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
|
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
|
3
|
Gural B, Kirkland L, Hockett A, Sandroni P, Zhang J, Rosa-Garrido M, Swift SK, Chapski D, Flinn MA, O'Meara CC, Vondriska TM, Patterson M, Jensen BC, Rau CD. Novel Insights into Post-Myocardial Infarction Cardiac Remodeling through Algorithmic Detection of Cell-Type Composition Shifts. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.09.607400. [PMID: 39149394 PMCID: PMC11326268 DOI: 10.1101/2024.08.09.607400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Background Recent advances in single cell sequencing have led to an increased focus on the role of cell-type composition in phenotypic presentation and disease progression. Cell-type composition research in the heart is challenging due to large, frequently multinucleated cardiomyocytes that preclude most single cell approaches from obtaining accurate measurements of cell composition. Our in silico studies reveal that ignoring cell type composition when calculating differentially expressed genes (DEGs) can have significant consequences. For example, a relatively small change in cell abundance of only 10% can result in over 25% of DEGs being false positives. Methods We have implemented an algorithmic approach that uses snRNAseq datasets as a reference to accurately calculate cell type compositions from bulk RNAseq datasets through robust data cleaning, gene selection, and multi-sample cross-subject and cross-cell-type deconvolution. We applied our approach to cardiomyocyte-specific α1A adrenergic receptor (CM-α1A-AR) knockout mice. 8-12 week-old mice (either WT or CM-α1A-KO) were subjected to permanent left coronary artery (LCA) ligation or sham surgery (n=4 per group). Transcriptomes from the infarct border zones were collected 3 days later and analyzed using our algorithm to determine cell-type abundances, corrected differential expression calculations using DESeq2, and validated these findings using RNAscope. Results Uncorrected DEGs for the CM-α1A-KO X LCA interaction term featured many cell-type specific genes such as Timp4 (fibroblasts) and Aplnr (cardiomyocytes) and overall GO enrichment for terms pertaining to cardiomyocyte differentiation (P=3.1E-4). Using our algorithm, we observe a striking loss of cardiomyocytes and gain in fibroblasts in the α1A-KO + LCA mice that was not recapitulated in WT + LCA animals, although we did observe a similar increase in macrophage abundance in both conditions. This recapitulates prior results that showed a much more severe heart failure phenotype in CM-α1A-KO + LCA mice. Following correction for cell-type, our DEGs now highlight a novel set of genes enriched for GO terms such as cardiac contraction (P=3.7E-5) and actin filament organization (P=6.3E-5). Conclusions Our algorithm identifies and corrects for cell-type abundance in bulk RNAseq datasets opening new avenues for research on novel genes and pathways as well as an improved understanding of the role of cardiac cell types in cardiovascular disease.
Collapse
Affiliation(s)
- Brian Gural
- Department of Genetics and Computational Medicine Program, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Logan Kirkland
- McAllister Heart Institute, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Medicine, Division of Cardiology, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Abbey Hockett
- Department of Genetics and Computational Medicine Program, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Peyton Sandroni
- Department of Pharmacology, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jiandong Zhang
- McAllister Heart Institute, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Medicine, Division of Cardiology, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Manuel Rosa-Garrido
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Samantha K Swift
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Douglas Chapski
- Departments of Anesthesiology & Perioperative Medicine, Medicine/Cardiology, and Physiology, David Geffen School of Medicine; Molecular Biology Institute; University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Michael A Flinn
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Caitlin C O'Meara
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Thomas M Vondriska
- Departments of Anesthesiology & Perioperative Medicine, Medicine/Cardiology, and Physiology, David Geffen School of Medicine; Molecular Biology Institute; University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Michaela Patterson
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Brian C Jensen
- McAllister Heart Institute, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Medicine, Division of Cardiology, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Pharmacology, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Christoph D Rau
- Department of Genetics and Computational Medicine Program, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
4
|
Chatham JC, Patel RP. Protein glycosylation in cardiovascular health and disease. Nat Rev Cardiol 2024; 21:525-544. [PMID: 38499867 DOI: 10.1038/s41569-024-00998-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/13/2024] [Indexed: 03/20/2024]
Abstract
Protein glycosylation, which involves the attachment of carbohydrates to proteins, is one of the most abundant protein co-translational and post-translational modifications. Advances in technology have substantially increased our knowledge of the biosynthetic pathways involved in protein glycosylation, as well as how changes in glycosylation can affect cell function. In addition, our understanding of the role of protein glycosylation in disease processes is growing, particularly in the context of immune system function, infectious diseases, neurodegeneration and cancer. Several decades ago, cell surface glycoproteins were found to have an important role in regulating ion transport across the cardiac sarcolemma. However, with very few exceptions, our understanding of how changes in protein glycosylation influence cardiovascular (patho)physiology remains remarkably limited. Therefore, in this Review, we aim to provide an overview of N-linked and O-linked protein glycosylation, including intracellular O-linked N-acetylglucosamine protein modification. We discuss our current understanding of how all forms of protein glycosylation contribute to normal cardiovascular function and their roles in cardiovascular disease. Finally, we highlight potential gaps in our knowledge about the effects of protein glycosylation on the heart and vascular system, highlighting areas for future research.
Collapse
Affiliation(s)
- John C Chatham
- Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Rakesh P Patel
- Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
5
|
Diab T, El-Shaer W, Ibrahim S, El-Barky E, Elezz AA. Does preoperative silodosin administration facilitate ureteral dilatation during flexible ureterorenoscopy? A randomized clinical trial. Int Urol Nephrol 2024; 56:839-846. [PMID: 37902925 PMCID: PMC10853317 DOI: 10.1007/s11255-023-03824-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 09/26/2023] [Indexed: 11/01/2023]
Abstract
PURPOSE To assess whether preoperative administration of silodosin can facilitate the placement of ureteral access sheath (UAS) prior to flexible ureteroscopy (F-URS) and reduce the occurrence of ureteric injury in challenging cases. METHODS This prospective randomized clinical trial was carried out on 147 patients diagnosed with upper ureteric stone or stone kidney, non-stented. The patients were randomly divided into two equal groups. Group A (silodosin group) included patients in whom F-URS was done with daily preoperative intake of 8 mg silodosin for 1 week and group B (placebo/control group) included patients in whom F-URS was done with daily preoperative intake of placebo tablets. RESULTS In group A, a total of 23 (33.3%) experienced ureteral wall injury following UAS insertion, while in group B, this occurred in 40 patients (59.7%). There was a statistically significant difference in the grade of ureteral wall injury between the two groups (P < 0.001). In the multiple regression analysis, age, operative time and silodosin were found to be significant risk factors for ureteral wall injury (P = 0.007, 0.041 and < 0.001, respectively). CONCLUSIONS The administration of silodosin prior to retrograde intrarenal surgery (RIRS) effectively prevented significant ureteral wall damage and reduced initial postoperative discomfort.
Collapse
Affiliation(s)
- Tamer Diab
- Urology Department, Faculty of Medicine, Benha University, Benha, Qalyubiyya Governorate, Egypt.
| | - Waleed El-Shaer
- Urology Department, Faculty of Medicine, Benha University, Benha, Qalyubiyya Governorate, Egypt
| | - Saad Ibrahim
- Urology Department, Faculty of Medicine, Benha University, Benha, Qalyubiyya Governorate, Egypt
| | - Ehab El-Barky
- Urology Department, Faculty of Medicine, Benha University, Benha, Qalyubiyya Governorate, Egypt
| | - Ahmed Abou Elezz
- Urology Department, Faculty of Medicine, Benha University, Benha, Qalyubiyya Governorate, Egypt
| |
Collapse
|
6
|
Sandroni PB, Schroder MA, Hawkins HT, Bailon JD, Huang W, Hagen JT, Montgomery M, Hong SJ, Chin AL, Zhang J, Rodrigo MC, Kim B, Simpson PC, Schisler JC, Ellis JM, Fisher-Wellman KH, Jensen BC. The alpha-1A adrenergic receptor regulates mitochondrial oxidative metabolism in the mouse heart. J Mol Cell Cardiol 2024; 187:101-117. [PMID: 38331556 PMCID: PMC10861168 DOI: 10.1016/j.yjmcc.2023.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 12/11/2023] [Accepted: 12/12/2023] [Indexed: 02/10/2024]
Abstract
AIMS The sympathetic nervous system regulates numerous critical aspects of mitochondrial function in the heart through activation of adrenergic receptors (ARs) on cardiomyocytes. Mounting evidence suggests that α1-ARs, particularly the α1A subtype, are cardioprotective and may mitigate the deleterious effects of chronic β-AR activation by shared ligands. The mechanisms underlying these adaptive effects remain unclear. Here, we tested the hypothesis that α1A-ARs adaptively regulate cardiomyocyte oxidative metabolism in both the uninjured and infarcted heart. METHODS We used high resolution respirometry, fatty acid oxidation (FAO) enzyme assays, substrate-specific electron transport chain (ETC) enzyme assays, transmission electron microscopy (TEM) and proteomics to characterize mitochondrial function comprehensively in the uninjured hearts of wild type and α1A-AR knockout mice and defined the effects of chronic β-AR activation and myocardial infarction on selected mitochondrial functions. RESULTS We found that isolated cardiac mitochondria from α1A-KO mice had deficits in fatty acid-dependent respiration, FAO, and ETC enzyme activity. TEM revealed abnormalities of mitochondrial morphology characteristic of these functional deficits. The selective α1A-AR agonist A61603 enhanced fatty-acid dependent respiration, fatty acid oxidation, and ETC enzyme activity in isolated cardiac mitochondria. The β-AR agonist isoproterenol enhanced oxidative stress in vitro and this adverse effect was mitigated by A61603. A61603 enhanced ETC Complex I activity and protected contractile function following myocardial infarction. CONCLUSIONS Collectively, these novel findings position α1A-ARs as critical regulators of cardiomyocyte metabolism in the basal state and suggest that metabolic mechanisms may underlie the protective effects of α1A-AR activation in the failing heart.
Collapse
Affiliation(s)
- Peyton B Sandroni
- Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America; McAllister Heart Institute, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America
| | - Melissa A Schroder
- McAllister Heart Institute, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America
| | - Hunter T Hawkins
- McAllister Heart Institute, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America
| | - Julian D Bailon
- McAllister Heart Institute, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America
| | - Wei Huang
- McAllister Heart Institute, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America
| | - James T Hagen
- Department of Physiology, East Carolina University, Brody School of Medicine, Greenville, NC, United States of America; East Carolina University Diabetes and Obesity Institute, East Carolina University, Brody School of Medicine, Greenville, NC, United States of America
| | - McLane Montgomery
- Department of Physiology, East Carolina University, Brody School of Medicine, Greenville, NC, United States of America; East Carolina University Diabetes and Obesity Institute, East Carolina University, Brody School of Medicine, Greenville, NC, United States of America
| | - Seok J Hong
- McAllister Heart Institute, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America
| | - Andrew L Chin
- McAllister Heart Institute, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America
| | - Jiandong Zhang
- McAllister Heart Institute, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America; Department of Medicine, Division of Cardiology, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America
| | - Manoj C Rodrigo
- Cytokinetics, Inc., South San Francisco, CA, United States of America
| | - Boa Kim
- McAllister Heart Institute, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America; Department of Cell Biology and Physiology, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America
| | - Paul C Simpson
- Department of Medicine and Research Service, San Francisco Veterans Affairs Medical Center, San Francisco, CA, United States of America; Cardiovascular Research Institute, University of California, San Francisco, CA, United States of America
| | - Jonathan C Schisler
- Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America; McAllister Heart Institute, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America
| | - Jessica M Ellis
- Department of Physiology, East Carolina University, Brody School of Medicine, Greenville, NC, United States of America; East Carolina University Diabetes and Obesity Institute, East Carolina University, Brody School of Medicine, Greenville, NC, United States of America
| | - Kelsey H Fisher-Wellman
- Department of Physiology, East Carolina University, Brody School of Medicine, Greenville, NC, United States of America; East Carolina University Diabetes and Obesity Institute, East Carolina University, Brody School of Medicine, Greenville, NC, United States of America
| | - Brian C Jensen
- Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America; McAllister Heart Institute, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America; Department of Medicine, Division of Cardiology, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America.
| |
Collapse
|
7
|
Fairuz S, Ang CW, Mraiche F, Goh JK. Current Targets and Future Directions of Positive Inotropes for Heart Failure. Curr Med Chem 2024; 31:6971-6991. [PMID: 37909442 DOI: 10.2174/0109298673262360231018193823] [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: 05/23/2023] [Revised: 06/23/2023] [Accepted: 09/11/2023] [Indexed: 11/03/2023]
Abstract
While a congestive heart failure patient will ultimately need an assist device or even a replacement heart as the disease progresses, not every patient is qualified for such advanced therapy. Such patients awaiting better circulatory support benefit from positive inotropes in the meantime as palliative care. These agents are often prescribed in patients with acute decompensated heart failure, with reduced left ventricular ejection fraction and symptoms of organ dysfunction. Although positive inotropes, for example, digoxin, dobutamine, milrinone, levosimendan, etc., are successfully marketed and in use, a lot of their adverse effects, like arrhythmias, hypotension, and even sudden cardiac death, are rather encouraging further research on the development of novel positive inotropes. This review has investigated the molecular mechanisms of some of these adverse effects in terms of the proteins they target, followed by research on newer targets. Studies from 2013-2023 that have reported new small molecules with positive inotropic effects have been revisited in order to determine the progress made so far in drug discovery.
Collapse
Affiliation(s)
- Shadreen Fairuz
- School of Science, Monash University, Jalan Lagoon Selatan, Bandar Sunway, 47500 Subang Jaya, Selangor, Malaysia
| | - Chee Wei Ang
- School of Science, Monash University, Jalan Lagoon Selatan, Bandar Sunway, 47500 Subang Jaya, Selangor, Malaysia
| | - Fatima Mraiche
- Department of Pharmacology, University of Alberta, 116 St & 85 Ave, Edmonton, ABT6G 2R3, Canada
| | - Joo Kheng Goh
- School of Science, Monash University, Jalan Lagoon Selatan, Bandar Sunway, 47500 Subang Jaya, Selangor, Malaysia
| |
Collapse
|
8
|
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
|
9
|
Ruan Y, Buonfiglio F, Gericke A. Adrenoceptors in the Eye - Physiological and Pathophysiological Relevance. Handb Exp Pharmacol 2024; 285:453-505. [PMID: 38082203 DOI: 10.1007/164_2023_702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
The autonomic nervous system plays a crucial role in the innervation of the eye. Consequently, it comes as no surprise that catecholamines and their corresponding receptors have been extensively studied and characterized in numerous ocular structures, including the cornea, conjunctiva, lacrimal gland, trabecular meshwork, uvea, and retina. These investigations have unveiled substantial clinical implications, particularly in the context of treating glaucoma, a progressive neurodegenerative disorder responsible for irreversible vision loss on a global scale. The primary therapeutic approaches for glaucoma frequently involve the modulation of α1-, α2-, and β-adrenoceptors, making them pivotal targets. In this chapter, we offer a comprehensive overview of the expression, distribution, and functional roles of adrenoceptors within various components of the eye and its associated structures. Additionally, we delve into the pivotal role of adrenoceptors in the pathophysiology of glaucoma. Furthermore, we provide a concise historical perspective on adrenoceptor research, examine the distinct contributions of individual adrenoceptor subtypes to the treatment of various ocular conditions, and propose potential future avenues of exploration in this field.
Collapse
Affiliation(s)
- Yue Ruan
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Francesco Buonfiglio
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Adrian Gericke
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany.
| |
Collapse
|
10
|
Li Z, Wang H, Bao X, Liu X, Yang J. Gene network analyses of Sepia esculenta larvae exposed to copper and cadmium: A comprehensive investigation of oxidative stress, immune response, and toxicological mechanisms. FISH & SHELLFISH IMMUNOLOGY 2023; 143:109230. [PMID: 37977542 DOI: 10.1016/j.fsi.2023.109230] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 10/12/2023] [Accepted: 11/11/2023] [Indexed: 11/19/2023]
Abstract
Copper (Cu) and Cadmium (Cd), prevalent heavy metals in marine environments, have known implications in oxidative stress, immune response, and toxicity in marine organisms. Sepia esculenta, a cephalopod of significant economic value along China's eastern coastline, experiences alterations in growth, mobility, and reproduction when subjected to these heavy metals. However, the specific mechanisms resulting from heavy metal exposure in S. esculenta remain largely uncharted. In this study, we utilized transcriptome and four oxidative, immunity, and toxicity indicators to assess the toxicological mechanism in S. esculenta larvae exposed to Cu and Cd. The measurements of Superoxide Dismutase (SOD), Malondialdehyde (MDA), Glutathione S-Transferase (GST), and Metallothioneins (MTs) revealed that Cu and Cd trigger substantial oxidative stress, immune response, and metal toxicity. Further, we performed an analysis on the transcriptome data through Weighted Gene Co-expression Network Analysis (WGCNA) and Protein-Protein Interaction (PPI) network analysis. Our findings indicate that exposure methods and duration influence the type and the extent of toxicity and oxidative stress within the S. esculenta larvae. We took an innovative approach in this research by integrating WGCNA and PPI network analysis with four significant physiological indicators to closely examine the toxicity and oxidative stress profiles of S. esculenta upon exposure to Cu and Cd. This investigation is vital in decoding the toxicological, immunological, and oxidative stress mechanisms within S. esculenta when subjected to heavy metals. It provides foundational insights capable of advancing invertebrate environmental toxicology and informs S. esculenta artificial breeding practices.
Collapse
Affiliation(s)
- Zan Li
- School of Agriculture, Ludong University, Yantai, 264025, China
| | - Haoyu Wang
- St. John's School, Vancouver, V6K 2J1, Canada
| | - Xiaokai Bao
- School of Agriculture, Ludong University, Yantai, 264025, China
| | - Xiumei Liu
- College of Life Sciences, Yantai University, Yantai, 264005, China
| | - Jianmin Yang
- School of Agriculture, Ludong University, Yantai, 264025, China.
| |
Collapse
|
11
|
Zhang J, Latour CD, Olawore O, Pate V, Friedlander DF, Stürmer T, Jonsson Funk M, Jensen BC. Cardiovascular Outcomes of α-Blockers vs 5-α Reductase Inhibitors for Benign Prostatic Hyperplasia. JAMA Netw Open 2023; 6:e2343299. [PMID: 37962887 PMCID: PMC10646730 DOI: 10.1001/jamanetworkopen.2023.43299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 10/04/2023] [Indexed: 11/15/2023] Open
Abstract
Importance The most prescribed class of medications for benign prostatic hyperplasia (BPH) is α-blockers (ABs). However, the cardiovascular safety profile of these medications among patients with BPH is not well understood. Objective To compare the safety of ABs vs 5-α reductase inhibitors (5-ARIs) for risk of adverse cardiovascular outcomes. Design, Setting, and Participants This active comparator, new-user cohort study was conducted using insurance claims data from a 20% random sample of Medicare beneficiaries from 2007 to 2019 to evaluate the 1-year risk of adverse cardiovascular outcomes. Males aged 66 to 90 years were indexed into the cohort at new use of an AB or 5-ARI. Twelve months of continuous enrollment and at least 1 diagnosis code for BPH within 12 months prior to initiation were required. Data were analyzed from January 2007 through December 2019. Exposures Exposure was defined by a qualifying prescription fill for an AB or 5-ARI after at least 12 months without a prescription for these drug classes. Main Outcomes and Measures Follow-up began at a qualified refill for the study drug. Primary study outcomes were hospitalization for heart failure (HF), composite major adverse cardiovascular events (MACE; hospitalization for stroke, myocardial infarction, or death), composite MACE or hospitalization for HF, and death. Inverse probability of treatment and censoring-weighted 1-year risks, risk ratios (RRs), and risk differences (RDs) were estimated for each outcome. Results Among 189 868 older adult males, there were 163 829 patients initiating ABs (mean [SD] age, 74.6 [6.2] years; 579 American Indian or Alaska Native [0.4%], 5890 Asian or Pacific Islander [3.6%], 9179 Black [5.6%], 10 610 Hispanic [6.5%], and 133 510 non-Hispanic White [81.5%]) and 26 039 patients initiating 5-ARIs (mean [SD] age, 75.3 [6.4] years; 76 American Indian or Alaska Native [0.3%], 827 Asian or Pacific Islander [3.2%], 1339 Black [5.1%], 1656 Hispanic [6.4%], and 21 605 non-Hispanic White [83.0%]). ABs compared with 5-ARIs were associated with an increased 1-year risk of MACE (8.95% [95% CI, 8.81%-9.09%] vs 8.32% [95% CI, 7.92%-8.72%]; RR = 1.08 [95% CI, 1.02-1.13]; RD per 1000 individuals = 6.26 [95% CI, 2.15-10.37]), composite MACE and HF (RR = 1.07; [95% CI, 1.03-1.12]; RD per 1000 individuals = 7.40 [95% CI, 2.88-11.93 ]), and death (RR = 1.07; [95% CI, 1.01-1.14]; RD per 1000 individuals = 3.85 [95% CI, 0.40-7.29]). There was no difference in risk for HF hospitalization alone. Conclusions and Relevance These results suggest that ABs may be associated with an increased risk of adverse cardiovascular outcomes compared with 5-ARIs. If replicated with more detailed confounder data, these results may have important public health implications given these medications' widespread use.
Collapse
Affiliation(s)
- Jiandong Zhang
- Division of Cardiology, School of Medicine, University of North Carolina at Chapel Hill
| | - Chase D. Latour
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill
- Cecil G. Sheps Center for Health Services Research, University of North Carolina at Chapel Hill
| | - Oluwasolape Olawore
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill
| | - Virginia Pate
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill
| | | | - Til Stürmer
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill
| | - Michele Jonsson Funk
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill
| | - Brian C. Jensen
- Division of Cardiology, School of Medicine, University of North Carolina at Chapel Hill
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill
| |
Collapse
|
12
|
Nakamura M, Keller MA, Fefelova N, Zhai P, Liu T, Tian Y, Ikeda S, Del Re DP, Li H, Xie LH, Sadoshima J. Ser14 phosphorylation of Bcl-xL mediates compensatory cardiac hypertrophy in male mice. Nat Commun 2023; 14:5805. [PMID: 37726310 PMCID: PMC10509265 DOI: 10.1038/s41467-023-41595-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 09/08/2023] [Indexed: 09/21/2023] Open
Abstract
The anti-apoptotic function of Bcl-xL in the heart during ischemia/reperfusion is diminished by K-Ras-Mst1-mediated phosphorylation of Ser14, which allows dissociation of Bcl-xL from Bax and promotes cardiomyocyte death. Here we show that Ser14 phosphorylation of Bcl-xL is also promoted by hemodynamic stress in the heart, through the H-Ras-ERK pathway. Bcl-xL Ser14 phosphorylation-resistant knock-in male mice develop less cardiac hypertrophy and exhibit contractile dysfunction and increased mortality during acute pressure overload. Bcl-xL Ser14 phosphorylation enhances the Ca2+ transient by blocking the inhibitory interaction between Bcl-xL and IP3Rs, thereby promoting Ca2+ release and activation of the calcineurin-NFAT pathway, a Ca2+-dependent mechanism that promotes cardiac hypertrophy. These results suggest that phosphorylation of Bcl-xL at Ser14 in response to acute pressure overload plays an essential role in mediating compensatory hypertrophy by inducing the release of Bcl-xL from IP3Rs, alleviating the negative constraint of Bcl-xL upon the IP3R-NFAT pathway.
Collapse
Affiliation(s)
- Michinari Nakamura
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers-New Jersey Medical School, 185 South Orange Ave, Newark, NJ, 07103, USA.
| | - Mariko Aoyagi Keller
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers-New Jersey Medical School, 185 South Orange Ave, Newark, NJ, 07103, USA
| | - Nadezhda Fefelova
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers-New Jersey Medical School, 185 South Orange Ave, Newark, NJ, 07103, USA
| | - Peiyong Zhai
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers-New Jersey Medical School, 185 South Orange Ave, Newark, NJ, 07103, USA
| | - Tong Liu
- Center for Advanced Proteomics Research, Department of Biochemistry & Molecular Biology, Rutgers New Jersey Medical School, Newark, NJ, 07103, USA
| | - Yimin Tian
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers-New Jersey Medical School, 185 South Orange Ave, Newark, NJ, 07103, USA
| | - Shohei Ikeda
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers-New Jersey Medical School, 185 South Orange Ave, Newark, NJ, 07103, USA
| | - Dominic P Del Re
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers-New Jersey Medical School, 185 South Orange Ave, Newark, NJ, 07103, USA
| | - Hong Li
- Center for Advanced Proteomics Research, Department of Biochemistry & Molecular Biology, Rutgers New Jersey Medical School, Newark, NJ, 07103, USA
| | - Lai-Hua Xie
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers-New Jersey Medical School, 185 South Orange Ave, Newark, NJ, 07103, USA
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers-New Jersey Medical School, 185 South Orange Ave, Newark, NJ, 07103, USA.
| |
Collapse
|
13
|
Mokrov GV. Multitargeting in cardioprotection: An example of biaromatic compounds. Arch Pharm (Weinheim) 2023; 356:e2300196. [PMID: 37345968 DOI: 10.1002/ardp.202300196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 05/23/2023] [Accepted: 05/26/2023] [Indexed: 06/23/2023]
Abstract
A multitarget drug design approach is actively developing in modern medicinal chemistry and pharmacology, especially with regard to multifactorial diseases such as cardiovascular diseases, cancer, and neurodegenerative diseases. A detailed study of many well-known drugs developed within the single-target approach also often reveals additional mechanisms of their real pharmacological action. One of the multitarget drug design approaches can be the identification of the basic pharmacophore models corresponding to a wide range of the required target ligands. Among such models in the group of cardioprotectors is the linked biaromatic system. This review develops the concept of a "basic pharmacophore" using the biaromatic pharmacophore of cardioprotectors as an example. It presents an analysis of possible biological targets for compounds corresponding to the biaromatic pharmacophore and an analysis of the spectrum of biological targets for the five most known and most studied cardioprotective drugs corresponding to this model, and their involvement in the biological effects of these drugs.
Collapse
|
14
|
Su M, Wang J, Xiang G, Do HN, Levitz J, Miao Y, Huang XY. Structural basis of agonist specificity of α 1A-adrenergic receptor. Nat Commun 2023; 14:4819. [PMID: 37563160 PMCID: PMC10415349 DOI: 10.1038/s41467-023-40524-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 07/27/2023] [Indexed: 08/12/2023] Open
Abstract
α1-adrenergic receptors (α1-ARs) play critical roles in the cardiovascular and nervous systems where they regulate blood pressure, cognition, and metabolism. However, the lack of specific agonists for all α1 subtypes has limited our understanding of the physiological roles of different α1-AR subtypes, and led to the stagnancy in agonist-based drug development for these receptors. Here we report cryo-EM structures of α1A-AR in complex with heterotrimeric G-proteins and either the endogenous common agonist epinephrine or the α1A-AR-specific synthetic agonist A61603. These structures provide molecular insights into the mechanisms underlying the discrimination between α1A-AR and α1B-AR by A61603. Guided by the structures and corresponding molecular dynamics simulations, we engineer α1A-AR mutants that are not responsive to A61603, and α1B-AR mutants that can be potently activated by A61603. Together, these findings advance our understanding of the agonist specificity for α1-ARs at the molecular level, opening the possibility of rational design of subtype-specific agonists.
Collapse
Affiliation(s)
- Minfei Su
- Department of Physiology and Biophysics, Weill Cornell Medical College of Cornell University, New York, NY, 10065, USA
| | - Jinan Wang
- Center for Computational Biology and Department of Molecular Biosciences, University of Kansas, Lawrence, KS, 66047, USA
| | - Guoqing Xiang
- Department of Biochemistry, Weill Cornell Medical College of Cornell University, New York, NY, 10065, USA
- Department of Psychiatry, Weill Cornell Medical College of Cornell University, New York, NY, 10065, USA
| | - Hung Nguyen Do
- Center for Computational Biology and Department of Molecular Biosciences, University of Kansas, Lawrence, KS, 66047, USA
| | - Joshua Levitz
- Department of Biochemistry, Weill Cornell Medical College of Cornell University, New York, NY, 10065, USA
- Department of Psychiatry, Weill Cornell Medical College of Cornell University, New York, NY, 10065, USA
| | - Yinglong Miao
- Center for Computational Biology and Department of Molecular Biosciences, University of Kansas, Lawrence, KS, 66047, USA.
| | - Xin-Yun Huang
- Department of Physiology and Biophysics, Weill Cornell Medical College of Cornell University, New York, NY, 10065, USA.
| |
Collapse
|
15
|
Rupert C, López JE, Cortez-Toledo E, De la Cruz Cabrera O, Chesler NC, Simpson PC, Campbell SG, Baker AJ. Increased length-dependent activation of human engineered heart tissue after chronic α 1A-adrenergic agonist treatment: testing a novel heart failure therapy. Am J Physiol Heart Circ Physiol 2023; 324:H293-H304. [PMID: 36637971 PMCID: PMC9886349 DOI: 10.1152/ajpheart.00279.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 12/06/2022] [Accepted: 12/23/2022] [Indexed: 01/14/2023]
Abstract
Chronic stimulation of cardiac α1A-adrenergic receptors (α1A-ARs) improves symptoms in multiple preclinical models of heart failure. However, the translational significance remains unclear. Human engineered heart tissues (EHTs) provide a means of quantifying the effects of chronic α1A-AR stimulation on human cardiomyocyte physiology. EHTs were created from thin slices of decellularized pig myocardium seeded with human induced pluripotent stem cell (iPSC)-derived cardiomyocytes and fibroblasts. With a paired experimental design, EHTs were cultured for 3 wk, mechanically tested, cultured again for 2 wk with α1A-AR agonist A61603 (10 nM) or vehicle control, and retested after drug washout for 24 h. Separate control experiments determined the effects of EHT age (3-5 wk) or repeat mechanical testing. We found that chronic A61603 treatment caused a 25% increase of length-dependent activation (LDA) of contraction compared with vehicle treatment (n = 7/group, P = 0.035). EHT force was not increased after chronic A61603 treatment. However, after vehicle treatment, EHT force was increased by 35% relative to baseline testing (n = 7/group, P = 0.022), suggesting EHT maturation. Control experiments suggested that increased EHT force resulted from repeat mechanical testing, not from EHT aging. RNA-seq analysis confirmed that the α1A-AR is expressed in human EHTs and found chronic A61603 treatment affected gene expression in biological pathways known to be activated by α1A-ARs, including the MAP kinase signaling pathway. In conclusion, increased LDA in human EHT after chronic A61603 treatment raises the possibility that chronic stimulation of the α1A-AR might be beneficial for increasing LDA in human myocardium and might be beneficial for treating human heart failure by restoring LDA.NEW & NOTEWORTHY Chronic stimulation of α1A-adrenergic receptors (α1A-ARs) is known to mediate therapeutic effects in animal heart failure models. To investigate the effects of chronic α1A-AR stimulation in human cardiomyocytes, we tested engineered heart tissue (EHT) created with iPSC-derived cardiomyocytes. RNA-seq analysis confirmed human EHT expressed α1A-ARs. Chronic (2 wk) α1A-AR stimulation with A61603 (10 nM) increased length-dependent activation (LDA) of contraction. Chronic α1A-AR stimulation might be beneficial for treating human heart failure by restoring LDA.
Collapse
Affiliation(s)
- C. Rupert
- Propria LLC, Branford, Connecticut, United States
| | - J. E. López
- Division of Cardiovascular Medicine, Department of Internal Medicine,
University of California Davis, Davis, California, United States
| | - E. Cortez-Toledo
- Division of Cardiovascular Medicine, Department of Internal Medicine,
University of California Davis, Davis, California, United States
| | | | - N. C. Chesler
- Edwards Lifesciences Foundation Cardiovascular Innovation Research Center, Irvine, California, United States
- Department of Biomedical Engineering, University of California, Irvine, California, United States
| | - P. C. Simpson
- Cardiology Division, Veterans Affairs Medical Center, San Francisco, California, United States
- Department of Medicine, University of California, San Francisco, California, United States
| | - S. G. Campbell
- Departments of Biomedical Engineering and Cellular and Molecular Physiology, Yale University, New Haven, Connecticut, United States
| | - A. J. Baker
- Cardiology Division, Veterans Affairs Medical Center, San Francisco, California, United States
- Department of Medicine, University of California, San Francisco, California, United States
| |
Collapse
|
16
|
Mechanisms for the α-Adrenoceptor-Mediated Positive Inotropy in Mouse Ventricular Myocardium: Enhancing Effect of Action Potential Prolongation. Int J Mol Sci 2023; 24:ijms24043926. [PMID: 36835338 PMCID: PMC9964142 DOI: 10.3390/ijms24043926] [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: 12/27/2022] [Revised: 02/09/2023] [Accepted: 02/13/2023] [Indexed: 02/17/2023] Open
Abstract
Mechanisms for the α-adrenoceptor-mediated positive inotropy in neonatal mouse ventricular myocardium were studied with isolated myocardial preparations. The phenylephrine-induced positive inotropy was suppressed by prazosin, nifedipine, and chelerythrine, a protein kinase C inhibitor, but not by SEA0400, a selective Na+/Ca2+ exchanger inhibitor. Phenylephrine increased the L-type Ca2+ channel current and prolonged the action potential duration, while the voltage-dependent K+ channel current was not influenced. In the presence of cromakalim, an ATP-sensitive K+ channel opener, the phenylephrine-induced prolongation of action potential duration, as well as the positive inotropy, were smaller than in the absence of cromakalim. These results suggest that the α-adrenoceptor-mediated positive inotropy is mediated by an increase in Ca2+ influx through the L-type Ca2+ channel, and the concomitant increase in action potential duration acts as an enhancing factor.
Collapse
|
17
|
Altara R, Booz G. Central role for BRAF in cardiac hypertrophy: rethinking the pathological-physiological divide. Clin Sci (Lond) 2023; 137:143-148. [PMID: 36651286 PMCID: PMC9873497 DOI: 10.1042/cs20220776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/23/2022] [Accepted: 01/05/2023] [Indexed: 01/19/2023]
Abstract
The RAF/MEK/ERK1/2 signaling cascade has been implicated in pathological cardiac hypertrophy downstream of some Gq-coupled receptors. The RAF family of kinases consists of three isoforms (ARAF, BRAF, and CRAF) and until recently most studies on this signaling pathway in the heart have focused on RAF1 (CRAF). In a recent issue of Clinical Science, Alharbi et al. utilized an inducible cardiac myocyte targeted knockout mouse model to define the role of BRAF in pathological versus physiological hypertrophy using angiotensin II and phenylephrine (PE) infusion, respectively. They reported that loss of BRAF attenuated both pathological cardiac hypertrophy and interstitial fibrosis. BRAF knockout decreased cardiac function with PE in male mice and enhanced both interstitial and perivascular cardiac fibrosis but had no effect on hypertrophy. In contrast, loss of BRAF attenuated physiological hypertrophy in female mice but had no effect on fibrosis or contractility. These observations extend those previously made by this group assessing the consequences of expressing an inducible activating mutant of BRAF in the heart and the benefit of enhancing RAF/MEK/ERK1/2 signaling by exploiting the 'RAF paradox'. Additional studies are needed to better define the role of BRAF under conditions reflective of chronic stress on the heart due to the biomechanical stimulation exerted by hypertension. In addition, the role of BRAF and its activation in overt heart failure remains to be established. Nevertheless, the new findings highlight the potential importance of additional signaling events, perhaps related to RAF1 or ERK1/2 activation, in shaping BRAF signaling in a sex- and context-dependent manner.
Collapse
Affiliation(s)
- Raffaele Altara
- Department of Pathology, School of Medicine, University of Mississippi Medical Center, Jackson, MS, U.S.A
- Department of Anatomy and Embryology, Maastricht University, Maastricht, The Netherlands
| | - George W. Booz
- Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center, Jackson, MS, U.S.A
- Correspondence: George W. Booz ()
| |
Collapse
|
18
|
Żmudzka E, Lustyk K, Siwek A, Wolak M, Gałuszka A, Jaśkowska J, Kołaczkowski M, Sapa J, Pytka K. Novel Arylpiperazine Derivatives of Salicylamide with α 1-Adrenolytic Properties Showed Antiarrhythmic and Hypotensive Properties in Rats. Int J Mol Sci 2022; 24:ijms24010293. [PMID: 36613736 PMCID: PMC9820316 DOI: 10.3390/ijms24010293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/09/2022] [Accepted: 12/21/2022] [Indexed: 12/28/2022] Open
Abstract
Cardiovascular diseases remain one of the leading causes of death worldwide. Unfortunately, the available pharmacotherapeutic options have limited effectiveness. Therefore, developing new drug candidates remains very important. We selected six novel arylpiperazine alkyl derivatives of salicylamide to investigate their cardiovascular effects. Having in mind the beneficial role of α1-adrenergic receptors in restoring sinus rhythm and regulating blood pressure, first, using radioligand binding assays, we evaluated the affinity of the tested compounds for α-adrenergic receptors. Our experiments revealed their high to moderate affinity for α1- but not α2-adrenoceptors. Next, we aimed to determine the antiarrhythmic potential of novel derivatives in rat models of arrhythmia induced by adrenaline, calcium chloride, or aconitine. All compounds showed potent prophylactic antiarrhythmic activity in the adrenaline-induced arrhythmia model and no effects in calcium chloride- or aconitine-induced arrhythmias. Moreover, the tested compounds demonstrated therapeutic antiarrhythmic activity, restoring a normal sinus rhythm immediately after the administration of the arrhythmogen adrenaline. Notably, none of the tested derivatives affected the normal electrocardiogram (ECG) parameters in rodents, which excludes their proarrhythmic potential. Finally, all tested compounds decreased blood pressure in normotensive rats and reversed the pressor response to methoxamine, suggesting that their hypotensive mechanism of action is connected with the blockade of α1-adrenoceptors. Our results confirm the antiarrhythmic and hypotensive activities of novel arylpiperazine derivatives and encourage their further investigation as model structures for potential drugs.
Collapse
Affiliation(s)
- Elżbieta Żmudzka
- Department of Social Pharmacy, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Krakow, Poland
- Correspondence:
| | - Klaudia Lustyk
- Department of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Krakow, Poland
| | - Agata Siwek
- Department of Pharmacobiology, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Krakow, Poland
| | - Małgorzata Wolak
- Department of Pharmacobiology, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Krakow, Poland
| | - Adam Gałuszka
- Department of Automatic Control and Robotics, Silesian University of Technology, Akademicka 2A, 44-100 Gliwice, Poland
| | - Jolanta Jaśkowska
- Department of Organic Chemistry and Technology, Faculty of Chemical and Engineering and Technology, Cracow University of Technology, Warszawska 24, 31-155 Krakow, Poland
| | - Marcin Kołaczkowski
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Krakow, Poland
| | - Jacek Sapa
- Department of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Krakow, Poland
| | - Karolina Pytka
- Department of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Krakow, Poland
| |
Collapse
|
19
|
Associations between β-Blocker Therapy at Discharge and Long-Term Follow-Up Outcomes in Patients with Unstable Angina Pectoris. Cardiol Res Pract 2022; 2022:5287566. [PMID: 36213457 PMCID: PMC9537036 DOI: 10.1155/2022/5287566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 07/12/2022] [Accepted: 07/14/2022] [Indexed: 11/18/2022] Open
Abstract
Background The effects of β-blockers in patients with unstable angina pectoris (UAP) are unclear. We tried to evaluate associations between β-blockers in UAP and long-term outcomes. Methods We enrolled 5591 UAP patients and divided them into 2 groups based on β-blockers at discharge: 3790 did β-blockers and 1801 did not used them. Propensity score matching at 1 : 1 was performed to select 1786 patients from each group. The primary endpoint was major adverse cardiac and cerebral events (MACCE) during the long-term follow-up period. Results 67.8% of patients were on β-blockers at discharge; these patients were more likely to have CHD risk factors, lower ejection fraction, and severity of the coronary artery lesions. Over a median of 25.0 years, the incidence of MACCE was 25.5%. The risk was not significantly different between those on and those not on β-blocker treatment. The multivariate Cox regression analysis showed that no β-blocker use at discharge was not an independent risk factor for MACCE and sequence secondary endpoints. After propensity score matching, the results were similar. Conclusions β-blocker use was not associated with lower MACCE and other secondary composite endpoints in long-term outcomes. This result adds to the increasing body of evidence that the routine prescription of β-blockers might not be indicated in patients with UAP. Trial registration had retrospectively registered.
Collapse
|
20
|
Mazurara GR, Dallagnol JCC, Chatenet D, Allen BG, Hébert TE. The complicated lives of GPCRs in cardiac fibroblasts. Am J Physiol Cell Physiol 2022; 323:C813-C822. [PMID: 35938678 DOI: 10.1152/ajpcell.00120.2022] [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: 11/22/2022]
Abstract
The role of different G protein-coupled receptors (GPCRs) in the cardiovascular system is well understood in cardiomyocytes and vascular smooth muscle cells (VSMCs). In the former, stimulation of Gs-coupled receptors leads to increases in contractility, while stimulation of Gq-coupled receptors modulates cellular survival and hypertrophic responses. In VSMCs, stimulation of GPCRs also modulates contractile and cell growth phenotypes. Here, we will focus on the relatively less well studied effects of GPCRs in cardiac fibroblasts, focusing on key signalling events involved in the activation and differentiation of these cells. We also review the hierarchy of signalling events driving the fibrotic response and the communications between fibroblasts and other cells in the heart. We discuss how such events may be distinct depending on where the GPCRs and their associated signalling machinery are localized in these cells with an emphasis on nuclear membrane-localized receptors. Finally, we explore what such connections between cell surface and nuclear GPCR signalling might mean for cardiac fibrosis.
Collapse
Affiliation(s)
- Grace R Mazurara
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada
| | - Juliana C C Dallagnol
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada.,Institut National de la Recherche Scientifique, Centre Armand-Frappier Santé Biotechnologie, Groupe de Recherche en Ingénierie des Peptides et en Pharmacothérapie (GRIPP), Université du Québec, Laval, Québec, Canada.,Research Center, Montreal Heart Institute, Montreal, Quebec, Canada
| | - David Chatenet
- Institut National de la Recherche Scientifique, Centre Armand-Frappier Santé Biotechnologie, Groupe de Recherche en Ingénierie des Peptides et en Pharmacothérapie (GRIPP), Université du Québec, Laval, Québec, Canada
| | - Bruce G Allen
- Research Center, Montreal Heart Institute, Montreal, Quebec, Canada
| | - Terence E Hébert
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada
| |
Collapse
|
21
|
Pluteanu F, Kirchhefer U. Reply to Cristóbal et al. Am J Physiol Heart Circ Physiol 2022; 323:H67-H68. [PMID: 35648098 DOI: 10.1152/ajpheart.00236.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 05/16/2022] [Indexed: 11/22/2022]
Affiliation(s)
- Florentina Pluteanu
- Department of Anatomy, Animal Physiology and Biophysics, University of Bucharest, Bucharest, Romania
| | - Uwe Kirchhefer
- Institute of Pharmacology and Toxicology, University of Muenster, Muenster, Germany
| |
Collapse
|
22
|
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: 0.7] [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
|
23
|
Li J, Yang Z, Zhang C, Hu Y, Li H, Zhang M, Bu P, Wang S, Zhang C, Li W. Chinese Classical Music Lowers Blood Pressure and Improves Left Ventricular Hypertrophy in Spontaneously Hypertensive Rats. Front Pharmacol 2022; 13:826669. [PMID: 35586054 PMCID: PMC9108543 DOI: 10.3389/fphar.2022.826669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 04/19/2022] [Indexed: 11/13/2022] Open
Abstract
High blood pressure (BP) plays an important role in the pathogenesis and development of cardiovascular diseases and multi-organ damages. Music has been well known to elicit emotional changes, such as anxiolytic effects. However, whether music therapy lowers BP in spontaneously hypertensive rats (SHR) and the potential mechanism remains unknown. SHRs were, respectively exposed to white noise (WN), Western classical music (WM), Chinese classical music (CCM), rock music (RM), and bisoprolol treatment. WN and WM did not lower systemic BP, but CCM and RM significantly lowered BPs in SHRs. The effects of CCM therapy on lowering systemic BPs is comparable to that of bisoprolol at the dose of low to medium. Combination of CCM treatment with bisoprolol further improved systemic BPs and myocardial hypertrophy in SHRs, compared to CCM treatment or bisoprolol alone. Furthermore, IHC and WB analysis indicated that CCM therapy inhibited the β1/cAMP/PKA and α1/PLC/PKC signalings, but didn’t alter the β2/PI3K/Akt signaling. Above all, CCM therapy lowers systemic BPs and alleviates myocardial hypertrophy in hypertensive rats, which may be caused by the inhibitions of β1/cAMP/PKA and α1/PLC/PKC signalings.
Collapse
Affiliation(s)
- Jingyuan Li
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Zhi Yang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Chunmei Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yang Hu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Hongxuan Li
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Meng Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Peili Bu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Shuangxi Wang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Cheng Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wenjing Li
- Fine Arts School of Shandong University, Jinan, China
- *Correspondence: Wenjing Li,
| |
Collapse
|
24
|
Modification of Ischemia/Reperfusion-Induced Alterations in Subcellular Organelles by Ischemic Preconditioning. Int J Mol Sci 2022; 23:ijms23073425. [PMID: 35408783 PMCID: PMC8998910 DOI: 10.3390/ijms23073425] [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: 02/16/2022] [Revised: 03/17/2022] [Accepted: 03/18/2022] [Indexed: 02/07/2023] Open
Abstract
It is now well established that ischemia/reperfusion (I/R) injury is associated with the compromised recovery of cardiac contractile function. Such an adverse effect of I/R injury in the heart is attributed to the development of oxidative stress and intracellular Ca2+-overload, which are known to induce remodeling of subcellular organelles such as sarcolemma, sarcoplasmic reticulum, mitochondria and myofibrils. However, repeated episodes of brief periods of ischemia followed by reperfusion or ischemic preconditioning (IP) have been shown to improve cardiac function and exert cardioprotective actions against the adverse effects of prolonged I/R injury. This protective action of IP in attenuating myocardial damage and subcellular remodeling is likely to be due to marked reductions in the occurrence of oxidative stress and intracellular Ca2+-overload in cardiomyocytes. In addition, the beneficial actions of IP have been attributed to the depression of proteolytic activities and inflammatory levels of cytokines as well as the activation of the nuclear factor erythroid factor 2-mediated signal transduction pathway. Accordingly, this review is intended to describe some of the changes in subcellular organelles, which are induced in cardiomyocytes by I/R for the occurrence of oxidative stress and intracellular Ca2+-overload and highlight some of the mechanisms for explaining the cardioprotective effects of IP.
Collapse
|
25
|
Nicin L, Schroeter SM, Glaser SF, Schulze-Brüning R, Pham MD, Hille SS, Yekelchyk M, Kattih B, Abplanalp WT, Tombor L, Müller OJ, Braun T, Meder B, Reich C, Arsalan M, Holubec T, Walther T, Emrich F, Krishnan J, Zeiher AM, John D, Dimmeler S. A human cell atlas of the pressure-induced hypertrophic heart. NATURE CARDIOVASCULAR RESEARCH 2022; 1:174-185. [PMID: 39195989 PMCID: PMC11357985 DOI: 10.1038/s44161-022-00019-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 01/05/2022] [Indexed: 08/29/2024]
Abstract
Pathological cardiac hypertrophy is a leading cause of heart failure, but knowledge of the full repertoire of cardiac cells and their gene expression profiles in the human hypertrophic heart is missing. Here, by using large-scale single-nucleus transcriptomics, we present the transcriptional response of human cardiomyocytes to pressure overload caused by aortic valve stenosis and describe major alterations in cardiac cellular crosstalk. Hypertrophied cardiomyocytes had reduced input from endothelial cells and fibroblasts. Genes encoding Eph receptor tyrosine kinases, particularly EPHB1, were significantly downregulated in cardiomyocytes of the hypertrophied heart. Consequently, EPHB1 activation by its ligand ephrin (EFN)B2, which is mainly expressed by endothelial cells, was reduced. EFNB2 inhibited cardiomyocyte hypertrophy in vitro, while silencing its expression in endothelial cells induced hypertrophy in co-cultured cardiomyocytes. Our human cell atlas of the hypertrophied heart highlights the importance of intercellular crosstalk in disease pathogenesis and provides a valuable resource.
Collapse
Affiliation(s)
- Luka Nicin
- Institute for Cardiovascular Regeneration, Goethe University Frankfurt, Frankfurt, Germany
- German Center for Cardiovascular Research (DZHK), partner site Frankfurt Rhine-Main, Berlin, Germany
- Cardiopulmonary Institute, Goethe University Frankfurt, Frankfurt, Germany
| | - Sam Michael Schroeter
- Institute for Cardiovascular Regeneration, Goethe University Frankfurt, Frankfurt, Germany
- German Center for Cardiovascular Research (DZHK), partner site Frankfurt Rhine-Main, Berlin, Germany
- Cardiopulmonary Institute, Goethe University Frankfurt, Frankfurt, Germany
| | - Simone Franziska Glaser
- Institute for Cardiovascular Regeneration, Goethe University Frankfurt, Frankfurt, Germany
- German Center for Cardiovascular Research (DZHK), partner site Frankfurt Rhine-Main, Berlin, Germany
- Cardiopulmonary Institute, Goethe University Frankfurt, Frankfurt, Germany
| | - Ralf Schulze-Brüning
- Institute for Cardiovascular Regeneration, Goethe University Frankfurt, Frankfurt, Germany
| | - Minh-Duc Pham
- Cardiopulmonary Institute, Goethe University Frankfurt, Frankfurt, Germany
- Cardiac Metabolism Group, Department of Cardiology, Goethe University Frankfurt, Frankfurt, Germany
| | - Susanne S Hille
- Department of Internal Medicine III, University Hospital Schleswig-Holstein, Kiel, Germany
- German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Berlin, Germany
| | - Michail Yekelchyk
- Cardiopulmonary Institute, Goethe University Frankfurt, Frankfurt, Germany
- Department of Cardiac Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Badder Kattih
- Institute for Cardiovascular Regeneration, Goethe University Frankfurt, Frankfurt, Germany
- German Center for Cardiovascular Research (DZHK), partner site Frankfurt Rhine-Main, Berlin, Germany
- Cardiopulmonary Institute, Goethe University Frankfurt, Frankfurt, Germany
| | - Wesley Tyler Abplanalp
- Institute for Cardiovascular Regeneration, Goethe University Frankfurt, Frankfurt, Germany
- German Center for Cardiovascular Research (DZHK), partner site Frankfurt Rhine-Main, Berlin, Germany
- Cardiopulmonary Institute, Goethe University Frankfurt, Frankfurt, Germany
| | - Lukas Tombor
- Institute for Cardiovascular Regeneration, Goethe University Frankfurt, Frankfurt, Germany
- German Center for Cardiovascular Research (DZHK), partner site Frankfurt Rhine-Main, Berlin, Germany
- Cardiopulmonary Institute, Goethe University Frankfurt, Frankfurt, Germany
| | - Oliver J Müller
- Department of Internal Medicine III, University Hospital Schleswig-Holstein, Kiel, Germany
- German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Berlin, Germany
| | - Thomas Braun
- German Center for Cardiovascular Research (DZHK), partner site Frankfurt Rhine-Main, Berlin, Germany
- Cardiopulmonary Institute, Goethe University Frankfurt, Frankfurt, Germany
- Department of Cardiac Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Benjamin Meder
- Institute for Cardiomyopathies, University Hospital Heidelberg, Heidelberg, Germany
- German Center for Cardiovascular Research (DZHK), partner site Heidelberg/Mannheim, Berlin, Germany
| | - Christoph Reich
- Institute for Cardiomyopathies, University Hospital Heidelberg, Heidelberg, Germany
- German Center for Cardiovascular Research (DZHK), partner site Heidelberg/Mannheim, Berlin, Germany
| | - Mani Arsalan
- Department of Cardiovascular Surgery, Goethe University Hospital, Frankfurt, Germany
| | - Tomas Holubec
- Department of Cardiovascular Surgery, Goethe University Hospital, Frankfurt, Germany
| | - Thomas Walther
- German Center for Cardiovascular Research (DZHK), partner site Frankfurt Rhine-Main, Berlin, Germany
- Cardiopulmonary Institute, Goethe University Frankfurt, Frankfurt, Germany
- Department of Cardiovascular Surgery, Goethe University Hospital, Frankfurt, Germany
| | - Fabian Emrich
- Department of Cardiovascular Surgery, Goethe University Hospital, Frankfurt, Germany
| | - Jaya Krishnan
- German Center for Cardiovascular Research (DZHK), partner site Frankfurt Rhine-Main, Berlin, Germany
- Cardiopulmonary Institute, Goethe University Frankfurt, Frankfurt, Germany
- Cardiac Metabolism Group, Department of Cardiology, Goethe University Frankfurt, Frankfurt, Germany
| | - Andreas M Zeiher
- Institute for Cardiovascular Regeneration, Goethe University Frankfurt, Frankfurt, Germany
- German Center for Cardiovascular Research (DZHK), partner site Frankfurt Rhine-Main, Berlin, Germany
- Cardiopulmonary Institute, Goethe University Frankfurt, Frankfurt, Germany
| | - David John
- Institute for Cardiovascular Regeneration, Goethe University Frankfurt, Frankfurt, Germany
- German Center for Cardiovascular Research (DZHK), partner site Frankfurt Rhine-Main, Berlin, Germany
- Cardiopulmonary Institute, Goethe University Frankfurt, Frankfurt, Germany
| | - Stefanie Dimmeler
- Institute for Cardiovascular Regeneration, Goethe University Frankfurt, Frankfurt, Germany.
- German Center for Cardiovascular Research (DZHK), partner site Frankfurt Rhine-Main, Berlin, Germany.
- Cardiopulmonary Institute, Goethe University Frankfurt, Frankfurt, Germany.
| |
Collapse
|
26
|
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: 2.0] [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
|
27
|
Abstract
PURPOSE OF REVIEW To discuss the use of vasopressors and inotropes in cardiogenic shock. RECENT FINDINGS The classic form or cardiogenic shock requires administration of inotropic and/or vasopressor agents to try to improve the impaired tissue perfusion. Among vasopressors various alpha-adrenergic agents, vasopressin derivatives and angiotensin can be used. The first-line therapy remains norepinephrine as it is associated with minimal adverse effects and appears to be associated by the best outcome in network meta-analyses. On the contrary, epinephrine is associated with an increased incidence of refractory shock and observational studies suggest an increased risk of death. Vasopressin may be an excellent alternative in tachycardiac patients or in the presence of pulmonary hypertension. Concerning inotropic agents, dobutamine is the first-line agent but levosimendan is an excellent alternative or additional agent in cases not responding to dobutamine. The impact on outcome of inotropic agents remains controversial. SUMMARY Recent studies have refined the position of the various vasopressor and inotropic agents. Norepinephrine is recommended as first-line vasopressor agent by various guidelines. Among inotropic agents, selection between the agents should be individualized and based on the hemodynamic response.
Collapse
|
28
|
Lymperopoulos A, Cora N, Maning J, Brill AR, Sizova A. Signaling and function of cardiac autonomic nervous system receptors: Insights from the GPCR signalling universe. FEBS J 2021; 288:2645-2659. [PMID: 33599081 DOI: 10.1111/febs.15771] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 02/02/2021] [Accepted: 02/16/2021] [Indexed: 12/16/2022]
Affiliation(s)
- Anastasios Lymperopoulos
- Laboratory for the Study of Neurohormonal Control of the Circulation Department of Pharmaceutical Sciences Nova Southeastern University Fort Lauderdale FL USA
| | - Natalie Cora
- Laboratory for the Study of Neurohormonal Control of the Circulation Department of Pharmaceutical Sciences Nova Southeastern University Fort Lauderdale FL USA
| | - Jennifer Maning
- Laboratory for the Study of Neurohormonal Control of the Circulation Department of Pharmaceutical Sciences Nova Southeastern University Fort Lauderdale FL USA
| | - Ava R. Brill
- Laboratory for the Study of Neurohormonal Control of the Circulation Department of Pharmaceutical Sciences Nova Southeastern University Fort Lauderdale FL USA
| | - Anastasiya Sizova
- Laboratory for the Study of Neurohormonal Control of the Circulation Department of Pharmaceutical Sciences Nova Southeastern University Fort Lauderdale FL USA
| |
Collapse
|
29
|
Govindsamy A, Ghoor S, Cerf ME. Programming With Varying Dietary Fat Content Alters Cardiac Insulin Receptor, Glut4 and FoxO1 Immunoreactivity in Neonatal Rats, Whereas High Fat Programming Alters Cebpa Gene Expression in Neonatal Female Rats. Front Endocrinol (Lausanne) 2021; 12:772095. [PMID: 35069436 PMCID: PMC8766637 DOI: 10.3389/fendo.2021.772095] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 11/01/2021] [Indexed: 12/20/2022] Open
Abstract
Fetal programming refers to an intrauterine stimulus or insult that shapes growth, development and health outcomes. Dependent on the quality and quantity, dietary fats can be beneficial or detrimental for the growth of the fetus and can alter insulin signaling by regulating the expression of key factors. The effects of varying dietary fat content on the expression profiles of factors in the neonatal female and male rat heart were investigated and analyzed in control (10% fat), 20F (20% fat), 30F (30% fat) and 40F (40% fat which was a high fat diet used to induce high fat programming) neonatal rats. The whole neonatal heart was immunostained for insulin receptor, glucose transporter 4 (Glut4) and forkhead box protein 1 (FoxO1), followed by image analysis. The expression of 84 genes, commonly associated with the insulin signaling pathway, were then examined in 40F female and 40F male offspring. Maintenance on diets, varying in fat content during fetal life, altered the expression of cardiac factors, with changes induced from 20% fat in female neonates, but from 30% fat in male neonates. Further, CCAAT/enhancer-binding protein alpha (Cebpa) was upregulated in 40F female neonates. There was, however, differential expression of several insulin signaling genes in 40F (high fat programmed) offspring, with some tending to significance but most differences were in fold changes (≥1.5 fold). The increased immunoreactivity for insulin receptor, Glut4 and FoxO1 in 20F female and 30F male neonatal rats may reflect a compensatory response to programming to maintain cardiac physiology. Cebpa was upregulated in female offspring maintained on a high fat diet, with fold increases in other insulin signaling genes viz. Aebp1, Cfd (adipsin), Adra1d, Prkcg, Igfbp, Retn (resistin) and Ucp1. In female offspring maintained on a high fat diet, increased Cebpa gene expression (concomitant with fold increases in other insulin signaling genes) may reflect cardiac stress and an adaptative response to cardiac inflammation, stress and/or injury, after high fat programming. Diet and the sex are determinants of cardiac physiology and pathophysiology, reflecting divergent mechanisms that are sex-specific.
Collapse
Affiliation(s)
- Annelene Govindsamy
- Discipline of Pharmaceutical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Samira Ghoor
- Biomedical Research and Innovation Platform, South African Medical Research Council, Cape Town, South Africa
| | - Marlon E. Cerf
- Biomedical Research and Innovation Platform, South African Medical Research Council, Cape Town, South Africa
- Grants, Innovation and Product Development, South African Medical Research Council, Cape Town, South Africa
- *Correspondence: Marlon E. Cerf,
| |
Collapse
|