1
|
Wei W, Smrcka AV. Internalized β2-Adrenergic Receptors Oppose PLC-Dependent Hypertrophic Signaling. Circ Res 2024; 135:e24-e38. [PMID: 38813686 PMCID: PMC11223973 DOI: 10.1161/circresaha.123.323201] [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: 06/07/2023] [Accepted: 05/16/2024] [Indexed: 05/31/2024]
Abstract
BACKGROUND Chronically elevated neurohumoral drive, and particularly elevated adrenergic tone leading to β-adrenergic receptor (β-AR) overstimulation in cardiac myocytes, is a key mechanism involved in the progression of heart failure. β1-AR (β1-adrenergic receptor) and β2-ARs (β2-adrenergic receptor) are the 2 major subtypes of β-ARs present in the human heart; however, they elicit different or even opposite effects on cardiac function and hypertrophy. For example, chronic activation of β1-ARs drives detrimental cardiac remodeling while β2-AR signaling is protective. The underlying molecular mechanisms for cardiac protection through β2-ARs remain unclear. METHODS β2-AR signaling mechanisms were studied in isolated neonatal rat ventricular myocytes and adult mouse ventricular myocytes using live cell imaging and Western blotting methods. Isolated myocytes and mice were used to examine the roles of β2-AR signaling mechanisms in the regulation of cardiac hypertrophy. RESULTS Here, we show that β2-AR activation protects against hypertrophy through inhibition of phospholipaseCε signaling at the Golgi apparatus. The mechanism for β2-AR-mediated phospholipase C inhibition requires internalization of β2-AR, activation of Gi and Gβγ subunit signaling at endosome and ERK (extracellular regulated kinase) activation. This pathway inhibits both angiotensin II and Golgi-β1-AR-mediated stimulation of phosphoinositide hydrolysis at the Golgi apparatus ultimately resulting in decreased PKD (protein kinase D) and histone deacetylase 5 phosphorylation and protection against cardiac hypertrophy. CONCLUSIONS This reveals a mechanism for β2-AR antagonism of the phospholipase Cε pathway that may contribute to the known protective effects of β2-AR signaling on the development of heart failure.
Collapse
Affiliation(s)
- Wenhui Wei
- Department of Pharmacology, University of Michigan School of Medicine, Ann Arbor, United States
| | - Alan V. Smrcka
- Department of Pharmacology, University of Michigan School of Medicine, Ann Arbor, United States
| |
Collapse
|
2
|
Grisanti LA. TRAIL and its receptors in cardiac diseases. Front Physiol 2023; 14:1256852. [PMID: 37621762 PMCID: PMC10445540 DOI: 10.3389/fphys.2023.1256852] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 07/28/2023] [Indexed: 08/26/2023] Open
Abstract
Cardiovascular disease is a leading cause of death worldwide. Loss of cardiomyocytes that occurs during many types of damage to the heart such as ischemic injury and stress caused by pressure overload, diminishes cardiac function due to their limited regenerative capacity and promotes remodeling, which further damages the heart. Cardiomyocyte death occurs through two primary mechanisms, necrosis and apoptosis. Apoptosis is a highly regulated form of cell death that can occur through intrinsic (mitochondrial) or extrinsic (receptor mediated) pathways. Extrinsic apoptosis occurs through a subset of Tumor Necrosis Receptor (TNF) family receptors termed "Death Receptors." While some ligands for death receptors have been extensively studied in the heart, such as TNF-α, others have been virtually unstudied. One poorly characterized cardiac TNF related ligand is TNF-Related Apoptosis Inducing Ligand (TRAIL). TRAIL binds to two apoptosis-inducing receptors, Death Receptor (DR) 4 and DR5. There are also three decoy TRAIL receptors, Decoy Receptor (DcR) 1, DcR2 and osteoprotegerin (OPG). While TRAIL has been extensively studied in the cancer field due to its ability to selectively induce apoptosis in transformed cell types, emerging clinical evidence points towards a role for TRAIL and its receptors in cardiac pathology. This article will highlight our current understanding of TRAIL and its receptors in normal and pathological conditions in the heart.
Collapse
Affiliation(s)
- Laurel A. Grisanti
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, United States
| |
Collapse
|
3
|
Wei W, Smrcka AV. Internalized β2-Adrenergic Receptors Inhibit Subcellular Phospholipase C-Dependent Cardiac Hypertrophic Signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.07.544153. [PMID: 37333278 PMCID: PMC10274790 DOI: 10.1101/2023.06.07.544153] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Chronically elevated neurohumoral drive, and particularly elevated adrenergic tone leading to β-adrenergic receptor (β-AR) overstimulation in cardiac myocytes, is a key mechanism involved in the progression of heart failure. β1-AR and β2-ARs are the two major subtypes of β-ARs present in the human heart, however, they elicit different or even opposite effects on cardiac function and hypertrophy. For example, chronic activation of β1ARs drives detrimental cardiac remodeling while β2AR signaling is protective. The underlying molecular mechanisms for cardiac protection through β2ARs remain unclear. Here we show that β2-AR protects against hypertrophy through inhibition of PLCε signaling at the Golgi apparatus. The mechanism for β2AR-mediated PLC inhibition requires internalization of β2AR, activation of Gi and Gβγ subunit signaling at endosomes and ERK activation. This pathway inhibits both angiotensin II and Golgi-β1-AR-mediated stimulation of phosphoinositide hydrolysis at the Golgi apparatus ultimately resulting in decreased PKD and HDAC5 phosphorylation and protection against cardiac hypertrophy. This reveals a mechanism for β2-AR antagonism of the PLCε pathway that may contribute to the known protective effects of β2-AR signaling on the development of heart failure.
Collapse
Affiliation(s)
- Wenhui Wei
- Department of Pharmacology, University of Michigan School of Medicine, Ann Arbor, United States
| | - Alan V. Smrcka
- Department of Pharmacology, University of Michigan School of Medicine, Ann Arbor, United States
| |
Collapse
|
4
|
Chang WT, Lin HW, Chang TC, Lin SH, Li YH. The association between tyrosine kinase inhibitors and fatal arrhythmia in patients with non-small cell lung cancer in Taiwan. Front Oncol 2023; 13:1172036. [PMID: 37139162 PMCID: PMC10150998 DOI: 10.3389/fonc.2023.1172036] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 04/04/2023] [Indexed: 05/05/2023] Open
Abstract
Objective As a standard therapy, tyrosine kinase inhibitors (TKIs) improved survival in patients with non-small cell lung cancer (NSCLC) and epidermal growth factor receptor (EGFR) mutation. However, treatment-related cardiotoxicity, particularly arrhythmia, cannot be ignored. With the prevalence of EGFR mutations in Asian populations, the risk of arrhythmia among patients with NSCLC remains unclear. Methods Using data from the Taiwanese National Health Insurance Research Database and National Cancer Registry, we identified patients with NSCLC from 2001 to 2014. Using Cox proportional hazards models, we analyzed outcomes of death and arrhythmia, including ventricular arrhythmia (VA), sudden cardiac death (SCD), and atrial fibrillation (AF). The follow-up duration was three years. Results In total, 3876 patients with NSCLC treated with TKIs were matched to 3876 patients treated with platinum analogues. After adjusting for age, sex, comorbidities, and anticancer and cardiovascular therapies, patients receiving TKIs had a significantly lower risk of death (adjusted HR: 0.767; CI: 0.729-0.807, p < 0.001) than those receiving platinum analogues. Given that approximately 80% of the studied population reached the endpoint of mortality, we also adjusted for mortality as a competing risk. Notably, we observed significantly increased risks of both VA (adjusted sHR: 2.328; CI: 1.592-3.404, p < 0.001) and SCD (adjusted sHR: 1.316; CI: 1.041-1.663, p = 0.022) among TKI users compared with platinum analogue users. Conversely, the risk of AF was similar between the two groups. In the subgroup analysis, the increasing risk of VA/SCD persisted regardless of sex and most cardiovascular comorbidities. Conclusions Collectively, we highlighted a higher risk of VA/SCD in TKI users than in patients receiving platinum analogues. Further research is needed to validate these findings.
Collapse
Affiliation(s)
- Wei-Ting Chang
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Division of Cardiology, Department of Internal Medicine, Chi-Mei Medical Center, Tainan, Taiwan
- Department of Biotechnology, Southern Taiwan University of Science and Technology, Tainan, Taiwan
| | - Hui-Wen Lin
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Biostatistics Consulting Center, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ting-Chia Chang
- Division of Pulmonology, Department of Internal Medicine, Chi Mei Medical Center, Tainan, Taiwan
| | - Sheng-Hsiang Lin
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Biostatistics Consulting Center, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Public Health, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yi-Heng Li
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
5
|
Steinberg SF. N-Tertaining a New Signaling Paradigm for the Cardiomyocyte β 1 -Adrenergic Receptor. J Cardiovasc Pharmacol 2022; 80:328-333. [PMID: 35099166 PMCID: PMC9170829 DOI: 10.1097/fjc.0000000000001194] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 11/20/2021] [Indexed: 01/31/2023]
Abstract
ABSTRACT β 1 -adrenergic receptors (β 1 ARs) are the principle mediators of catecholamine actions in cardiomyocytes. β 1 ARs rapidly adjust cardiac output and provide short-term hemodynamic support for the failing heart by activating a Gs-adenylyl cyclase pathway that increases 3'-5'-cyclic adenosine monophosphate and leads to the activation of protein kinase A and the phosphorylation of substrates involved in excitation-contraction coupling. However, chronic persistent β 1 AR activation in the setting of heart failure leads to a spectrum of maladaptive changes that contribute to the evolution of heart failure. The molecular basis for β 1 AR-driven maladaptive responses remains uncertain because chronic persistent β 1 AR activation has been linked to the activation of both proapoptotic and antiapoptotic signaling pathways. Of note, studies to date have been predicated on the assumption that β 1 ARs signal exclusively as full-length receptor proteins. Our recent studies show that β 1 ARs are detected as both full-length and N-terminally truncated species in cardiomyocytes, that N-terminal cleavage is regulated by O-glycan modifications at specific sites on the β 1 AR N-terminus, and that N-terminally truncated β 1 ARs remain signaling competent, but their signaling properties differ from those of the full-length β 1 AR. The N-terminally truncated form of the β 1 AR constitutively activates the protein kinase B signaling pathway and confers protection against doxorubicin-dependent apoptosis in cardiomyocytes. These studies identify a novel signaling paradigm for the β 1 AR, implicating the N-terminus as a heretofore-unrecognized structural determinant of β 1 AR responsiveness that could be pharmacologically targeted for therapeutic advantage.
Collapse
|
6
|
Wang J, Pani B, Gokhan I, Xiong X, Kahsai AW, Jiang H, Ahn S, Lefkowitz RJ, Rockman HA. β-Arrestin-Biased Allosteric Modulator Potentiates Carvedilol-Stimulated β Adrenergic Receptor Cardioprotection. Mol Pharmacol 2021; 100:568-579. [PMID: 34561298 DOI: 10.1124/molpharm.121.000359] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 09/20/2021] [Indexed: 01/14/2023] Open
Abstract
β 1 adrenergic receptors (β 1ARs) are central regulators of cardiac function and a drug target for cardiac disease. As a member of the G protein-coupled receptor family, β 1ARs activate cellular signaling by primarily coupling to Gs proteins to activate adenylyl cyclase, cAMP-dependent pathways, and the multifunctional adaptor-transducer protein β-arrestin. Carvedilol, a traditional β-blocker widely used in treating high blood pressure and heart failure by blocking β adrenergic receptor-mediated G protein activation, can selectively stimulate Gs-independent β-arrestin signaling of β adrenergic receptors, a process known as β-arrestin-biased agonism. Recently, a DNA-encoded small-molecule library screen against agonist-occupied β 2 adrenergic receptors (β 2ARs) identified Compound-6 (Cmpd-6) to be a positive allosteric modulator for agonists on β 2ARs. Intriguingly, it was further discovered that Cmpd-6 is positively cooperative with the β-arrestin-biased ligand carvedilol at β 2ARs. Here we describe the surprising finding that at β 1ARs unlike β 2ARs, Cmpd-6 is cooperative only with carvedilol and not agonists. Cmpd-6 increases the binding affinity of carvedilol for β 1ARs and potentiates carvedilol-stimulated, β-arrestin-dependent β 1AR signaling, such as epidermal growth factor receptor transactivation and extracellular signal-regulated kinase activation, whereas it does not have an effect on Gs-mediated cAMP generation. In vivo, Cmpd-6 enhances the antiapoptotic, cardioprotective effect of carvedilol in response to myocardial ischemia/reperfusion injury. This antiapoptotic role of carvedilol is dependent on β-arrestins since it is lost in mice with myocyte-specific deletion of β-arrestins. Our findings demonstrate that Cmpd-6 is a selective β-arrestin-biased allosteric modulator of β 1ARs and highlight its potential clinical utility in enhancing carvedilol-mediated cardioprotection against ischemic injury. SIGNIFICANCE STATEMENT: This study demonstrates the positive cooperativity of Cmpd-6 on β1ARs as a β-arrestin-biased positive allosteric modulator. Cmpd-6 selectively enhances the affinity and cellular signaling of carvedilol, a known β-arrestin-biased β-blocker for β1ARs, whereas it has minimal effect on other ligands tested. Importantly, Cmpd-6 enhances the β-arrestin-dependent in vivo cardioprotective effect of carvedilol during ischemia/reperfusion injury-induced apoptosis. The data support the potential therapeutic application of Cmpd-6 to enhance the clinical benefits of carvedilol in the treatment of cardiac disease.
Collapse
Affiliation(s)
- Jialu Wang
- Departments of Medicine (J.W., B.P, I.G., X.X., A.W.K., H.J., S.A., R.J.L., H.A.R.), Biochemistry (R.J.W.), Howard Hughes Medical Institute (R.J.L.), and Cell Biology (H.A.R.), Duke University Medical Center, Durham, North Carolina
| | - Biswaranjan Pani
- Departments of Medicine (J.W., B.P, I.G., X.X., A.W.K., H.J., S.A., R.J.L., H.A.R.), Biochemistry (R.J.W.), Howard Hughes Medical Institute (R.J.L.), and Cell Biology (H.A.R.), Duke University Medical Center, Durham, North Carolina
| | - Ilhan Gokhan
- Departments of Medicine (J.W., B.P, I.G., X.X., A.W.K., H.J., S.A., R.J.L., H.A.R.), Biochemistry (R.J.W.), Howard Hughes Medical Institute (R.J.L.), and Cell Biology (H.A.R.), Duke University Medical Center, Durham, North Carolina
| | - Xinyu Xiong
- Departments of Medicine (J.W., B.P, I.G., X.X., A.W.K., H.J., S.A., R.J.L., H.A.R.), Biochemistry (R.J.W.), Howard Hughes Medical Institute (R.J.L.), and Cell Biology (H.A.R.), Duke University Medical Center, Durham, North Carolina
| | - Alem W Kahsai
- Departments of Medicine (J.W., B.P, I.G., X.X., A.W.K., H.J., S.A., R.J.L., H.A.R.), Biochemistry (R.J.W.), Howard Hughes Medical Institute (R.J.L.), and Cell Biology (H.A.R.), Duke University Medical Center, Durham, North Carolina
| | - Haoran Jiang
- Departments of Medicine (J.W., B.P, I.G., X.X., A.W.K., H.J., S.A., R.J.L., H.A.R.), Biochemistry (R.J.W.), Howard Hughes Medical Institute (R.J.L.), and Cell Biology (H.A.R.), Duke University Medical Center, Durham, North Carolina
| | - Seungkirl Ahn
- Departments of Medicine (J.W., B.P, I.G., X.X., A.W.K., H.J., S.A., R.J.L., H.A.R.), Biochemistry (R.J.W.), Howard Hughes Medical Institute (R.J.L.), and Cell Biology (H.A.R.), Duke University Medical Center, Durham, North Carolina
| | - Robert J Lefkowitz
- Departments of Medicine (J.W., B.P, I.G., X.X., A.W.K., H.J., S.A., R.J.L., H.A.R.), Biochemistry (R.J.W.), Howard Hughes Medical Institute (R.J.L.), and Cell Biology (H.A.R.), Duke University Medical Center, Durham, North Carolina
| | - Howard A Rockman
- Departments of Medicine (J.W., B.P, I.G., X.X., A.W.K., H.J., S.A., R.J.L., H.A.R.), Biochemistry (R.J.W.), Howard Hughes Medical Institute (R.J.L.), and Cell Biology (H.A.R.), Duke University Medical Center, Durham, North Carolina
| |
Collapse
|
7
|
Tanner MA, Grisanti LA. A Dual Role for Death Receptor 5 in Regulating Cardiac Fibroblast Function. Front Cardiovasc Med 2021; 8:699102. [PMID: 34527710 PMCID: PMC8437145 DOI: 10.3389/fcvm.2021.699102] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 08/06/2021] [Indexed: 12/30/2022] Open
Abstract
The fibrotic response is involved in nearly all forms of heart failure and dysregulated responses can lead to enhanced cardiac dysfunction. TNF-related apoptosis-inducing ligand (TRAIL) and its receptor, death receptor (DR) 5, are associated with multiple forms of heart failure, but their role in the heart is poorly defined. Our previous study identified DR5 expression on cardiac fibroblasts however, the impact of DR5 on fibroblast function remains unexplored. To investigate the role of DR5 in cardiac fibroblasts, a variety of fibroblast functions were examined following treatment with the endogenous ligand, TRAIL, or small molecule agonist, bioymifi. DR5 activation did not induce apoptosis in naïve fibroblasts but activated ERK1/2 signaling to increase proliferation. However, upon activation and differentiation to myofibroblasts, DR5 expression was elevated, and DR5 agonists induced caspase 3 activation resulting in myofibroblast apoptosis. To investigate the impact of DR5 regulation of fibroblasts in vivo, a chronic isoproterenol administration model of heart failure was used. Wild-type (WT) mice receiving isoproterenol had increased hypertrophy, cardiomyocyte death, and fibrosis and decreased contractility compared to vehicle treated animals. DR5 knockout (KO) mice had no overt baseline phenotype however, following isoproterenol infusion, increased cardiomyocyte death and hypertrophy in comparison to isoproterenol treated WT animals was observed. DR5KO mice had an augmented fibrotic response with isoproterenol treatment compared with WT, which corresponded with additional decreases in contractility. These findings identify a dual role for DR5 in cardiac fibroblast function through enhanced naïve fibroblast proliferation, which switches to a pro-apoptotic function upon differentiation to myofibroblasts. This is important in heart failure where DR5 activation suppresses maladaptive remodeling and may represent a novel therapeutic target for the treatment of heart failure.
Collapse
Affiliation(s)
- Miles A Tanner
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, United States
| | - Laurel A Grisanti
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, United States
| |
Collapse
|
8
|
Palanisamy S, Xue C, Ishiyama S, Naga Prasad SV, Gabrielson K. GPCR-ErbB transactivation pathways and clinical implications. Cell Signal 2021; 86:110092. [PMID: 34303814 DOI: 10.1016/j.cellsig.2021.110092] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 07/18/2021] [Accepted: 07/19/2021] [Indexed: 11/18/2022]
Abstract
Cell surface receptors including the epidermal growth factor receptor (EGFR) family and G-protein coupled receptors (GPCRs) play quintessential roles in physiology, and in diseases, including cardiovascular diseases. While downstream signaling from these individual receptor families has been well studied, the cross-talk between EGF and GPCR receptor families is still incompletely understood. Including members of both receptor families, the number of receptor and ligand combinations for unique interactions is vast, offering a frontier of pharmacologic targets to explore for preventing and treating disease. This molecular cross-talk, called receptor transactivation, is reviewed here with a focus on the cardiovascular system featuring the well-studied GPCR receptors, but also discussing less-studied receptors from both families for a broad understanding of context of expansile interactions, repertoire of cellular signaling, and disease consequences. Attention is given to cell type, level of chronicity, and disease context given that transactivation and comorbidities, including diabetes, hypertension, coronavirus infection, impact cardiovascular disease and health outcomes.
Collapse
Affiliation(s)
| | - Carolyn Xue
- University of California, Los Angeles, 101 Hershey Hall, 612 Charles E. Young Drive South, Los Angeles, CA 90095, USA.
| | - Shun Ishiyama
- Sidney Kimmel Cancer Center, Department of Surgery, Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Coloproctological Surgery, Juntendo University School of Medicine, Tokyo, Japan.
| | - Sathyamangla Venkata Naga Prasad
- NB50, Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, 1, Cleveland, OH 44195, USA.
| | - Kathleen Gabrielson
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University, School of Medicine, 733 North Broadway, Miller Research Building, Room 807, Baltimore, MD 21205-2196, USA.
| |
Collapse
|
9
|
Guo S, Okyere AD, McEachern E, Strong JL, Carter RL, Patwa VC, Thomas TP, Landy M, Song J, Lucchese AM, Martin TG, Gao E, Rajan S, Kirk JA, Koch WJ, Cheung JY, Tilley DG. Epidermal growth factor receptor-dependent maintenance of cardiac contractility. Cardiovasc Res 2021; 118:1276-1288. [PMID: 33892492 DOI: 10.1093/cvr/cvab149] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 02/16/2021] [Accepted: 04/21/2021] [Indexed: 01/22/2023] Open
Abstract
AIMS Epidermal growth factor receptor (EGFR) is essential to the development of multiple tissues and organs and is a target of cancer therapeutics. Due to the embryonic lethality of global EGFR deletion and conflicting reports of cardiac-overexpressed EGFR mutants, its specific impact on the adult heart, normally or in response to chronic stress, has not been established. Using complimentary genetic strategies to modulate cardiomyocyte-specific EGFR expression, we aim to define its role in the regulation of cardiac function and remodeling. METHODS AND RESULTS A floxed EGFR mouse model with α-myosin heavy chain-Cre-mediated cardiomyocyte-specific EGFR downregulation (CM-EGFR-KD mice) developed contractile dysfunction by 9 weeks of age, marked by impaired diastolic relaxation, as monitored via echocardiographic, hemodynamic and isolated cardiomyocyte contractility analyses. This contractile defect was maintained over time without overt cardiac remodeling until 10 months of age, after which the mice ultimately developed severe heart failure and reduced lifespan. Acute downregulation of EGFR in adult floxed EGFR mice with adeno-associated virus 9 (AAV9)-encoded Cre with a cardiac troponin T promoter (AAV9-cTnT-Cre) recapitulated the CM-EGFR-KD phenotype, while AAV9-cTnT-EGFR treatment of adult CM-EGFR-KD mice rescued the phenotype. Notably, chronic administration of the β-adrenergic receptor (βAR) agonist isoproterenol effectively and reversibly compensated for the contractile dysfunction in the absence of cardiomyocyte hypertrophy in CM-EGFR-KD mice. Mechanistically, EGFR downregulation reduced the expression of protein phosphatase 2 A (PP2A) regulatory subunit Ppp2r3a/PR72, which was associated with decreased phosphorylation of phospholamban (PLB) and Ca2+ clearance, and whose re-expression via AAV9-cTnT-PR72 rescued the CM-EGFR-KD phenotype. CONCLUSIONS Altogether our study highlights a previously unrecognized role for EGFR in maintaining contractile homeostasis under physiologic conditions in the adult heart via regulation of PR72 expression. TRANSLATIONAL PERSPECTIVE Our study highlights a previously unrecognized role for EGFR in maintaining contractile homeostasis under physiologic conditions in the adult heart via regulation of PR72, a PP2A regulatory subunit with an unknown impact on cardiac function. Further, we have shown that cardiomyocyte-expressed EGFR is required for the promotion of cardiac hypertrophy under conditions of chronic catecholamine stress. Altogether, our study provides new insight into the dynamic nature of cardiomyocyte-specific EGFR.
Collapse
Affiliation(s)
- Shuchi Guo
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Ama Dedo Okyere
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Erin McEachern
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Joshua L Strong
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Rhonda L Carter
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Viren C Patwa
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Toby P Thomas
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Melissa Landy
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Jianliang Song
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Ana Maria Lucchese
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Thomas G Martin
- Loyola University Chicago, Department of Cell and Molecular Physiology, Chicago, Illinois, USA
| | - Erhe Gao
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Sudarsan Rajan
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Jonathan A Kirk
- Loyola University Chicago, Department of Cell and Molecular Physiology, Chicago, Illinois, USA
| | - Walter J Koch
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Joseph Y Cheung
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Douglas G Tilley
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| |
Collapse
|
10
|
Zhu J, Steinberg SF. β 1-adrenergic receptor N-terminal cleavage by ADAM17; the mechanism for redox-dependent downregulation of cardiomyocyte β 1-adrenergic receptors. J Mol Cell Cardiol 2021; 154:70-79. [PMID: 33556394 DOI: 10.1016/j.yjmcc.2021.01.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 01/09/2021] [Accepted: 01/30/2021] [Indexed: 12/15/2022]
Abstract
β1-adrenergic receptors (β1ARs) are the principle mediators of catecholamine action in cardiomyocytes. We previously showed that the β1AR extracellular N-terminus is a target for post-translational modifications that impact on signaling responses. Specifically, we showed that the β1AR N-terminus carries O-glycan modifications at Ser37/Ser41, that O-glycosylation prevents β1AR N-terminal cleavage, and that N-terminal truncation influences β1AR signaling to downstream effectors. However, the site(s) and mechanism for β1AR N-terminal cleavage in cells was not identified. This study shows that β1ARs are expressed in cardiomyocytes and other cells types as both full-length and N-terminally truncated species and that the truncated β1AR species is formed as a result of an O-glycan regulated N-terminal cleavage by ADAM17 at R31↓L32. We identify Ser41 as the major O-glycosylation site on the β1AR N-terminus and show that an O-glycan modification at Ser41 prevents ADAM17-dependent cleavage of the β1-AR N-terminus at S41↓L42, a second N-terminal cleavage site adjacent to this O-glycan modification (and it attenuates β1-AR N-terminal cleavage at R31↓L32). We previously reported that oxidative stress leads to a decrease in β1AR expression and catecholamine responsiveness in cardiomyocytes. This study shows that redox-inactivation of cardiomyocyte β1ARs is via a mechanism involving N-terminal truncation at R31↓L32 by ADAM17. In keeping with the previous observation that N-terminally truncated β1ARs constitutively activate an AKT pathway that affords protection against doxorubicin-dependent apoptosis, overexpression of a cleavage resistant β1AR mutant exacerbates doxorubicin-dependent apoptosis. These studies identify the β1AR N-terminus as a structural determinant of β1AR responses that can be targeted for therapeutic advantage.
Collapse
Affiliation(s)
- Jing Zhu
- Department of Pharmacology, Columbia University, New York, NY 10032, United States of America
| | - Susan F Steinberg
- Department of Pharmacology, Columbia University, New York, NY 10032, United States of America.
| |
Collapse
|
11
|
Kilpatrick LE, Hill SJ. Transactivation of G protein-coupled receptors (GPCRs) and receptor tyrosine kinases (RTKs): Recent insights using luminescence and fluorescence technologies. CURRENT OPINION IN ENDOCRINE AND METABOLIC RESEARCH 2021; 16:102-112. [PMID: 33748531 PMCID: PMC7960640 DOI: 10.1016/j.coemr.2020.10.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Alterations in signalling due to bidirectional transactivation of G protein-coupled receptor (GPCRs) and receptor tyrosine kinases (RTKs) are well established. Transactivation significantly diversifies signalling networks within a cell and has been implicated in promoting both advantageous and disadvantageous physiological and pathophysiological outcomes, making the GPCR/RTK interactions attractive new targets for drug discovery programmes. Transactivation has been observed for a plethora of receptor pairings in multiple cell types; however, the precise molecular mechanisms and signalling effectors involved can vary with receptor pairings and cell type. This short review will discuss the recent applications of proximity-based assays, such as resonance energy transfer and fluorescence-based imaging in investigating the dynamics of GPCR/RTK complex formation, subsequent effector protein recruitment and the cellular locations of complexes in living cells.
Collapse
Key Words
- 5-hydroxytryptamine receptor 1A, (5-HT1A)
- Endocytosis
- Förster Resonance Energy Transfer, (FRET)
- G protein-coupled receptor
- G protein-coupled receptors, (GPCRs)
- GPCR kinases, (GRKs)
- Oligomeric complexes
- Receptor tyrosine kinase
- Resonance energy transfer
- Transactivation
- adrenoceptors, (AR)
- bioluminescence resonance energy transfer, (BRET)
- cannabinoid receptor 2, (CB2R)
- disintegrin and metalloproteinases, (ADAMs)
- epidermal growth factor receptor, (EGFR)
- epidermal growth factor, (EGF)
- fibroblast growth factor receptor, (FGFR)
- fluorescence correlation spectroscopy, (FCS)
- formyl peptide receptor, (FPR)
- free fatty acid, (FFA)
- heparin binding EGF, (Hb-EGF)
- hepatocyte growth factor, (HGF)
- human umbilical vein endothelial cells, (HUVECs)
- insulin growth factor receptor-1, (IGFR-1)
- insulin receptor, (IR)
- lysophosphatidic acid receptor 1, (LPA)
- matrix metalloproteinases, (MMPs)
- platelet-derived growth factor receptor, (PDGFR)
- proximity ligation assay, (PLA)
- reactive oxygen species, (ROS)
- receptor tyrosine kinases, (RTKs)
- sphingosine-1-phosphate receptor, (S1PR)
- tetrahydrocannabinol, (THC)
- total internal reflection fluorescence microscopy, (TIRF-M)
- vascular endothelial growth factor receptor 2, (VEGFR2)
- vascular endothelial growth factor, (VEGF)
- vasopressin 2 receptor, (V2R)
Collapse
Affiliation(s)
- Laura E. Kilpatrick
- Division of Bimolecular Sciences and Medicinal Chemistry, Biodiscovery Institute, School of Pharmacy, University of Nottingham, Nottingham, NG7 2RD, UK
- Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, The Midlands, NG7 2UH, UK
| | - Stephen J. Hill
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, NG7 2UH, UK
- Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, The Midlands, NG7 2UH, UK
| |
Collapse
|
12
|
Patwa V, Guo S, Carter RL, Kraus L, Einspahr J, Teplitsky D, Sabri A, Tilley DG. Epidermal growth factor receptor association with β1-adrenergic receptor is mediated via its juxtamembrane domain. Cell Signal 2020; 78:109846. [PMID: 33238186 DOI: 10.1016/j.cellsig.2020.109846] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 11/19/2020] [Accepted: 11/20/2020] [Indexed: 01/20/2023]
Abstract
β1-adrenergic receptor (β1AR)-mediated transactivation of epidermal growth factor receptor (EGFR) engages downstream signaling events that impact numerous cellular processes including growth and survival. While association of these receptors has been shown to occur basally and be important for relaying transactivation-specific intracellular events, the mechanism by which they do so is unclear and elucidation of which would aid in understanding the consequence of disrupting their interaction. Using fluorescence resonance energy transfer (FRET) and immunoprecipitation (IP) analyses, we evaluated the impact of C-terminal truncations of EGFR on its ability to associate with β1AR. While loss of the last 230 amino acid C-terminal phosphotyrosine-rich domain did not disrupt the ability of EGFR to associate with β1AR, truncation of the entire intracellular domain of EGFR resulted in almost complete loss of its interaction with β1AR, suggesting that either the kinase domain or juxtamembrane domain (JMD) may be required for this association. Treatment with the EGFR antagonist gefitinib did not prevent β1AR-EGFR association, however, treatment with a palmitoylated peptide encoding the first 20 amino acids of the JMD domain (JMD-A) disrupted β1AR-EGFR association over time and prevented β1AR-mediated ERK1/2 phosphorylation, both in general and specifically in association with EGFR. Conversely, neither a mutated JMD-A peptide nor a palmitoylated peptide fragment consisting of the subsequent 18 amino acids of the JMD domain (JMD-B) were capable of doing so. Altogether, the proximal region of the JMD of EGFR is responsible for its association with β1AR, and its disruption prevents β1AR-mediated transactivation, thus providing a new tool to study the functional consequences of disrupting β1AR-EGFR downstream signaling.
Collapse
Affiliation(s)
- Viren Patwa
- Center for Translational Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Shuchi Guo
- Center for Translational Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Rhonda L Carter
- Center for Translational Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Lindsay Kraus
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Jeanette Einspahr
- Center for Translational Medicine, Temple University, Philadelphia, PA 19140, USA
| | - David Teplitsky
- Center for Translational Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Abdelkarim Sabri
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Douglas G Tilley
- Center for Translational Medicine, Temple University, Philadelphia, PA 19140, USA.
| |
Collapse
|
13
|
Tanner MA, Thomas TP, Maitz CA, Grisanti LA. β2-Adrenergic Receptors Increase Cardiac Fibroblast Proliferation Through the Gαs/ERK1/2-Dependent Secretion of Interleukin-6. Int J Mol Sci 2020; 21:ijms21228507. [PMID: 33198112 PMCID: PMC7697911 DOI: 10.3390/ijms21228507] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/04/2020] [Accepted: 11/10/2020] [Indexed: 12/18/2022] Open
Abstract
Fibroblasts are an important resident cell population in the heart involved in maintaining homeostasis and structure during normal conditions. They are also crucial in disease states for sensing signals and initiating the appropriate repair responses to maintain the structural integrity of the heart. This sentinel role of cardiac fibroblasts occurs, in part, through their ability to secrete cytokines. β-adrenergic receptors (βAR) are also critical regulators of cardiac function in the normal and diseased state and a major therapeutic target clinically. βAR are known to influence cytokine secretion in various cell types and they have been shown to be involved in cytokine production in the heart, but their role in regulating cytokine production in cardiac fibroblasts is not well understood. Thus, we hypothesized that βAR activation on cardiac fibroblasts modulates cytokine production to influence fibroblast function. Using primary fibroblast cultures from neonatal rats and adult mice, increased interleukin (IL)-6 expression and secretion occurred following β2AR activation. The use of pharmacological inhibitors and genetic manipulations showed that IL-6 elevations occurred through the Gαs-mediated activation of ERK1/2 and resulted in increased fibroblast proliferation. In vivo, a lack of β2AR resulted in increased infarct size following myocardial infarction and impaired wound closure in a murine dermal wound healing assay. These findings identify an important role for β2AR in regulating fibroblast proliferation through Gαs/ERK1/2-dependent alterations in IL-6 and may lead to the development of improved heart failure therapies through targeting fibrotic function of β2AR.
Collapse
Affiliation(s)
- Miles A. Tanner
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO 65211, USA; (M.A.T.); (T.P.T.)
| | - Toby P. Thomas
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO 65211, USA; (M.A.T.); (T.P.T.)
| | - Charles A. Maitz
- Department of Veterinary Medicine and Surgery, College of Veterinary Medicine, University of Missouri, Columbia, MO 65211, USA;
| | - Laurel A. Grisanti
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO 65211, USA; (M.A.T.); (T.P.T.)
- Correspondence: ; Tel.: +573-884-8852
| |
Collapse
|
14
|
Scrimgeour LA, Potz BA, Aboul Gheit A, Liu Y, Shi G, Pfeiffer M, Colantuono BJ, Sodha NR, Abid MR, Sellke FW. Intravenous injection of extracellular vesicles to treat chronic myocardial ischemia. PLoS One 2020; 15:e0238879. [PMID: 32915887 PMCID: PMC7485873 DOI: 10.1371/journal.pone.0238879] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 08/25/2020] [Indexed: 01/21/2023] Open
Abstract
Background Mesenchymal stem cell-derived extracellular vesicles (EVs) appear to be a very exciting treatment option for heart disease. Here, we used a swine model of chronic myocardial ischemia to evaluate the efficacy of a less-invasive method of injection of EVs via a peripheral intravenous route. Methods Sixteen Yorkshire swine underwent placement of an ameroid constrictor on the left circumflex (LCx) artery at age 11 weeks to induce chronic myocardial ischemia. Two weeks later, they were divided into two groups: control (CON; n = 8), and intravenous injection of EVs (EVIV; n = 8). At 18 weeks of age, animals underwent final analysis and euthanasia. The chronically ischemic myocardium (LCx territory) was harvested for analysis. Results Intravenous injection (IV) of EVs induced several pro-angiogenic markers such as MAPK, JNK but not Akt. Whereas IV injections of EVs decreased VEGFR2 expression and inhibited apoptotic signaling (caspase 3), they increased expression of VEGFR1 that is believed to be anti-angiogenic. Injection of EVs did not result in an increase in vessel density and blood flow when compared to the control group. Conclusions Although IV injection of EVs upregulated several pro-angiogenic signaling pathways, it failed to induce changes in vascular density in the chronically ischemic myocardium. Thus, a lack of increase in vascular density at the doses tested failed to elicit a functional response in ischemic myocardium.
Collapse
Affiliation(s)
- Laura A. Scrimgeour
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Brown University Warren Alpert Medical School, Providence, RI, United States of America
| | - Brittany A. Potz
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Brown University Warren Alpert Medical School, Providence, RI, United States of America
| | - Ahmad Aboul Gheit
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Brown University Warren Alpert Medical School, Providence, RI, United States of America
| | - Yuhong Liu
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Brown University Warren Alpert Medical School, Providence, RI, United States of America
| | - Guangbin Shi
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Brown University Warren Alpert Medical School, Providence, RI, United States of America
| | - Melissa Pfeiffer
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Brown University Warren Alpert Medical School, Providence, RI, United States of America
| | - Bonnie J. Colantuono
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Brown University Warren Alpert Medical School, Providence, RI, United States of America
| | - Neel R. Sodha
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Brown University Warren Alpert Medical School, Providence, RI, United States of America
| | - M. Ruhul Abid
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Brown University Warren Alpert Medical School, Providence, RI, United States of America
| | - Frank W. Sellke
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Brown University Warren Alpert Medical School, Providence, RI, United States of America
- * E-mail:
| |
Collapse
|
15
|
Tanner MA, Thomas TP, Grisanti LA. Death receptor 5 contributes to cardiomyocyte hypertrophy through epidermal growth factor receptor transactivation. J Mol Cell Cardiol 2019; 136:1-14. [PMID: 31473246 DOI: 10.1016/j.yjmcc.2019.08.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 08/26/2019] [Accepted: 08/27/2019] [Indexed: 12/30/2022]
Abstract
Cardiomyocyte survival and death contributes to many cardiac diseases. A common mechanism of cardiomyocyte death is through apoptosis however, numerous death receptors (DR) have been virtually unstudied in the context of cardiovascular disease. Previous studies have identified TNF-related apoptosis inducing ligand (TRAIL) and its receptor, DR5, as being altered in a chronic catecholamine administration model of heart failure, and suggest a role of non-canonical signaling in cardiomyocytes. Furthermore, multiple clinical studies have identified TRAIL or DR5 as biomarkers in the prediction of severity and mortality following myocardial infarction and in heart failure development risk suggesting a role of DR5 signaling in the heart. While TRAIL/DR5 have been extensively studied as a potential cancer therapeutic due to their ability to selectively activate apoptosis in cancer cells, TRAIL and DR5 are highly expressed in the heart where their function is uncharacterized. However, many non-transformed cell types are resistant to TRAIL-induced apoptosis suggesting non-canonical functions in non-cancerous cell types. Our goal was to determine the role of DR5 in the heart with the hypothesis that DR5 does not induce cardiomyocyte apoptosis but initiates non-canonical signaling to promote cardiomyocyte growth and survival. Histological analysis of hearts from mice treated with a DR5 agonists showed increased hypertrophy with no differences in cardiomyocyte death, fibrosis or function. Mechanistic studies in the heart and isolated cardiomyocytes identified ERK1/2 activation with DR5 agonist treatment which contributed to hypertrophy. Furthermore, epidermal growth factor receptor (EGFR) was activated following DR5 agonist treatment through activation of MMP and HB-EGFR cleavage and specific inhibitors of MMP and EGFR prevented DR5-mediated ERK1/2 signaling and hypertrophy. Taken together, these studies identify a previously unidentified role for DR5 in the heart, which does not promote apoptosis but acts through non-canonical MMP-EGFR-ERK1/2 signaling mechanisms to contribute to cardiomyocyte hypertrophy.
Collapse
Affiliation(s)
- Miles A Tanner
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA
| | - Toby P Thomas
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA
| | - Laurel A Grisanti
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA.
| |
Collapse
|
16
|
Brand CS, Lighthouse JK, Trembley MA. Protective transcriptional mechanisms in cardiomyocytes and cardiac fibroblasts. J Mol Cell Cardiol 2019; 132:1-12. [PMID: 31042488 DOI: 10.1016/j.yjmcc.2019.04.023] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 04/19/2019] [Accepted: 04/22/2019] [Indexed: 12/13/2022]
Abstract
Heart failure is the leading cause of morbidity and mortality worldwide. Several lines of evidence suggest that physical activity and exercise can pre-condition the heart to improve the response to acute cardiac injury such as myocardial infarction or ischemia/reperfusion injury, preventing the progression to heart failure. It is becoming more apparent that cardioprotection is a concerted effort between multiple cell types and converging signaling pathways. However, the molecular mechanisms of cardioprotection are not completely understood. What is clear is that the mechanisms underlying this protection involve acute activation of transcriptional activators and their corresponding gene expression programs. Here, we review the known stress-dependent transcriptional programs that are activated in cardiomyocytes and cardiac fibroblasts to preserve function in the adult heart after injury. Focus is given to prominent transcriptional pathways such as mechanical stress or reactive oxygen species (ROS)-dependent activation of myocardin-related transcription factors (MRTFs) and transforming growth factor beta (TGFβ), and gene expression that positively regulates protective PI3K/Akt signaling. Together, these pathways modulate both beneficial and pathological responses to cardiac injury in a cell-specific manner.
Collapse
Affiliation(s)
- Cameron S Brand
- Department of Pharmacology, School of Medicine, University of California - San Diego, 9500 Gilman Drive, Biomedical Sciences Building, La Jolla, CA 92093, USA.
| | - Janet K Lighthouse
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Box CVRI, Rochester, NY 14624, USA.
| | - Michael A Trembley
- Department of Cardiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA.
| |
Collapse
|
17
|
Grisanti LA, Thomas TP, Carter RL, de Lucia C, Gao E, Koch WJ, Benovic JL, Tilley DG. Pepducin-mediated cardioprotection via β-arrestin-biased β2-adrenergic receptor-specific signaling. Theranostics 2018; 8:4664-4678. [PMID: 30279730 PMCID: PMC6160776 DOI: 10.7150/thno.26619] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 08/21/2018] [Indexed: 12/20/2022] Open
Abstract
Reperfusion as a therapeutic intervention for acute myocardial infarction-induced cardiac injury itself induces further cardiomyocyte death. β-arrestin (βarr)-biased β-adrenergic receptor (βAR) activation promotes survival signaling responses in vitro; thus, we hypothesize that this pathway can mitigate cardiomyocyte death at the time of reperfusion to better preserve function. However, a lack of efficacious βarr-biased orthosteric small molecules has prevented investigation into whether this pathway relays protection against ischemic injury in vivo. We recently demonstrated that the pepducin ICL1-9, a small lipidated peptide fragment designed from the first intracellular loop of β2AR, allosterically engaged pro-survival signaling cascades in a βarr-dependent manner in vitro. Thus, in this study we tested whether ICL1-9 relays cardioprotection against ischemia/reperfusion (I/R)-induced injury in vivo. Methods: Wild-type (WT) C57BL/6, β2AR knockout (KO), βarr1KO and βarr2KO mice received intracardiac injections of either ICL1-9 or a scrambled control pepducin (Scr) at the time of ischemia (30 min) followed by reperfusion for either 24 h, to assess infarct size and cardiomyocyte death, or 4 weeks, to monitor the impact of ICL1-9 on long-term cardiac structure and function. Neonatal rat ventricular myocytes (NRVM) were used to assess the impact of ICL1-9 versus Scr pepducin on cardiomyocyte survival and mitochondrial superoxide formation in response to either serum deprivation or hypoxia/reoxygenation (H/R) in vitro and to investigate the associated mechanism(s). Results: Intramyocardial injection of ICL1-9 at the time of I/R reduced infarct size, cardiomyocyte death and improved cardiac function in a β2AR- and βarr-dependent manner, which led to improved contractile function early and less fibrotic remodeling over time. Mechanistically, ICL1-9 attenuated mitochondrial superoxide production and promoted cardiomyocyte survival in a RhoA/ROCK-dependent manner. RhoA activation could be detected in cardiomyocytes and whole heart up to 24 h post-treatment, demonstrating the stability of ICL1-9 effects on βarr-dependent β2AR signaling. Conclusion: Pepducin-based allosteric modulation of βarr-dependent β2AR signaling represents a novel therapeutic approach to reduce reperfusion-induced cardiac injury and relay long-term cardiac remodeling benefits.
Collapse
|
18
|
Wang H, Ma S, Li J, Zhao M, Huo X, Sun J, Sun L, Hu J, Liu Q. ADAM17 participates in the protective effect of paeoniflorin on mouse brain microvascular endothelial cells. J Cell Physiol 2018; 233:9320-9329. [DOI: 10.1002/jcp.26308] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 12/01/2017] [Indexed: 12/13/2022]
Affiliation(s)
- Haifang Wang
- Laboratory Center of Shaanxi Provincial People's HospitalXi'anChina
| | - Shuhui Ma
- Department of Clinical Traditional Chinese Medicine‐Western MedicineXi'an JiaoTong University School of MedicineXi'anChina
| | - Jing Li
- Department of Traditional Chinese MedicineShaanxi Provincial People's HospitalXi'anChina
| | - Miaomiao Zhao
- Department of Clinical Traditional Chinese Medicine‐Western MedicineXi'an JiaoTong University School of MedicineXi'anChina
| | - Xueping Huo
- Laboratory Center of Shaanxi Provincial People's HospitalXi'anChina
| | - Jingying Sun
- Laboratory Center of Shaanxi Provincial People's HospitalXi'anChina
| | - Lijun Sun
- Laboratory Center of Shaanxi Provincial People's HospitalXi'anChina
| | - Jun Hu
- Laboratory Center of Shaanxi Provincial People's HospitalXi'anChina
| | - Qinshe Liu
- Medical Experiment Center and Shaanxi Key Laboratory of Integrated Traditional and Western Medicine for Prevention and Treatment of Cardiovascular DiseasesShaanxi University of Chinese MedicineXi'anChina
| |
Collapse
|
19
|
Abstract
G protein-coupled receptors (GPCRs) remain primary therapeutic targets for numerous cardiovascular disorders, including heart failure (HF), because of their influence on cardiac remodeling in response to elevated neurohormone signaling. GPCR blockers have proven to be beneficial in the treatment of HF by reducing chronic G protein activation and cardiac remodeling, thereby extending the lifespan of patients with HF. Unfortunately, this effect does not persist indefinitely, thus next-generation therapeutics aim to selectively block harmful GPCR-mediated pathways while simultaneously promoting beneficial signaling. Transactivation of epidermal growth factor receptor (EGFR) has been shown to be mediated by an expanding repertoire of GPCRs in the heart, and promotes cardiomyocyte survival, thus may offer a new avenue of HF therapeutics. However, GPCR-dependent EGFR transactivation has also been shown to regulate cardiac hypertrophy and fibrosis by different GPCRs and through distinct molecular mechanisms. Here, we discuss the mechanisms and impact of GPCR-mediated EGFR transactivation in the heart, focusing on angiotensin II, urotensin II, and β-adrenergic receptor systems, and highlight areas of research that will help us to determine whether this pathway can be engaged as future therapeutic strategy.
Collapse
|
20
|
Mallet RT, Olivencia-Yurvati AH, Bünger R. Pyruvate enhancement of cardiac performance: Cellular mechanisms and clinical application. Exp Biol Med (Maywood) 2017; 243:198-210. [PMID: 29154687 DOI: 10.1177/1535370217743919] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Cardiac contractile function is adenosine-5'-triphosphate (ATP)-intensive, and the myocardium's high demand for oxygen and energy substrates leaves it acutely vulnerable to interruptions in its blood supply. The myriad cardioprotective properties of the natural intermediary metabolite pyruvate make it a potentially powerful intervention against the complex injury cascade ignited by myocardial ischemia-reperfusion. A readily oxidized metabolic substrate, pyruvate augments myocardial free energy of ATP hydrolysis to a greater extent than the physiological fuels glucose, lactate and fatty acids, particularly when it is provided at supra-physiological plasma concentrations. Pyruvate also exerts antioxidant effects by detoxifying reactive oxygen and nitrogen intermediates, and by increasing nicotinamide adenine dinucleotide phosphate reduced form (NADPH) production to maintain glutathione redox state. These enhancements of free energy and antioxidant defenses combine to augment sarcoplasmic reticular Ca2+ release and re-uptake central to cardiac mechanical performance and to restore β-adrenergic signaling of ischemically stunned myocardium. By minimizing Ca2+ mismanagement and oxidative stress, pyruvate suppresses inflammation in post-ischemic myocardium. Thus, pyruvate administration stabilized cardiac performance, augmented free energy of ATP hydrolysis and glutathione redox systems, and/or quelled inflammation in a porcine model of cardiopulmonary bypass, a canine model of cardiac arrest-resuscitation, and a caprine model of hypovolemia and hindlimb ischemia-reperfusion. Pyruvate's myriad benefits in preclinical models provide the mechanistic framework for its clinical application as metabolic support for myocardium at risk. Phase one trials have demonstrated pyruvate's safety and efficacy for intravenous resuscitation for septic shock, intracoronary infusion for heart failure and as a component of cardioplegia for cardiopulmonary bypass. The favorable outcomes of these trials, which argue for expanded, phase three investigations of pyruvate therapy, mirror findings in isolated, perfused hearts, underscoring the pivotal role of preclinical research in identifying clinical interventions for cardiovascular diseases. Impact statement This article reviews pyruvate's cardioprotective properties as an energy-yielding metabolic fuel, antioxidant and anti-inflammatory agent in mammalian myocardium. Preclinical research has shown these properties make pyruvate a powerful intervention to curb the complex injury cascade ignited by ischemia and reperfusion. In ischemically stunned isolated hearts and in large mammal models of cardiopulmonary bypass, cardiac arrest-resuscitation and hypovolemia, intracoronary pyruvate supports recovery of myocardial contractile function, intracellular Ca2+ homeostasis and free energy of ATP hydrolysis, and its antioxidant actions restore β-adrenergic signaling and suppress inflammation. The first clinical trials of pyruvate for cardiopulmonary bypass, fluid resuscitation and intracoronary intervention for congestive heart failure have been reported. Receiver operating characteristic analyses show remarkable concordance between pyruvate's beneficial functional and metabolic effects in isolated, perfused hearts and in patients recovering from cardiopulmonary bypass in which they received pyruvate- vs. L-lactate-fortified cardioplegia. This research exemplifies the translation of mechanism-oriented preclinical studies to clinical application and outcomes.
Collapse
Affiliation(s)
- Robert T Mallet
- 1 Department of Integrative Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX 76107-2699, USA
| | - Albert H Olivencia-Yurvati
- 1 Department of Integrative Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX 76107-2699, USA.,2 Department of Medical Education, University of North Texas Health Science Center, Fort Worth, TX 76107-2699, USA
| | - Rolf Bünger
- 3 Emeritus Member of the American Physiological Society, McLean, VA 22101, USA
| |
Collapse
|
21
|
Kumar S, Jahangir Alam M, Prabhakar P, Ahmad S, Maulik SK, Sharma M, Goswami SK. Proteomic analysis of the protective effects of aqueous bark extract of Terminalia arjuna (Roxb.) on isoproterenol-induced cardiac hypertrophy in rats. JOURNAL OF ETHNOPHARMACOLOGY 2017; 198:98-108. [PMID: 28063919 DOI: 10.1016/j.jep.2016.12.050] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 12/12/2016] [Accepted: 12/31/2016] [Indexed: 06/06/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Aqueous bark extract of Terminalia arjuna (TA) has been in use as an ethnomedicine for cardiovascular ailments in the Indian subcontinent for centuries. Studies using hemodynamic, ROS scavenging and anti-inflammatory parameters in animal models have shown its anti-atherogenic, hypotensive, inotropic, anti-inflammatory effects. However, details analysis on its effects on established molecular and cell biological markers are a prerequisite for its wider acceptance to the medical community. AIMS OF THE STUDY To test the efficacy of TA extract in ameliorating cardiac hypertrophy induced by ISO in rats. METHODS Cardiac hypertrophy was induced by ISO (5mg/kg/day s.c. for 14 days) in rats and a standardized aqueous extract of TA stem bark was orally administered by gavage. Total RNA and protein were isolated from control, ISO, ISO plus TA and TA treated rat hearts and analyzed for the transcripts for the markers of hypertrophy, signaling kinases, transcription factors and total protein profile. RESULTS TA extract reversed the induction of fetal genes like β-myosin heavy chain, skeletal α-actin and brain natriuretic peptide in hypertrophic rat hearts. While ISO slightly increased the level of phospho-ERK, TA repressed it to about one third of the base line level. Survival kinase Akt, ER stress marker Grp78 and epigenetic regulator HDAC5 were augmented by ISO and TA restored them by various extents. ISO administration moderately increased the transcription factor NFκB binding activity, while coadministration of TA further increased it. AP-1 binding activity was largely unchanged by ISO treatment but it was upregulated when administered along with TA. MEF2D binding activity was increased by ISO and TA restored it to the baseline level. Global proteomic analysis revealed that TA treatment restored a subset of proteins up- and down-regulated in the hypertrophied hearts. Amongst those restored by TA were purinergic receptor X, myosin light chain 3, tropomyosin, and kininogen; suggesting a nodal role of TA in modulating cardiac function. CONCLUSIONS This study for the first time reveals that TA partially or completely restores the marker mRNAs, signaling kinases, transcription factors and total protein profile in rat heart, thereby demonstrating its efficacy in preventing ISO-induced cardiac hypertrophy.
Collapse
Affiliation(s)
- Santosh Kumar
- School of Life Sciences, Jawaharlal Nehru University, New Mehrauli Road, New Delhi 110067, India.
| | - Md Jahangir Alam
- School of Life Sciences, Jawaharlal Nehru University, New Mehrauli Road, New Delhi 110067, India.
| | - Pankaj Prabhakar
- Department of Pharmacology, All India Institute of Medical Sciences (A.I.I.M.S.), Ansari Nagar, 110029, New Delhi, India.
| | - Sayeed Ahmad
- Department of Pharmacognosy and Phytochemistry, Faculty of Pharmacy, Jamia Hamdard, Hamdard Nagar, New Delhi 110062, India.
| | - Subir K Maulik
- Department of Pharmacology, All India Institute of Medical Sciences (A.I.I.M.S.), Ansari Nagar, 110029, New Delhi, India.
| | - Manish Sharma
- Defence Institute of Physiology and Allied Sciences (DIPAS), Defence Research and Development Organization, Lucknow Road, Timarpur, Delhi 110054, India.
| | - Shyamal K Goswami
- School of Life Sciences, Jawaharlal Nehru University, New Mehrauli Road, New Delhi 110067, India.
| |
Collapse
|
22
|
Transactivation of the epidermal growth factor receptor in responses to myocardial stress and cardioprotection. Int J Biochem Cell Biol 2017; 83:97-110. [DOI: 10.1016/j.biocel.2016.12.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 12/25/2016] [Accepted: 12/26/2016] [Indexed: 12/20/2022]
|
23
|
Öztürk T, Vural K, Tuğlu İ, Var A, Kurdal T, Aydemir I. Acute and Chronic Pretreatment With Atenolol Attenuates Intestinal Ischemia and Reperfusion Injury in Hypercholesterolemic Rats. J Cardiothorac Vasc Anesth 2016; 30:985-92. [PMID: 27521968 DOI: 10.1053/j.jvca.2016.03.140] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Indexed: 01/19/2023]
Abstract
OBJECTIVE To evaluate the protective effects of preinjury atenolol (acute v chronic) on apoptosis, contractility, oxidative stress, and inflammatory markers in hypercholesterolemic rats undergoing intestinal ischemia-reperfusion (I/R) injury. DESIGN Prospective, experimental animal study. SETTING University laboratory. PARTICIPANTS Male Wistar rats (n = 32). INTERVENTIONS Rats were divided into the following 4 groups: 1 group was fed a normal diet (ND) (group ND+NoAT [no atenolol]), and the other 3 groups were fed a high-cholesterol diet (HCD)-group HCD+NoAT, group HCD+ChAT (chronic atenolol, 3 mg/kg/day for 8 weeks), and group HCD+AcAT (acute atenolol, 1.5 mg/kg, given 5 minutes before intestinal clamping). All rats underwent I/R injury. The superior mesenteric artery was clamped for 60 minutes, then opened for 120 minutes (reperfusion). Apoptotic cells and stimulated contractions of ileal segments were examined. Tissue markers of intestinal I/R injury were examined. Intestinal malondialdehyde, superoxide dismutase, and nitrate/nitrite levels were measured. MEASUREMENTS AND MAIN RESULTS The chronic atenolol group had fewer apoptotic cells and higher superoxide dismutase activity compared with the other groups. Intestinal contraction was higher in both atenolol pretreatment groups compared with the NoAT groups. Chronic and acute atenolol resulted in lower ileal levels of malondialdehyde and immunolabeling-positive cells (intestinal inducible nitric oxide synthase, endothelial nitric oxide synthase, interleukin-1, and interleukin-8) after I/R injury compared with the no atenolol groups. CONCLUSIONS Both chronic and acute pre-I/R injury treatment with atenolol attenuated I/R injury in this hypercholesterolemic rat model. These findings should encourage future studies of atenolol in hypercholesterolemic patients undergoing procedures with a high risk of intestinal ischemia.
Collapse
Affiliation(s)
- Tülün Öztürk
- Departments of Anaesthesiology and Reanimation, Celal Bayar University, Faculty of Medicine, Uncubozköy, Manisa, Turkey.
| | - Kamil Vural
- Pharmacology, Celal Bayar University, Faculty of Medicine, Uncubozköy, Manisa, Turkey
| | - İbrahim Tuğlu
- Histology, Celal Bayar University, Faculty of Medicine, Uncubozköy, Manisa, Turkey
| | - Ahmet Var
- Biochemistry, Celal Bayar University, Faculty of Medicine, Uncubozköy, Manisa, Turkey
| | - Taner Kurdal
- Cardiovascular Surgery, Celal Bayar University, Faculty of Medicine, Uncubozköy, Manisa, Turkey
| | - Işıl Aydemir
- Pharmacology, Celal Bayar University, Faculty of Medicine, Uncubozköy, Manisa, Turkey; Histology, Celal Bayar University, Faculty of Medicine, Uncubozköy, Manisa, Turkey
| |
Collapse
|
24
|
Grisanti LA, Gumpert AM, Traynham CJ, Gorsky JE, Repas AA, Gao E, Carter RL, Yu D, Calvert JW, García AP, Ibáñez B, Rabinowitz JE, Koch WJ, Tilley DG. Leukocyte-Expressed β2-Adrenergic Receptors Are Essential for Survival After Acute Myocardial Injury. Circulation 2016; 134:153-67. [PMID: 27364164 DOI: 10.1161/circulationaha.116.022304] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 05/17/2016] [Indexed: 11/16/2022]
Abstract
BACKGROUND Immune cell-mediated inflammation is an essential process for mounting a repair response after myocardial infarction (MI). The sympathetic nervous system is known to regulate immune system function through β-adrenergic receptors (βARs); however, their role in regulating immune cell responses to acute cardiac injury is unknown. METHODS Wild-type (WT) mice were irradiated followed by isoform-specific βAR knockout (βARKO) or WT bone-marrow transplantation (BMT) and after full reconstitution underwent MI surgery. Survival was monitored over time, and alterations in immune cell infiltration after MI were examined through immunohistochemistry. Alterations in splenic function were identified through the investigation of altered adhesion receptor expression. RESULTS β2ARKO BMT mice displayed 100% mortality resulting from cardiac rupture within 12 days after MI compared with ≈20% mortality in WT BMT mice. β2ARKO BMT mice displayed severely reduced post-MI cardiac infiltration of leukocytes with reciprocally enhanced splenic retention of the same immune cell populations. Splenic retention of the leukocytes was associated with an increase in vascular cell adhesion molecule-1 expression, which itself was regulated via β-arrestin-dependent β2AR signaling. Furthermore, vascular cell adhesion molecule-1 expression in both mouse and human macrophages was sensitive to β2AR activity, and spleens from human tissue donors treated with β-blocker showed enhanced vascular cell adhesion molecule-1 expression. The impairments in splenic retention and cardiac infiltration of leukocytes after MI were restored to WT levels via lentiviral-mediated re-expression of β2AR in β2ARKO bone marrow before transplantation, which also resulted in post-MI survival rates comparable to those in WT BMT mice. CONCLUSIONS Immune cell-expressed β2AR plays an essential role in regulating the early inflammatory repair response to acute myocardial injury by facilitating cardiac leukocyte infiltration.
Collapse
Affiliation(s)
- Laurel A Grisanti
- From Center for Translational Medicine (L.A.G., A.M.G., C.J.T., J.E.G., A.A.R., E.G., R.L.C., J.E.R., W.J.K., D.G.T.), Department of Pharmacology (E.G., J.E.R., W.J.K., D.G.T.), and Department of Clinical Sciences (D.Y.), Temple University School of Medicine, Philadelphia, PA; Department of Surgery, Division of Cardiothoracic Surgery, Emory University School of Medicine and Carlyle Fraser Heart Center, Atlanta, GA (J.W.C.); and Spanish National Center for Cardiovascular Research, Madrid, Spain (A.P.G., B.I.)
| | - Anna M Gumpert
- From Center for Translational Medicine (L.A.G., A.M.G., C.J.T., J.E.G., A.A.R., E.G., R.L.C., J.E.R., W.J.K., D.G.T.), Department of Pharmacology (E.G., J.E.R., W.J.K., D.G.T.), and Department of Clinical Sciences (D.Y.), Temple University School of Medicine, Philadelphia, PA; Department of Surgery, Division of Cardiothoracic Surgery, Emory University School of Medicine and Carlyle Fraser Heart Center, Atlanta, GA (J.W.C.); and Spanish National Center for Cardiovascular Research, Madrid, Spain (A.P.G., B.I.)
| | - Christopher J Traynham
- From Center for Translational Medicine (L.A.G., A.M.G., C.J.T., J.E.G., A.A.R., E.G., R.L.C., J.E.R., W.J.K., D.G.T.), Department of Pharmacology (E.G., J.E.R., W.J.K., D.G.T.), and Department of Clinical Sciences (D.Y.), Temple University School of Medicine, Philadelphia, PA; Department of Surgery, Division of Cardiothoracic Surgery, Emory University School of Medicine and Carlyle Fraser Heart Center, Atlanta, GA (J.W.C.); and Spanish National Center for Cardiovascular Research, Madrid, Spain (A.P.G., B.I.)
| | - Joshua E Gorsky
- From Center for Translational Medicine (L.A.G., A.M.G., C.J.T., J.E.G., A.A.R., E.G., R.L.C., J.E.R., W.J.K., D.G.T.), Department of Pharmacology (E.G., J.E.R., W.J.K., D.G.T.), and Department of Clinical Sciences (D.Y.), Temple University School of Medicine, Philadelphia, PA; Department of Surgery, Division of Cardiothoracic Surgery, Emory University School of Medicine and Carlyle Fraser Heart Center, Atlanta, GA (J.W.C.); and Spanish National Center for Cardiovascular Research, Madrid, Spain (A.P.G., B.I.)
| | - Ashley A Repas
- From Center for Translational Medicine (L.A.G., A.M.G., C.J.T., J.E.G., A.A.R., E.G., R.L.C., J.E.R., W.J.K., D.G.T.), Department of Pharmacology (E.G., J.E.R., W.J.K., D.G.T.), and Department of Clinical Sciences (D.Y.), Temple University School of Medicine, Philadelphia, PA; Department of Surgery, Division of Cardiothoracic Surgery, Emory University School of Medicine and Carlyle Fraser Heart Center, Atlanta, GA (J.W.C.); and Spanish National Center for Cardiovascular Research, Madrid, Spain (A.P.G., B.I.)
| | - Erhe Gao
- From Center for Translational Medicine (L.A.G., A.M.G., C.J.T., J.E.G., A.A.R., E.G., R.L.C., J.E.R., W.J.K., D.G.T.), Department of Pharmacology (E.G., J.E.R., W.J.K., D.G.T.), and Department of Clinical Sciences (D.Y.), Temple University School of Medicine, Philadelphia, PA; Department of Surgery, Division of Cardiothoracic Surgery, Emory University School of Medicine and Carlyle Fraser Heart Center, Atlanta, GA (J.W.C.); and Spanish National Center for Cardiovascular Research, Madrid, Spain (A.P.G., B.I.)
| | - Rhonda L Carter
- From Center for Translational Medicine (L.A.G., A.M.G., C.J.T., J.E.G., A.A.R., E.G., R.L.C., J.E.R., W.J.K., D.G.T.), Department of Pharmacology (E.G., J.E.R., W.J.K., D.G.T.), and Department of Clinical Sciences (D.Y.), Temple University School of Medicine, Philadelphia, PA; Department of Surgery, Division of Cardiothoracic Surgery, Emory University School of Medicine and Carlyle Fraser Heart Center, Atlanta, GA (J.W.C.); and Spanish National Center for Cardiovascular Research, Madrid, Spain (A.P.G., B.I.)
| | - Daohai Yu
- From Center for Translational Medicine (L.A.G., A.M.G., C.J.T., J.E.G., A.A.R., E.G., R.L.C., J.E.R., W.J.K., D.G.T.), Department of Pharmacology (E.G., J.E.R., W.J.K., D.G.T.), and Department of Clinical Sciences (D.Y.), Temple University School of Medicine, Philadelphia, PA; Department of Surgery, Division of Cardiothoracic Surgery, Emory University School of Medicine and Carlyle Fraser Heart Center, Atlanta, GA (J.W.C.); and Spanish National Center for Cardiovascular Research, Madrid, Spain (A.P.G., B.I.)
| | - John W Calvert
- From Center for Translational Medicine (L.A.G., A.M.G., C.J.T., J.E.G., A.A.R., E.G., R.L.C., J.E.R., W.J.K., D.G.T.), Department of Pharmacology (E.G., J.E.R., W.J.K., D.G.T.), and Department of Clinical Sciences (D.Y.), Temple University School of Medicine, Philadelphia, PA; Department of Surgery, Division of Cardiothoracic Surgery, Emory University School of Medicine and Carlyle Fraser Heart Center, Atlanta, GA (J.W.C.); and Spanish National Center for Cardiovascular Research, Madrid, Spain (A.P.G., B.I.)
| | - Andrés Pun García
- From Center for Translational Medicine (L.A.G., A.M.G., C.J.T., J.E.G., A.A.R., E.G., R.L.C., J.E.R., W.J.K., D.G.T.), Department of Pharmacology (E.G., J.E.R., W.J.K., D.G.T.), and Department of Clinical Sciences (D.Y.), Temple University School of Medicine, Philadelphia, PA; Department of Surgery, Division of Cardiothoracic Surgery, Emory University School of Medicine and Carlyle Fraser Heart Center, Atlanta, GA (J.W.C.); and Spanish National Center for Cardiovascular Research, Madrid, Spain (A.P.G., B.I.)
| | - Borja Ibáñez
- From Center for Translational Medicine (L.A.G., A.M.G., C.J.T., J.E.G., A.A.R., E.G., R.L.C., J.E.R., W.J.K., D.G.T.), Department of Pharmacology (E.G., J.E.R., W.J.K., D.G.T.), and Department of Clinical Sciences (D.Y.), Temple University School of Medicine, Philadelphia, PA; Department of Surgery, Division of Cardiothoracic Surgery, Emory University School of Medicine and Carlyle Fraser Heart Center, Atlanta, GA (J.W.C.); and Spanish National Center for Cardiovascular Research, Madrid, Spain (A.P.G., B.I.)
| | - Joseph E Rabinowitz
- From Center for Translational Medicine (L.A.G., A.M.G., C.J.T., J.E.G., A.A.R., E.G., R.L.C., J.E.R., W.J.K., D.G.T.), Department of Pharmacology (E.G., J.E.R., W.J.K., D.G.T.), and Department of Clinical Sciences (D.Y.), Temple University School of Medicine, Philadelphia, PA; Department of Surgery, Division of Cardiothoracic Surgery, Emory University School of Medicine and Carlyle Fraser Heart Center, Atlanta, GA (J.W.C.); and Spanish National Center for Cardiovascular Research, Madrid, Spain (A.P.G., B.I.)
| | - Walter J Koch
- From Center for Translational Medicine (L.A.G., A.M.G., C.J.T., J.E.G., A.A.R., E.G., R.L.C., J.E.R., W.J.K., D.G.T.), Department of Pharmacology (E.G., J.E.R., W.J.K., D.G.T.), and Department of Clinical Sciences (D.Y.), Temple University School of Medicine, Philadelphia, PA; Department of Surgery, Division of Cardiothoracic Surgery, Emory University School of Medicine and Carlyle Fraser Heart Center, Atlanta, GA (J.W.C.); and Spanish National Center for Cardiovascular Research, Madrid, Spain (A.P.G., B.I.)
| | - Douglas G Tilley
- From Center for Translational Medicine (L.A.G., A.M.G., C.J.T., J.E.G., A.A.R., E.G., R.L.C., J.E.R., W.J.K., D.G.T.), Department of Pharmacology (E.G., J.E.R., W.J.K., D.G.T.), and Department of Clinical Sciences (D.Y.), Temple University School of Medicine, Philadelphia, PA; Department of Surgery, Division of Cardiothoracic Surgery, Emory University School of Medicine and Carlyle Fraser Heart Center, Atlanta, GA (J.W.C.); and Spanish National Center for Cardiovascular Research, Madrid, Spain (A.P.G., B.I.).
| |
Collapse
|
25
|
Feldman AM, Gordon J, Wang J, Song J, Zhang XQ, Myers VD, Tilley DG, Gao E, Hoffman NE, Tomar D, Madesh M, Rabinowitz J, Koch WJ, Su F, Khalili K, Cheung JY. BAG3 regulates contractility and Ca(2+) homeostasis in adult mouse ventricular myocytes. J Mol Cell Cardiol 2016; 92:10-20. [PMID: 26796036 PMCID: PMC4789075 DOI: 10.1016/j.yjmcc.2016.01.015] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Revised: 01/14/2016] [Accepted: 01/17/2016] [Indexed: 12/22/2022]
Abstract
Bcl2-associated athanogene 3 (BAG3) is a 575 amino acid anti-apoptotic protein that is constitutively expressed in the heart. BAG3 mutations, including mutations leading to loss of protein, are associated with familial cardiomyopathy. Furthermore, BAG3 levels have been found to be reduced in end-stage non-familial failing myocardium. In contrast to neonatal myocytes in which BAG3 is found in the cytoplasm and involved in protein quality control and apoptosis, in adult mouse left ventricular (LV) myocytes BAG3 co-localized with Na(+)-K(+)-ATPase and L-type Ca(2+) channels in the sarcolemma and t-tubules. BAG3 co-immunoprecipitated with β1-adrenergic receptor, L-type Ca(2+) channels and phospholemman. To simulate decreased BAG3 protein levels observed in human heart failure, we targeted BAG3 by shRNA (shBAG3) in adult LV myocytes. Reducing BAG3 by 55% resulted in reduced contraction and [Ca(2+)]i transient amplitudes in LV myocytes stimulated with isoproterenol. L-type Ca(2+) current (ICa) and sarcoplasmic reticulum (SR) Ca(2+) content but not Na(+)/Ca(2+) exchange current (INaCa) or SR Ca(2+) uptake were reduced in isoproterenol-treated shBAG3 myocytes. Forskolin or dibutyryl cAMP restored ICa amplitude in shBAG3 myocytes to that observed in WT myocytes, consistent with BAG3 having effects upstream and at the level of the receptor. Resting membrane potential and action potential amplitude were unaffected but APD50 and APD90 were prolonged in shBAG3 myocytes. Protein levels of Ca(2+) entry molecules and other important excitation-contraction proteins were unchanged in myocytes with lower BAG3. Our findings that BAG3 is localized at the sarcolemma and t-tubules while modulating myocyte contraction and action potential duration through specific interaction with the β1-adrenergic receptor and L-type Ca(2+) channel provide novel insight into the role of BAG3 in cardiomyopathies and increased arrhythmia risks in heart failure.
Collapse
MESH Headings
- Action Potentials/drug effects
- Adaptor Proteins, Signal Transducing/biosynthesis
- Adaptor Proteins, Signal Transducing/genetics
- Animals
- Apoptosis Regulatory Proteins/biosynthesis
- Apoptosis Regulatory Proteins/genetics
- Arrhythmias, Cardiac/genetics
- Arrhythmias, Cardiac/metabolism
- Arrhythmias, Cardiac/pathology
- Calcium/metabolism
- Calcium Channels, L-Type/metabolism
- Cardiomyopathy, Dilated/genetics
- Cardiomyopathy, Dilated/metabolism
- Cardiomyopathy, Dilated/pathology
- Excitation Contraction Coupling
- Heart Failure/genetics
- Heart Failure/metabolism
- Heart Failure/pathology
- Heart Ventricles/metabolism
- Heart Ventricles/pathology
- Homeostasis
- Humans
- Isoproterenol/administration & dosage
- Membrane Proteins/metabolism
- Mice
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Phosphoproteins/metabolism
- RNA, Small Interfering/genetics
- Receptors, Adrenergic, beta-1/metabolism
- Sarcolemma/metabolism
- Sodium-Potassium-Exchanging ATPase/metabolism
Collapse
Affiliation(s)
- Arthur M Feldman
- Department of Medicine, Temple University School of Medicine, Philadelphia, PA 19140, USA; Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Jennifer Gordon
- Comprehensive NeuroAIDS Center, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - JuFang Wang
- Center of Translational Medicine, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Jianliang Song
- Center of Translational Medicine, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Xue-Qian Zhang
- Center of Translational Medicine, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Valerie D Myers
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Douglas G Tilley
- Center of Translational Medicine, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Erhe Gao
- Center of Translational Medicine, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Nicholas E Hoffman
- Center of Translational Medicine, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Dhanendra Tomar
- Center of Translational Medicine, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Muniswamy Madesh
- Center of Translational Medicine, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Joseph Rabinowitz
- Center of Translational Medicine, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Walter J Koch
- Center of Translational Medicine, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Feifei Su
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA 19140, USA; Department of Cardiology, Tangdu Hospital, the Fourth Military Medical University, Xi'an, China
| | - Kamel Khalili
- Comprehensive NeuroAIDS Center, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Joseph Y Cheung
- Center of Translational Medicine, Temple University School of Medicine, Philadelphia, PA 19140, USA; Department of Medicine, Temple University School of Medicine, Philadelphia, PA 19140, USA.
| |
Collapse
|
26
|
Sysa-Shah P, Tocchetti CG, Gupta M, Rainer PP, Shen X, Kang BH, Belmonte F, Li J, Xu Y, Guo X, Bedja D, Gao WD, Paolocci N, Rath R, Sawyer DB, Naga Prasad SV, Gabrielson K. Bidirectional cross-regulation between ErbB2 and β-adrenergic signalling pathways. Cardiovasc Res 2015; 109:358-73. [PMID: 26692570 DOI: 10.1093/cvr/cvv274] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 12/01/2015] [Indexed: 12/31/2022] Open
Abstract
AIMS Despite the observation that ErbB2 regulates sensitivity of the heart to doxorubicin or ErbB2-targeted cancer therapies, mechanisms that regulate ErbB2 expression and activity have not been studied. Since isoproterenol up-regulates ErbB2 in kidney and salivary glands and β2AR and ErbB2 complex in brain and heart, we hypothesized that β-adrenergic receptors (AR) modulate ErbB2 signalling status. METHODS AND RESULTS ErbB2 transfection of HEK293 cells up-regulates β2AR, and β2AR transfection of HEK293 up-regulates ErbB2. Interestingly, cardiomyocytes isolated from myocyte-specific ErbB2-overexpressing (ErbB2(tg)) mice have amplified response to selective β2-agonist zinterol, and right ventricular trabeculae baseline force generation is markedly reduced with β2-antagonist ICI-118 551. Consistently, receptor binding assays and western blotting demonstrate that β2ARs levels are markedly increased in ErbB2(tg) myocardium and reduced by EGFR/ErbB2 inhibitor, lapatinib. Intriguingly, acute treatment of mice with β1- and β2-AR agonist isoproterenol resulted in myocardial ErbB2 increase, while inhibition with either β1- or β2-AR antagonist did not completely prevent isoproterenol-induced ErbB2 expression. Furthermore, inhibition of ErbB2 kinase predisposed mice hearts to injury from chronic isoproterenol treatment while significantly reducing isoproterenol-induced pAKT and pERK levels, suggesting ErbB2's role in transactivation in the heart. CONCLUSION Our studies show that myocardial ErbB2 and βAR signalling are linked in a feedback loop with βAR activation leading to increased ErbB2 expression and activity, and increased ErbB2 activity regulating β2AR expression. Most importantly, ErbB2 kinase activity is crucial for cardioprotection in the setting of β-adrenergic stress, suggesting that this mechanism is important in the pathophysiology and treatment of cardiomyopathy induced by ErbB2-targeting antineoplastic drugs.
Collapse
Affiliation(s)
- Polina Sysa-Shah
- Department of Molecular and Comparative Pathobiology, Johns Hopkins Medical Institutions, MRB 807, 733 N. Broadway, Baltimore, MD 21205, USA
| | - Carlo G Tocchetti
- Division of Internal Medicine, Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Manveen Gupta
- Department of Molecular Cardiology, Cleveland Clinic, Lerner Research Institute, Cleveland, OH, USA
| | - Peter P Rainer
- Division of Cardiology, Department of Medicine, Medical University of Graz, Graz, Austria Division of Cardiology, Department of Medicine, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Xiaoxu Shen
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Byung-Hak Kang
- Department of Oncology, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Frances Belmonte
- Department of Molecular and Comparative Pathobiology, Johns Hopkins Medical Institutions, MRB 807, 733 N. Broadway, Baltimore, MD 21205, USA
| | - Jian Li
- Clinical Laboratory, Chinese PLA General Hospital, Beijing, China
| | - Yi Xu
- Department of Molecular and Comparative Pathobiology, Johns Hopkins Medical Institutions, MRB 807, 733 N. Broadway, Baltimore, MD 21205, USA
| | - Xin Guo
- Department of Molecular and Comparative Pathobiology, Johns Hopkins Medical Institutions, MRB 807, 733 N. Broadway, Baltimore, MD 21205, USA
| | - Djahida Bedja
- Department of Molecular and Comparative Pathobiology, Johns Hopkins Medical Institutions, MRB 807, 733 N. Broadway, Baltimore, MD 21205, USA
| | - Wei Dong Gao
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Nazareno Paolocci
- Division of Cardiology, Department of Medicine, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Rutwik Rath
- Cardiovascular Services, Maine Medical Center, Portland, ME, USA
| | - Douglas B Sawyer
- Cardiovascular Services, Maine Medical Center, Portland, ME, USA
| | | | - Kathleen Gabrielson
- Department of Molecular and Comparative Pathobiology, Johns Hopkins Medical Institutions, MRB 807, 733 N. Broadway, Baltimore, MD 21205, USA
| |
Collapse
|
27
|
Jung G, Fajardo G, Ribeiro AJS, Kooiker KB, Coronado M, Zhao M, Hu DQ, Reddy S, Kodo K, Sriram K, Insel PA, Wu JC, Pruitt BL, Bernstein D. Time-dependent evolution of functional vs. remodeling signaling in induced pluripotent stem cell-derived cardiomyocytes and induced maturation with biomechanical stimulation. FASEB J 2015; 30:1464-79. [PMID: 26675706 DOI: 10.1096/fj.15-280982] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 11/30/2015] [Indexed: 01/13/2023]
Abstract
Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) are a powerful platform for uncovering disease mechanisms and assessing drugs for efficacy/toxicity. However, the accuracy with which hiPSC-CMs recapitulate the contractile and remodeling signaling of adult cardiomyocytes is not fully known. We used β-adrenergic receptor (β-AR) signaling as a prototype to determine the evolution of signaling component expression and function during hiPSC-CM maturation. In "early" hiPSC-CMs (less than or equal to d 30), β2-ARs are a primary source of cAMP/PKA signaling. With longer culture, β1-AR signaling increases: from 0% of cAMP generation at d 30 to 56.8 ± 6.6% by d 60. PKA signaling shows a similar increase: 15.7 ± 5.2% (d 30), 49.8 ± 0.5% (d 60), and 71.0 ± 6.1% (d 90). cAMP generation increases 9-fold from d 30 to 60, with enhanced coupling to remodeling pathways (e.g., Akt and Ca(2+)/calmodulin-dependent protein kinase type II) and development of caveolin-mediated signaling compartmentalization. By contrast, cardiotoxicity induced by chronic β-AR stimulation, a major component of heart failure, develops much later: 5% cell death at d 30vs 55% at d 90. Moreover, β-AR maturation can be accelerated by biomechanical stimulation. The differential maturation of β-AR functionalvs remodeling signaling in hiPSC-CMs has important implications for their use in disease modeling and drug testing. We propose that assessment of signaling be added to the indices of phenotypic maturation of hiPSC-CMs.-Jung, G., Fajardo, G., Ribeiro, A. J. S., Kooiker, K. B., Coronado, M., Zhao, M., Hu, D.-Q., Reddy, S., Kodo, K., Sriram, K., Insel, P. A., Wu, J. C., Pruitt, B. L., Bernstein, D. Time-dependent evolution of functionalvs remodeling signaling in induced pluripotent stem cell-derived cardiomyocytes and induced maturation with biomechanical stimulation.
Collapse
Affiliation(s)
- Gwanghyun Jung
- *Division of Cardiology, Department of Pediatrics, Stanford Cardiovascular Institute, Division of Cardiovascular Medicine, Department of Medicine, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA; Department of Mechanical Engineering, Stanford University School of Engineering, Stanford, California, USA; and Departments of Pharmacology and Medicine, University of California, San Diego, San Diego, California, USA
| | - Giovanni Fajardo
- *Division of Cardiology, Department of Pediatrics, Stanford Cardiovascular Institute, Division of Cardiovascular Medicine, Department of Medicine, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA; Department of Mechanical Engineering, Stanford University School of Engineering, Stanford, California, USA; and Departments of Pharmacology and Medicine, University of California, San Diego, San Diego, California, USA
| | - Alexandre J S Ribeiro
- *Division of Cardiology, Department of Pediatrics, Stanford Cardiovascular Institute, Division of Cardiovascular Medicine, Department of Medicine, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA; Department of Mechanical Engineering, Stanford University School of Engineering, Stanford, California, USA; and Departments of Pharmacology and Medicine, University of California, San Diego, San Diego, California, USA
| | - Kristina Bezold Kooiker
- *Division of Cardiology, Department of Pediatrics, Stanford Cardiovascular Institute, Division of Cardiovascular Medicine, Department of Medicine, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA; Department of Mechanical Engineering, Stanford University School of Engineering, Stanford, California, USA; and Departments of Pharmacology and Medicine, University of California, San Diego, San Diego, California, USA
| | - Michael Coronado
- *Division of Cardiology, Department of Pediatrics, Stanford Cardiovascular Institute, Division of Cardiovascular Medicine, Department of Medicine, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA; Department of Mechanical Engineering, Stanford University School of Engineering, Stanford, California, USA; and Departments of Pharmacology and Medicine, University of California, San Diego, San Diego, California, USA
| | - Mingming Zhao
- *Division of Cardiology, Department of Pediatrics, Stanford Cardiovascular Institute, Division of Cardiovascular Medicine, Department of Medicine, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA; Department of Mechanical Engineering, Stanford University School of Engineering, Stanford, California, USA; and Departments of Pharmacology and Medicine, University of California, San Diego, San Diego, California, USA
| | - Dong-Qing Hu
- *Division of Cardiology, Department of Pediatrics, Stanford Cardiovascular Institute, Division of Cardiovascular Medicine, Department of Medicine, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA; Department of Mechanical Engineering, Stanford University School of Engineering, Stanford, California, USA; and Departments of Pharmacology and Medicine, University of California, San Diego, San Diego, California, USA
| | - Sushma Reddy
- *Division of Cardiology, Department of Pediatrics, Stanford Cardiovascular Institute, Division of Cardiovascular Medicine, Department of Medicine, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA; Department of Mechanical Engineering, Stanford University School of Engineering, Stanford, California, USA; and Departments of Pharmacology and Medicine, University of California, San Diego, San Diego, California, USA
| | - Kazuki Kodo
- *Division of Cardiology, Department of Pediatrics, Stanford Cardiovascular Institute, Division of Cardiovascular Medicine, Department of Medicine, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA; Department of Mechanical Engineering, Stanford University School of Engineering, Stanford, California, USA; and Departments of Pharmacology and Medicine, University of California, San Diego, San Diego, California, USA
| | - Krishna Sriram
- *Division of Cardiology, Department of Pediatrics, Stanford Cardiovascular Institute, Division of Cardiovascular Medicine, Department of Medicine, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA; Department of Mechanical Engineering, Stanford University School of Engineering, Stanford, California, USA; and Departments of Pharmacology and Medicine, University of California, San Diego, San Diego, California, USA
| | - Paul A Insel
- *Division of Cardiology, Department of Pediatrics, Stanford Cardiovascular Institute, Division of Cardiovascular Medicine, Department of Medicine, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA; Department of Mechanical Engineering, Stanford University School of Engineering, Stanford, California, USA; and Departments of Pharmacology and Medicine, University of California, San Diego, San Diego, California, USA
| | - Joseph C Wu
- *Division of Cardiology, Department of Pediatrics, Stanford Cardiovascular Institute, Division of Cardiovascular Medicine, Department of Medicine, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA; Department of Mechanical Engineering, Stanford University School of Engineering, Stanford, California, USA; and Departments of Pharmacology and Medicine, University of California, San Diego, San Diego, California, USA
| | - Beth L Pruitt
- *Division of Cardiology, Department of Pediatrics, Stanford Cardiovascular Institute, Division of Cardiovascular Medicine, Department of Medicine, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA; Department of Mechanical Engineering, Stanford University School of Engineering, Stanford, California, USA; and Departments of Pharmacology and Medicine, University of California, San Diego, San Diego, California, USA
| | - Daniel Bernstein
- *Division of Cardiology, Department of Pediatrics, Stanford Cardiovascular Institute, Division of Cardiovascular Medicine, Department of Medicine, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA; Department of Mechanical Engineering, Stanford University School of Engineering, Stanford, California, USA; and Departments of Pharmacology and Medicine, University of California, San Diego, San Diego, California, USA
| |
Collapse
|
28
|
Forrester SJ, Kawai T, O'Brien S, Thomas W, Harris RC, Eguchi S. Epidermal Growth Factor Receptor Transactivation: Mechanisms, Pathophysiology, and Potential Therapies in the Cardiovascular System. Annu Rev Pharmacol Toxicol 2015; 56:627-53. [PMID: 26566153 DOI: 10.1146/annurev-pharmtox-070115-095427] [Citation(s) in RCA: 111] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Epidermal growth factor receptor (EGFR) activation impacts the physiology and pathophysiology of the cardiovascular system, and inhibition of EGFR activity is emerging as a potential therapeutic strategy to treat diseases including hypertension, cardiac hypertrophy, renal fibrosis, and abdominal aortic aneurysm. The capacity of G protein-coupled receptor (GPCR) agonists, such as angiotensin II (AngII), to promote EGFR signaling is called transactivation and is well described, yet delineating the molecular processes and functional relevance of this crosstalk has been challenging. Moreover, these critical findings are dispersed among many different fields. The aim of our review is to highlight recent advancements in defining the signaling cascades and downstream consequences of EGFR transactivation in the cardiovascular renal system. We also focus on studies that link EGFR transactivation to animal models of the disease, and we discuss potential therapeutic applications.
Collapse
Affiliation(s)
- Steven J Forrester
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania 19140;
| | - Tatsuo Kawai
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania 19140;
| | - Shannon O'Brien
- The School of Biomedical Sciences, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Walter Thomas
- The School of Biomedical Sciences, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Raymond C Harris
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee 37232
| | - Satoru Eguchi
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania 19140;
| |
Collapse
|
29
|
De Giusti VC, Orlowski A, Ciancio MC, Espejo MS, Gonano LA, Caldiz CI, Vila Petroff MG, Villa-Abrille MC, Aiello EA. Aldosterone stimulates the cardiac sodium/bicarbonate cotransporter via activation of the g protein-coupled receptor gpr30. J Mol Cell Cardiol 2015; 89:260-7. [PMID: 26497404 DOI: 10.1016/j.yjmcc.2015.10.024] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 10/07/2015] [Accepted: 10/20/2015] [Indexed: 12/22/2022]
Abstract
Some cardiac non-genomic effects of aldosterone (Ald) are reported to be mediated through activation of the classic mineralocorticoid receptor (MR). However, in the last years, it was proposed that activation of the novel G protein-coupled receptor GPR30 mediates certain non-genomic effects of Ald. The aim of this study was to elucidate if the sodium/bicarbonate cotransporter (NBC) is stimulated by Ald and if the activation of GPR30 mediates this effect. NBC activity was evaluated in rat cardiomyocytes perfused with HCO3(-)/CO2 solution in the continuous presence of HOE642 (sodium/hydrogen exchanger blocker) during recovery from acidosis using intracellular fluorescence measurements. Ald enhanced NBC activity (% of ΔJHCO3(-); control: 100±5.82%, n=7 vs Ald: 151.88±11.02%, n=5; P<0.05), which was prevented by G15 (GPR30 blocker, 90.53±7.81%, n=7). Further evidence for the involvement of GPR30 was provided by G1 (GPR30 agonist), which stimulated NBC (185.13±18.28%, n=6; P<0.05) and this effect was abrogated by G15 (124.19±10.96%, n=5). Ald- and G1-induced NBC stimulation was abolished by the reactive oxygen species (ROS) scavenger MPG and by the NADPH oxidase inhibitor apocynin. In addition, G15 prevented Ald- and G1-induced ROS production. Pre-incubation of myocytes with wortmannin (PI3K-AKT pathway blocker) prevented Ald- or G1-induced NBC stimulation. In summary, Ald stimulates NBC by GPR30 activation, ROS production and AKT stimulation.
Collapse
Affiliation(s)
- Verónica C De Giusti
- Centro de Investigaciones Cardiovasculares, Facultad de Ciencias Médicas, Universidad Nacional de La Plata-CONICET, La Plata, Argentina
| | - Alejandro Orlowski
- Centro de Investigaciones Cardiovasculares, Facultad de Ciencias Médicas, Universidad Nacional de La Plata-CONICET, La Plata, Argentina
| | - María C Ciancio
- Centro de Investigaciones Cardiovasculares, Facultad de Ciencias Médicas, Universidad Nacional de La Plata-CONICET, La Plata, Argentina
| | - María S Espejo
- Centro de Investigaciones Cardiovasculares, Facultad de Ciencias Médicas, Universidad Nacional de La Plata-CONICET, La Plata, Argentina
| | - Luis A Gonano
- Centro de Investigaciones Cardiovasculares, Facultad de Ciencias Médicas, Universidad Nacional de La Plata-CONICET, La Plata, Argentina
| | - Claudia I Caldiz
- Centro de Investigaciones Cardiovasculares, Facultad de Ciencias Médicas, Universidad Nacional de La Plata-CONICET, La Plata, Argentina
| | - Martín G Vila Petroff
- Centro de Investigaciones Cardiovasculares, Facultad de Ciencias Médicas, Universidad Nacional de La Plata-CONICET, La Plata, Argentina
| | - María C Villa-Abrille
- Centro de Investigaciones Cardiovasculares, Facultad de Ciencias Médicas, Universidad Nacional de La Plata-CONICET, La Plata, Argentina
| | - Ernesto A Aiello
- Centro de Investigaciones Cardiovasculares, Facultad de Ciencias Médicas, Universidad Nacional de La Plata-CONICET, La Plata, Argentina
| |
Collapse
|
30
|
LIU MIHUA, LIN XIAOLONG, ZHANG YUAN, HE JUN, TAN TIANPING, WU SHAOJIAN, LIU JUN, TIAN WEI, CHEN LI, YU SHAN, LI JIAN, YUAN CONG. Hydrogen sulfide attenuates doxorubicin-induced cardiotoxicity by inhibiting reactive oxygen species-activated extracellular signal-regulated kinase 1/2 in H9c2 cardiac myocytes. Mol Med Rep 2015; 12:6841-8. [DOI: 10.3892/mmr.2015.4234] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Accepted: 07/21/2015] [Indexed: 11/06/2022] Open
|
31
|
Orphan Nuclear Receptor Nur77 Inhibits Cardiac Hypertrophic Response to Beta-Adrenergic Stimulation. Mol Cell Biol 2015. [PMID: 26195821 DOI: 10.1128/mcb.00229-15] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The orphan nuclear receptor Nur77 plays critical roles in cardiovascular diseases, and its expression is markedly induced in the heart after beta-adrenergic receptor (β-AR) activation. However, the functional significance of Nur77 in β-AR signaling in the heart remains unclear. By using Northern blot, Western blot, and immunofluorescent staining assays, we showed that Nur77 expression was markedly upregulated in cardiomyocytes in response to multiple hypertrophic stimuli, including isoproterenol (ISO), phenylephrine (PE), and endothelin-1 (ET-1). In a time- and dose-dependent manner, ISO increases Nur77 expression in the nuclei of cardiomyocytes. Overexpression of Nur77 markedly inhibited ISO-induced cardiac hypertrophy by inducing nuclear translocation of Nur77 in cardiomyocytes. Furthermore, cardiac overexpression of Nur77 by intramyocardial injection of Ad-Nur77 substantially inhibited cardiac hypertrophy and ameliorated cardiac dysfunction after chronic infusion of ISO in mice. Mechanistically, we demonstrated that Nur77 functionally interacts with NFATc3 and GATA4 and inhibits their transcriptional activities, which are critical for the development of cardiac hypertrophy. These results demonstrate for the first time that Nur77 is a novel negative regulator for the β-AR-induced cardiac hypertrophy through inhibiting the NFATc3 and GATA4 transcriptional pathways. Targeting Nur77 may represent a potentially novel therapeutic strategy for preventing cardiac hypertrophy and heart failure.
Collapse
|
32
|
Takayanagi T, Kawai T, Forrester SJ, Obama T, Tsuji T, Fukuda Y, Elliott KJ, Tilley DG, Davisson RL, Park JY, Eguchi S. Role of epidermal growth factor receptor and endoplasmic reticulum stress in vascular remodeling induced by angiotensin II. Hypertension 2015; 65:1349-55. [PMID: 25916723 DOI: 10.1161/hypertensionaha.115.05344] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 04/01/2015] [Indexed: 12/18/2022]
Abstract
The mechanisms by which angiotensin II (AngII) elevates blood pressure and enhances end-organ damage seem to be distinct. However, the signal transduction cascade by which AngII specifically mediates vascular remodeling such as medial hypertrophy and perivascular fibrosis remains incomplete. We have previously shown that AngII-induced epidermal growth factor receptor (EGFR) transactivation is mediated by disintegrin and metalloproteinase domain 17 (ADAM17), and that this signaling is required for vascular smooth muscle cell hypertrophy but not for contractile signaling in response to AngII. Recent studies have implicated endoplasmic reticulum (ER) stress in hypertension. Interestingly, EGFR is capable of inducing ER stress. The aim of this study was to test the hypothesis that activation of EGFR and ER stress are critical components required for vascular remodeling but not hypertension induced by AngII. Mice were infused with AngII for 2 weeks with or without treatment of EGFR inhibitor, erlotinib, or ER chaperone, 4-phenylbutyrate. AngII infusion induced vascular medial hypertrophy in the heart, kidney and aorta, and perivascular fibrosis in heart and kidney, cardiac hypertrophy, and hypertension. Treatment with erlotinib as well as 4-phenylbutyrate attenuated vascular remodeling and cardiac hypertrophy but not hypertension. In addition, AngII infusion enhanced ADAM17 expression, EGFR activation, and ER/oxidative stress in the vasculature, which were diminished in both erlotinib-treated and 4-phenylbutyrate-treated mice. ADAM17 induction and EGFR activation by AngII in vascular cells were also prevented by inhibition of EGFR or ER stress. In conclusion, AngII induces vascular remodeling by EGFR activation and ER stress via a signaling mechanism involving ADAM17 induction independent of hypertension.
Collapse
Affiliation(s)
- Takehiko Takayanagi
- From the Department of Physiology, Cardiovascular Research Center (T. Takayanagi, T.K., S.J.F., T.O., T. Tsuji, Y.F., K.J.E., J.-Y.P., S.E.) and Department of Pharmacology, Center for Translational Medicine (D.G.T.), Temple University School of Medicine, Philadelphia, PA; Department of Kinesiology, Temple University College of Public Health, Philadelphia, PA (S.J.F., J.-Y.P.); and Department of Biomedical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY (R.L.D.)
| | - Tatsuo Kawai
- From the Department of Physiology, Cardiovascular Research Center (T. Takayanagi, T.K., S.J.F., T.O., T. Tsuji, Y.F., K.J.E., J.-Y.P., S.E.) and Department of Pharmacology, Center for Translational Medicine (D.G.T.), Temple University School of Medicine, Philadelphia, PA; Department of Kinesiology, Temple University College of Public Health, Philadelphia, PA (S.J.F., J.-Y.P.); and Department of Biomedical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY (R.L.D.)
| | - Steven J Forrester
- From the Department of Physiology, Cardiovascular Research Center (T. Takayanagi, T.K., S.J.F., T.O., T. Tsuji, Y.F., K.J.E., J.-Y.P., S.E.) and Department of Pharmacology, Center for Translational Medicine (D.G.T.), Temple University School of Medicine, Philadelphia, PA; Department of Kinesiology, Temple University College of Public Health, Philadelphia, PA (S.J.F., J.-Y.P.); and Department of Biomedical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY (R.L.D.)
| | - Takashi Obama
- From the Department of Physiology, Cardiovascular Research Center (T. Takayanagi, T.K., S.J.F., T.O., T. Tsuji, Y.F., K.J.E., J.-Y.P., S.E.) and Department of Pharmacology, Center for Translational Medicine (D.G.T.), Temple University School of Medicine, Philadelphia, PA; Department of Kinesiology, Temple University College of Public Health, Philadelphia, PA (S.J.F., J.-Y.P.); and Department of Biomedical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY (R.L.D.)
| | - Toshiyuki Tsuji
- From the Department of Physiology, Cardiovascular Research Center (T. Takayanagi, T.K., S.J.F., T.O., T. Tsuji, Y.F., K.J.E., J.-Y.P., S.E.) and Department of Pharmacology, Center for Translational Medicine (D.G.T.), Temple University School of Medicine, Philadelphia, PA; Department of Kinesiology, Temple University College of Public Health, Philadelphia, PA (S.J.F., J.-Y.P.); and Department of Biomedical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY (R.L.D.)
| | - Yamato Fukuda
- From the Department of Physiology, Cardiovascular Research Center (T. Takayanagi, T.K., S.J.F., T.O., T. Tsuji, Y.F., K.J.E., J.-Y.P., S.E.) and Department of Pharmacology, Center for Translational Medicine (D.G.T.), Temple University School of Medicine, Philadelphia, PA; Department of Kinesiology, Temple University College of Public Health, Philadelphia, PA (S.J.F., J.-Y.P.); and Department of Biomedical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY (R.L.D.)
| | - Katherine J Elliott
- From the Department of Physiology, Cardiovascular Research Center (T. Takayanagi, T.K., S.J.F., T.O., T. Tsuji, Y.F., K.J.E., J.-Y.P., S.E.) and Department of Pharmacology, Center for Translational Medicine (D.G.T.), Temple University School of Medicine, Philadelphia, PA; Department of Kinesiology, Temple University College of Public Health, Philadelphia, PA (S.J.F., J.-Y.P.); and Department of Biomedical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY (R.L.D.)
| | - Douglas G Tilley
- From the Department of Physiology, Cardiovascular Research Center (T. Takayanagi, T.K., S.J.F., T.O., T. Tsuji, Y.F., K.J.E., J.-Y.P., S.E.) and Department of Pharmacology, Center for Translational Medicine (D.G.T.), Temple University School of Medicine, Philadelphia, PA; Department of Kinesiology, Temple University College of Public Health, Philadelphia, PA (S.J.F., J.-Y.P.); and Department of Biomedical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY (R.L.D.)
| | - Robin L Davisson
- From the Department of Physiology, Cardiovascular Research Center (T. Takayanagi, T.K., S.J.F., T.O., T. Tsuji, Y.F., K.J.E., J.-Y.P., S.E.) and Department of Pharmacology, Center for Translational Medicine (D.G.T.), Temple University School of Medicine, Philadelphia, PA; Department of Kinesiology, Temple University College of Public Health, Philadelphia, PA (S.J.F., J.-Y.P.); and Department of Biomedical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY (R.L.D.)
| | - Joon-Young Park
- From the Department of Physiology, Cardiovascular Research Center (T. Takayanagi, T.K., S.J.F., T.O., T. Tsuji, Y.F., K.J.E., J.-Y.P., S.E.) and Department of Pharmacology, Center for Translational Medicine (D.G.T.), Temple University School of Medicine, Philadelphia, PA; Department of Kinesiology, Temple University College of Public Health, Philadelphia, PA (S.J.F., J.-Y.P.); and Department of Biomedical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY (R.L.D.)
| | - Satoru Eguchi
- From the Department of Physiology, Cardiovascular Research Center (T. Takayanagi, T.K., S.J.F., T.O., T. Tsuji, Y.F., K.J.E., J.-Y.P., S.E.) and Department of Pharmacology, Center for Translational Medicine (D.G.T.), Temple University School of Medicine, Philadelphia, PA; Department of Kinesiology, Temple University College of Public Health, Philadelphia, PA (S.J.F., J.-Y.P.); and Department of Biomedical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY (R.L.D.).
| |
Collapse
|
33
|
Del Olmo-Turrubiarte A, Calzada-Torres A, Díaz-Rosas G, Palma-Lara I, Sánchez-Urbina R, Balderrábano-Saucedo NA, González-Márquez H, Garcia-Alonso P, Contreras-Ramos A. Mouse models for the study of postnatal cardiac hypertrophy. IJC HEART & VASCULATURE 2015; 7:131-140. [PMID: 28785661 PMCID: PMC5497247 DOI: 10.1016/j.ijcha.2015.02.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Revised: 01/19/2015] [Accepted: 02/01/2015] [Indexed: 12/16/2022]
Abstract
The main objective of this study was to create a postnatal model for cardiac hypertrophy (CH), in order to explain the mechanisms that are present in childhood cardiac hypertrophy. Five days after implantation, intraperitoneal (IP) isoproterenol (ISO) was injected for 7 days to pregnant female mice. The fetuses were obtained at 15, 17 and 19 dpc from both groups, also newborns (NB), neonates (7-15 days) and young adults (6 weeks of age). Histopathological exams were done on the hearts. Immunohistochemistry and western blot demonstrated GATA4 and PCNA protein expression, qPCR real time the mRNA of adrenergic receptors (α-AR and β-AR), alpha and beta myosins (α-MHC, β-MHC) and GATA4. After the administration of ISO, there was no change in the number of offsprings. We observed significant structural changes in the size of the offspring hearts. Morphometric analysis revealed an increase in the size of the left ventricular wall and interventricular septum (IVS). Histopathological analysis demonstrated loss of cellular compaction and presence of left ventricular small fibrous foci after birth. Adrenergic receptors might be responsible for changing a physiological into a pathological hypertrophy. However GATA4 seemed to be the determining factor in the pathology. A new animal model was established for the study of pathologic CH in early postnatal stages.
Collapse
Affiliation(s)
- A Del Olmo-Turrubiarte
- Laboratorio de Investigación de Biología del Desarrollo y Teratogénesis Experimental, Hospital Infantil de México Federico Gómez (HIMFG), Mexico.,Posgrado en Biología Experimental, Universidad Autónoma Metropolitana, Mexico
| | - A Calzada-Torres
- Laboratorio de Investigación de Biología del Desarrollo y Teratogénesis Experimental, Hospital Infantil de México Federico Gómez (HIMFG), Mexico
| | - G Díaz-Rosas
- Laboratorio de Investigación de Biología del Desarrollo y Teratogénesis Experimental, Hospital Infantil de México Federico Gómez (HIMFG), Mexico
| | | | - R Sánchez-Urbina
- Laboratorio de Investigación de Biología del Desarrollo y Teratogénesis Experimental, Hospital Infantil de México Federico Gómez (HIMFG), Mexico
| | | | - H González-Márquez
- Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana, Mexico
| | | | - A Contreras-Ramos
- Laboratorio de Investigación de Biología del Desarrollo y Teratogénesis Experimental, Hospital Infantil de México Federico Gómez (HIMFG), Mexico
| |
Collapse
|
34
|
Copik AJ, Baldys A, Nguyen K, Sahdeo S, Ho H, Kosaka A, Dietrich PJ, Fitch B, Raymond JR, Ford APDW, Button D, Milla ME. Isoproterenol acts as a biased agonist of the alpha-1A-adrenoceptor that selectively activates the MAPK/ERK pathway. PLoS One 2015; 10:e0115701. [PMID: 25606852 PMCID: PMC4301629 DOI: 10.1371/journal.pone.0115701] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2014] [Accepted: 11/26/2014] [Indexed: 11/24/2022] Open
Abstract
The α1A-AR is thought to couple predominantly to the Gαq/PLC pathway and lead to phosphoinositide hydrolysis and calcium mobilization, although certain agonists acting at this receptor have been reported to trigger activation of arachidonic acid formation and MAPK pathways. For several G protein-coupled receptors (GPCRs) agonists can manifest a bias for activation of particular effector signaling output, i.e. not all agonists of a given GPCR generate responses through utilization of the same signaling cascade(s). Previous work with Gαq coupling-defective variants of α1A-AR, as well as a combination of Ca2+ channel blockers, uncovered cross-talk between α1A-AR and β2-AR that leads to potentiation of a Gαq-independent signaling cascade in response to α1A-AR activation. We hypothesized that molecules exist that act as biased agonists to selectively activate this pathway. In this report, isoproterenol (Iso), typically viewed as β-AR-selective agonist, was examined with respect to activation of α1A-AR. α1A-AR selective antagonists were used to specifically block Iso evoked signaling in different cellular backgrounds and confirm its action at α1A-AR. Iso induced signaling at α1A-AR was further interrogated by probing steps along the Gαq /PLC, Gαs and MAPK/ERK pathways. In HEK-293/EBNA cells transiently transduced with α1A-AR, and CHO_α1A-AR stable cells, Iso evoked low potency ERK activity as well as Ca2+ mobilization that could be blocked by α1A-AR selective antagonists. The kinetics of Iso induced Ca2+ transients differed from typical Gαq- mediated Ca2+ mobilization, lacking both the fast IP3R mediated response and the sustained phase of Ca2+ re-entry. Moreover, no inositol phosphate (IP) accumulation could be detected in either cell line after stimulation with Iso, but activation was accompanied by receptor internalization. Data are presented that indicate that Iso represents a novel type of α1A-AR partial agonist with signaling bias toward MAPK/ERK signaling cascade that is likely independent of coupling to Gαq.
Collapse
Affiliation(s)
- Alicja. J. Copik
- Biochemical Pharmacology, Inflammation Discovery, Roche Palo Alto LLC, 3401 Hillview Drive, Palo Alto, CA 94304, United States of America
| | - Aleksander Baldys
- Nephrology Division, Department of Medicine, Medical University of South Carolina, and Medical and Research Services, Ralph H Johnson Veterans Affairs Medical Center, Charleston, South Carolina 29425, United States of America
| | - Khanh Nguyen
- Discovery Technologies, Roche Palo Alto LLC, 3401 Hillview Drive, Palo Alto, CA 94304, United States of America
| | - Sunil Sahdeo
- Biochemical Pharmacology, Inflammation Discovery, Roche Palo Alto LLC, 3401 Hillview Drive, Palo Alto, CA 94304, United States of America
| | - Hoangdung Ho
- Discovery Technologies, Roche Palo Alto LLC, 3401 Hillview Drive, Palo Alto, CA 94304, United States of America
| | - Alan Kosaka
- Discovery Technologies, Roche Palo Alto LLC, 3401 Hillview Drive, Palo Alto, CA 94304, United States of America
| | - Paul J. Dietrich
- Discovery Technologies, Roche Palo Alto LLC, 3401 Hillview Drive, Palo Alto, CA 94304, United States of America
| | - Bill Fitch
- Discovery Technologies, Roche Palo Alto LLC, 3401 Hillview Drive, Palo Alto, CA 94304, United States of America
| | - John R. Raymond
- Nephrology Division, Department of Medicine, Medical University of South Carolina, and Medical and Research Services, Ralph H Johnson Veterans Affairs Medical Center, Charleston, South Carolina 29425, United States of America
| | - Anthony P. D. W. Ford
- Biochemical Pharmacology, Inflammation Discovery, Roche Palo Alto LLC, 3401 Hillview Drive, Palo Alto, CA 94304, United States of America
| | - Donald Button
- Biochemical Pharmacology, Inflammation Discovery, Roche Palo Alto LLC, 3401 Hillview Drive, Palo Alto, CA 94304, United States of America
| | - Marcos E. Milla
- Biochemical Pharmacology, Inflammation Discovery, Roche Palo Alto LLC, 3401 Hillview Drive, Palo Alto, CA 94304, United States of America
- * E-mail:
| |
Collapse
|
35
|
Grisanti LA, Repas AA, Talarico JA, Gold JI, Carter RL, Koch WJ, Tilley DG. Temporal and gefitinib-sensitive regulation of cardiac cytokine expression via chronic β-adrenergic receptor stimulation. Am J Physiol Heart Circ Physiol 2014; 308:H316-30. [PMID: 25485901 DOI: 10.1152/ajpheart.00635.2014] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Chronic stimulation of β-adrenergic receptors (βAR) can promote survival signaling via transactivation of epidermal growth factor receptor (EGFR) but ultimately alters cardiac structure and contractility over time, in part via enhanced cytokine signaling. We hypothesized that chronic catecholamine signaling will have a temporal impact on cardiac transcript expression in vivo, in particular cytokines, and that EGFR transactivation plays a role in this process. C57BL/6 mice underwent infusion with vehicle or isoproterenol (Iso)±gefitinib (Gef) for 1 or 2 wk. Cardiac contractility decreased following 2 wk of Iso treatment, while cardiac hypertrophy, fibrosis, and apoptosis were enhanced at both timepoints. Inclusion of Gef preserved contractility, blocked Iso-induced apoptosis, and prevented hypertrophy at the 2-wk timepoint, but caused fibrosis on its own. RNAseq analysis revealed hundreds of cardiac transcripts altered by Iso at each timepoint with subsequent RT-quantitative PCR validation confirming distinct temporal patterns of transcript regulation, including those involved in cardiac remodeling and survival signaling, as well as numerous cytokines. Although Gef infusion alone did not significantly alter cytokine expression, it abrogated the Iso-mediated changes in a majority of the βAR-sensitive cytokines, including CCL2 and TNF-α. Additionally, the impact of βAR-dependent EGFR transactivation on the acute regulation of cytokine transcript expression was assessed in isolated cardiomyocytes and in cardiac fibroblasts, where the majority of Iso-dependent, and EGFR-sensitive, changes in cytokines occurred. Overall, coincident with changes in cardiac structure and contractility, βAR stimulation dynamically alters cardiac transcript expression over time, including numerous cytokines that are regulated via EGFR-dependent signaling.
Collapse
Affiliation(s)
- Laurel A Grisanti
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania; and
| | - Ashley A Repas
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania; and
| | - Jennifer A Talarico
- Center for Translational Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Jessica I Gold
- Center for Translational Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Rhonda L Carter
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania; and
| | - Walter J Koch
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania; and
| | - Douglas G Tilley
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania; and
| |
Collapse
|
36
|
Talarico JA, Carter RL, Grisanti LA, Yu JE, Repas AA, Tilley DG. β-adrenergic receptor-dependent alterations in murine cardiac transcript expression are differentially regulated by gefitinib in vivo. PLoS One 2014; 9:e99195. [PMID: 24901703 PMCID: PMC4047088 DOI: 10.1371/journal.pone.0099195] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Accepted: 05/12/2014] [Indexed: 11/18/2022] Open
Abstract
β-adrenergic receptor (βAR)-mediated transactivation of epidermal growth factor receptor (EGFR) has been shown to promote cardioprotection in a mouse model of heart failure and we recently showed that this mechanism leads to enhanced cell survival in part via regulation of apoptotic transcript expression in isolated primary rat neonatal cardiomyocytes. Thus, we hypothesized that this process could regulate cardiac transcript expression in vivo. To comprehensively assess cardiac transcript alterations in response to acute βAR-dependent EGFR transactivation, we performed whole transcriptome analysis of hearts from C57BL/6 mice given i.p. injections of the βAR agonist isoproterenol in the presence or absence of the EGFR antagonist gefitinib for 1 hour. Total cardiac RNA from each treatment group underwent transcriptome analysis, revealing a substantial number of transcripts regulated by each treatment. Gefitinib alone significantly altered the expression of 405 transcripts, while isoproterenol either alone or in conjunction with gefitinib significantly altered 493 and 698 distinct transcripts, respectively. Further statistical analysis was performed, confirming 473 transcripts whose regulation by isoproterenol were significantly altered by gefitinib (isoproterenol-induced up/downregulation antagonized/promoted by gefinitib), including several known to be involved in the regulation of numerous processes including cell death and survival. Thus, βAR-dependent regulation of cardiac transcript expression in vivo can be modulated by the EGFR antagonist gefitinib.
Collapse
Affiliation(s)
- Jennifer A. Talarico
- Center for Translational Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Rhonda L. Carter
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Laurel A. Grisanti
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Justine E. Yu
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Ashley A. Repas
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Douglas G. Tilley
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
- Department of Pharmacology, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
37
|
Alpha1a-adrenoceptor genetic variant induces cardiomyoblast-to-fibroblast-like cell transition via distinct signaling pathways. Cell Signal 2014; 26:1985-97. [PMID: 24835978 DOI: 10.1016/j.cellsig.2014.05.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Revised: 05/08/2014] [Accepted: 05/09/2014] [Indexed: 12/21/2022]
Abstract
The role of naturally occurring human α1a-Adrenergic Receptor (α1aAR) genetic variants associated with cardiovascular disorders is poorly understood. Here, we present the novel findings that expression of human α1aAR-247R (247R) genetic variant in cardiomyoblasts leads to transition of cardiomyoblasts into a fibroblast-like phenotype, evidenced by morphology and distinct de novo expression of characteristic genes. These fibroblast-like cells exhibit constitutive, high proliferative capacity and agonist-induced hypertrophy compared with cells prior to transition. We demonstrate that constitutive, synergistic activation of EGFR, Src and ERK kinases is the potential molecular mechanism of this transition. We also demonstrate that 247R triggers two distinct EGFR transactivation-dependent signaling pathways: 1) constitutive Gq-independent β-arrestin-1/Src/MMP/EGFR/ERK-dependent hyperproliferation and 2) agonist-induced Gq- and EGFR/STAT-dependent hypertrophy. Interestingly, in cardiomyoblasts agonist-independent hyperproliferation is MMP-dependent, but in fibroblast-like cells it is MMP-independent, suggesting that expression of α1aAR genetic variant in cardiomyocytes may trigger extracellular matrix remodeling. Thus, these novel findings demonstrate that EGFR transactivation by α1aAR-247R leads to hyperproliferation, hypertrophy and alterations in cardiomyoblasts, suggesting that these unique genetically-mediated alterations in signaling pathways and cellular function may lead to myocardial fibrosis. Such extracellular matrix remodeling may contribute to the genesis of arrhythmias in certain types of heart failure.
Collapse
|