin significantly improved ISO-induced cardiomyocyte hypertrophy, decreased the levels of cell size, protein content of single cell, reactive oxygen species, atrial natriuretic peptide (ANP), brain natriuretic peptide (BNP), beta-myosin heavy chain (β-MHC). Moreover, in vivo, changes in histopathological of the animals caused by ISO are improved by dioscin. And dioscin decreased the index of CH and the levels of CK, MDA, LDH, and increased the levels of GSH, SOD and GSH-Px. Mechanism research showed that dioscin inhibited the expression levels of PKCε, and affected the expression levels of p-MEK, p-ERK, Nrf2, Keap1 and HO-1 to inhibit oxidative stress. In addition, the results of ISO-induced CH in PKCε siRNA transfected H9C2 cells and C57BL/6 mice further showed that the protective effect of dioscin on CH, which was mediated by inhibition of PKCε/ERK signal pathway. In summary, dioscin can effectively inhibit CH by regulating PKCε-mediated oxidative stress, which should be considered as one potent candidate for new drug research and development to treat CH in the future.
Collapse
Affiliation(s)
- Ruomiao Li
- College of Pharmacy, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian, China
| | - Yan Qi
- College of Pharmacy, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian, China
| | - Qianhui Yuan
- College of Pharmacy, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian, China
| | - Lina Xu
- College of Pharmacy, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian, China
| | - Meng Gao
- College of Pharmacy, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian, China
| | - Youwei Xu
- College of Pharmacy, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian, China
| | - Xu Han
- College of Pharmacy, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian, China
| | - Lianhong Yin
- College of Pharmacy, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian, China.
| | - Chuntong Liu
- Pharmaceutical Department, The Second Hospital of Dalian Medical University, 467 Zhongshan Road, Dalian, 116023, China.
| |
Collapse
|
21
|
Impact of Aldosterone on the Failing Myocardium: Insights from Mitochondria and Adrenergic Receptors Signaling and Function. Cells 2021; 10:cells10061552. [PMID: 34205363 PMCID: PMC8235589 DOI: 10.3390/cells10061552] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 06/08/2021] [Accepted: 06/16/2021] [Indexed: 02/06/2023] Open
Abstract
The mineralocorticoid aldosterone regulates electrolyte and blood volume homeostasis, but it also adversely modulates the structure and function of the chronically failing heart, through its elevated production in chronic human post-myocardial infarction (MI) heart failure (HF). By activating the mineralocorticoid receptor (MR), a ligand-regulated transcription factor, aldosterone promotes inflammation and fibrosis of the heart, while increasing oxidative stress, ultimately induding mitochondrial dysfunction in the failing myocardium. To reduce morbidity and mortality in advanced stage HF, MR antagonist drugs, such as spironolactone and eplerenone, are used. In addition to the MR, aldosterone can bind and stimulate other receptors, such as the plasma membrane-residing G protein-coupled estrogen receptor (GPER), further complicating it signaling properties in the myocardium. Given the salient role that adrenergic receptor (ARs)—particularly βARs—play in cardiac physiology and pathology, unsurprisingly, that part of the impact of aldosterone on the failing heart is mediated by its effects on the signaling and function of these receptors. Aldosterone can significantly precipitate the well-documented derangement of cardiac AR signaling and impairment of AR function, critically underlying chronic human HF. One of the main consequences of HF in mammalian models at the cellular level is the presence of mitochondrial dysfunction. As such, preventing mitochondrial dysfunction could be a valid pharmacological target in this condition. This review summarizes the current experimental evidence for this aldosterone/AR crosstalk in both the healthy and failing heart, and the impact of mitochondrial dysfunction in HF. Recent findings from signaling studies focusing on MR and AR crosstalk via non-conventional signaling of molecules that normally terminate the signaling of ARs in the heart, i.e., the G protein-coupled receptor-kinases (GRKs), are also highlighted.
Collapse
|
22
|
Saadeh K, Chadda KR, Ahmad S, Valli H, Nanthakumar N, Fazmin IT, Edling CE, Huang CLH, Jeevaratnam K. Molecular basis of ventricular arrhythmogenicity in a Pgc-1α deficient murine model. Mol Genet Metab Rep 2021; 27:100753. [PMID: 33898262 PMCID: PMC8059080 DOI: 10.1016/j.ymgmr.2021.100753] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 04/03/2021] [Indexed: 11/17/2022] Open
Abstract
Mitochondrial dysfunction underlying metabolic disorders such as obesity and diabetes mellitus is strongly associated with cardiac arrhythmias. Murine Pgc-1α-/- hearts replicate disrupted mitochondrial function and model the associated pro-arrhythmic electrophysiological abnormalities. Quantitative PCR, western blotting and histological analysis were used to investigate the molecular basis of the electrophysiological changes associated with mitochondrial dysfunction. qPCR analysis implicated downregulation of genes related to Na+-K+ ATPase activity (Atp1b1), surface Ca2+ entry (Cacna1c), action potential repolarisation (Kcnn1), autonomic function (Adra1d, Adcy4, Pde4d, Prkar2a), and morphological properties (Myh6, Tbx3) in murine Pgc-1α-/- ventricles. Western blotting revealed reduced NaV1.5 but normal Cx43 expression. Histological analysis revealed increased tissue fibrosis in the Pgc-1α-/- ventricles. These present findings identify altered transcription amongst a strategically selected set of genes established as encoding proteins involved in cardiac electrophysiological activation and therefore potentially involved in alterations in ventricular activation and Ca2+ homeostasis in arrhythmic substrate associated with Pgc-1α deficiency. They complement and complete previous studies examining such expression characteristics in the atria and ventricles of Pgc-1 deficient murine hearts.
Collapse
Affiliation(s)
- Khalil Saadeh
- Faculty of Health and Medical Sciences, University of Surrey, GU2 7AL Guildford, United Kingdom
- School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Karan R. Chadda
- Faculty of Health and Medical Sciences, University of Surrey, GU2 7AL Guildford, United Kingdom
| | - Shiraz Ahmad
- Faculty of Health and Medical Sciences, University of Surrey, GU2 7AL Guildford, United Kingdom
- Physiological Laboratory and Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Haseeb Valli
- Physiological Laboratory and Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Nakulan Nanthakumar
- Faculty of Health and Medical Sciences, University of Surrey, GU2 7AL Guildford, United Kingdom
- Bristol Medical School. University of Bristol, Bristol, United Kingdom
| | - Ibrahim T. Fazmin
- Faculty of Health and Medical Sciences, University of Surrey, GU2 7AL Guildford, United Kingdom
- School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Charlotte E. Edling
- Faculty of Health and Medical Sciences, University of Surrey, GU2 7AL Guildford, United Kingdom
| | - Christopher L.-H. Huang
- Faculty of Health and Medical Sciences, University of Surrey, GU2 7AL Guildford, United Kingdom
- Physiological Laboratory and Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Kamalan Jeevaratnam
- Faculty of Health and Medical Sciences, University of Surrey, GU2 7AL Guildford, United Kingdom
| |
Collapse
|
23
|
Gilani A, De Caterina R, Papacosta O, Lennon LT, Whincup PH, Wannamethee SG. Excessive Orthostatic Changes in Blood Pressure Are Associated With Incident Heart Failure in Older Men: A Prospective Analysis From the BRHS. Hypertension 2021; 77:1481-1489. [PMID: 33719509 DOI: 10.1161/hypertensionaha.120.15817] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Artaza Gilani
- University College London Research Department of Primary Care and Population Health, Royal Free Hospital, United Kingdom (A.G., O.P., L.T.L., S.G.W.)
| | - Raffaele De Caterina
- Cardiovascular Division, Pisa University Hospital, University of Pisa, Italy (R.D.E.C.).,Fondazione VillaSerena per la Ricerca, Città Sant'Angelo, Pescara, Italy (R.D.E.C.)
| | - Olia Papacosta
- University College London Research Department of Primary Care and Population Health, Royal Free Hospital, United Kingdom (A.G., O.P., L.T.L., S.G.W.)
| | - Lucy T Lennon
- University College London Research Department of Primary Care and Population Health, Royal Free Hospital, United Kingdom (A.G., O.P., L.T.L., S.G.W.)
| | - Peter H Whincup
- Population Health Research Institute, St George's, University of London, Cranmer Terrace, United Kingdom (P.H.W.)
| | - S Goya Wannamethee
- University College London Research Department of Primary Care and Population Health, Royal Free Hospital, United Kingdom (A.G., O.P., L.T.L., S.G.W.)
| |
Collapse
|
24
|
Joyce W, Scholman KT, Jensen B, Wang T, Boukens BJ. α 1-adrenergic stimulation increases ventricular action potential duration in the intact mouse heart. Facets (Ott) 2021. [DOI: 10.1139/facets-2020-0081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The role of α1-adrenergic receptors (α-ARs) in the regulation of myocardial function is less well-understood than that of β-ARs. Previous reports in the mouse heart have described that α1-adrenergic stimulation shortens action potential duration in isolated cells or tissues, in contrast to prolongation of the action potential reported in most other mammalian hearts. It has since become appreciated, however, that the mouse heart exhibits marked variation in inotropic response to α1-adrenergic stimulation between ventricles and even individual cardiomyocytes. We investigated the effects of α1-adrenergic stimulation on action potential duration at 80% of repolarization in the right and left ventricles of Langendorff-perfused mouse hearts using optical mapping. In hearts under β-adrenergic blockade (propranolol), phenylephrine or noradrenaline perfusion both increased action potential duration in both ventricles. The increased action potential duration was partially reversed by subsequent perfusion with the α-adrenergic antagonist phentolamine (1 μmol L−1). These data show that α1-receptor stimulation may lead to a prolonging of action potential in the mouse heart and thereby refine our understanding of how action potential duration adjusts during sympathetic stimulation.
Collapse
Affiliation(s)
- William Joyce
- Department of Biology—Zoophysiology, Aarhus University, DK-8000 Aarhus C, Denmark
- Department of Biology, University of Ottawa, 30 Marie Curie, Ottawa, ON K1N 6N5, Canada
| | - Koen T. Scholman
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, 11005 AZ Amsterdam, the Netherlands
| | - Bjarke Jensen
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, 11005 AZ Amsterdam, the Netherlands
| | - Tobias Wang
- Department of Biology—Zoophysiology, Aarhus University, DK-8000 Aarhus C, Denmark
| | - Bastiaan J. Boukens
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, 11005 AZ Amsterdam, the Netherlands
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, 1100 DD Amsterdam, the Netherlands
| |
Collapse
|
25
|
Zhang J, Simpson PC, Jensen BC. Cardiac α1A-adrenergic receptors: emerging protective roles in cardiovascular diseases. Am J Physiol Heart Circ Physiol 2020; 320:H725-H733. [PMID: 33275531 DOI: 10.1152/ajpheart.00621.2020] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
α1-Adrenergic receptors (ARs) are catecholamine-activated G protein-coupled receptors (GPCRs) that are expressed in mouse and human myocardium and vasculature, and play essential roles in the regulation of cardiovascular physiology. Though α1-ARs are less abundant in the heart than β1-ARs, activation of cardiac α1-ARs results in important biologic processes such as hypertrophy, positive inotropy, ischemic preconditioning, and protection from cell death. Data from the Antihypertensive and Lipid-Lowering Treatment to Prevent Heart Attack Trial (ALLHAT) indicate that nonselectively blocking α1-ARs is associated with a twofold increase in adverse cardiac events, including heart failure and angina, suggesting that α1-AR activation might also be cardioprotective in humans. Mounting evidence implicates the α1A-AR subtype in these adaptive effects, including prevention and reversal of heart failure in animal models by α1A agonists. In this review, we summarize recent advances in our understanding of cardiac α1A-ARs.
Collapse
Affiliation(s)
- Jiandong Zhang
- McAllister Heart Institute, University of North Carolina, School of Medicine, Chapel Hill, North Carolina
| | - Paul C Simpson
- Department of Medicine and Research Service, San Francisco Veterans Affairs Medical Center and Cardiovascular Research Institute, University of California, San Francisco, California
| | - Brian C Jensen
- McAllister Heart Institute, University of North Carolina, School of Medicine, Chapel Hill, North Carolina
| |
Collapse
|
26
|
Kaykı-Mutlu G, Papazisi O, Palmen M, Danser AHJ, Michel MC, Arioglu-Inan E. Cardiac and Vascular α 1-Adrenoceptors in Congestive Heart Failure: A Systematic Review. Cells 2020; 9:E2412. [PMID: 33158106 PMCID: PMC7694190 DOI: 10.3390/cells9112412] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/29/2020] [Accepted: 11/02/2020] [Indexed: 02/07/2023] Open
Abstract
As heart failure (HF) is a devastating health problem worldwide, a better understanding and the development of more effective therapeutic approaches are required. HF is characterized by sympathetic system activation which stimulates α- and β-adrenoceptors (ARs). The exposure of the cardiovascular system to the increased locally released and circulating levels of catecholamines leads to a well-described downregulation and desensitization of β-ARs. However, information on the role of α-AR is limited. We have performed a systematic literature review examining the role of both cardiac and vascular α1-ARs in HF using 5 databases for our search. All three α1-AR subtypes (α1A, α1B and α1D) are expressed in human and animal hearts and blood vessels in a tissue-dependent manner. We summarize the changes observed in HF regarding the density, signaling and responses of α1-ARs. Conflicting findings arise from different studies concerning the influence that HF has on α1-AR expression and function; in contrast to β-ARs there is no consistent evidence for down-regulation or desensitization of cardiac or vascular α1-ARs. Whether α1-ARs are a therapeutic target in HF remains a matter of debate.
Collapse
Affiliation(s)
- Gizem Kaykı-Mutlu
- Department of Pharmacology, Faculty of Pharmacy, Ankara University, 06560 Ankara, Turkey; (G.K.-M.); (E.A.-I.)
| | - Olga Papazisi
- Department of Cardiothoracic Surgery, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (O.P.); (M.P.)
| | - Meindert Palmen
- Department of Cardiothoracic Surgery, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (O.P.); (M.P.)
| | - A. H. Jan Danser
- Department of Internal Medicine, Division of Pharmacology, Erasmus Medical Center, 3000 CA Rotterdam, The Netherlands;
| | - Martin C. Michel
- Department of Pharmacology, Johannes Gutenberg University, 55131 Mainz, Germany
| | - Ebru Arioglu-Inan
- Department of Pharmacology, Faculty of Pharmacy, Ankara University, 06560 Ankara, Turkey; (G.K.-M.); (E.A.-I.)
| |
Collapse
|
27
|
Alluri SR, Kim SW, Volkow ND, Kil KE. PET Radiotracers for CNS-Adrenergic Receptors: Developments and Perspectives. Molecules 2020; 25:molecules25174017. [PMID: 32899124 PMCID: PMC7504810 DOI: 10.3390/molecules25174017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/29/2020] [Accepted: 09/01/2020] [Indexed: 12/30/2022] Open
Abstract
Epinephrine (E) and norepinephrine (NE) play diverse roles in our body’s physiology. In addition to their role in the peripheral nervous system (PNS), E/NE systems including their receptors are critical to the central nervous system (CNS) and to mental health. Various antipsychotics, antidepressants, and psychostimulants exert their influence partially through different subtypes of adrenergic receptors (ARs). Despite the potential of pharmacological applications and long history of research related to E/NE systems, research efforts to identify the roles of ARs in the human brain taking advantage of imaging have been limited by the lack of subtype specific ligands for ARs and brain penetrability issues. This review provides an overview of the development of positron emission tomography (PET) radiotracers for in vivo imaging of AR system in the brain.
Collapse
Affiliation(s)
- Santosh Reddy Alluri
- University of Missouri Research Reactor, University of Missouri, Columbia, MO 65211-5110, USA;
| | - Sung Won Kim
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892-1013, USA;
| | - Nora D. Volkow
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892-1013, USA;
- National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD 20892-1013, USA
- Correspondence: (N.D.V.); (K.-E.K.); Tel.: +1-(301)-443-6480 (N.D.V.); +1-(573)-884-7885 (K.-E.K.)
| | - Kun-Eek Kil
- University of Missouri Research Reactor, University of Missouri, Columbia, MO 65211-5110, USA;
- Department of Veterinary Medicine and Surgery, University of Missouri, Columbia, MO 65211, USA
- Correspondence: (N.D.V.); (K.-E.K.); Tel.: +1-(301)-443-6480 (N.D.V.); +1-(573)-884-7885 (K.-E.K.)
| |
Collapse
|
28
|
Hemodynamic, respiratory and sedative effects of progressively increasing doses of acepromazine in conscious dogs. Vet Anaesth Analg 2020; 47:447-453. [PMID: 32362549 DOI: 10.1016/j.vaa.2020.02.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 02/25/2020] [Accepted: 02/27/2020] [Indexed: 11/21/2022]
Abstract
OBJECTIVE To evaluate the effects of progressively increasing doses of acepromazine on cardiopulmonary variables and sedation in conscious dogs. STUDY DESIGN Prospective, experimental study. ANIMALS A group of six healthy, adult, mixed-breed dogs weighing 16.5 ± 5.0 kg (mean ± standard deviation). METHODS Dogs were instrumented with thermodilution and arterial catheters for evaluation of hemodynamics and arterial blood gases. On a single occasion, acepromazine was administered intravenously to each dog at 10, 15, 25 and 50 μg kg-1 at 20 minute intervals, resulting in cumulative acepromazine doses of 10 μg kg-1 (ACP10), 25 μg kg-1 (ACP25), 50 μg kg-1 (ACP50) and 100 μg kg-1 (ACP100). Hemodynamic data and sedation scores were recorded before (baseline) and 20 minutes after each acepromazine dose. RESULTS Compared with baseline, all acepromazine doses significantly decreased stroke index (SI), mean arterial pressure (MAP) and arterial oxygen content (CaO2) with maximum decreases of 16%, 17% and 21%, respectively. Cardiac index (CI) decreased by up to 19% but not significantly. Decreases of 26-38% were recorded for oxygen delivery index (DO2I), with significant differences for ACP50 and ACP100. Systemic vascular resistance index (SVRI) and heart rate did not change significantly. No significant difference was found among acepromazine doses for hemodynamic data. After ACP10, mild sedation was observed in five/six dogs and moderate sedation in one/six dogs, whereas after ACP25, ACP50 and ACP100, moderate sedation was observed in five/six or six/six dogs. CONCLUSIONS AND CLINICAL RELEVANCE In conscious dogs, acepromazine decreased MAP, SI, CaO2 and DO2I, but no significant dose effect was detected. SVRI was not significantly changed, suggesting that the reduction in MAP resulted from decreased CI. The ACP25, ACP50 and ACP100 doses resulted in moderate sedation in most dogs; ACP10 resulted in only mild sedation.
Collapse
|
29
|
MacDonald EA, Rose RA, Quinn TA. Neurohumoral Control of Sinoatrial Node Activity and Heart Rate: Insight From Experimental Models and Findings From Humans. Front Physiol 2020; 11:170. [PMID: 32194439 PMCID: PMC7063087 DOI: 10.3389/fphys.2020.00170] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 02/13/2020] [Indexed: 12/22/2022] Open
Abstract
The sinoatrial node is perhaps one of the most important tissues in the entire body: it is the natural pacemaker of the heart, making it responsible for initiating each-and-every normal heartbeat. As such, its activity is heavily controlled, allowing heart rate to rapidly adapt to changes in physiological demand. Control of sinoatrial node activity, however, is complex, occurring through the autonomic nervous system and various circulating and locally released factors. In this review we discuss the coupled-clock pacemaker system and how its manipulation by neurohumoral signaling alters heart rate, considering the multitude of canonical and non-canonical agents that are known to modulate sinoatrial node activity. For each, we discuss the principal receptors involved and known intracellular signaling and protein targets, highlighting gaps in our knowledge and understanding from experimental models and human studies that represent areas for future research.
Collapse
Affiliation(s)
- Eilidh A. MacDonald
- Department of Physiology and Biophysics, Dalhousie University, Halifax, NS, Canada
| | - Robert A. Rose
- Cumming School of Medicine, Libin Cardiovascular Institute of Alberta, University of Calgary, Calgary, AB, Canada
| | - T. Alexander Quinn
- Department of Physiology and Biophysics, Dalhousie University, Halifax, NS, Canada
- School of Biomedical Engineering, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
30
|
Stimulation of Alpha 1-Adrenergic Receptor Ameliorates Cellular Functions of Multiorgans beyond Vasomotion through PPAR δ. PPAR Res 2020; 2020:3785137. [PMID: 32099540 PMCID: PMC7016472 DOI: 10.1155/2020/3785137] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 12/02/2019] [Accepted: 01/03/2020] [Indexed: 02/07/2023] Open
Abstract
Cells can shift their metabolism between glycolysis and oxidative phosphorylation to enact their cell fate program in response to external signals. Widely distributed α1-adrenergic receptors (ARs) are physiologically stimulated during exercise, were reported to associate with the activating energetic AMPK pathway, and are expected to have biological effects beyond their hemodynamic effects. To investigate the effects and mechanism of AR stimulation on the physiology of the whole body, various in vitro and in vivo experiments were conducted using the AR agonist midodrine, 2-amino-N-[2-(2,5-dimethoxyphenyl)-2-hydroxy-ethyl]-acetamide. The expression of various biomarkers involved in ATP production was estimated through Western blotting, reverse transcription polymerase chain reaction, oxygen consumption rate, enzyme-linked immunosorbent assay (ELISA), fluorescence staining, and Oil red O staining in several cell lines (skeletal muscle, cardiac muscle, liver, macrophage, vascular endothelial, and adipose cells). In spontaneously hypertensive rats, blood pressure, blood analysis, organ-specific biomarkers, and general biomolecules related to ATP production were measured with Western blot analysis, immunohistochemistry, ELISA, and echocardiography. Pharmacological activation of α1-adrenergic receptors in C2C12 skeletal muscle cells promoted mitochondrial oxidative phosphorylation and ATP production by increasing the expression of catabolic molecules, including PPARδ, AMPK, and PGC-1α, through cytosolic calcium signaling and increased GLUT4 expression, as seen in exercise. It also activated those energetic molecules and mitochondrial oxidative phosphorylation with cardiomyocytes, endothelial cells, adipocytes, macrophages, and hepatic cells and affected their relevant cell-specific biological functions. All of those effects occurred around 3 h (and peaked 6 h) after midodrine treatment. In spontaneously hypertensive rats, α1-adrenergic receptor stimulation affected mitochondrial oxidative phosphorylation and ATP production by activating PPARδ, AMPK, and PGC-1α and the relevant biologic functions of multiple organs, suggesting organ crosstalk. The treatment lowered blood pressure, fat and body weight, cholesterol levels, and inflammatory activity; increased ATP content and insulin sensitivity in skeletal muscles; and increased cardiac contractile function without exercise training. These results suggest that the activation of α1-adrenergic receptor stimulates energetic reprogramming via PPARδ that increases mitochondrial oxidative phosphorylation and has healthy and organ-specific biological effects in multiple organs, including skeletal muscle, beyond its vasomotion effect. In addition, the action mechanism of α1-adrenergic receptor may be mainly exerted via PPARδ.
Collapse
|
31
|
Ramchandra R, Xing DT, Matear M, Lambert G, Allen AM, May CN. Neurohumoral interactions contributing to renal vasoconstriction and decreased renal blood flow in heart failure. Am J Physiol Regul Integr Comp Physiol 2019; 317:R386-R396. [DOI: 10.1152/ajpregu.00026.2019] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In heart failure (HF), increases in renal sympathetic nerve activity (RSNA), renal norepinephrine spillover, and renin release cause renal vasoconstriction, which may contribute to the cardiorenal syndrome. To increase our understanding of the mechanisms causing renal vasoconstriction in HF, we investigated the interactions between the increased activity of the renal nerves and the renal release of norepinephrine and renin in an ovine pacing-induced model of HF compared with healthy sheep. In addition, we determined the level of renal angiotensin type-1 receptors and the renal vascular responsiveness to stimulation of the renal nerves and α1-adrenoceptors. In conscious sheep with mild HF (ejection fraction 35%–40%), renal blood flow (276 ± 13 to 185 ± 18 mL/min) and renal vascular conductance (3.8 ± 0.2 to 3.1 ± 0.2 mL·min−1·mmHg−1) were decreased compared with healthy sheep. There were increases in the burst frequency of RSNA (27%), renal norepinephrine spillover (377%), and plasma renin activity (141%), whereas the density of renal medullary angiotensin type-1 receptors decreased. In anesthetized sheep with HF, the renal vasoconstrictor responses to electrical stimulation of the renal nerves or to phenylephrine were attenuated. Irbesartan improved the responses to nerve stimulation, but not to phenylephrine, in HF and reduced the responses in normal sheep. In summary, in HF, the increases in renal norepinephrine spillover and plasma renin activity are augmented compared with the increase in RSNA. The vasoconstrictor effect of the increased renal norepinephrine and angiotensin II is offset by reduced levels of renal angiotensin type-1 receptors and reduced renal vasoconstrictor responsiveness to α1-adrenoceptor stimulation.
Collapse
Affiliation(s)
- Rohit Ramchandra
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
- Department of Physiology, University of Auckland, Auckland, New Zealand
| | - Daniel T. Xing
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Marcus Matear
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
- Department of Physiology, University of Melbourne, Parkville, VIC, Australia
| | - Gavin Lambert
- Iverson Health Innovation Research Institute and Faculty of Health, Arts and Design, Swinburne University of Technology, Melbourne, VIC, Australia
| | - Andrew M. Allen
- Department of Physiology, University of Melbourne, Parkville, VIC, Australia
| | - Clive N. May
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
32
|
Cowley PM, Wang G, Swigart PM, Raghunathan A, Reddy N, Dulam P, Lovett DH, Simpson PC, Baker AJ. Reversal of right ventricular failure by chronic α 1A-subtype adrenergic agonist therapy. Am J Physiol Heart Circ Physiol 2019; 316:H224-H232. [PMID: 30412439 PMCID: PMC6859419 DOI: 10.1152/ajpheart.00507.2018] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 10/01/2018] [Accepted: 11/06/2018] [Indexed: 12/31/2022]
Abstract
Right ventricular (RV) failure (RVF) is a serious disease with no effective treatment available. We recently reported a disease prevention study showing that chronic stimulation of α1A-adrenergic receptors (α1A-ARs), started at the time of RV injury, prevented the development of RVF. The present study used a clinically relevant disease reversal design to test if chronic α1A-AR stimulation, started after RVF was established, could reverse RVF. RVF was induced surgically by pulmonary artery constriction in mice. Two weeks after pulmonary artery constriction, in vivo RV fractional shortening as assessed by MRI was reduced by half relative to sham-operated controls (25 ± 2%, n = 27, vs. 52 ± 2%, n = 13, P < 10-11). Subsequent chronic treatment with the α1A-AR agonist A61603 for a further 2 wk resulted in a substantial recovery of RV fractional shortening (to 41 ± 2%, n = 17, P < 10-7 by a paired t-test) along with recovery of voluntary exercise capacity. Mechanistically, chronic A61603 treatment resulted in increased activation of the prosurvival kinase ERK, increased abundance of the antiapoptosis factor Bcl-2, and decreased myocyte necrosis evidenced by a decreased serum level of cardiac troponin. Moreover, A61603 treatment caused increased abundance of the antioxidant glutathione peroxidase-1, decreased level of reactive oxygen species, and decreased oxidative modification (carbonylation) of myofilament proteins. Consistent with these effects, A61603 treatment resulted in increased force development by cardiac myofilaments, which might have contributed to increased RV function. These findings suggest that the α1A-AR is a therapeutic target to reverse established RVF. NEW & NOTEWORTHY Currently, there are no effective therapies for right ventricular (RV) failure (RVF). This project evaluated a novel therapy for RVF. In a mouse model of RVF, chronic stimulation of α1A-adrenergic receptors with the agonist A61603 resulted in recovery of in vivo RV function, improved exercise capacity, reduced oxidative stress-related carbonylation of contractile proteins, and increased myofilament force generation. These results suggest that the α1A-adrenergic receptor is a therapeutic target to treat RVF.
Collapse
Affiliation(s)
- Patrick M Cowley
- Veterans Affairs Medical Center, San Francisco, California, and Department of Medicine, University of California , San Francisco, California
| | - Guanying Wang
- Veterans Affairs Medical Center, San Francisco, California, and Department of Medicine, University of California , San Francisco, California
| | - Philip M Swigart
- Veterans Affairs Medical Center, San Francisco, California, and Department of Medicine, University of California , San Francisco, California
| | - Anaha Raghunathan
- Veterans Affairs Medical Center, San Francisco, California, and Department of Medicine, University of California , San Francisco, California
| | - Nikitha Reddy
- Veterans Affairs Medical Center, San Francisco, California, and Department of Medicine, University of California , San Francisco, California
| | - Pranavi Dulam
- Veterans Affairs Medical Center, San Francisco, California, and Department of Medicine, University of California , San Francisco, California
| | - David H Lovett
- Veterans Affairs Medical Center, San Francisco, California, and Department of Medicine, University of California , San Francisco, California
| | - Paul C Simpson
- Veterans Affairs Medical Center, San Francisco, California, and Department of Medicine, University of California , San Francisco, California
| | - Anthony J Baker
- Veterans Affairs Medical Center, San Francisco, California, and Department of Medicine, University of California , San Francisco, California
| |
Collapse
|
33
|
Jhun BS, O-Uchi J, Adaniya SM, Cypress MW, Yoon Y. Adrenergic Regulation of Drp1-Driven Mitochondrial Fission in Cardiac Physio-Pathology. Antioxidants (Basel) 2018; 7:antiox7120195. [PMID: 30567380 PMCID: PMC6316402 DOI: 10.3390/antiox7120195] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Revised: 12/13/2018] [Accepted: 12/14/2018] [Indexed: 12/28/2022] Open
Abstract
Abnormal mitochondrial morphology, especially fragmented mitochondria, and mitochondrial dysfunction are hallmarks of a variety of human diseases including heart failure (HF). Although emerging evidence suggests a link between mitochondrial fragmentation and cardiac dysfunction, it is still not well described which cardiac signaling pathway regulates mitochondrial morphology and function under pathophysiological conditions such as HF. Mitochondria change their shape and location via the activity of mitochondrial fission and fusion proteins. This mechanism is suggested as an important modulator for mitochondrial and cellular functions including bioenergetics, reactive oxygen species (ROS) generation, spatiotemporal dynamics of Ca2+ signaling, cell growth, and death in the mammalian cell- and tissue-specific manners. Recent reports show that a mitochondrial fission protein, dynamin-like/related protein 1 (DLP1/Drp1), is post-translationally modified via cell signaling pathways, which control its subcellular localization, stability, and activity in cardiomyocytes/heart. In this review, we summarize the possible molecular mechanisms for causing post-translational modifications (PTMs) of DLP1/Drp1 in cardiomyocytes, and further discuss how these PTMs of DLP1/Drp1 mediate abnormal mitochondrial morphology and mitochondrial dysfunction under adrenergic signaling activation that contributes to the development and progression of HF.
Collapse
Affiliation(s)
- Bong Sook Jhun
- Lillehei Heart Institute, Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Jin O-Uchi
- Lillehei Heart Institute, Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Stephanie M Adaniya
- Lillehei Heart Institute, Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA.
- Cardiovascular Research Center, Rhode Island Hospital, Providence, RI 02903, USA.
- Department of Medicine, Division of Cardiology, the Alpert Medical School of Brown University, Providence, RI 02903, USA.
| | - Michael W Cypress
- Lillehei Heart Institute, Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Yisang Yoon
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA.
| |
Collapse
|
34
|
Viswanathan G, Mamazhakypov A, Schermuly RT, Rajagopal S. The Role of G Protein-Coupled Receptors in the Right Ventricle in Pulmonary Hypertension. Front Cardiovasc Med 2018; 5:179. [PMID: 30619886 PMCID: PMC6305072 DOI: 10.3389/fcvm.2018.00179] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 11/30/2018] [Indexed: 12/14/2022] Open
Abstract
Pressure overload of the right ventricle (RV) in pulmonary arterial hypertension (PAH) leads to RV remodeling and failure, an important determinant of outcome in patients with PAH. Several G protein-coupled receptors (GPCRs) are differentially regulated in the RV myocardium, contributing to the pathogenesis of RV adverse remodeling and dysfunction. Many pharmacological agents that target GPCRs have been demonstrated to result in beneficial effects on left ventricular (LV) failure, such as beta-adrenergic receptor and angiotensin receptor antagonists. However, the role of such drugs on RV remodeling and performance is not known at this time. Moreover, many of these same receptors are also expressed in the pulmonary vasculature, which could result in complex effects in PAH. This manuscript reviews the role of GPCRs in the RV remodeling and dysfunction and discusses activating and blocking GPCR signaling to potentially attenuate remodeling while promoting improvements of RV function in PAH.
Collapse
Affiliation(s)
- Gayathri Viswanathan
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC, United States
| | - Argen Mamazhakypov
- Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Justus Liebig University of Giessen, Giessen, Germany
| | - Ralph T Schermuly
- Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Justus Liebig University of Giessen, Giessen, Germany
| | - Sudarshan Rajagopal
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC, United States
| |
Collapse
|
35
|
Janssen PM, Canan BD, Kilic A, Whitson BA, Baker AJ. Human Myocardium Has a Robust α1A-Subtype Adrenergic Receptor Inotropic Response. J Cardiovasc Pharmacol 2018; 72:136-142. [PMID: 29923888 PMCID: PMC6126952 DOI: 10.1097/fjc.0000000000000604] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Recent studies report that a single subtype of α1-adrenergic receptor (α1-AR), the α1A-subtype, mediates robust cardioprotective effects in multiple experimental models of heart failure, suggesting that the α1A-subtype is a potential therapeutic target for an agonist to treat heart failure. Moreover, we recently found that the α1A-subtype is present in human heart. The goal of this study was to assess the inotropic response mediated by the α1A-subtype in human myocardium, and to determine whether the response is downregulated in myocardium from failing human heart. We measured in vitro contractile responses of cardiac muscle preparations (trabeculae) isolated from the right ventricle from nonfailing and failing human hearts. Addition of the α1A-subtype agonist A61603 (100 nM) resulted in a large positive inotropic response (force increased ≈ 2-fold). This response represented ≈70% of the response mediated by the β-adrenergic receptor agonist isoproterenol (1 μM). Moreover, in myocardium from failing hearts, α1A-subtype responses remained robust, and only slightly reduced relative to nonfailing hearts. We conclude that α1A-subtype-mediated inotropy could represent a significant source of inotropic support in the human heart. Furthermore, the α1A-subtype remains functional in myocardium from failing human hearts and thus, might be a therapeutic target to support cardioprotective effects in patients with heart failure.
Collapse
Affiliation(s)
- Paul M.L. Janssen
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, Ohio
| | - Benjamin D. Canan
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, Ohio
| | - Ahmet Kilic
- Department of Surgery, The Ohio State University, Columbus, Ohio
| | - Bryan A Whitson
- Department of Surgery, The Ohio State University, Columbus, Ohio
| | - Anthony J. Baker
- Veterans Affairs Medical Center, San Francisco, and Department of Medicine, Univ. Calif. San Francisco, San Francisco
| |
Collapse
|
36
|
Zhang S, Takahashi R, Yamashita N, Teraoka H, Kitazawa T. Αlpha 1B-adrenoceptor-mediated positive inotropic and positive chronotropic actions in the mouse atrium. Eur J Pharmacol 2018; 839:82-88. [PMID: 30172786 DOI: 10.1016/j.ejphar.2018.08.038] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 08/21/2018] [Accepted: 08/29/2018] [Indexed: 11/15/2022]
Abstract
Modulation of cardiac contractility by α-adrenoceptor is well known in several mammals. Mice are useful experimental animals, but α-adrenoceptor-mediated responses have been examined only in the ventricles. To determine function of α-adrenoceptors in the atrium, effects of α-adrenoceptor agonists on spontaneous contraction and electrical-field stimulation (EFS)-induced contraction were examined. In addition, expression of α1A, α1B, α1D and β1-adrenoceptor mRNAs were examined. In the right atrium, noradrenaline and phenylephrine caused positive inotropic and positive chronotropic actions. However, methoxamine, clonidine and xylazine caused positive inotropic actions, but contractile frequency was decreased at high concentrations. Phenylephrine-induced positive inotropic and chronotropic actions were partially decreased by propranolol, and both actions remained in the presence of propranolol were inhibited by phentolamine or prazosin. A low concentration of silodosin (<100 nM) did not but a high concentration (1 μM) decreased the phenylephrine-induced chronotropic actions. Negative chronotropic actions of clonidine and xylazine were insensitive to propranolol and phentolamine. The EFS-induced contraction of the left atrium was potentiated by noradrenaline, phenylephrine and methoxamine but was not changed by clonidine or xylazine. Propranolol partially decreased the actions of phenylephrine, and prazosin caused additional inhibition. Expression of β1-, α1A-, α1B- and α1D-adrenoceptor mRNAs was found in the atrium, and the expression level of β1-adrenoceptor was the highest. Of α1-adrenoceptors, the expression level of α1B was higher than that of α1A and α1D. In conclusion, α1B-adrenoceptors are expressed in the mouse atrium and mediate both positive chronotropic and inotropic actions. In contrast, the α2-adrenoceptor is not functional in the isolated atrium.
Collapse
Affiliation(s)
- Shuangyi Zhang
- Veterinary Pharmacology, Department of Veterinary Medicine, School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Hokkaido 069-8501, Japan
| | - Rena Takahashi
- Comparative Animal Pharmacology, Department of Veterinary Science, School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Hokkaido 069-8501, Japan
| | - Natsumi Yamashita
- Veterinary Pharmacology, Department of Veterinary Medicine, School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Hokkaido 069-8501, Japan
| | - Hiroki Teraoka
- Veterinary Pharmacology, Department of Veterinary Medicine, School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Hokkaido 069-8501, Japan
| | - Takio Kitazawa
- Veterinary Pharmacology, Department of Veterinary Medicine, School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Hokkaido 069-8501, Japan; Comparative Animal Pharmacology, Department of Veterinary Science, School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Hokkaido 069-8501, Japan.
| |
Collapse
|
37
|
Lai YC, Li N, Lawrence W, Wang S, Levine A, Burchhardt DM, Pautler RG, Valderrábano M, Wehrens XH, Anderson AE. Myocardial remodeling and susceptibility to ventricular tachycardia in a model of chronic epilepsy. Epilepsia Open 2018; 3:213-223. [PMID: 29881800 PMCID: PMC5983128 DOI: 10.1002/epi4.12107] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/02/2018] [Indexed: 01/08/2023] Open
Abstract
Objective Sympathetic predominance and ventricular repolarization abnormalities represent epilepsy‐associated cardiac alterations and may underlie seizure‐induced ventricular arrhythmias. Myocardial ion channel and electrical remodeling have been described early in epilepsy development and may contribute to ventricular repolarization abnormalities and excitability. Using the pilocarpine‐induced acquired epilepsy model we sought to examine whether altered myocardial ion channel levels and electrophysiological changes also occur in animals with long‐standing epilepsy. Methods We examined myocardial adrenergic receptor and ion channel protein levels of epileptic and age‐matched sham rats (9–20 months old) using western blotting. Cardiac electrical properties were examined using optical mapping ex vivo and electrophysiology in vivo. We investigated the propensity for ventricular tachycardia (VT) and the effects of β‐adrenergic blockade on ventricular electrical properties and excitability in vivo. Results In animals with long‐standing epilepsy, we observed decreased myocardial voltage‐gated K+ channels Kv4.2 and Kv4.3, which are known to underlie early ventricular repolarization in rodents. Decreased β1 and increased α1A adrenergic receptor protein levels occurred in the myocardium of chronically epileptic animals consistent with elevated sympathetic tone. These animals exhibited many cardiac electrophysiological abnormalities, represented by longer QRS and corrected QT (QTc) intervals in vivo, slower conduction velocity ex vivo, and stimulation‐induced VT. Administration of a β‐adrenergic antagonist late in epilepsy was beneficial, as the therapy shortened the QTc interval and decreased stimulation‐induced VT. Significance Our findings demonstrate that myocardial ion channel remodeling and sympathetic predominance, risk factors for increased ventricular excitability and arrhythmias, persist in chronic epilepsy. The beneficial effects of β‐adrenergic antagonist treatment late in the course of epilepsy suggest that attenuating elevated sympathetic tone may represent a therapeutic target for ameliorating epilepsy‐associated cardiac morbidity.
Collapse
Affiliation(s)
- Yi-Chen Lai
- Department of Pediatrics Baylor College of Medicine Houston Texas U.S.A
| | - Na Li
- Department of Molecular Physiology and Biophysics Baylor College of Medicine Houston Texas U.S.A
| | - William Lawrence
- Department of Molecular Physiology and Biophysics Baylor College of Medicine Houston Texas U.S.A
| | - Sufen Wang
- DeBakey Heart and Vascular Center Methodist Hospital Research Institute Houston Texas U.S.A
| | - Amber Levine
- Department of Neuroscience Baylor College of Medicine Houston Texas U.S.A
| | | | - Robia G Pautler
- Department of Molecular Physiology and Biophysics Baylor College of Medicine Houston Texas U.S.A
| | - Miguel Valderrábano
- DeBakey Heart and Vascular Center Methodist Hospital Research Institute Houston Texas U.S.A
| | - Xander H Wehrens
- Department of Molecular Physiology and Biophysics Baylor College of Medicine Houston Texas U.S.A
| | - Anne E Anderson
- Department of Pediatrics Baylor College of Medicine Houston Texas U.S.A.,Department of Neuroscience Baylor College of Medicine Houston Texas U.S.A.,Department of Neurology Baylor College of Medicine Houston Texas U.S.A
| |
Collapse
|
38
|
An inactivating mutation in the histone deacetylase SIRT6 causes human perinatal lethality. Genes Dev 2018; 32:373-388. [PMID: 29555651 PMCID: PMC5900711 DOI: 10.1101/gad.307330.117] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 02/26/2018] [Indexed: 12/27/2022]
Abstract
Ferrer et al. demonstrate that a homozygous inactivating mutation in the histone deacetylase SIRT6 results in severe congenital anomalies and perinatal lethality in four affected fetuses. Human induced pluripotent stem cells derived from D63H homozygous fetuses fail to differentiate into embryoid bodies, functional cardiomyocytes, and neural progenitor cells due to a failure to repress pluripotent genes. It has been well established that histone and DNA modifications are critical to maintaining the equilibrium between pluripotency and differentiation during early embryogenesis. Mutations in key regulators of DNA methylation have shown that the balance between gene regulation and function is critical during neural development in early years of life. However, there have been no identified cases linking epigenetic regulators to aberrant human development and fetal demise. Here, we demonstrate that a homozygous inactivating mutation in the histone deacetylase SIRT6 results in severe congenital anomalies and perinatal lethality in four affected fetuses. In vitro, the amino acid change at Asp63 to a histidine results in virtually complete loss of H3K9 deacetylase and demyristoylase functions. Functionally, SIRT6 D63H mouse embryonic stem cells (mESCs) fail to repress pluripotent gene expression, direct targets of SIRT6, and exhibit an even more severe phenotype than Sirt6-deficient ESCs when differentiated into embryoid bodies (EBs). When terminally differentiated toward cardiomyocyte lineage, D63H mutant mESCs maintain expression of pluripotent genes and fail to form functional cardiomyocyte foci. Last, human induced pluripotent stem cells (iPSCs) derived from D63H homozygous fetuses fail to differentiate into EBs, functional cardiomyocytes, and neural progenitor cells due to a failure to repress pluripotent genes. Altogether, our study described a germline mutation in SIRT6 as a cause for fetal demise, defining SIRT6 as a key factor in human development and identifying the first mutation in a chromatin factor behind a human syndrome of perinatal lethality.
Collapse
|
39
|
Kanae H, Hamaguchi S, Wakasugi Y, Kusakabe T, Kato K, Namekata I, Tanaka H. Pathological prolongation of action potential duration as a cause of the reduced alpha-adrenoceptor-mediated negative inotropy in streptozotocin-induced diabetic mice myocardium. J Pharmacol Sci 2017; 135:131-133. [DOI: 10.1016/j.jphs.2017.10.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 10/09/2017] [Accepted: 10/12/2017] [Indexed: 10/18/2022] Open
|
40
|
Abbott GW. β Subunits Control the Effects of Human Kv4.3 Potassium Channel Phosphorylation. Front Physiol 2017; 8:646. [PMID: 28919864 PMCID: PMC5585193 DOI: 10.3389/fphys.2017.00646] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 08/16/2017] [Indexed: 11/17/2022] Open
Abstract
The transient outward K+ current, Ito, activates early in the cardiac myocyte action potential, to begin repolarization. Human Ito is generated primarily by two Kv4.3 potassium channel α subunit splice variants (Kv4.3L and Kv4.3S) that diverge only by a C-terminal, membrane-proximal, 19-residue stretch unique to Kv4.3L. Protein kinase C (PKC) phosphorylation of threonine 504 within the Kv4.3L-specific 19-residues mediates α-adrenergic inhibition of Ito in human heart. Kv4.3 is regulated in human heart by various β subunits, including cytosolic KChIP2b and transmembrane KCNEs, yet their impact on the functional effects of human Kv4.3 phosphorylation has not been reported. Here, this gap in knowledge was addressed using human Kv4.3 splice variants, T504 mutants, and human β subunits. Subunits were co-expressed in Xenopus laevis oocytes and analyzed by two-electrode voltage-clamp, using phorbol 12-myristate 13-acetate (PMA) to stimulate PKC. Unexpectedly, KChIP2b removed the inhibitory effect of PKC on Kv4.3L (but not Kv4.3L threonine phosphorylation by PKC per-se), while co-expression with KCNE2, but not KCNE4, restored PKC-dependent inhibition of Kv4.3L-KChIP2b to quantitatively resemble previously reported effects of α-adrenergic modulation of human ventricular Ito. In addition, PKC accelerated recovery from inactivation of Kv4.3L-KChIP2b channels and, interestingly, of both Kv4.3L and Kv4.3S alone. Thus, β subunits regulate the response of human Kv4.3 to PKC phosphorylation and provide a potential mechanism for modifying the response of Ito to α-adrenergic regulation in vivo.
Collapse
Affiliation(s)
- Geoffrey W Abbott
- Bioelectricity Laboratory, Department of Physiology and Biophysics, School of Medicine, University of California, IrvineIrvine, CA, United States
| |
Collapse
|
41
|
Caso S, Maric D, Arambasic M, Cotecchia S, Diviani D. AKAP-Lbc mediates protection against doxorubicin-induced cardiomyocyte toxicity. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1864:2336-2346. [PMID: 28923249 DOI: 10.1016/j.bbamcr.2017.09.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 08/30/2017] [Accepted: 09/14/2017] [Indexed: 12/31/2022]
Abstract
Doxorubicin (DOX) is a chemotherapic agent that is widely used to treat hematological and solid tumors. Despite its efficacy, DOX displays significant cardiac toxicity associated with cardiomyocytes death and heart failure. Cardiac toxicity is mainly associated with the ability of DOX to alter mitochondrial function. The current lack of treatments to efficiently prevent DOX cardiotoxicity underscores the need of new therapeutic approaches. Our current findings show that stimulation of cardiomyocytes with the α1-adrenergic receptor (AR) agonist phenylephrine (PE) significantly inhibits the apoptotic effect of DOX. Importantly, our results indicate that AKAP-Lbc is critical for transducing protective signals downstream of α1-ARs. In particular, we could show that suppression of AKAP-Lbc expression by infecting primary cultures of ventricular myocytes with lentiviruses encoding AKAP-Lbc specific short hairpin (sh) RNAs strongly impairs the ability of PE to reduce DOX-induced apoptosis. AKAP-Lbc-mediated cardiomyocyte protection requires the activation of anchored protein kinase D1 (PKD1)-dependent prosurvival pathways that promote the expression of the anti-apoptotic protein Bcl2 and inhibit the translocation of the pro-apoptotic protein Bax to mitochondria. In conclusion, AKAP-Lbc emerges as a coordinator of signals that protect cardiomyocytes against the toxic effects of DOX.
Collapse
Affiliation(s)
- Stefania Caso
- Département de Pharmacologie et de Toxicologie, Faculté de Biologie et de Médecine, Lausanne 1005, Switzerland; Dipartimento di Bioscienze, Biotecnologie e Biofarmaceutica, Università di Bari, Via Orabona 4, 70125 Bari, Italy
| | - Darko Maric
- Département de Pharmacologie et de Toxicologie, Faculté de Biologie et de Médecine, Lausanne 1005, Switzerland
| | - Miroslav Arambasic
- Département de Pharmacologie et de Toxicologie, Faculté de Biologie et de Médecine, Lausanne 1005, Switzerland
| | - Susanna Cotecchia
- Dipartimento di Bioscienze, Biotecnologie e Biofarmaceutica, Università di Bari, Via Orabona 4, 70125 Bari, Italy
| | - Dario Diviani
- Département de Pharmacologie et de Toxicologie, Faculté de Biologie et de Médecine, Lausanne 1005, Switzerland.
| |
Collapse
|
42
|
Cowley PM, Wang G, Joshi S, Swigart PM, Lovett DH, Simpson PC, Baker AJ. α 1A-Subtype adrenergic agonist therapy for the failing right ventricle. Am J Physiol Heart Circ Physiol 2017; 313:H1109-H1118. [PMID: 28822963 DOI: 10.1152/ajpheart.00153.2017] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 07/06/2017] [Accepted: 08/10/2017] [Indexed: 11/22/2022]
Abstract
Failure of the right ventricle (RV) is a serious disease with a poor prognosis and limited treatment options. Signaling by α1-adrenergic receptors (α1-ARs), in particular the α1A-subtype, mediate cardioprotective effects in multiple heart failure models. Recent studies have shown that chronic treatment with the α1A-subtype agonist A61603 improves function and survival in a model of left ventricular failure. The goal of the present study was to determine if chronic A61603 treatment is beneficial in a RV failure model. We used tracheal instillation of the fibrogenic antibiotic bleomycin in mice to induce pulmonary fibrosis, pulmonary hypertension, and RV failure within 2 wk. Some mice were chronically treated with a low dose of A61603 (10 ng·kg-1·day-1). In the bleomycin model of RV failure, chronic A61603 treatment was associated with improved RV fractional shortening and greater in vitro force development by RV muscle preparations. Cell injury markers were reduced with A61603 treatment (serum cardiac troponin I, RV fibrosis, and expression of matrix metalloproteinase-2). RV oxidative stress was reduced (using the probes dihydroethidium and 4-hydroxynonenal). Consistent with lowered RV oxidative stress, A61603 was associated with an increased level of the cellular antioxidant superoxide dismutase 1 and a lower level of the prooxidant NAD(P)H oxidase isoform NOX4. In summary, in the bleomycin model of RV failure, chronic A61603 treatment reduced RV oxidative stress, RV myocyte necrosis, and RV fibrosis and increased both RV function and in vitro force development. These findings suggest that in the context of pulmonary fibrosis, the α1A-subtype is a potential therapeutic target to treat the failing RV.NEW & NOTEWORTHY Right ventricular (RV) failure is a serious disease with a poor prognosis and no effective treatments. In the mouse bleomycin model of RV failure, we tested the efficacy of a treatment using the α1A-adrenergic receptor subtype agonist A61603. Chronic A61603 treatment improved RV contraction and reduced multiple indexes of RV injury, suggesting that the α1A-subtype is a therapeutic target to treat RV failure.
Collapse
Affiliation(s)
- Patrick M Cowley
- Veterans Affairs Medical Center, San Francisco, California, and Department of Medicine, University of California, San Francisco, California
| | - Guanying Wang
- Veterans Affairs Medical Center, San Francisco, California, and Department of Medicine, University of California, San Francisco, California
| | - Sunil Joshi
- Veterans Affairs Medical Center, San Francisco, California, and Department of Medicine, University of California, San Francisco, California
| | - Philip M Swigart
- Veterans Affairs Medical Center, San Francisco, California, and Department of Medicine, University of California, San Francisco, California
| | - David H Lovett
- Veterans Affairs Medical Center, San Francisco, California, and Department of Medicine, University of California, San Francisco, California
| | - Paul C Simpson
- Veterans Affairs Medical Center, San Francisco, California, and Department of Medicine, University of California, San Francisco, California
| | - Anthony J Baker
- Veterans Affairs Medical Center, San Francisco, California, and Department of Medicine, University of California, San Francisco, California
| |
Collapse
|
43
|
Heart-rate response to alpha 2-adrenergic receptor antagonism by antipsychotics. Clin Auton Res 2017; 27:407-410. [PMID: 28674870 DOI: 10.1007/s10286-017-0444-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2017] [Accepted: 06/20/2017] [Indexed: 12/12/2022]
Abstract
PURPOSE To explore the relationship between antipsychotic-associated antagonism of alpha2-adrenergic receptors and resting heart rate in individuals with schizophrenia. METHODS Thirty-one inpatients treated with antipsychotics were included in this exploratory analysis. Antipsychotic doses were converted to haloperidol equivalents for alpha2-adrenergic receptor antagonism. Resting heart rate was measured with the patient in the seated upright posture. RESULTS After controlling for confounding variables, the relationship between alpha2-adrenergic receptor antagonism and resting heart rate demonstrated a positive linear effect (P = 0.002) as well as a nonlinear effect that accounted for an additional 14% of the variability in resting heart rate (P = 0.005). CONCLUSION The observed inverted-U relationship between alpha2-adrenergic receptor antagonism and resting heart rate can possibly be attributed to an altered response of beta1-adrenergic receptors to increased norepinephrine release. Further investigations are required to confirm this exploratory finding, taking into account additional variables that include other receptors which either directly or indirectly influence heart rate. CLINICALTRIALS. GOV IDENTIFIER NCT01392885.
Collapse
|
44
|
Long-term effect of a chronic low-protein multideficient diet on the heart: Hypertension and heart failure in chronically malnourished young adult rats. Int J Cardiol 2017; 238:43-56. [DOI: 10.1016/j.ijcard.2017.03.110] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 03/16/2017] [Accepted: 03/24/2017] [Indexed: 01/21/2023]
|
45
|
Arce C, Vicente D, Segura V, Flacco N, Montó F, Almenar L, Agüero J, Rueda J, Jiménez-Altayó F, Vila E, Noguera MA, D'Ocon P, Ivorra MD. Activation of α 1A -adrenoceptors desensitizes the rat aorta response to phenylephrine through a neuronal NOS pathway, a mechanism lost with ageing. Br J Pharmacol 2017; 174:2015-2030. [PMID: 28369791 DOI: 10.1111/bph.13800] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 03/07/2017] [Accepted: 03/09/2017] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND AND PURPOSE A NO-mediated desensitization of vasoconstrictor responses evoked by stimulation of α1 -adrenoceptors has been reported in different vessels. We investigated the involvement of each α1 -adrenoceptor subtype and constitutive NOS isoforms and the influence of ageing and hypertension on this process. EXPERIMENTAL APPROACH Wistar and spontaneously hypertensive rats (SHR), 16, 32, 52 and 72 weeks-old, were used to evaluate the desensitization process. Expression of α1 -adrenoceptor subtypes, endothelial NOS (eNOS) and neuronal NOS (nNOS) were determined in rat aorta and left ventricle (LV). Expression levels were also evaluated in LV of a group of heart failure patients with a wide age range. KEY RESULTS Repeated application of phenylephrine decreased subsequent α1 -adrenoceptor-mediated vasoconstriction by increasing nNOS protein expression in aorta, but not in tail or mesenteric resistance arteries, where mRNA levels of nNOS were undetectable. This desensitization process disappeared in the absence of endothelium or in the presence of L-NAME (100 μM), nNOS inhibitors, SMTC (1 μM) and TRIM (100 μM), and 5-methylurapidil (100 nM, α1A -antagonist), but not BMY7378 (10 nM, α1D -antagonist). The α1A /nNOS-mediated desensitization was absent in aged SHR and Wistar animals, where the expression of α1A -adrenoceptors was reduced in aorta and LV. In human LV, a negative correlation was found between age and α1A -adrenoceptor expression. CONCLUSIONS AND IMPLICATIONS The α1A -adrenoceptor subtype, through endothelial nNOS-derived NO, may act as a physiological 'brake' against the detrimental effects of excessive α1 -adrenoceptor-mediated vasoconstriction. Reduced α1A -adrenoceptor- and nNOS-mediated desensitization in aged patients could be involved in the age-dependent elevation of adrenergic activity.
Collapse
Affiliation(s)
- Cristina Arce
- Departamento de Farmacología, Facultad de Farmacia, Universitat de València, Burjassot, Spain.,Estructura de Recerca Interdisciplinar en Biotecnologia i Biomedicina (ERI BIOTECMED), Universitat de València, Burjassot, Spain
| | - Diana Vicente
- Departamento de Farmacología, Facultad de Farmacia, Universitat de València, Burjassot, Spain
| | - Vanessa Segura
- Departamento de Farmacología, Facultad de Farmacia, Universitat de València, Burjassot, Spain
| | - Nicla Flacco
- Departamento de Farmacología, Facultad de Farmacia, Universitat de València, Burjassot, Spain
| | - Fermi Montó
- Departamento de Farmacología, Facultad de Farmacia, Universitat de València, Burjassot, Spain.,Estructura de Recerca Interdisciplinar en Biotecnologia i Biomedicina (ERI BIOTECMED), Universitat de València, Burjassot, Spain
| | - Luis Almenar
- Unidad de Insuficiencia Cardiaca y Trasplantes, Servicio de Cardiología, Hospital Universitario La Fe, Valencia, Spain
| | - Jaime Agüero
- Unidad de Insuficiencia Cardiaca y Trasplantes, Servicio de Cardiología, Hospital Universitario La Fe, Valencia, Spain
| | - Joaquín Rueda
- Unidad de Insuficiencia Cardiaca y Trasplantes, Servicio de Cardiología, Hospital Universitario La Fe, Valencia, Spain
| | - Francesc Jiménez-Altayó
- Facultat de Medicina, Departament de Farmacologia, Terapèutica i Toxicologia, Institut de Neurociències, Universitat Autònoma de Barcelona, Bellaterra (Cerdanyola del Vallès), Spain
| | - Elisabet Vila
- Facultat de Medicina, Departament de Farmacologia, Terapèutica i Toxicologia, Institut de Neurociències, Universitat Autònoma de Barcelona, Bellaterra (Cerdanyola del Vallès), Spain
| | - Maria Antonia Noguera
- Departamento de Farmacología, Facultad de Farmacia, Universitat de València, Burjassot, Spain.,Estructura de Recerca Interdisciplinar en Biotecnologia i Biomedicina (ERI BIOTECMED), Universitat de València, Burjassot, Spain
| | - Pilar D'Ocon
- Departamento de Farmacología, Facultad de Farmacia, Universitat de València, Burjassot, Spain.,Estructura de Recerca Interdisciplinar en Biotecnologia i Biomedicina (ERI BIOTECMED), Universitat de València, Burjassot, Spain
| | - Maria Dolores Ivorra
- Departamento de Farmacología, Facultad de Farmacia, Universitat de València, Burjassot, Spain.,Estructura de Recerca Interdisciplinar en Biotecnologia i Biomedicina (ERI BIOTECMED), Universitat de València, Burjassot, Spain
| |
Collapse
|
46
|
García-Canales A, Peña-Juárez RA, Sandoval-Franco LDM. [Vasopressors and inotropes: use in paediatrics]. ARCHIVOS DE CARDIOLOGIA DE MEXICO 2017; 88:39-50. [PMID: 28336302 DOI: 10.1016/j.acmx.2017.02.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 02/14/2017] [Accepted: 02/15/2017] [Indexed: 11/16/2022] Open
Abstract
The cardiovascular system is a dynamic system, which is required to ensure adequate delivery of oxygen, nutrients, and hormones to the tissues that are necessary for cell metabolism. It also synthesises and modifies the vasoactive components that regulate vascular tone and myocardial function. These vasoactive components have demonstrated their beneficial effects in the management of paediatric patients in a critical condition with heart failure and shock. However, their use and abuse brings harmful effects, increases mortality, and is associated with arrhythmias. An increase in myocardial oxygen consumption favours the presence of ischaemia, therefore it is necessary to know the mechanism of action and indications of these drugs to minimise their harmful effects. The purpose of this review is to describe the pharmacology and clinical applications of inotropic and vasopressor agents in the paediatric patient in acritical condition.
Collapse
Affiliation(s)
- Adrián García-Canales
- Departamento de Terapia Intensiva Pediátrica, Hospital Regional Valentín Gómez Farías, ISSSTE, Zapopan, Jalisco, México.
| | | | | |
Collapse
|
47
|
Lynch JJ, Van Vleet TR, Mittelstadt SW, Blomme EAG. Potential functional and pathological side effects related to off-target pharmacological activity. J Pharmacol Toxicol Methods 2017; 87:108-126. [PMID: 28216264 DOI: 10.1016/j.vascn.2017.02.020] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 01/24/2017] [Accepted: 02/15/2017] [Indexed: 12/22/2022]
Abstract
Most pharmaceutical companies test their discovery-stage proprietary molecules in a battery of in vitro pharmacology assays to try to determine off-target interactions. During all phases of drug discovery and development, various questions arise regarding potential side effects associated with such off-target pharmacological activity. Here we present a scientific literature curation effort undertaken to determine and summarize the most likely functional and pathological outcomes associated with interactions at 70 receptors, enzymes, ion channels and transporters with established links to adverse effects. To that end, the scientific literature was reviewed using an on-line database, and the most commonly reported effects were summarized in tabular format. The resultant table should serve as a practical guide for research scientists and clinical investigators for the prediction and interpretation of adverse side effects associated with molecules interacting with components of this screening battery.
Collapse
Affiliation(s)
- James J Lynch
- AbbVie Inc., 1 North Waukegan Road, North Chicago, IL 60064, USA.
| | | | | | - Eric A G Blomme
- AbbVie Inc., 1 North Waukegan Road, North Chicago, IL 60064, USA
| |
Collapse
|
48
|
Abstract
There are 2 α1-ARs on cardiac myocytes: α1A and α1B. α1A adrenergic receptors serve important cardioprotective roles and do not mediate cardiac hypertrophy. Dabuzalgron, an oral α1A-AR agonist developed for the treatment of urinary incontinence and tolerated well in Phase 2 clinical trials, protects against doxorubicin-induced cardiotoxicity in vivo. Dabuzalgron enhances contractile function, regulates transcription of genes related to energy production and mitochondrial function, and preserves myocardial ATP content after doxorubicin. Activation of α1A-ARs on cardiomyocytes protects against doxorubicin cytotoxicity and enhances mitochondrial function in vitro. These cytoprotective effects likely are mediated by activation of ERK 1/2. Future studies will explore whether dabuzalgron, a well-tolerated oral α1A-AR agonist, might be repurposed to treat heart failure.
Alpha-1 adrenergic receptors (α1-ARs) play adaptive and protective roles in the heart. Dabuzalgron is an oral selective α1A-AR agonist that was well tolerated in multiple clinical trials of treatment for urinary incontinence, but has never been used to treat heart disease in humans or animal models. In this study, the authors administered dabuzalgron to mice treated with doxorubicin (DOX), a widely used chemotherapeutic agent with dose-limiting cardiotoxicity that can lead to heart failure (HF). Dabuzalgron protected against DOX-induced cardiotoxicity, likely by preserving mitochondrial function. These results suggest that activating cardiac α1A-ARs with dabuzalgron, a well-tolerated oral agent, might represent a novel approach to treating HF.
Collapse
|
49
|
Montgomery MD, Chan T, Swigart PM, Myagmar BE, Dash R, Simpson PC. An Alpha-1A Adrenergic Receptor Agonist Prevents Acute Doxorubicin Cardiomyopathy in Male Mice. PLoS One 2017; 12:e0168409. [PMID: 28081170 PMCID: PMC5231318 DOI: 10.1371/journal.pone.0168409] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 11/29/2016] [Indexed: 01/12/2023] Open
Abstract
Alpha-1 adrenergic receptors mediate adaptive effects in the heart and cardiac myocytes, and a myocyte survival pathway involving the alpha-1A receptor subtype and ERK activation exists in vitro. However, data in vivo are limited. Here we tested A61603 (N-[5-(4,5-dihydro-1H-imidazol-2-yl)-2-hydroxy-5,6,7,8-tetrahydronaphthalen-1-yl]methanesulfonamide), a selective imidazoline agonist for the alpha-1A. A61603 was the most potent alpha-1-agonist in activating ERK in neonatal rat ventricular myocytes. A61603 activated ERK in adult mouse ventricular myocytes and protected the cells from death caused by the anthracycline doxorubicin. A low dose of A61603 (10 ng/kg/d) activated ERK in the mouse heart in vivo, but did not change blood pressure. In male mice, concurrent subcutaneous A61603 infusion at 10 ng/kg/d for 7 days after a single intraperitoneal dose of doxorubicin (25 mg/kg) increased survival, improved cardiac function, heart rate, and cardiac output by echocardiography, and reduced cardiac cell necrosis and apoptosis and myocardial fibrosis. All protective effects were lost in alpha-1A-knockout mice. In female mice, doxorubicin at doses higher than in males (35-40 mg/kg) caused less cardiac toxicity than in males. We conclude that the alpha-1A-selective agonist A61603, via the alpha-1A adrenergic receptor, prevents doxorubicin cardiomyopathy in male mice, supporting the theory that alpha-1A adrenergic receptor agonists have potential as novel heart failure therapies.
Collapse
Affiliation(s)
- Megan D. Montgomery
- Department of Medicine, Cardiology Division, VA Medical Center, San Francisco, CA, United States of America
- Department of Medicine and Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, United States of America
| | - Trevor Chan
- Department of Medicine, Cardiology Division, VA Medical Center, San Francisco, CA, United States of America
- Department of Medicine and Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, United States of America
| | - Philip M. Swigart
- Department of Medicine, Cardiology Division, VA Medical Center, San Francisco, CA, United States of America
- Department of Medicine and Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, United States of America
| | - Bat-erdene Myagmar
- Department of Medicine, Cardiology Division, VA Medical Center, San Francisco, CA, United States of America
- Department of Medicine and Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, United States of America
| | - Rajesh Dash
- Department of Medicine, Cardiology Division, VA Medical Center, San Francisco, CA, United States of America
- Department of Medicine and Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, United States of America
| | - Paul C. Simpson
- Department of Medicine, Cardiology Division, VA Medical Center, San Francisco, CA, United States of America
- Department of Medicine and Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, United States of America
| |
Collapse
|
50
|
Willis MS, Ilaiwy A, Montgomery MD, Simpson PC, Jensen BC. The alpha-1A adrenergic receptor agonist A61603 reduces cardiac polyunsaturated fatty acid and endocannabinoid metabolites associated with inflammation in vivo. Metabolomics 2016; 12:155. [PMID: 28533737 PMCID: PMC5437747 DOI: 10.1007/s11306-016-1097-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
INTRODUCTION Alpha-1-adrenergic receptors (α1-ARs) are G-protein coupled receptors (GPCRs) with three highly homologous subtypes (α1A, α1B, and α1D). Of these three subtypes, only the α1A and α1B are expressed in the heart. Multiple pre-clinical models of heart injury demonstrate cardioprotective roles for the α1A. Non-selective α1-AR activation promotes glycolysis in the heart, but the functional α1-AR subtype and broader metabolic effects have not been studied. OBJECTIVES Given the high metabolic demands of the heart and previous evidence indicating benefit from α1A activation, we chose to investigate the effects of α1A activation on the cardiac metabolome in vivo. METHODS Mice were treated for one week with a low, subpressor dose of A61603, a highly selective and potent α1A agonist. Cardiac tissue and serum were analyzed using a non-targeted metabolomics approach. RESULTS We identified previously unrecognized metabolic responses to α1A activation, most notably broad reduction in the abundance of polyunsaturated fatty acids (PUFAs) and endocannabinoids (ECs). CONCLUSION Given the well characterized roles of PUFAs and ECs in inflammatory pathways, these findings suggest a possible role for cardiac α1A-ARs in the regulation of inflammation and may offer novel insight into the mechanisms underlying the cardioprotective benefit of selective pharmacologic α1A activation.
Collapse
Affiliation(s)
- Monte S. Willis
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC USA
- Department of Pathology & Laboratory Medicine, University of North Carolina, Chapel Hill, NC USA
- Department of Pharmacology, University of North Carolina, Chapel Hill, NC USA
| | - Amro Ilaiwy
- Sarah W. Stedman Nutrition and Metabolism Center, Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA
- Division of Endocrinology, Metabolism, and Nutrition, Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | | | - Paul C. Simpson
- VA Medical Center and University of California, San Francisco, CA, USA
| | - Brian C. Jensen
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC USA
- Department of Internal Medicine, Division of Cardiology University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|